1
|
Kumai T, Shinomiya H, Shibata H, Takahashi H, Kishikawa T, Okada R, Fujieda S, Sakashita M. Translational research in head and neck cancer: Molecular and immunological updates. Auris Nasus Larynx 2024; 51:391-400. [PMID: 37640594 DOI: 10.1016/j.anl.2023.08.006] [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: 06/19/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) has a poor prognosis. Each year, approximately 880,000 patients are newly diagnosed with HNSCC worldwide, and 450,000 patients with HNSCC die. Risk factors for developing HNSCC have been identified, with cigarette smoking, alcohol consumption, and viral infections being the major factors. Owing to the prevalence of human papillomavirus infection, the number of HNSCC cases is increasing considerably. Surgery and chemoradiotherapy are the primary treatments for HNSCC. With advancements in tumor biology, patients are eligible for novel treatment modalities, namely targeted therapies, immunotherapy, and photoimmunotherapy. Because this area of research has rapidly progressed, clinicians should understand the basic biology of HNSCC to choose an appropriate therapy in the upcoming era of personalized medicine. This review summarized recent developments in tumor biology, focusing on epidemiology, genetic/epigenetic factors, the tumor microenvironment, microbiota, immunity, and photoimmunotherapy in HNSCC, as well as how these findings can be translated into clinical settings.
Collapse
Affiliation(s)
- Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Midorigaoka-Higashi 2-1-1-1, Asahikawa 078-8510, Japan.
| | - Hirotaka Shinomiya
- Department of Otolaryngology-Head and Neck Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Hirofumi Shibata
- Department of Otolaryngology-Head and Neck Surgery, Gifu University Graduate School of Medicine, Gifu, Japan.
| | - Hideaki Takahashi
- Department of Otorhinolaryngology, Head and Neck Surgery, School of Medicine, Yokohama City University, Yokohama, Japan.
| | - Toshihiro Kishikawa
- Department of Head and Neck Surgery, Aichi Cancer Center Hospital, Nagoya, Japan.
| | - Ryuhei Okada
- Department of Head and Neck Surgery, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| | - Masafumi Sakashita
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| |
Collapse
|
2
|
Wang D, Li H, Zeng T, Chen Q, Huang W, Huang Y, Liao Y, Jiang Q. Exosome-transmitted ANGPTL1 suppresses angiogenesis in glioblastoma by inhibiting the VEGFA/VEGFR2/Akt/eNOS pathway. J Neuroimmunol 2024; 387:578266. [PMID: 38150891 DOI: 10.1016/j.jneuroim.2023.578266] [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: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE Glioblastoma (GBM) is a highly vascularized malignancy that relies on new vessel generation, and thus targeting angiogenesis has been a promising anti-GBM approach. ANGPTL1 is well-known for its anti-angiogenic property; nevertheless, its role in GBM is yet to be explored. Recently, the crucial role of exosomes (Exos) as intercellular communication mediators has gained prominence in GBM therapy. This work aimed to explore the role of exosomal ANGPTL1 in GBM angiogenesis and its mechanisms. METHODS Bioinformatic analysis was performed to evaluate ANGPTL expression in GBM. Human GBM cell lines (U87 and U251) and a xenograft mouse model were employed. Exos were isolated from oe-NC- and oe-ANGPTL-transfected bone mesenchymal stem cells and identified. Cell proliferation, migration, and apoptosis were detected. Immunofluorescence, qRT-PCR, western blotting, co-immunoprecipitation, and immunohistochemistry were used to determine the molecular mechanisms underlying exosomal ANGPTL1 against GBM angiogenesis. Besides, tube generation and transmission electron microscope assays were conducted to assess GBM angiogenesis. RESULTS Low ANGPTL1 expression was observed in GBM tumor tissues and cells. Functionally, e-ANGPTL-Exos inhibited GBM malignant progression and angiogenesis in vitro and in vivo. Mechanically, e-ANGPTL-Exos reduced VEGFA expression and blocked the VEGFR2/Akt/eNOS pathway in GBM cells and tumor tissues. Co-immunoprecipitation revealed a link between ANGPTL1 and VEGFA in GBM cells. Notably, oe-VEGFA abolished the suppressive functions of e-ANGPTL-Exos in GBM progression and angiogenesis and the VEGFR2/Akt/eNOS axis. The VEGFR2 inhibitor, vandetanib, eliminated the promotive effects of oe-VEGFA on GBM angiogenesis with suppressed VEGFR2/Akt/eNOS pathway. CONCLUSIONS Exosomal ANGPTL1 suppressed GBM angiogenesis by inhibiting the VEGFA/VEGFR2/Akt/eNOS axis.
Collapse
Affiliation(s)
- Dong Wang
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| | - Huichen Li
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Tianxiang Zeng
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Qiang Chen
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Weilong Huang
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Yujing Huang
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Yuqing Liao
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Qiuhua Jiang
- Department of Neurosurgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
3
|
Long Z, Grandis JR, Johnson DE. Emerging tyrosine kinase inhibitors for head and neck cancer. Expert Opin Emerg Drugs 2022; 27:333-344. [PMID: 36131561 PMCID: PMC9987561 DOI: 10.1080/14728214.2022.2125954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/14/2022] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Conventional regimens for head and neck squamous cell carcinoma (HNSCC) are limited in efficacy and are associated with adverse toxicities. Food and Drug Administration (FDA) approved molecular targeting agents include the HER1 (EGFR)-directed monoclonal antibody cetuximab and the immune checkpoint inhibitors nivolumab and pembrolizumab. However, clinical benefit is only seen in roughly 15-20% of HNSCC patients treated with these agents. New molecular targeting agents are needed that either act with monotherapeutic activity against HNSCC tumors or enhance the activities of current therapies, particularly immunotherapy. Small-molecule tyrosine kinase inhibitors (TKIs) represent a viable option toward this goal. AREAS COVERED This review provides an update on TKIs currently under investigation in HNSCC. We focus our review on data obtained and trials underway in HNSCC, including salivary gland cancers and nasopharyngeal carcinomas, but excluding thyroid cancer and esophageal cancer. EXPERT OPINION While some emerging TKIs have shown clinical benefit, the positive effects have, largely, been modest. The design of clinical trials of TKIs has been hampered by a lack of understanding of biomarkers that can be used to define patient populations most likely to respond. Further preclinical and translational studies to define biomarkers of TKI response will be critically important.
Collapse
Affiliation(s)
- Zhen Long
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, California, USA
| | - Jennifer R. Grandis
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, California, USA
| | - Daniel E. Johnson
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, California, USA
| |
Collapse
|
4
|
Kordbacheh F, Farah CS. Current and Emerging Molecular Therapies for Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13215471. [PMID: 34771633 PMCID: PMC8582411 DOI: 10.3390/cancers13215471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/09/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022] Open
Abstract
Head and neck cancer affects nearly 750,000 patients, with more than 300,000 deaths annually. Advances in first line surgical treatment have improved survival rates marginally particularly in developed countries, however survival rates for aggressive locally advanced head and neck cancer are still poor. Recurrent and metastatic disease remains a significant problem for patients and the health system. As our knowledge of the genomic landscape of the head and neck cancers continues to expand, there are promising developments occurring in molecular therapies available for advanced or recalcitrant disease. The concept of precision medicine is underpinned by our ability to accurately sequence tumour samples to best understand individual patient genomic variations and to tailor targeted therapy for them based on such molecular profiling. Not only is their purported response to therapy a factor of their genomic variation, but so is their inclusion in biomarker-driven personalised medicine therapeutic trials. With the ever-expanding number of molecular druggable targets explored through advances in next generation sequencing, the number of clinical trials assessing these targets has significantly increased over recent years. Although some trials are focussed on first-line therapeutic approaches, a greater majority are focussed on locally advanced, recurrent or metastatic disease. Similarly, although single agent monotherapy has been found effective in some cases, it is the combination of drugs targeting different signalling pathways that seem to be more beneficial to patients. This paper outlines current and emerging molecular therapies for head and neck cancer, and updates readers on outcomes of the most pertinent clinical trials in this area while also summarising ongoing efforts to bring more molecular therapies into clinical practice.
Collapse
Affiliation(s)
- Farzaneh Kordbacheh
- Broad Institute of MIT and Harvard, Boston, MA 02142, USA;
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, ACT 0200, Australia
| | - Camile S. Farah
- The Australian Centre for Oral Oncology Research & Education, Nedlands, WA 6009, Australia
- Genomics for Life, Milton, QLD 4064, Australia
- Anatomical Pathology, Australian Clinical Labs, Subiaco, WA 6009, Australia
- Head and Neck Cancer Signalling Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Correspondence:
| |
Collapse
|
5
|
González A, Alonso-González C, González-González A, Menéndez-Menéndez J, Cos S, Martínez-Campa C. Melatonin as an Adjuvant to Antiangiogenic Cancer Treatments. Cancers (Basel) 2021; 13:3263. [PMID: 34209857 PMCID: PMC8268559 DOI: 10.3390/cancers13133263] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023] Open
Abstract
Melatonin is a hormone with different functions, antitumor actions being one of the most studied. Among its antitumor mechanisms is its ability to inhibit angiogenesis. Melatonin shows antiangiogenic effects in several types of tumors. Combination of melatonin and chemotherapeutic agents have a synergistic effect inhibiting angiogenesis. One of the undesirable effects of chemotherapy is the induction of pro-angiogenic factors, whilst the addition of melatonin is able to overcome these undesirable effects. This protective effect of the pineal hormone against angiogenesis might be one of the mechanisms underlying its anticancer effect, explaining, at least in part, why melatonin administration increases the sensitivity of tumors to the inhibitory effects exerted by ordinary chemotherapeutic agents. Melatonin has the ability to turn cancer totally resistant to chemotherapeutic agents into a more sensitive chemotherapy state. Definitely, melatonin regulates the expression and/or activity of many factors involved in angiogenesis which levels are affected (either positively or negatively) by chemotherapeutic agents. In addition, the pineal hormone has been proposed as a radiosensitizer, increasing the oncostatic effects of radiation on tumor cells. This review serves as a synopsis of the interaction between melatonin and angiogenesis, and we will outline some antiangiogenic mechanisms through which melatonin sensitizes cancer cells to treatments, such as radiotherapy or chemotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain; (A.G.); (A.G.-G.); (J.M.-M.); (C.M.-C.)
| | | |
Collapse
|
6
|
Fasano M, Della Corte CM, Viscardi G, Di Liello R, Paragliola F, Sparano F, Iacovino ML, Castrichino A, Doria F, Sica A, Morgillo F, Colella G, Tartaro G, Cappabianca S, Testa D, Motta G, Ciardiello F. Head and neck cancer: the role of anti-EGFR agents in the era of immunotherapy. Ther Adv Med Oncol 2021; 13:1758835920949418. [PMID: 33767760 PMCID: PMC7953226 DOI: 10.1177/1758835920949418] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/15/2020] [Indexed: 01/08/2023] Open
Abstract
Head and neck cancers (HNC) represent the seventh most frequent cancer worldwide, with squamous cell carcinomas as the most frequent histologic subtype. Standard treatment for early stage diseases is represented by single modality surgery or radiotherapy, whereas in the locally advanced and recurrent or metastatic settings a more aggressive multi-modal approach is needed with locoregional intervention and/or systemic therapies. Epidermal Growth Factor Receptor (EGFR) plays an important role in HNC biology and has been studied extensively in preclinical and clinical settings. In this scenario, anti-EGFR targeted agent cetuximab, introduced in clinical practice a decade ago, represents the only approved targeted therapy to date, while the development of immune-checkpoint inhibitors has recently changed the available treatment options. In this review, we focus on the current role of anti-EGFR therapies in HNCs, underlying available clinical data and mechanisms of resistance, and highlight future perspectives regarding their role in the era of immunotherapy.
Collapse
Affiliation(s)
- Morena Fasano
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli. Via Sergio Pansini 5, Naples, 80131, Italy
| | - Carminia Maria Della Corte
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuseppe Viscardi
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Raimondo Di Liello
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Fernando Paragliola
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Sparano
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Lucia Iacovino
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Francesca Doria
- Centro radiologico Vega, Centro radiologico fisica e terapia fisica Morrone, Caserta, Italy
| | - Antonello Sica
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Floriana Morgillo
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuseppe Colella
- Maxillo-Facial Surgery Department, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giampaolo Tartaro
- Maxillo-Facial Surgery Department, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, Radiology Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Domenico Testa
- Department of Anesthesiology, Surgical and Emergency Science, Clinic of Otorhinolaryngology, Head and Neck Surgery Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gaetano Motta
- Department of Anesthesiology, Surgical and Emergency Science, Clinic of Otorhinolaryngology, Head and Neck Surgery Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
7
|
Emdadi A, Eslahchi C. Auto-HMM-LMF: feature selection based method for prediction of drug response via autoencoder and hidden Markov model. BMC Bioinformatics 2021; 22:33. [PMID: 33509079 PMCID: PMC7844991 DOI: 10.1186/s12859-021-03974-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Predicting the response of cancer cell lines to specific drugs is an essential problem in personalized medicine. Since drug response is closely associated with genomic information in cancer cells, some large panels of several hundred human cancer cell lines are organized with genomic and pharmacogenomic data. Although several methods have been developed to predict the drug response, there are many challenges in achieving accurate predictions. This study proposes a novel feature selection-based method, named Auto-HMM-LMF, to predict cell line-drug associations accurately. Because of the vast dimensions of the feature space for predicting the drug response, Auto-HMM-LMF focuses on the feature selection issue for exploiting a subset of inputs with a significant contribution. RESULTS This research introduces a novel method for feature selection of mutation data based on signature assignments and hidden Markov models. Also, we use the autoencoder models for feature selection of gene expression and copy number variation data. After selecting features, the logistic matrix factorization model is applied to predict drug response values. Besides, by comparing to one of the most powerful feature selection methods, the ensemble feature selection method (EFS), we showed that the performance of the predictive model based on selected features introduced in this paper is much better for drug response prediction. Two datasets, the Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Cell Line Encyclopedia (CCLE) are used to indicate the efficiency of the proposed method across unseen patient cell-line. Evaluation of the proposed model showed that Auto-HMM-LMF could improve the accuracy of the results of the state-of-the-art algorithms, and it can find useful features for the logistic matrix factorization method. CONCLUSIONS We depicted an application of Auto-HMM-LMF in exploring the new candidate drugs for head and neck cancer that showed the proposed method is useful in drug repositioning and personalized medicine. The source code of Auto-HMM-LMF method is available in https://github.com/emdadi/Auto-HMM-LMF .
Collapse
Affiliation(s)
- Akram Emdadi
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
| | - Changiz Eslahchi
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran.
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), 193955746, Tehran, Iran.
| |
Collapse
|
8
|
Combined treatment with acetazolamide and cisplatin enhances the chemosensitivity of human head and neck squamous cell carcinoma TU868 cells. Arch Oral Biol 2020; 119:104905. [PMID: 32947166 DOI: 10.1016/j.archoralbio.2020.104905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/21/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022]
Abstract
AIMS To investigate whether combination of acetazolamide and cisplatin can enhance the chemosensitivity of human head and neck squamous cell carcinoma (HNSCC) cell line TU868. METHODS MTT assay was performed to determine the effect of acetazolamide, cisplatin and their combination on the proliferation of TU868 cells. Then the effect of these 2 drugs on the expression of proliferation-related and apoptosis-related proteins was detected by Western blot. Moreover, the effect of acetazolamide and cisplatin on the expression of aquaporin-1 was detected by RT-qPCR. Loss-of-function assays was performed to assess whether the effect of acetazolamide and cisplatin on TU868 cells was mediated by aquaporin-1. The effect of acetazolamide and cisplatin on tumor cell growth was confirmed in mice by testing the tumor growth size. RESULTS Acetazolamide and cisplatin treatment displayed synergistic effects on the inhibition of TU868 cell growth compared with the drugs used alone. Moreover, the acetazolamide/cisplatin combination could decrease the level of PCNA but increase the level of p53; decrease the ratio of Bcl-2/Bax and increase the expression of caspase-3 compared with the single drug treated group. Moreover, we found that the combination also significantly inhibits aquaporin-1 expression. Loss-of-function assays suggested that the anti-tumor effect of these 2 drugs was achieved via affecting aquaporin-1. Consistent with the in vitro assays, combined treatment with acetazolamide and cisplatin significantly inhibits the tumor growth in mice compared with the single drug treated group. CONCLUSION These results demonstrated that combined treatment with acetazolamide and cisplatin could synergistically inhibit the malignant development of HNSCC cells.
Collapse
|
9
|
Kitamura S, Maeda T, Yanagi T. Vandetanib inhibits cell growth in EGFR-expressing cutaneous squamous cell carcinoma. Biochem Biophys Res Commun 2020; 531:396-401. [PMID: 32800552 DOI: 10.1016/j.bbrc.2020.07.111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022]
Abstract
Advanced cutaneous squamous cell carcinoma (SCC) responds poorly to chemotherapy, leading to significant morbidity or death. Overexpression of epidermal growth factor receptor (EGFR) is frequently observed in advanced cutaneous SCC. Vandetanib is a multiple tyrosine kinase targeting vascular endothelial growth factor receptor-2 (VEGFR2), EGFR, and the rearranged during transfection (RET) proto-oncogene. Vandetanib has been reported to inhibit tumor growth in head and neck SCC. However, the efficacy of vandetanib against cutaneous SCC has not been thoroughly investigated. The aim of this study is to evaluate the efficacy of vandetanib against cutaneous SCC in vitro and in vivo. Vandetanib is found to inhibit the proliferation of cutaneous SCC cells as assessed by cell viability and clonogenic assay. Cell death analysis indicates that vandetanib induces cell death in SCC cells but not in normal human keratinocytes or fibroblasts. The in vivo anti-tumor effect of vandetanib is shown in xenograft tumor models using A431 SCC cells. Mechanistically, vandetanib suppresses the phosphorylation of EGFR in SCC cells. Clinically, EGFR expression levels are elevated in cutaneous SCC specimens, relative to normal epidermis. In conclusion, we identified vandetanib as a novel therapeutic option for cutaneous SCC, especially in tumors with high EGFR expression.
Collapse
Affiliation(s)
- Shinya Kitamura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Takuya Maeda
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan.
| |
Collapse
|
10
|
Lorusso G, Rüegg C, Kuonen F. Targeting the Extra-Cellular Matrix-Tumor Cell Crosstalk for Anti-Cancer Therapy: Emerging Alternatives to Integrin Inhibitors. Front Oncol 2020; 10:1231. [PMID: 32793493 PMCID: PMC7387567 DOI: 10.3389/fonc.2020.01231] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network composed of a multitude of different macromolecules. ECM components typically provide a supportive structure to the tissue and engender positional information and crosstalk with neighboring cells in a dynamic reciprocal manner, thereby regulating tissue development and homeostasis. During tumor progression, tumor cells commonly modify and hijack the surrounding ECM to sustain anchorage-dependent growth and survival, guide migration, store pro-tumorigenic cell-derived molecules and present them to enhance receptor activation. Thereby, ECM potentially supports tumor progression at various steps from initiation, to local growth, invasion, and systemic dissemination and ECM-tumor cells interactions have long been considered promising targets for cancer therapy. Integrins represent key surface receptors for the tumor cell to sense and interact with the ECM. Yet, attempts to therapeutically impinge on these interactions using integrin inhibitors have failed to deliver anticipated results, and integrin inhibitors are still missing in the emerging arsenal of drugs for targeted therapies. This paradox situation should urge the field to reconsider the role of integrins in cancer and their targeting, but also to envisage alternative strategies. Here, we review the therapeutic targets implicated in tumor cell adhesion to the ECM, whose inhibitors are currently in clinical trials and may offer alternatives to integrin inhibition.
Collapse
Affiliation(s)
- Girieca Lorusso
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Curzio Rüegg
- Experimental and Translational Oncology, Department of Oncology Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - François Kuonen
- Department of Dermatology and Venereology, Hôpital de Beaumont, Lausanne University Hospital Center, Lausanne, Switzerland
| |
Collapse
|
11
|
Wang J, Cui R, Clement CG, Nawgiri R, Powell DW, Pinchuk IV, Watts TL. Activation PDGFR-α/AKT Mediated Signaling Pathways in Oral Squamous Cell Carcinoma by Mesenchymal Stem/Stromal Cells Promotes Anti-apoptosis and Decreased Sensitivity to Cisplatin. Front Oncol 2020; 10:552. [PMID: 32411595 PMCID: PMC7199219 DOI: 10.3389/fonc.2020.00552] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/27/2020] [Indexed: 01/31/2023] Open
Abstract
Desmoplasia, a hallmark of a head and neck cancer, has both biologic and physiologic effects on cancer progression and chemotherapeutic response. Mesenchymal stem/stromal cells (MSCs), also known as mesenchymal stromal progenitor cells, have been shown to play a role in cancer progression, alter apoptotic responses, and confer resistance to chemotherapy in various carcinomas. The pathophysiology of MSCs with respect to tumorigenesis is widely reported in other cancers and is sparsely reported in oral squamous cell carcinomas (OSCCs). We previously reported paracrine mediated PDGF-AA/PDGFR-α signaling to underlie MSCs chemotaxis in OSCC. Given the poor clinical response to primary chemotherapy, we hypothesized that MSCs may alter cancer cell sensitivity to cisplatin through activation of PDGFR-α mediated signaling pathways. Co-culture of MSCs with human derived OSCC cell lines, JHU-012 and −019, resulted in a significant increase in the production of PDGF-AA and MCP-1 compared to cancer cells grown alone (p < 0.005) and was accompanied by an increase in the phosphorylation state of PDGFR-α (p < 0.02) and downstream target AKT at S473 (p < 0.025) and T308 (p < 0.02). JHU-012 and −019 cancer cells grown in co-culture were significantly less apoptotic (p < 0.001), expressed significantly higher levels of Bcl-2 (p < 0.04) with a concomitant significant decrease in bid expression (p < 0.001) compared to cancer cells grown alone. There was a significant increase in the cisplatin dose response curve in cancer cell clones derived from JHU-012 and 019 cancer cells grown in co-culture with MSCs compared to clones derived from cancer cells grown alone (p < 0.001). Moreover clones derived from JHU-012 cells grown in co-culture with MSCs were significantly more susceptible to cisplatin following pretreatment with, crenolanib, a PDGFR inhibitor, compared to cancer cells grown alone or in co-culture with MSCs (p < 0.0001). These findings suggest that crosstalk between cancer cells and MSCs is mediated, at least in part, by activation of autocrine PDGF-AA/PDGFR-α loop driving AKT-mediated signaling pathways, resulting in reduced cancer cell sensitivity to cisplatin through alterations in apoptosis.
Collapse
Affiliation(s)
- Jia Wang
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ruwen Cui
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, TX, United States
| | - Cecila G Clement
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ranjana Nawgiri
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Don W Powell
- Department of Internal Medicine, Division of Gastroenterology, University of Texas Medical Branch, Galveston, TX, United States
| | - Irina V Pinchuk
- Department of Internal Medicine, Division of Gastroenterology, University of Texas Medical Branch, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Tammara L Watts
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
12
|
Mehta M, Griffith J, Panneerselvam J, Babu A, Mani J, Herman T, Ramesh R, Munshi A. Regorafenib sensitizes human breast cancer cells to radiation by inhibiting multiple kinases and inducing DNA damage. Int J Radiat Biol 2020; 97:1109-1120. [PMID: 32052681 PMCID: PMC7882427 DOI: 10.1080/09553002.2020.1730012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/12/2019] [Accepted: 01/30/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is the most challenging and aggressive subtype of breast cancer with limited treatment options because of tumor heterogeneity, lack of druggable targets and therapy resistance. TNBCs are characterized by overexpression of growth factor receptors such as epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), and platelet derived growth factor receptor (PDGFR) making them promising therapeutic targets. Regorafenib is an FDA approved oral multi-kinase inhibitor that blocks the activity of multiple protein kinases including those involved in the regulation of tumor angiogenesis [VEGFR1-3, TIE2], tumor microenvironment [PDGFR-β, FGFR] and oncogenesis (KIT, RET, RAF-1, BRAF). In the current study, we examined the radiosensitizing effects of Regorafenib on TNBC cell lines and explored the mechanism by which Regorafenib enhances radiosensitivity. METHODS MDA-MB-231 and SUM159PT (human TNBC cell lines) and MCF 10a (human mammary epithelial cell line) were treated with Regorafenib, ionizing radiation or a combination of both. Following treatment with Regorafenib and radiation we conducted clonogenic assay to determine radiosensitivity, immunoblot analysis to assess the effect on key signaling targets, tube formation to evaluate effect on angiogenesis and comet assay as well as western blot for γH2AX to assess DNA damage response (DDR). RESULTS Regorafenib reduced cell proliferation and enhanced radiosensitivity of MDA-MB-231 and SUM159PT cell lines but had no effect on the MCF 10a cells. Clonogenic survival assays showed that the surviving fraction at 2 Gy for both MDA-MB-231 and SUM159PT was reduced from 66.4 ± 8.9 and 88.2 ± 1.7 in controls to 38.1 ± 4.9 and 75.1 ± 1.1 following a 24 hr pretreatment with 10 μM and 5 μM Regorafenib, respectively. A marked reduction in the expression of VEGFR, PDGFR, EGFR and the downstream target, ERK, was observed with Regorafenib treatment alone or in combination with radiation. We also observed a significant inhibition of VEGF-A production in the TNBC cell lines following treatment with Regorafenib. Further, the addition of conditioned medium from Regorafenib-treated tumor cells onto human umbilical vein endothelial cells (HUVEC) suppressed tube formation, indicating an inhibition of tumor angiogenesis. Regorafenib also decreased migration of TNBC cells and suppressed radiation-induced DNA damage repair in a time-dependent manner. CONCLUSIONS Our findings demonstrate that Regorafenib enhanced radiosensitivity of breast cancer cells by inhibiting the expression of multiple receptor tyrosine kinases, VEGF-mediated angiogenesis and DNA damage response in TNBC. Therefore, combining Regorafenib with radiation and antiangiogenic agents will be beneficial and effective in controlling TNBC.
Collapse
Affiliation(s)
- Meghna Mehta
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - James Griffith
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Janani Panneerselvam
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anish Babu
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jonathan Mani
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Terence Herman
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rajagopal Ramesh
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anupama Munshi
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
13
|
Sato S, Vasaikar S, Eskaros A, Kim Y, Lewis JS, Zhang B, Zijlstra A, Weaver AM. EPHB2 carried on small extracellular vesicles induces tumor angiogenesis via activation of ephrin reverse signaling. JCI Insight 2019; 4:132447. [PMID: 31661464 DOI: 10.1172/jci.insight.132447] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is a key process that allows nutrient uptake and cellular trafficking and is coopted in cancer to enable tumor growth and metastasis. Recently, extracellular vesicles (EVs) have been shown to promote angiogenesis; however, it is unclear what unique features EVs contribute to the process. Here, we studied the role of EVs derived from head and neck squamous cell carcinoma (HNSCC) in driving tumor angiogenesis. Small EVs (SEVs), in the size range of exosomes (50-150 nm), induced angiogenesis both in vitro and in vivo. Proteomic analysis of HNSCC SEVs revealed the cell-to-cell signaling receptor ephrin type B receptor 2 (EPHB2) as a promising candidate cargo to promote angiogenesis. Analysis of patient data further identified EPHB2 overexpression in HNSCC tumors to be associated with poor patient prognosis and tumor angiogenesis, especially in the context of overexpression of the exosome secretion regulator cortactin. Functional experiments revealed that EPHB2 expression in SEVs regulated angiogenesis both in vitro and in vivo and that EPHB2 carried by SEVs stimulates ephrin-B reverse signaling, inducing STAT3 phosphorylation. A STAT3 inhibitor greatly reduced SEV-induced angiogenesis. These data suggest a model in which EVs uniquely promote angiogenesis by transporting Eph transmembrane receptors to nonadjacent endothelial cells to induce ephrin reverse signaling.
Collapse
Affiliation(s)
- Shinya Sato
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Suhas Vasaikar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Adel Eskaros
- Department of Pathology, Microbiology and Immunology, and
| | - Young Kim
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James S Lewis
- Department of Pathology, Microbiology and Immunology, and
| | - Bing Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | | | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Pathology, Microbiology and Immunology, and
| |
Collapse
|
14
|
Abdelwahed A, Ahmed M. Rare epithelial breast cancer: surgery and adjuvant therapy. Transl Cancer Res 2019; 8:S479-S492. [PMID: 35117126 PMCID: PMC8797705 DOI: 10.21037/tcr.2019.05.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/10/2019] [Indexed: 01/30/2023]
Abstract
Breast cancer is a heterogenous disease, exhibiting a wide range of morphological phenotypes shaping its prognosis and clinical course. However, optimal management of rarer breast cancer subtypes is often undefined and controversial in literature due to the lack of large studies and randomised trials. This review aims to discuss the treatment of 13 rare epithelial subtypes, focussing on surgery and adjuvant therapies.
Collapse
Affiliation(s)
| | - Muneer Ahmed
- King's College London, Division of Cancer, Research Oncology, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
15
|
Liu L, Chen J, Cai X, Yao Z, Huang J. Progress in targeted therapeutic drugs for oral squamous cell carcinoma. Surg Oncol 2019; 31:90-97. [PMID: 31550560 DOI: 10.1016/j.suronc.2019.09.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/13/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
With the rapid development of biomedicine, people have a deeper understanding with the biological characteristics of malignant tumors, and begin to notice that in most tumors, there are over-expression of several molecules such as epidermal growth factor receptor(EGFR), vascular endothelial growth factor (VEGF) and its receptors,mammalian target of rapamycin(mTOR),programmed cell death receptor-1(PD-1),cyclin-dependent kinases(CDKs) and so on, whose levels are closely related to the prognosis of tumors. It has been found that the drugs targeting the above molecules can significantly improve the survival rate of cancer patients, and have the advantages of high selectivity, low toxicity and high therapeutic index. Targeted drugs, as new ones in the field of cancer, have achieved good efficacy in most tumor treatments. Oral cancer is an aggressive malignant tumour that is prone to relapse and metastasis. More than 90% of them are squamous cell carcinoma, and the 5-year survival rate remains at about 50%-60%.The proposing of targeted therapy opens up a new way for the treatment of oral cancer and brings dawn to patients with advanced diseases. Currently,a variety of targeted therapeutic drugs are being tested in various clinical trials in patients with oral squamous cell carcinoma(OSCC)·In this paper, we discuss the research progress of targeted therapeutic drugs in the treatment of OSCC in recent years.
Collapse
Affiliation(s)
- Lian Liu
- Department of Oral Pathology, Xiangya Stomatological Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Jili Chen
- Department of Oral Pathology, Xiangya Stomatological Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Xinjia Cai
- Department of Oral Pathology, Xiangya Stomatological Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Zhigang Yao
- Department of Oral Pathology, Xiangya Stomatological Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Junhui Huang
- Department of Oral Pathology, Xiangya Stomatological Hospital, Central South University, 410078, Changsha, Hunan, China.
| |
Collapse
|
16
|
Sola AM, Johnson DE, Grandis JR. Investigational multitargeted kinase inhibitors in development for head and neck neoplasms. Expert Opin Investig Drugs 2019; 28:351-363. [PMID: 30753792 DOI: 10.1080/13543784.2019.1581172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Despite advances in treatment, head and neck squamous cell carcinoma (HNSCC) survival rates remain stagnant. Current treatment is associated with significant toxicities and includes chemotherapy, radiation, surgery, and few targeted treatments. Targeted treatments, epidermal growth factor receptor (EGFR)-targeted agent, cetuximab, and immune checkpoint inhibitors, pembrolizumab and nivolumab, show improved toxicity profiles and modestly improved survival in select patients. An urgent need remains to identify novel targeted treatments for single-agent or combined therapy use. AREAS COVERED Multitargeted kinase inhibitors are small molecule inhibitors with limited toxicity. This review will focus on early-stage investigations of multitargeted tyrosine kinase inhibitors (m-TKIs) (those that target at least two tyrosine kinases) for HNSCC. Preclinical and early trials investigating m-TKIs for various disease settings of HNSCC will be evaluated for efficacy, identification of significant biomarkers and potential for combination therapy. EXPERT OPINION Few single agent m-TKIs have demonstrated efficacy in unselected HNSCC populations. The most promising clinical results have been obtained when m-TKIs are tested in combination with other therapies, including immunotherapy, or in mutation-defined subgroups of patients. The future success of m-TKIs will rely on identification, in preclinical models and clinical trials, of predictive biomarkers of response and mechanisms of innate and acquired resistance.
Collapse
Affiliation(s)
- Ana Marija Sola
- a Department of Otolaryngology - Head and Neck Surgery , University of California , San Francisco , CA , USA
| | - Daniel E Johnson
- a Department of Otolaryngology - Head and Neck Surgery , University of California , San Francisco , CA , USA
| | - Jennifer R Grandis
- a Department of Otolaryngology - Head and Neck Surgery , University of California , San Francisco , CA , USA
| |
Collapse
|
17
|
Iron-oxide nanoparticles target intracellular HSP90 to induce tumor radio-sensitization. Biochim Biophys Acta Gen Subj 2019; 1863:857-869. [PMID: 30794824 DOI: 10.1016/j.bbagen.2019.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Nanoparticle-based therapies have emerged as a promising approach to overcome limitations of conventional chemotherapy. Present study investigates the potential of oleic acid-functionalized iron-oxide nanoparticles (MN-OA) to enhance the radiation response of fibrosarcoma tumor and elucidates its underlying mechanism. METHODS Various cellular and molecular assays (e.g. MTT, clonogenic, cell cycle analysis, cell death, DNA damage/repair) and tumor growth kinetics were employed to investigate the mechanism of MN-OA induced radio-sensitization. RESULTS Mouse (WEHI-164) and human (HT-1080) fibrosarcoma cells treated with MN-OA and gamma-radiation (2 Gy) showed a significant decrease in the cell proliferation. Combination treatment showed significant decrease in clonogenic survival of WEHI-164 cells and was found to induce cell cycle arrest, apoptosis and mitotic catastrophe. The mechanism of radio-sensitization was found to involve binding of MN-OA with HSP90, resulting in down-regulation of its client proteins, involved in cell cycle progression (Cyclin B1 and CDC2) and DNA-double strand break repair (e.g. RAD51 and BRCA1). Consistently, longer persistence of DNA damage in cells treated with MN-OA and radiation was observed in the form of γ-H2AX foci. The efficacy and mechanism of MN-OA-induced radio-sensitization was also validated in an immuno-competent murine fibrosarcoma model. CONCLUSION This study reveals the key role of HSP90 in the mechanism of tumor radio-sensitization by MN-OA. GENERAL SIGNIFICANCE Present work provides a deeper understanding about the mechanism of MN-OA-induced tumor radiosensitization, highlighting the role of HSP90 protein. In addition to diagnostic and magnetic hyperthermia abilities, present remarkable radiosensitizing activity of MN-OA would further excite the clinicians to test its anti-cancer potential.
Collapse
|
18
|
Hum NR, Martin KA, Malfatti MA, Haack K, Buchholz BA, Loots GG. Tracking Tumor Colonization in Xenograft Mouse Models Using Accelerator Mass Spectrometry. Sci Rep 2018; 8:15013. [PMID: 30302019 PMCID: PMC6178347 DOI: 10.1038/s41598-018-33368-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Here we introduce an Accelerator Mass Spectrometry (AMS)-based high precision method for quantifying the number of cancer cells that initiate metastatic tumors, in xenograft mice. Quantification of 14C per cell prior to injection into animals, and quantification of 14C in whole organs allows us to extrapolate the number of cancer cells available to initiate metastatic tumors. The 14C labeling was optimized such that 1 cancer cell was detected among 1 million normal cells. We show that ~1–5% of human cancer cells injected into immunodeficient mice form subcutaneous tumors, and even fewer cells initiate metastatic tumors. Comparisons of metastatic site colonization between a highly metastatic (PC3) and a non-metastatic (LnCap) cell line showed that PC3 cells colonize target tissues in greater quantities at 2 weeks post-delivery, and by 12 weeks post-delivery no 14C was detected in LnCap xenografts, suggesting that all metastatic cells were cleared. The 14C-signal correlated with the presence and the severity of metastatic tumors. AMS measurements of 14C-labeled cells provides a highly-sensitive, quantitative assay to experimentally evaluate metastasis and colonization of target tissues in xenograft mouse models. This approach can potentially be used to evaluate tumor aggressiveness and assist in making informed decisions regarding treatment.
Collapse
Affiliation(s)
- Nicholas R Hum
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Livermore, CA, USA
| | - Kelly A Martin
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Livermore, CA, USA.,Georgetown University, Department of Biochemistry & Molecular Biology, Washington, DC, USA
| | - Michael A Malfatti
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Livermore, CA, USA
| | - Kurt Haack
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Livermore, CA, USA
| | - Bruce A Buchholz
- Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Gabriela G Loots
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Livermore, CA, USA. .,UC Merced, School of Natural Sciences, Merced, CA, USA.
| |
Collapse
|
19
|
Cort�s‑Mateus K, Holub K, Racca F, Grau J, Capdevila J. Concurrent palliative external radiotherapy with sorafenib or doxorubicin for bulky differentiated thyroid carcinoma: A case report. Oncol Lett 2018; 16:4085-4089. [DOI: 10.3892/ol.2018.9069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 06/19/2018] [Indexed: 11/05/2022] Open
Affiliation(s)
- Karen Cort�s‑Mateus
- Medical Oncology Department and Radiation Oncology Department, University of Barcelona, IDIBAPS, Hospital Clinic Barcelona, 08036 Barcelona, Spain
| | - Katarzyna Holub
- Medical Oncology Department and Radiation Oncology Department, University of Barcelona, IDIBAPS, Hospital Clinic Barcelona, 08036 Barcelona, Spain
| | - Fabricio Racca
- Vall Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Juan Grau
- Medical Oncology Department and Radiation Oncology Department, University of Barcelona, IDIBAPS, Hospital Clinic Barcelona, 08036 Barcelona, Spain
| | - Jaume Capdevila
- Vall Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| |
Collapse
|
20
|
Abstract
OPINION STATEMENT The survival rate for patients with advanced stages of squamous cell carcinoma of the head and neck (SCCHN) remains poor despite multimodal treatment options. Cetuximab, an anti-EGFR inhibitor, is the only FDA-approved targeted agent for this disease. Recent findings have implicated modifications of the microenvironment and, consequently, phenotypical modifications of the cancer cell, in treatment resistance mechanisms. For many years, cancer research has focused mainly on targetable sites on or inside the cancer cell. Nowadays, in preclinical and clinical studies, a greater emphasis is being placed on drugs that target the tumor microenvironment. Potential targets relate to tumor vascularization, immunology, extracellular matrix components, or cancer-associated fibroblasts. The combination of these new agents with standard treatment options is of particular interest to overcome resistance mechanisms and/or to increase treatment efficacy. Whereas antiangiogenic agents show poor clinical activity, immunotherapy seems to be a more promising tool with an objective response rate (ORR) of 20 % in patients with recurrent and/or metastatic squamous cell carcinoma (R/M SCC). Other targets, located inside the extracellular matrix or on cancer associated fibroblasts, are under preclinical investigation. These new agents all need to be tested in clinical trials alone, or in combination with standard treatment modalities, based on preclinical data. To increase our knowledge of the complex network between the cancer cell and its environment, preclinical studies should consider co-culture models, and clinical studies should incorporate a translational research objective.
Collapse
|
21
|
Ban K, Feng S, Shao L, Ittmann M. RET Signaling in Prostate Cancer. Clin Cancer Res 2017; 23:4885-4896. [PMID: 28490466 DOI: 10.1158/1078-0432.ccr-17-0528] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/24/2017] [Accepted: 05/04/2017] [Indexed: 12/14/2022]
Abstract
Purpose: Large diameter perineural prostate cancer is associated with poor outcomes. GDNF, with its coreceptor GFRα1, binds RET and activates downstream pro-oncogenic signaling. Because both GDNF and GFRα1 are secreted by nerves, we examined the role of RET signaling in prostate cancer.Experimental Design: Expression of RET, GDNF, and/or GFRα1 was assessed. The impact of RET signaling on proliferation, invasion and soft agar colony formation, perineural invasion, and growth in vivo was determined. Cellular signaling downstream of RET was examined by Western blotting.Results: RET is expressed in all prostate cancer cell lines. GFRα1 is only expressed in 22Rv1 cells, which is the only line that responds to exogenous GDNF. In contrast, all cell lines respond to GDNF plus GFRα1. Conditioned medium from dorsal root ganglia contains secreted GFRα1 and promotes transformation-related phenotypes, which can be blocked by anti-GFRα1 antibody. Perineural invasion in the dorsal root ganglion assay is inhibited by anti-GFRα antibody and RET knockdown. In vivo, knockdown of RET inhibits tumor growth. RET signaling activates ERK or AKT signaling depending on context, but phosphorylation of p70S6 kinase is markedly increased in all cases. Knockdown of p70S6 kinase markedly decreases RET induced transformed phenotypes. Finally, RET is expressed in 18% of adenocarcinomas and all three small-cell carcinomas examined.Conclusions: RET promotes transformation associated phenotypes, including perineural invasion in prostate cancer via activation of p70S6 kinase. GFRα1, which is secreted by nerves, is a limiting factor for RET signaling, creating a perineural niche where RET signaling can occur. Clin Cancer Res; 23(16); 4885-96. ©2017 AACR.
Collapse
Affiliation(s)
- Kechen Ban
- Department of Pathology & Immunology, Baylor College of Medicine and Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas
| | - Shu Feng
- Department of Pathology & Immunology, Baylor College of Medicine and Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas
| | - Longjiang Shao
- Department of Pathology & Immunology, Baylor College of Medicine and Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas
| | - Michael Ittmann
- Department of Pathology & Immunology, Baylor College of Medicine and Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas.
| |
Collapse
|
22
|
Wang T, Hu P, Li B, Zhang JP, Cheng YF, Liang YM. Role of Nrf2 signaling pathway in the radiation tolerance of patients with head and neck squamous cell carcinoma: an in vivo and in vitro study. Onco Targets Ther 2017; 10:1809-1819. [PMID: 28367064 PMCID: PMC5370066 DOI: 10.2147/ott.s122803] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We aimed to investigate the relationship between the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway and the radiation tolerance of patients with head and neck squamous cell carcinoma (HNSCC). From January 2015 to January 2016, 117 patients with HNSCC were enrolled in our study and assigned into the sensitive and tolerance groups based on curative effect. Immunohistochemistry (IHC) was conducted to measure protein expressions of Nrf2, heme oxygenase-1 (HO1), NADPH quinine oxidoreductase 1 (NQO1) and glutathione S-transferase (GST). Human squamous cell carcinoma cell line, HSC-4, was induced by radiation to construct the HSC-4-radiation resistance (RR) cell line. HSC-4 and HSC-4-RR were also assigned into the blank, negative control (NC) and Nrf2 siRNA groups. Cell Counting Kit-8 (CCK-8), quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were employed to detect cell viability, mRNA expression and protein expression, respectively, of Nrf2, HO1, NQO1 and GST. A total of 40 nude mice were equally assigned into the untreated, Nrf2 siRNA, radiation therapy (RT) and RT + Nrf2 siRNA groups. Compared with the sensitive group, patients in the tolerance group had upregulated Nrf2, HO1, NQO1 and GST expressions. HSC-4-RR cell line had improved cell viability and higher protein and mRNA expressions of Nrf2, HO1, NQO1 and GST compared with HSC-4 cell line. Compared with the HSC-4-NC and HSC-4-blank groups, the HSC-4-Nrf2 siRNA group had downregulated cell viability. Compared with the HSC-4-RR-NC and HSC-4-RR-blank groups, the HSC-4-RR-Nrf2 siRNA group had lower cell viability. However, the HSC-4-RR-Nrf2 siRNA group had elevated cell viability than the HSC-4-Nrf2 siRNA group. Tumor volume and tumor weight in the RT and RT + Nrf2 siRNA groups decreased evidently. The RT + Nrf2 siRNA group exhibited decreased tumor volume and tumor weight in comparison with the RT group. Our data demonstrated that downregulation of HO1, NQO1 and GST via inhibiting Nrf2 signaling pathway reduces the radiation tolerance of patients with HNSCC.
Collapse
Affiliation(s)
- Tao Wang
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Peng Hu
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Bo Li
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Jun-Peng Zhang
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Yu-Feng Cheng
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Ye-Min Liang
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| |
Collapse
|
23
|
Vazifedan V, Mousavi SH, Sargolzaei J, Soleymanifard S, Fani Pakdel A. Study of Crocin & Radiotherapy-induced Cytotoxicity and Apoptosis in the Head and Neck Cancer (HN-5) Cell Line. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2017; 16:230-237. [PMID: 28496478 PMCID: PMC5423250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Malignant tumors of head and neck carcinomas are the sixth most common type of cancer. Current systemic therapies for cancer show side effects in normal tissues and short-term efficacy due to drug resistance. Consequently, there is much interest in identifying new drugs for cancer treatment. Crocin (an active ingredient of saffron) has been shown to have cytotoxic effects on cancer cell lines. Chemo radiotherapy is the standard treatment for head and neck cancer. In the present study, the cytotoxic effects, inducing apoptosis and the radiation sensitivity of crocin were evaluated in the head and neck cancer cell line (HN-5). HN-5 cells were cultured in a DMEM medium and incubated with different concentrations of crocin (12.5-1000 µg/mL). They were exposed to 2 Gy γ-rays. Cell viability was quantified by the MTT assay. Apoptotic cells were determined using PI staining of DNA fragmentation by flowcytometry (sub-G1 peak). Crocin decreased cell viability in HN-5 cells in a time and concentration dependent manner. Crocin also induced a sub-G1 peak in the flowcytometery histogram of treated cells compared with the control, suggesting that apoptotic cell death is caused by its toxicity. Crocin was also shown to sensitize cells to radiation-induced toxicity and apoptosis. The simultaneous use of crocin and radiation therefore increases radiation sensitivity and cell death. Thus, after further study crocin can be considered as a potential drug and sensitizer in cancer treatment.
Collapse
Affiliation(s)
- Vahid Vazifedan
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran .
| | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Javad Sargolzaei
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | - Shokouhozaman Soleymanifard
- Omid Hospital Medical Physics Department, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Physics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Azar Fani Pakdel
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad , Iran.
| |
Collapse
|
24
|
Effect of postoperative radiotherapy for squamous cell cancer of the breast in a surveillance epidemiology and end results population-based study. Oncotarget 2016; 7:10684-93. [PMID: 26863453 PMCID: PMC4891151 DOI: 10.18632/oncotarget.7222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/24/2016] [Indexed: 11/25/2022] Open
Abstract
The therapeutic value of postoperative radiotherapy (RT) for squamous cell cancer of the breast (SCCB) is unclear. This retrospective study used a population-based national registry to determine the impact of postoperative RT on survival of women with SCCB. The Surveillance Epidemiology and End Results (SEER) database was used to identify females with SCCB who underwent primary surgical resection from 1973 to 2012. Kaplan–Meier survival analysis and Cox regression proportional hazard methods were used to determine the impact of RT following resection associated with cause-specific survival (CSS) and overall survival (OS). A total of 523 patients met the eligibility criteria. The median follow-up time was 55 months, the 10-year CSS and OS rates were 65.6%, and 46.0%, respectively. A total of 167 patients (31.9%) received postoperative RT. Multivariate analysis indicated that advanced pT and pN stage, and no postoperative RT were independently associated with poor OS; advanced pT and pN stage were independently associated with poor CSS. Postoperative RT was significantly associated with improved 10-year OS (54.5% vs. 42.0%, P =.001), but had no effect on CSS (P =.217). Analysis of patients with different stages of SCCB indicated that RT was associated with improved CSS (P =.047) and OS (P <.001) in those with stage II cancer and improved OS in patients with stage pN0 cancer (P <.001). Postoperative RT improved the survival of SCCB patients, especially in those with stage II and stage pN0 cancer.
Collapse
|
25
|
Saga R, Monzen S, Chiba M, Yoshino H, Nakamura T, Hosokawa Y. Anti-tumor and anti-invasion effects of a combination of 4-methylumbelliferone and ionizing radiation in human fibrosarcoma cells. Oncol Lett 2016; 13:410-416. [PMID: 28123575 DOI: 10.3892/ol.2016.5385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 10/03/2016] [Indexed: 11/06/2022] Open
Abstract
Hyaluronan (HA) is a major component of the extracellular matrix that is synthesized in excess in cancer tissues. 4-methylumbelliferone (MU) inhibits the synthesis of HA and is closely related to the invasion and metastasis of cancer. However, the effects of MU in conjunction with cancer radiotherapy remain unknown. The present study assessed the anti-tumor and anti-invasion effects of the concomitant use of ionizing radiation (IR) and 100 µM MU on human fibrosarcoma HT1080 cells. Cell viability and cellular invasion potency assays were performed. There was a greater decrease in the viability of cells cultured with a combination of 2 Gy IR and MU compared with untreated control cells. In addition, cell cycle distribution analysis demonstrated that a higher proportion of these cells were in the sub-G1 phase and higher fractions of annexin-V positive, propidium iodide positive cells (i.e., apoptotic cells) were observed. HA concentration in the 2 Gy irradiated culture was similar to that in the non-irradiated control culture, however, it significantly decreased following the administration of both MU alone and 2 Gy IR with MU. Furthermore, treatment with 2 Gy IR and MU resulted in a significant decrease in the invasion rate and matrix metalloproteinase (MMP)-2 and MPP-9 expression. Taken together, these results suggest that the administration of MU with 2 Gy IR is effective at reducing HA production, cell invasion and the metastatic potential of cancer cells.
Collapse
Affiliation(s)
- Ryo Saga
- Department of Radiological Life Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Satoru Monzen
- Department of Radiological Life Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Mitsuru Chiba
- Department of Biomedical Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Hironori Yoshino
- Department of Radiological Life Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Toshiya Nakamura
- Department of Biomedical Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Yoichiro Hosokawa
- Department of Radiological Life Sciences, Division of Medical Life Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| |
Collapse
|
26
|
Koole K, Peeters T, van Hengel ORJ, Brinkhuis A, van Es RJJ, Willems SM. RET protein is overexpressed in oral squamous cell carcinoma. Histopathology 2016; 69:1085-1087. [PMID: 27344057 DOI: 10.1111/his.13023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Koos Koole
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Department of Head and Neck Surgical Oncology, UMC Utrecht Cancer Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Ton Peeters
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Oscar R J van Hengel
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Anja Brinkhuis
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Robert J J van Es
- Department of Head and Neck Surgical Oncology, UMC Utrecht Cancer Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Stefan M Willems
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Ma W, Yu J, Qi X, Liang L, Zhang Y, Ding Y, Lin X, Li G, Ding Y. Radiation-induced microRNA-622 causes radioresistance in colorectal cancer cells by down-regulating Rb. Oncotarget 2016; 6:15984-94. [PMID: 25961730 PMCID: PMC4599251 DOI: 10.18632/oncotarget.3762] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/06/2015] [Indexed: 02/06/2023] Open
Abstract
The standard treatment for patients with locally advanced rectal cancer is preoperative 5-fluorouracil-based chemoradiotherapy followed by total mesorectal excision. However, tumor response to standard dose radiation varies. In this study, we found that miR-622 was increased significantly in ionizing radiation-treated colorectal cancer (CRC) cells compared to the cells cultured with irradiated medium, and persisted stably in surviving cells treated with continuous low-dose radiation. Overexpression of miR-622 induced the radioresistance in vitro. In addition, miR-622 inhibited Rb expression by directly targeting RB1-3′UTR. Overexpression of Rb reversed miR-622-induced radioresistance in vitro. In response to ionizing radiation, the Rb-E2F1-P/CAF complex activated proapoptotic genes. Importantly, miR-622 was highly expressed in tumors of rectal cancer patients with non-regression after standard dose radiotherapy. In conclusion, miR-622 overexpressing cells are induced or selected by radiotherapy, causing in turn radioresistance and poor response to further therapy. MiR-622 is a potential biomarker of responders for radiotherapy and a potential therapeutic target.
Collapse
Affiliation(s)
- Wenhui Ma
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolong Qi
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Zhang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoshan Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Takahashi Y, Lee JG, Pickering C, Bell D, Jiffar TW, Myers JN, Hanna EY, Kupferman ME. Human epidermal growth factor receptor 2/neu as a novel therapeutic target in sinonasal undifferentiated carcinoma. Head Neck 2016; 38 Suppl 1:E1926-34. [PMID: 26752332 PMCID: PMC6453572 DOI: 10.1002/hed.24350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/27/2015] [Accepted: 10/17/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Sinonasal undifferentiated carcinoma (SNUC) is a rare and aggressive cancer. Despite multimodal therapy, the prognosis in SNUC remains poor, and new therapies are needed. Thus, the purpose of this study was to explore potential therapeutic targets in SNUC. METHODS Using the human-derived SNUC MDA8788-6 cell line, we performed whole genome single nucleotide polymorphism (SNP) analysis to identify copy number changes in this line. Protein expression levels were evaluated by Western blotting. Cell growth inhibition was assessed by methylthiazol tetrazolium (MTT) and clonogenic assays. The mouse flank model was used to examine the effect of growth inhibition in vivo. RESULTS The ERBB2 gene was highly amplified and cell extracts showed human epidermal growth factor receptor 2 (HER2) was overexpressed and phosphorylated in MDA8788-6. Lapatinib effectively inhibited the HER2 signaling pathway in our SNUC cell line. HER2 inhibition successfully suppressed the cell growth of MDA8788-6 cells both in vitro and in vivo. CONCLUSION Targeting HER2 may be a promising avenue for the development of novel therapies for SNUC. © 2016 Wiley Periodicals, Inc. Head Neck 38: E1926-E1934, 2016.
Collapse
Affiliation(s)
- Yoko Takahashi
- Departments of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - June Goo Lee
- Departments of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Curtis Pickering
- Departments of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Diana Bell
- Departments of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tilahun W. Jiffar
- Departments of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey N. Myers
- Departments of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ehab Y. Hanna
- Departments of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael E. Kupferman
- Departments of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
29
|
Mu X, Ma J, Zhang Z, Zhou H, Xu S, Qin Y, Huang J, Yang K, Wu G. Famitinib enhances nasopharyngeal cancer cell radiosensitivity by attenuating radiation-induced phosphorylation of platelet-derived growth factor receptor and c-kit and inhibiting microvessel formation. Int J Radiat Biol 2015; 91:771-6. [PMID: 26073526 DOI: 10.3109/09553002.2015.1062574] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE Famitinib is a novel tyrosine kinase inhibitor. We investigated the effects of famitinib on the radiosensitivity of human nasopharyngeal carcinoma (NPC) cell radiosensitivity in vitro and in vivo, and explored its possible mechanisms. MATERIALS AND METHODS Human nasopharyngeal carcinoma cell line (CNE-2) were treated with famitinib and radiation, and analyzed by3-(4,5-dimethylthaizol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), clonogenic survival assay, and Western blot. A xenograft model using CNE-2 cells was established to analyze the effects of famitinib and radiation on tumor volume and microvessel density (MVD). RESULTS Famitinib dose-dependently inhibited CNE-2 cells growth and significantly reduced clonogenic survival (p < 0.05), with a sensitivity enhancement ratio (SER) of 1.45. The tumor inhibition rate of the combined treatment group was 91%, which was significantly higher than the radiation group (35%, p < 0.05) and famitinib group (46%, p < 0.05). Famitinib attenuated radiation-induced phosphorylation of the platelet-derived growth factor receptor (PDGFR) and stem cell factor (c-kit) at 0, 30, 60 min after radiation treatment. Furthermore, radiation combined with famitinib decreased tumor MVD (p < 0.05). CONCLUSIONS Famitinib significantly increased CNE-2 cell radiosensitivity in vitro and in vivo by attenuating radiation-induced PDGFR and c-kit phosphorylation and by inhibiting microvessel formation.
Collapse
Affiliation(s)
- Xiaoqian Mu
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China.,b Department of Medical Oncology , Affiliated Cancer Hospital of Zhengzhou University , Zhengzhou , Henan, China
| | - Jia Ma
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Zhanjie Zhang
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Hongxia Zhou
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Shuangbing Xu
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - You Qin
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Jing Huang
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Kunyu Yang
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Gang Wu
- a Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
30
|
Ventelä S, Sittig E, Mannermaa L, Mäkelä JA, Kulmala J, Löyttyniemi E, Strauss L, Cárpen O, Toppari J, Grénman R, Westermarck J. CIP2A is an Oct4 target gene involved in head and neck squamous cell cancer oncogenicity and radioresistance. Oncotarget 2015; 6:144-58. [PMID: 25474139 PMCID: PMC4381584 DOI: 10.18632/oncotarget.2670] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 11/02/2014] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy is a mainstay for treatment of many human cancer types, including head and neck squamous cell carcinoma (HNSCC). Thereby, it is clinically very relevant to understand the mechanisms determining radioresistance. Here, we identify CIP2A as an Oct4 target gene and provide evidence that they co-operate in radioresistance. Oct4 positively regulates CIP2A expression both in testicular cancer cell lines as well as in embryonic stem cells. To expand the relevance of these findings we show that Oct4 and CIP2A are co-expressed in CD24 positive side-population of patient-derived HNSCC cell lines. Most importantly, all Oct4 positive HNSCC patient samples were CIP2A positive and this double positivity was linked to poor differentiation level, and predicted for decreased patient survival among radiotherapy treated HNSCC patients. Oct4 and CIP2A expression was also linked with increased aggressiveness and radioresistancy in HNSCC cell lines. Together we demonstrate that CIP2A is a novel Oct4 target gene in stem cells and in human cancer cell lines. Clinically these results suggest that diagnostic evaluation of HNSCC tumors for Oct4 or Oct4/CIP2A positivity might help to predict HNSCC tumor radioresistancy. These results also identify both Oct4 and CIP2A as potential targets for radiosensitation.
Collapse
Affiliation(s)
- Sami Ventelä
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland. Department of Physiology, University of Turku, Finland. Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital, Turku, Finland
| | - Eleonora Sittig
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland
| | - Leni Mannermaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland
| | | | - Jarmo Kulmala
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | | | - Leena Strauss
- Institute of Biomedicine and Turku Center for Disease Modeling, University of Turku, Finland
| | - Olli Cárpen
- Department of Pathology, University of Turku, Finland
| | - Jorma Toppari
- Department of Physiology, University of Turku, Finland. Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Reidar Grénman
- Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital, Turku, Finland
| | - Jukka Westermarck
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland. Department of Pathology, University of Turku, Finland
| |
Collapse
|
31
|
Lukianova-Hleb EY, Lapotko DO. Rapid detection and destruction of squamous cell carcinoma of the head and neck by nano-quadrapeutics. Head Neck 2015; 37:1547-55. [DOI: 10.1002/hed.24018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 10/12/2014] [Accepted: 02/06/2015] [Indexed: 01/28/2023] Open
Affiliation(s)
| | - Dmitri O. Lapotko
- Department of BioSciences at Rice; Rice University; Houston Texas
- Department of Physics and Astronomy; Rice University; Houston Texas
| |
Collapse
|
32
|
Abstract
In recent years, our knowledge and understanding of head and neck squamous cell carcinoma (HNSCC) has expanded dramatically. New high-throughput sequencing technologies have accelerated these discoveries since the first reports of whole-exome sequencing of HNSCC tumors in 2011. In addition, the discovery of human papillomavirus in relationship with oropharyngeal squamous cell carcinoma has shifted our molecular understanding of the disease. New investigation into the role of immune evasion in HNSCC has also led to potential novel therapies based on immune-specific systemic therapies.
Collapse
Affiliation(s)
- Theresa Guo
- Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins Medical Institutions, 1550 Orleans St, Baltimore, MD 21231, USA.
| | - Joseph A Califano
- Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins Medical Institutions, 1550 Orleans St, Baltimore, MD 21231, USA
| |
Collapse
|
33
|
Papadimitrakopoulou VA, Frank SJ, Cohen EW, Hirsch FR, Myers JN, Heymach JV, Lin H, Tran HT, Chen CR, Jimeno A, Nedzi L, Vasselli JR, Lowe ES, Raben D. Phase I study of vandetanib with radiation therapy with or without cisplatin in locally advanced head and neck squamous cell carcinoma. Head Neck 2015; 38:439-47. [PMID: 25352401 DOI: 10.1002/hed.23922] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Vandetanib, added to cisplatin and radiation therapy (RT) overcomes chemoradiation therapy (CRT) and epidermal growth factor receptor (EGFR) inhibitor resistance in head and neck squamous cell carcinoma (HNSCC) lines and models. METHODS Patients with previously untreated HNSCC received vandetanib daily for 14 days (starting dose 100 mg) and then vandetanib + RT (2.2 Gy/day, 5 days/week) for 6 weeks (regimen 1) or vandetanib + RT (2 Gy/day, 5 days/week) + cisplatin (30 mg/m(2) weekly) for 7 weeks (regimen 2). The primary objective was the maximum tolerated dose (MTD) of vandetanib with RT +/- cisplatin. RESULTS Of 33 treated patients, 30 completed therapy (regimen 1, n = 12; regimen 2, n = 18). MTD in regimen 2 was 100 mg (3 dose limiting toxicities [DLTs] at 200 mg), whereas regimen 1 was stopped because of poor recruitment (1 DLT at 200 mg). Most common grade ≥3 adverse events (AEs) were dysphagia (30%), stomatitis (33%), and mucosal inflammation (27%). Five patients discontinued vandetanib because of AEs. CONCLUSION Vandetanib with CRT was feasible.
Collapse
Affiliation(s)
| | - Steven J Frank
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Ezra W Cohen
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Fred R Hirsch
- University of Colorado Cancer Center, Aurora, Colorado
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - John V Heymach
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Heather Lin
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas
| | - Hai T Tran
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Changhu R Chen
- Department of Radiation Oncology, University of Toledo, Toledo, Ohio
| | - Antonio Jimeno
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Lucien Nedzi
- Department of Radiation Oncology, Southwestern Medical Center, Dallas, Texas
| | | | | | - David Raben
- Department of Oncology, University of Colorado, Aurora, Colorado
| |
Collapse
|
34
|
Price KAR, Cohen EEW. Mechanisms of and therapeutic approaches for overcoming resistance to epidermal growth factor receptor (EGFR)-targeted therapy in squamous cell carcinoma of the head and neck (SCCHN). Oral Oncol 2015; 51:399-408. [PMID: 25725588 DOI: 10.1016/j.oraloncology.2015.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 01/19/2015] [Accepted: 01/28/2015] [Indexed: 01/07/2023]
Abstract
The majority of squamous cell carcinoma of the head and neck (SCCHN) overexpress epidermal growth factor receptor (EGFR), which has been associated with poor treatment response and survival. However, only modest success has been achieved with the use of single agents that target EGFR, possibly due to primary and acquired resistance. This review will discuss key mechanisms of and therapeutic approaches to overcoming resistance to EGFR-targeted therapy in SCCHN. Recent preclinical and clinical investigations have demonstrated that other ErbB family receptors (eg, HER2 and HER3) and other horizontal resistance mechanisms, as well as activation of downstream signaling pathways, epigenetic events, and nuclear EGFR, are possible mediators of resistance to anti-EGFR therapeutics. Key downstream pathways that may be implicated in EGFR resistance include phosphatidylinositol-3-kinase/protein kinase B, vascular endothelial growth factor (VEGF), and mammalian target of rapamycin (mTOR). Multiple agents that target EGFR and other ErbB family members (ie, lapatinib, afatinib, and dacomitinib), as well as combination therapies that target EGFR and selected other pathways (eg, VEGF, mTOR, and c-Met) are being investigated clinically. In addition, several phase II and III trials continue to investigate strategies to enhance the efficacy of EGFR-targeted therapy in SCCHN.
Collapse
Affiliation(s)
| | - Ezra E W Cohen
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
35
|
Lukianova-Hleb EY, Ren X, Sawant RR, Wu X, Torchilin VP, Lapotko DO. On-demand intracellular amplification of chemoradiation with cancer-specific plasmonic nanobubbles. Nat Med 2014; 20:778-784. [PMID: 24880615 PMCID: PMC4271542 DOI: 10.1038/nm.3484] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 10/24/2013] [Indexed: 02/08/2023]
Abstract
Chemoradiation-resistant cancers limit treatment efficacy and safety. We show here the cancer cell-specific, on-demand intracellular amplification of chemotherapy and chemoradiation therapy via gold nanoparticle- and laser pulse-induced mechanical intracellular impact. Cancer aggressiveness promotes the clustering of drug nanocarriers and gold nanoparticles in cancer cells. This cluster, upon exposure to a laser pulse, generates a plasmonic nanobubble, the mechanical explosion that destroys the host cancer cell or ejects the drug into its cytoplasm by disrupting the liposome and endosome. The same cluster locally amplifies external X-rays. Intracellular synergy of the mechanical impact of plasmonic nanobubble, ejected drug and amplified X-rays improves the efficacy of standard chemoradiation in resistant and aggressive head and neck cancer by 100-fold in vitro and 17-fold in vivo, reduces the effective entry doses of drugs and X-rays to 2-6% of their clinical doses and efficiently spares normal cells. The developed quadrapeutics technology combines four clinically validated components and transforms a standard macrotherapy into an intracellular on-demand theranostic microtreatment with radically amplified therapeutic efficacy and specificity.
Collapse
Affiliation(s)
| | - Xiaoyang Ren
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rupa R Sawant
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts, USA
| | - Xiangwei Wu
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts, USA
| | - Dmitri O Lapotko
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, USA
- Department of Physics and Astronomy, Rice University, Houston, Texas, USA
| |
Collapse
|
36
|
Melancon MP, Zhou M, Zhang R, Xiong C, Allen P, Wen X, Huang Q, Wallace M, Myers JN, Stafford RJ, Liang D, Ellington AD, Li C. Selective uptake and imaging of aptamer- and antibody-conjugated hollow nanospheres targeted to epidermal growth factor receptors overexpressed in head and neck cancer. ACS NANO 2014; 8:4530-8. [PMID: 24754567 PMCID: PMC4046795 DOI: 10.1021/nn406632u] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 04/22/2014] [Indexed: 05/26/2023]
Abstract
The purpose of this study was to compare the binding affinity and selective targeting of aptamer- and antibody-coated hollow gold nanospheres (HAuNS) targeted to epidermal growth factor receptors (EGFR). EGFR-targeting aptamers were conjugated to HAuNS (apt-HAuNS) by attaching a thiol-terminated single-stranded DNA to the HAuNS and then adding the complementary RNA targeted to EGFR. Apt-HAuNS was characterized in terms of size, surface charge, absorption, and number of aptamers per particle. The in vivo pharmacokinetics, in vivo biodistribution, and micro-SPECT/CT imaging of (111)In-labeled apt-HAuNS and anti-EGFR antibody (C225)-conjugated HAuNS were evaluated in nude mice bearing highly malignant human OSC-19 oral tumors. (111)In-labeled PEG-HAuNS was used as a control (n = 5/group). Apt-HAuNS did not have an altered absorbance profile or size (λmax = 800 nm; diameter = 55 nm) compared to C225-HAuNS or PEG-HAuNS. The surface charge became more negative upon conjugation of the aptamer (-51.4 vs -19.0 for PEG-HAuNS and -25.0 for C225-HAuNS). The number of aptamers/particle was ∼250. In vitro cell binding and in vivo biodistribution showed selective binding of the apt-HAuNS to EGFR. μSPECT/CT imaging confirmed that there was more tumor uptake of apt-HAuNS than C225-HAuNS. Aptamer is a promising ligand for image-guided delivery of nanoparticles for treatment of tumor cells overexpressing EGFR.
Collapse
Affiliation(s)
- Marites Pasuelo Melancon
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Min Zhou
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Rui Zhang
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Chiyi Xiong
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Peter Allen
- Department of Chemistry and Biochemistry, The University of Texas at Austin, 1 University Station, Austin, Texas 78712, United States
| | - Xiaoxia Wen
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Qian Huang
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Michael Wallace
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Jeffrey N. Myers
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - R. Jason Stafford
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Dong Liang
- Department of Pharmaceutical Sciences, Texas Southern University, 3100 Cleburne Street, Houston, Texas 77004, United States
| | - Andrew D. Ellington
- Department of Chemistry and Biochemistry, The University of Texas at Austin, 1 University Station, Austin, Texas 78712, United States
| | - Chun Li
- Departments of Interventional Radiology, Cancer Systems Imaging, Head and Neck Surgery, and Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| |
Collapse
|
37
|
Integration of molecular targeted therapy with radiation in head and neck cancer. Pharmacol Ther 2014; 142:88-98. [DOI: 10.1016/j.pharmthera.2013.11.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/01/2013] [Indexed: 12/20/2022]
|
38
|
Yamashita AS, Baia GS, Ho JSY, Velarde E, Wong J, Gallia GL, Belzberg AJ, Kimura ET, Riggins GJ. Preclinical evaluation of the combination of mTOR and proteasome inhibitors with radiotherapy in malignant peripheral nerve sheath tumors. J Neurooncol 2014; 118:83-92. [PMID: 24668609 DOI: 10.1007/s11060-014-1422-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 03/10/2014] [Indexed: 12/18/2022]
Abstract
About one half of malignant peripheral nerve sheath tumors (MPNST) have Neurofibromin 1 (NF1) mutations. NF1 is a tumor suppressor gene essential for negative regulation of RAS signaling. Survival for MPNST patients is poor and we sought to identify an effective combination therapy. Starting with the mTOR inhibitors rapamycin and everolimus, we screened for synergy in 542 FDA approved compounds using MPNST cells with a native NF1 loss in both alleles. We further analyzed the cell cycle and signal transduction. In vivo growth effects of the drug combination with local radiation therapy (RT) were assessed in MPNST xenografts. The synergistic combination of mTOR inhibitors with bortezomib yielded a reduction in MPNST cell proliferation. The combination of mTOR inhibitors and bortezomib also enhanced the anti-proliferative effect of radiation in vitro. In vivo, the combination of mTOR inhibitor (everolimus) and bortezomib with RT decreased tumor growth and proliferation, and augmented apoptosis. The combination of approved mTOR and proteasome inhibitors with radiation showed a significant reduction of tumor growth in an animal model and should be investigated and optimized further for MPNST therapy.
Collapse
Affiliation(s)
- A S Yamashita
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Johns Hopkins University, Koch Building Rm. 257, 1550 Orleans Street, Baltimore, MD, 21231, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Fu ZZ, Sun XD, Li P, Zhang Z, Li GZ, Gu T, Shao SS. Relationship between serum VEGF level and radiosensitivity of patients with nonsmall cell lung cancer among asians: a meta-analysis. DNA Cell Biol 2014; 33:426-37. [PMID: 24628278 DOI: 10.1089/dna.2013.2249] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
This meta-analysis aims to evaluate the relationships between serum vascular endothelial growth factor (VEGF) level and radiosensitivity in patients with nonsmall cell lung cancer (NSCLC) among Asians. We searched CISCOM, CINAHL, Web of Science, PubMed, Google Scholar, EBSCO, Cochrane Library, and CBM databases from their inception through October 1, 2013. Meta-analysis was performed using the STATA 12.0 software. Fourteen clinical studies were included in this meta-analysis, including five case-control studies and nine cohort studies. Our meta-analysis results revealed that levels of serum VEGF in NSCLC patients were higher than that of healthy controls. There was a significant difference in serum VEGF levels between before and after radiotherapy in NSCLC patients. Further, we found significant differences in serum VEGF levels between effective and noneffective clinical response groups pre- and postradiotherapy. Serum VEGF levels showed no significant associations with tumor-node-metastasis (TNM) stage and histologic grade in NSCLC patients. NSCLC patients with positive VEGF expression had shorter overall survival than those with negative VEGF expression. Our meta-analysis suggests that serum VEGF level may be a useful biomarker in predicting radiosensitivity and prognosis of NSCLC patients among Asians.
Collapse
Affiliation(s)
- Zhan-Zhao Fu
- 1 Department of Oncology, The First Hospital of Qinhuangdao , Qinhuangdao, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
Tao X, Hill KS, Gaziova I, Sastry SK, Qui S, Szaniszlo P, Fennewald S, Resto VA, Elferink LA. Silencing Met receptor tyrosine kinase signaling decreased oral tumor growth and increased survival of nude mice. Oral Oncol 2013; 50:104-12. [PMID: 24268630 DOI: 10.1016/j.oraloncology.2013.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/04/2013] [Accepted: 10/18/2013] [Indexed: 11/19/2022]
Abstract
OBJECTIVES The hepatocyte growth factor receptor (Met) is frequently overexpressed in Head and Neck Squamous Cell Carcinoma (HNSCC), correlating positively with high-grade tumors and shortened patient survival. As such, Met may represent an important therapeutic target. The purpose of this study was to explore the role of Met signaling for HNSCC growth and locoregional dissemination. MATERIALS AND METHODS Using a lentiviral system for RNA interference, we knocked down Met in established HNSCC cell lines that express high levels of the endogenous receptor. The effect of Met silencing on in vitro proliferation, cell survival and migration was examined using western analysis, immunohistochemistry and live cell imaging. In vivo tumor growth, dissemination and mouse survival was assessed using an orthotopic tongue mouse model for HNSCC. RESULTS We show that Met knockdown (1) impaired activation of downstream MAPK signaling; (2) reduced cell viability and anchorage independent growth; (3) abrogated HGF-induced cell motility on laminin; (4) reduced in vivo tumor growth by increased cell apoptosis; (5) caused reduced incidence of tumor dissemination to regional lymph nodes and (6) increased the survival of nude mice with orthotopic xenografts. CONCLUSION Met signaling is important for HNSCC growth and locoregional dissemination in vivo and that targeting Met may be an important strategy for therapy.
Collapse
Affiliation(s)
- X Tao
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States
| | - K S Hill
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States
| | - I Gaziova
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States
| | - S K Sastry
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States; Department of Sealy Center for Cancer Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States
| | - S Qui
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States; Department of Sealy Center for Cancer Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States
| | - P Szaniszlo
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States
| | - S Fennewald
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States
| | - V A Resto
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States; Department of Sealy Center for Cancer Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States
| | - L A Elferink
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States; Department of Sealy Center for Cancer Biology, University of Texas Medical Branch, Galveston, TX 77555-1074, United States.
| |
Collapse
|
41
|
Hsu HW, Wall NR, Hsueh CT, Kim S, Ferris RL, Chen CS, Mirshahidi S. Combination antiangiogenic therapy and radiation in head and neck cancers. Oral Oncol 2013; 50:19-26. [PMID: 24269532 DOI: 10.1016/j.oraloncology.2013.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/24/2013] [Accepted: 10/02/2013] [Indexed: 02/02/2023]
Abstract
Tumor angiogenesis is a hallmark of advanced cancers and promotes invasion and metastasis. Over 90% of head and neck squamous cell carcinomas (HNSCC) express angiogenic factors such as vascular endothelial growth factor (VEGF). Several preclinical studies support the prognostic implications of angiogenic markers for HNSCC and currently this is an attractive treatment target in solid tumors. Since radiotherapy is one of the most commonly used treatments for HNSCC, it is imperative to identify the interactions between antiangiogenic therapy and radiotherapy, and to develop combination therapy to improve clinical outcome. The mechanisms between antiangiogenic agents and ionizing radiation are complicated and involve many interactions between the vasculature, tumor stroma and tumor cells. The proliferation and metastasis of tumor cells rely on angiogenesis/blood vessel formation. Rapid growing tumors will cause hypoxia, which up-regulates tumor cell survival factors, such as hypoxia-inducing factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF), giving rise to more tumor proliferation, angiogenesis and increased radioresistance. Thus, agents that target tumor vasculature and new tumor vessel formation can modulate the tumor microenvironment to improve tumor blood flow and oxygenation, leading to enhanced radiosensitivity. In this review, we discuss the mechanisms of how antiangiogenic therapies improve tumor response to radiation and data that support this combination strategy as a promising method for the treatment of HNSCC in the future.
Collapse
Affiliation(s)
- Heng-Wei Hsu
- Department of Pharmacology, Loma Linda University, Loma Linda, CA, USA; Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA
| | - Nathan R Wall
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA, USA
| | - Chung-Tsen Hsueh
- Division of Oncology & Hematology, Loma Linda University, Loma Linda, CA, USA
| | - Seungwon Kim
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chien-Shing Chen
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA; Division of Oncology & Hematology, Loma Linda University, Loma Linda, CA, USA
| | - Saied Mirshahidi
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA; Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA; LLU Cancer Center Biospecimen Laboratory, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
42
|
Bossi P, Locati L, Licitra L. Emerging tyrosine kinase inhibitors for head and neck cancer. Expert Opin Emerg Drugs 2013; 18:445-59. [DOI: 10.1517/14728214.2013.842976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Schmitz S, Ang KK, Vermorken J, Haddad R, Suarez C, Wolf GT, Hamoir M, Machiels JP. Targeted therapies for squamous cell carcinoma of the head and neck: current knowledge and future directions. Cancer Treat Rev 2013; 40:390-404. [PMID: 24176789 DOI: 10.1016/j.ctrv.2013.09.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 08/26/2013] [Accepted: 09/04/2013] [Indexed: 12/19/2022]
Abstract
Despite progress in the therapeutic management of patients with squamous cell carcinoma of the head and neck (SCCHN), the mortality rate of patients presenting with advanced disease remains high. One approach to improve treatment efficacy is to add novel molecular targeted agents to the classical treatment regimens. Monoclonal antibodies targeting the epidermal growth factor receptor (EGFR) have shown clinical benefits in palliative and curative settings. However, only a minority of patients presenting with recurrent or metastatic (R/M) SCCHN have meaningful tumor regression with these agents and virtually all who do develop acquired tumor resistance after a few months of treatment. For these reasons, other inhibitors of EGFR or molecules that interfere with known molecular pathways activated in SCCHN are of considerable interest, either as single agents or in combination with other treatment modalities. In this review, we discuss the different molecular therapeutic approaches explored in SCCHN. We also briefly outline new trial designs that could be used to accelerate the investigation of emerging therapeutic agents in this disease.
Collapse
Affiliation(s)
- Sandra Schmitz
- Cancer Center, Department of Medical Oncology and Head and Neck Surgery, Cliniques Universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale (Pole MIRO), Université Catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium.
| | - Kie Kian Ang
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Jan Vermorken
- Antwerp University Hospital, Department of Medical Oncology, Wilrijkstraat 10, 2650 Edegem, Belgium.
| | - Robert Haddad
- Department of Medical Oncology, Head and Neck Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA.
| | - Carlos Suarez
- Department of Otolaryngology, Hospital Universitario Central de Asturias Oviedo, Celestino Villamil SN, 33006 Oviedo, Asturias, Spain.
| | - Gregory T Wolf
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, Ann Arbor, MI 48109, USA.
| | - Marc Hamoir
- Cancer Center, Department of Medical Oncology and Head and Neck Surgery, Cliniques Universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale (Pole MIRO), Université Catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium.
| | - Jean-Pascal Machiels
- Cancer Center, Department of Medical Oncology and Head and Neck Surgery, Cliniques Universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale (Pole MIRO), Université Catholique de Louvain, Avenue Hippocrate 10, 1200 Brussels, Belgium.
| |
Collapse
|
44
|
Sweeny L, Hartman YE, Zinn KR, Prudent JR, Marshall DJ, Shekhani MS, Rosenthal EL. A novel extracellular drug conjugate significantly inhibits head and neck squamous cell carcinoma. Oral Oncol 2013; 49:991-7. [PMID: 23920309 DOI: 10.1016/j.oraloncology.2013.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 07/13/2013] [Accepted: 07/17/2013] [Indexed: 12/01/2022]
Abstract
OBJECTIVES Despite advances in treatment modalities, head and neck squamous cell carcinoma (HNSCC) remains a challenge to treat with poor survival and high morbidity, necessitating a therapy with greater efficacy. EDC22 is an extracellular drug conjugate of the monoclonal antibody targeting CD147 (glycoprotein highly expressed on HNSCC cells) linked with a small drug molecule inhibitor of Na, K-ATPase. In this study, EDC22's potential as a treatment modality for HNSCC was performed. MATERIALS AND METHODS HNSCC cell lines (FADU, OSC-19, Cal27, SCC-1) were cultured in vitro and proliferation and cell viability were assessed following treatment with a range of concentrations of EDC22 (0.25-5.00μg/mL). Mice bearing HNSCC xenografts (OSC-19, SCC-1) were treated with either EDC22 (3-10mg/kg), anti-CD147 monoclonal antibody, cisplatin (1mg/kg) or radiation therapy (2Gy/week) monotherapy or in combination. RESULTS In vitro, treatment with minimal concentration of EDC22 (0.25μg/mL) significantly decreased cellular proliferation and cell viability (p<0.0001). In vivo, systemic treatment with EDC22 significantly decreased primary tumor growth rate in both an orthotopic mouse model (OSC-19) and a flank tumor mouse model (SCC-1) (p<0.05). In addition, EDC22 therapy resulted in a greater reduction in tumor growth in vivo compared to radiation monotherapy (p<0.05) and a similar reduction in tumor growth compared to cisplatin monotherapy. Combination therapy provided no significant further reduction in tumor growth relative to EDC22 monotherapy. CONCLUSION EDC22 is a potent inhibitor of HNSCC cell proliferation in vitro and in vivo, warranting further investigations of its clinical potential in the treatment of HNSCC.
Collapse
Affiliation(s)
- Larissa Sweeny
- University of Alabama at Birmingham, Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, 1670 University Boulevard, Volker Hall G082, Birmingham, AL 35233, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Takahashi H, Hyakusoku H, Horii C, Takahashi M, Nishimura G, Taguchi T, Kondo N, Sakakibara A, Urata Y, Sano D. Telomerase-specific oncolytic adenovirus: antitumor effects on radiation-resistant head and neck squamous cell carcinoma cells. Head Neck 2013; 36:411-8. [PMID: 23728900 DOI: 10.1002/hed.23309] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2013] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Radioresistance remains a critical issue in the use of radiotherapy for the treatment of head and neck squamous cell carcinoma (HNSCC). This study evaluated the efficacy of combination treatment with OBP-301, a telomerase-specific replication-selective adenovirus, and radiotherapy in overcoming radioresistance by examining its effect on radiation-resistant HNSCC cells. METHODS Radiation-resistant HNSCC cells were treated with OBP-301 and radiation in vitro and in an orthotopic nude mouse model in vivo and synergism was assessed. Apoptosis and expression of MRN complex, which plays a key role in DNA repair machinery, were also analyzed. RESULTS Infection with OBP-301 was found to enhance the antitumor efficacy of radiation both in vitro and in vivo by inhibiting MRN complex expression and increasing apoptosis induction. CONCLUSION Combined OBP-301 and radiation therapy seems to overcome radioresistance in HNSCC cells by inhibiting DNA repair machinery, and may thus be a novel therapeutic strategy for treating HNSCC.
Collapse
Affiliation(s)
- Hideaki Takahashi
- Department of Otorhinolaryngology and Head and Neck Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pendleton KP, Grandis JR. Cisplatin-Based Chemotherapy Options for Recurrent and/or Metastatic Squamous Cell Cancer of the Head and Neck. ACTA ACUST UNITED AC 2013; 2013. [PMID: 24273416 DOI: 10.4137/cmt.s10409] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Recurrent and/or metastatic head and neck squamous cell carcinoma (R/M HNSCC) is a devastating malignancy with a poor prognosis. Treatment is limited to chemotherapeutic approaches. Cisplatin is an established and effective treatment for R/M HNSCC, and many studies have investigated cisplatin treatment in combination with other agents. Even when being treated with first line therapy (cisplatin + 5-fluorouracil + cetuximab), overall survival is only 10 months, indicating the need for novel chemotherapeutics and treatment regimens. Current research is focused on molecular targeting therapies inhibiting epidermal growth factor receptor, phosphoinositide-3-kinase/Akt/mammalian target of rapamycin, and vascular endothelial growth factor pathways. A variety of clinical trials have been completed and are currently underway with encouraging results. Finally, future directions of cisplatin-based R/M HNSCC treatment may include targeting specific pathways known to induce cisplatin resistance, such as nucleotide excision repair and inhibition of apoptosis, in hopes to enhance response to cisplatin therapy.
Collapse
Affiliation(s)
- Kelsey P Pendleton
- University of Pittsburgh, Department of Otolaryngology, Eye and Ear Institute, Pittsburgh, PA
| | | |
Collapse
|
47
|
Farnebo L, Tiefenböck K, Ansell A, Thunell LK, Garvin S, Roberg K. Strong expression of survivin is associated with positive response to radiotherapy and improved overall survival in head and neck squamous cell carcinoma patients. Int J Cancer 2013; 133:1994-2003. [PMID: 23564498 DOI: 10.1002/ijc.28200] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/22/2013] [Indexed: 02/06/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a malignancy that is associated with severe mortality despite advances in therapy. Today's standard treatment most commonly includes radiotherapy, often combined with chemotherapy or surgery. There are so far no established biomarkers to predict response to radiation, and thus the aim of this study was to investigate a series of markers that could potentially identify HNSCC patients who would benefit from radiotherapy. The selected markers, both proteins (epidermal growth factor receptor, survivin and p53), and single nucleotide polymorphisms (SNPs) in the genes of XRCC3, XRCC1, XPC, XPD, MDM2, p53 and FGFR4 were correlated to the response to radiotherapy and overall survival. Investigations were performed on pretreatment tumor biopsies from patients classified as responders or nonresponders to radiotherapy. Protein expression was examined using immunohistochemistry and the genotyping of specific SNPs was analyzed using PCR-RFLP or pyrosequencing. We found that survivin expression was significantly stronger in the responder group (p = 0.003) and that patients with a strong survivin expression had a significantly better overall survival (p < 0.001). Moreover, downregulation of survivin by siRNA in two HNSCC cell lines significantly decreased their sensitivity to radiation. Among the SNPs analyzed, patients with the XPD Lys751Gln SNP had a significantly shorter overall survival (p = 0.048), and patients with the FGFR4 Gly388Arg SNP had a significantly longer overall survival (p = 0.010). In conclusion, our results suggest that survivin plays an important role in the response to radiotherapy and may be a useful marker for predicting radiotherapy response in patients with HNSCC.
Collapse
Affiliation(s)
- Lovisa Farnebo
- Department of ENT-Head and Neck Surgery, UHL, County Council of Östergötland, Linköping, Sweden
| | | | | | | | | | | |
Collapse
|
48
|
Hoeben A, Martin D, Clement PM, Cools J, Gutkind JS. Role of GRB2-associated binder 1 in epidermal growth factor receptor-induced signaling in head and neck squamous cell carcinoma. Int J Cancer 2013; 132:1042-50. [PMID: 22865653 PMCID: PMC3498529 DOI: 10.1002/ijc.27763] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 06/20/2012] [Indexed: 11/05/2022]
Abstract
The epidermal growth factor receptor (EGFR) plays an important role in the pathogenesis of head and neck squamous cell carcinoma (HNSCC). Despite the high expression of EGFR in HNSCC, EGFR inhibitors have only limited success as monotherapy. The Grb2-associated binder (GAB) family of adaptor proteins acts as docking/scaffolding molecules downstream of tyrosine kinase receptors. We hypothesized that GAB1 may amplify EGFR-induced signaling in HNSCCs and therefore could play a role in the reduced sensitivity of HNSCC to EGFR inhibitors. We used representative human HNSCC cell lines overexpressing wild type EGFR, and expressing GAB1 but not GAB2. We demonstrated that baseline Akt and MAPK signaling were reduced in HNSCC cells in which GAB1 expression was reduced. Furthermore, the maximal EGF-induced activation of the Akt and MAPK pathway was reduced and delayed, and the duration of the EGF-induced activation of these pathways was reduced in cells with GAB1 knock-down. In agreement with this, HNSCC cells in which GAB1 levels were reduced showed an increased sensitivity to the EGFR inhibitor gefitinib. Our work demonstrates that GAB1 plays an important role as part of the mechanism of by which EGFR induces induced activation of the MAPK and AKT pathway. Our results identify GAB1 as an amplifier of the EGFR-initiated signaling, which may also interfere with EGFR degradation. These findings support the emerging notion that reducing GAB1 function may sensitize HNSCC to EGFR inhibitors, hence representing a new therapeutic target HNSCC treatment in combination with EGFR targeting agents.
Collapse
Affiliation(s)
- A Hoeben
- General Medical Oncology, University Hospital Gasthuisberg, Leuven, Belgium
| | | | | | | | | |
Collapse
|
49
|
Sommariva M, de Cesare M, Meini A, Cataldo A, Zaffaroni N, Tagliabue E, Balsari A. High efficacy of CpG-ODN, cetuximab and cisplatin combination for very advanced ovarian xenograft tumors. J Transl Med 2013; 11:25. [PMID: 23360557 PMCID: PMC3571944 DOI: 10.1186/1479-5876-11-25] [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: 11/28/2012] [Accepted: 01/23/2013] [Indexed: 12/22/2022] Open
Abstract
Background To mimic clinical treatment situations in advanced human ovarian disease, we tested the efficacy of CpG-oligodeoxynucleotides (CpG-ODN), synthetic DNA sequences recognized by Toll-like receptor 9 and able to induce innate/adaptive immune responses, in combination with other possible therapeutic reagents in ovarian carcinoma ascites-bearing athymic mice. Methods Mice injected i.p. with IGROV-1 ovarian cancer cells were treated at different stages of ascites progression for 4 weeks with CpG-ODN, alone or in combination with Bevacizumab, Polyinosinic:Polycytidylic acid (Poly(I):Poly(C)), Gefitinib, Cetuximab and Cisplatin. Median survival time (MST) was calculated for each group. IGROV-1 cells treated or not with Cetuximab were assayed for antibody-dependent cellular cytotoxicity by 51Cr-release assay, and for macrophage antibody-dependent cell-mediated phagocytosis by flow cytometry. Results In mice treated when ascitic fluid began to accumulate, CpG-ODN combined with Bevacizumab, Poly(I):Poly(C) or Gefitinib did not significantly increase MST as compared with that using CpG-ODN alone, whereas MST in mice treated with CpG-ODN plus Cetuximab was significantly increased (>103 days for combination vs 62 days for CpG alone; P = 0.0008), with 4/8 mice alive at the end of the experiment. In experiments in mice showing increased abdominal volume and body weight (27.9 ± 0.8 g after vs 23 ± 1.1 g before tumor cell injection), treatment with Cisplatin in addition to CpG-ODN/Cetuximab led to significantly increased MST (105.5 days; P = 0.001), with all mice still alive at 85 days, over that using CpG-ODN/Cetuximab (66 days), Cetuximab/Cisplatin (18.5 days), Cisplatin (23 days) or saline (16 days). At a very advanced stage of disease (body weight: 31.4 ± 0.9 g), when more than half of control mice had to be sacrificed 6 days after starting treatments, the triple-combination therapy still increased MST (45 days; P = 0.0089) vs controls. Conclusions CpG-ODN combination therapies that enhance the immune response in the tumor microenvironment and concomitantly target tumor cells are highly efficacious even in experimental advanced malignancies. Although differences in the distribution of TLR9 in mice and humans and the enrichment of this receptor on innate immune cells of athymic mice must be considered, our results indicate a promising strategy to treat ovarian cancer patients with bulky ascites.
Collapse
Affiliation(s)
- Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
50
|
Denaro N, Russi EG, Colantonio I, Adamo V, Merlano MC. The role of antiangiogenic agents in the treatment of head and neck cancer. Oncology 2012; 83:108-16. [PMID: 22777379 DOI: 10.1159/000339542] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 05/02/2012] [Indexed: 01/25/2023]
Abstract
Despite progress in the treatment of locally advanced head and neck squamous cell cancer (HNSCC), the prognosis remains dismal and 5-year survival does not exceed 40%. In metastatic and recurrent disease, in spite of the introduction of cetuximab in combination with platinum and fluorouracil, the median overall survival rate remains lower than 11 months. There are many possible reasons for these disappointing results including acquired drug resistance and tumor hypoxia. Angiogenesis plays an important role in HNSCC development and proliferation. Promising preclinical results with antiangiogenic therapies have engendered a number of clinical trials, but so far there have not been any conclusive results on the value of such treatments. This paper aims to review the role of angiogenesis in head and neck cancer and to suggest future perspectives.
Collapse
Affiliation(s)
- N Denaro
- Department of Oncology, Santa Croce e Carle General Hospital, Cuneo, Italy.
| | | | | | | | | |
Collapse
|