51
|
Yamaoka T, Ohba M, Ohmori T. Molecular-Targeted Therapies for Epidermal Growth Factor Receptor and Its Resistance Mechanisms. Int J Mol Sci 2017; 18:ijms18112420. [PMID: 29140271 PMCID: PMC5713388 DOI: 10.3390/ijms18112420] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/11/2017] [Accepted: 11/12/2017] [Indexed: 12/20/2022] Open
Abstract
Cancer therapies targeting epidermal growth factor receptor (EGFR), such as small-molecule kinase inhibitors and monoclonal antibodies, have been developed as standard therapies for several cancers, such as non-small cell lung cancer, colorectal cancer, pancreatic cancer, breast cancer, and squamous cell carcinoma of the head and neck. Although these therapies can significantly prolong progression-free survival, curative effects are not often achieved because of intrinsic and/or acquired resistance. The resistance mechanisms to EGFR-targeted therapies can be categorized as resistant gene mutations, activation of alternative pathways, phenotypic transformation, and resistance to apoptotic cell death. Analysis of the processes that modulate EGFR signal transduction by EGFR-targeted inhibitors, such as tyrosine kinase inhibitors and monoclonal antibodies, has revealed new therapeutic opportunities and has elucidated novel mechanisms contributing to the discovery of more effective anticancer treatments. In this review, we discuss the roles of EGFR in cancer development, therapeutic strategies for targeting EGFR, and resistance mechanisms to EGFR-targeted therapies, with a focus on cancer therapies for individual patients.
Collapse
Affiliation(s)
- Toshimitsu Yamaoka
- Institute of Molecular Oncology, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | - Motoi Ohba
- Institute of Molecular Oncology, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | - Tohru Ohmori
- Institute of Molecular Oncology, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| |
Collapse
|
52
|
Mao F, Wang B, Xiao Q, Cheng F, Lei T, Guo D. LRIG proteins in glioma: Functional roles, molecular mechanisms, and potential clinical implications. J Neurol Sci 2017; 383:56-60. [PMID: 29246624 DOI: 10.1016/j.jns.2017.10.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/26/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022]
Abstract
Gliomas are the most common intracranial tumors of the nervous system. These tumors are characterized by unlimited cell proliferation and excessive invasiveness. Despite the advances in diagnostic imaging, microneurosurgical techniques, radiation therapy, and chemotherapy, significant increases in the progression free survival of glioma patients have not been achieved. Improvements in our understanding of the molecular subtypes of gliomas and the underlying alterations in specific signaling pathways may impact both the diagnosis and the treatment strategies for patients with gliomas. Growth factors and their corresponding receptor tyrosine kinases are associated with oncogenesis and development of tumors in numerous human cancer types, including glioma. Leucine-rich repeats and immunoglobulin-like domains (LRIG) are integral membrane proteins which contain three vertebrate members including LRIG1, LRIG2 and LRIG3. They mainly function as regulators of growth factor signaling. Specifically, LRIG1 has been identified as a tumor suppressor in human cancers. In contrast, LRIG2 appears to function as a tumor promoter, while LRIG3 appears to have a function similar to that of LRIG1. In the present review, we summarize the functional roles, molecular mechanisms, and clinical perspectives of LRIG proteins in gliomas and propose that these proteins may be useful in the future as targets for treatment and prognostication in glioma patients.
Collapse
Affiliation(s)
- Feng Mao
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baofeng Wang
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qungen Xiao
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangling Cheng
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Lei
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongsheng Guo
- Department of Neurosurgery and Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
53
|
Jahan N, Lee JM, Shah K, Wakimoto H. Therapeutic targeting of chemoresistant and recurrent glioblastoma stem cells with a proapoptotic variant of oncolytic herpes simplex virus. Int J Cancer 2017; 141:1671-1681. [PMID: 28567859 PMCID: PMC5796532 DOI: 10.1002/ijc.30811] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 02/21/2017] [Accepted: 05/15/2017] [Indexed: 01/14/2023]
Abstract
Temozolomide (TMZ) chemotherapy, in combination with maximal safe resection and radiotherapy, is the current standard of care for patients with glioblastoma (GBM). Despite this multimodal approach, GBM inevitably relapses primarily due to resistance to chemo-radiotherapy, and effective treatment is not available for recurrent disease. In this study we identified TMZ resistant patient-derived primary and previously treated recurrent GBM stem cells (GSC), and investigated the therapeutic activity of a pro-apoptotic variant of oHSV (oHSV-TRAIL) in vitro and in vivo. We show that oHSV-TRAIL modulates cell survival and MAP Kinase proliferation signaling pathways as well as DNA damage response pathways in both primary and recurrent TMZ-resistant GSC. Utilizing real time in vivo imaging and correlative immunohistochemistry, we show that oHSV-TRAIL potently inhibits tumor growth and extends survival of mice bearing TMZ-insensitive recurrent intracerebral GSC tumors via robust and selective induction of apoptosis-mediated death in tumor cells, resulting in cures in 40% of the treated mice. In comparison, the anti-tumor effects in a primary chemoresistant GSC GBM model exhibiting a highly invasive phenotype were significant but less prominent. This work thus demonstrates the ability of oHSV-TRAIL to overcome the therapeutic resistance and recurrence of GBM, and provides a basis for its testing in a GBM clinical trial.
Collapse
Affiliation(s)
- Nusrat Jahan
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Jae M. Lee
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Khalid Shah
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Hiroaki Wakimoto
- Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
54
|
Cytoplasmic p53 couples oncogene-driven glucose metabolism to apoptosis and is a therapeutic target in glioblastoma. Nat Med 2017; 23:1342-1351. [PMID: 29035366 PMCID: PMC5683421 DOI: 10.1038/nm.4418] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 09/08/2017] [Indexed: 02/06/2023]
Abstract
Cross-talk among oncogenic signaling and metabolic pathways may create
opportunities for novel therapeutic strategies in cancer. Here we show that
acute inhibition of EGFR-driven glucose metabolism induces minimal cell death,
yet lowers the apoptotic threshold in a subset of patient-derived glioblastoma
(GBM) cells. Mechanistic studies revealed that, following attenuated glucose
consumption, Bcl-xL blocks cytoplasmic p53 from triggering intrinsic apoptosis.
Consequently, pharmacological stabilization of p53 with the brain-penetrant
small molecule, Idasanutlin, in combination with targeting EGFR-driven glucose
metabolism promoted synthetic lethality in orthotopic xenograft models. Notably,
neither inhibition of EGFR signaling, nor genetic analysis of
EGFR, was sufficient to predict sensitivity to this new
therapeutic combination. Conversely, rapid changes in
18F-fluorodeoxyglucose (18F-FDG) uptake using non-invasive
positron emission tomography was an effective predictive biomarker of response
in vivo. Together, these studies identify a critical link between oncogene
signaling, glucose metabolism, and cytoplasmic p53, which could be exploited for
combination therapy in GBM and potentially, other malignancies.
Collapse
|
55
|
Li J, Zhou Y, Wang H, Gao Y, Li L, Hwang SH, Ji X, Hammock BD. COX-2/sEH dual inhibitor PTUPB suppresses glioblastoma growth by targeting epidermal growth factor receptor and hyaluronan mediated motility receptor. Oncotarget 2017; 8:87353-87363. [PMID: 29152086 PMCID: PMC5675638 DOI: 10.18632/oncotarget.20928] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 08/26/2017] [Indexed: 11/25/2022] Open
Abstract
Aims Cyclooxygenase-2 (COX-2)/soluble epoxide hydrolase (sEH) dual inhibitor, PTUPB, has been demonstrated to inhibit angiogenesis, primary tumor growth and metastasis. The aim of this study is to investigate the effects of PTUPB on glioblastoma cells and xenograft model. Results We show here that PTUPB inhibits glioblastoma cell proliferation and G1 phase cell cycle arrest in vitro, and suppresses the tumor growth and angiogenesis in vivo. The expression and activation of epidermal growth factor receptor (EGFR) and its downstream kinases, ERK1/2 and AKT, are reduced by PTUPB, indicating that the EGF/EGFR signaling pathway is a potential target. Moreover, PTUPB dramatically suppresses expression of hyaluronan mediated motility receptor (HMMR) in the glioblastoma cell lines and xenograft mouse model, suggesting that the HMMR is the other potential target. Materials and Methods Cellular immunofluorescence assays were used for cell staining of actin fibers and HMMR. CCK-8 kit was used for cell proliferation assay. Cell-cycle analysis was performed by flow cytometry. Quantitative real-time PCR assay was performed to test mRNA level. Western blot analysis was used to test protein expression. Immunohistochemical staining assay was used for xenograft tumor tissue staining of Ki-67, CD31 and HMMR. The SPSS version 17.0 software was applied for statistical analysis. Conclusions Our data demonstrate that PTUPB is a potential therapeutic agent to treat glioblastomas.
Collapse
Affiliation(s)
- Junyang Li
- Department of Neurosurgery, Jinling Hospital, Medical school of Nanjing University, Nanjing, 210002, China
| | - Yali Zhou
- Department of Neurosurgery, Jinling Hospital, Medical school of Nanjing University, Nanjing, 210002, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical school of Nanjing University, Nanjing, 210002, China
| | - Yongyue Gao
- Department of Neurosurgery, Jinling Hospital, Medical school of Nanjing University, Nanjing, 210002, China
| | - Liwen Li
- Department of Neurosurgery, Jinling Hospital, Medical school of Nanjing University, Nanjing, 210002, China
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Xiangjun Ji
- Department of Neurosurgery, Jinling Hospital, Medical school of Nanjing University, Nanjing, 210002, China
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| |
Collapse
|
56
|
Abstract
PURPOSE OF REVIEW This review summarizes the use of molecular diagnostics in glioma and its effect on the development of novel therapeutics and management decisions. RECENT FINDINGS Genomic and proteomic profiling of brain tumors has provided significant expansion of our understanding of oncogenesis, characterization, and prognostication of brain tumors. Molecular markers such as MGMT, EGFR, IDH, 1p19q, ATRX, TERT, FGFR-TACC, and BRAF are now being used to classify brain tumors as well as influence management decisions. Several of these markers are also being used as therapeutic targets. We review the use of several molecular diagnostics in gliomas and discuss their impact on drug development and clinical trial design. In the future, molecular characterization based on a specific genomic, proteomic as well as transcriptomes for bioformatics analysis will provide clinicians the ability to rationally select drugs with actionable targets for each patient.
Collapse
|
57
|
Tosoni A, Franceschi E, Poggi R, Brandes AA. Relapsed Glioblastoma: Treatment Strategies for Initial and Subsequent Recurrences. Curr Treat Options Oncol 2017; 17:49. [PMID: 27461038 DOI: 10.1007/s11864-016-0422-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OPINION STATEMENT At the time of glioblastoma (GBM) recurrence, a sharp analysis of prognostic factors, disease characteristics, response to adjuvant treatment, and clinical conditions should be performed. A prognostic assessment could allow a careful selection between patients that could be proposed to intensified approaches or palliative setting. Participation in clinical trials aims to improve outcome, and should be encouraged due to dismal prognosis of GBM patients after recurrence. Reoperation should be proposed if the tumor is amenable to a complete resection and if prognostic factors suggest that patient could benefit from a second surgery. Second-line chemotherapy should be chosen based on MGMT status, time to disease recurrence, and toxicity profile. If enrollment into a clinical trial is not possible, a nitrosourea-based regimen is the preferred choice, carefully evaluating any previous temozolomide (TMZ)-related toxicity. In MGMT-methylated patients relapsing after TMZ completion, a rechallenge could be proposed. After second progression, the clinical advantage of subsequent lines of chemotherapy still needs to be clarified. However, based on performance status, patients' preference, and disease behavior, a third-line treatment could be considered. Available treatments include nitrosoureas, bevacizumab, or carboplatin plus etoposide. However, more effective therapeutic options are needed.
Collapse
Affiliation(s)
- Alicia Tosoni
- Department of Medical Oncology, Bellaria Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Via Altura 4, Bologna, Italy
| | - Enrico Franceschi
- Department of Medical Oncology, Bellaria Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Via Altura 4, Bologna, Italy
| | - Rosalba Poggi
- Department of Medical Oncology, Bellaria Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Via Altura 4, Bologna, Italy
| | - Alba A Brandes
- Department of Medical Oncology, Bellaria Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Via Altura 4, Bologna, Italy.
| |
Collapse
|
58
|
Misek SA, Chen J, Schroeder L, Rattanasinchai C, Sample A, Sarkaria JN, Gallo KA. EGFR Signals through a DOCK180-MLK3 Axis to Drive Glioblastoma Cell Invasion. Mol Cancer Res 2017; 15:1085-1095. [PMID: 28487380 DOI: 10.1158/1541-7786.mcr-16-0318] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 03/14/2017] [Accepted: 05/05/2017] [Indexed: 11/16/2022]
Abstract
A hallmark of glioblastoma (GBM) tumors is their highly invasive behavior. Tumor dissemination into surrounding brain tissue is responsible for incomplete surgical resection, and subsequent tumor recurrence. Identification of targets that control GBM cell dissemination is critical for developing effective therapies to treat GBM. A majority of GBM tumors have dysregulated EGFR signaling, due most frequently to EGFR amplification or the presence of a constitutively active EGFRvIII mutant. Mixed lineage kinase 3 (MLK3) is a mitogen-activated protein kinase kinase kinase (MAP3K) that can activate multiple MAPK pathways. In this study, evidence is provided that MLK3 is essential for GBM cell migration and invasion, and that an MLK inhibitor blocks EGF-induced migration and invasion. MLK3 silencing or MLK inhibition blocks EGF-induced JNK activation, suggesting that MLK3-JNK signaling promotes invasion of GBM cells. Mechanistically, it is demonstrated that DOCK180, a RAC1 guanine nucleotide exchange factor (GEF) overexpressed in invasive GBM cells, activates the MLK3-JNK signaling axis in a RAC1-dependent manner. In summary, this investigation identifies an EGFR-DOCK180-RAC1-MLK3-JNK signaling axis that drives glioblastoma cell migration and dissemination.Implications: On the basis of these findings, MLK3 emerges as a potential therapeutic target for the treatment of glioblastoma. Mol Cancer Res; 15(8); 1085-95. ©2017 AACR.
Collapse
Affiliation(s)
- Sean A Misek
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Jian Chen
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Laura Schroeder
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Department of Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan
| | - Chotirat Rattanasinchai
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Department of Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan
| | - Ashley Sample
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, Michigan.
- Department of Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan
| |
Collapse
|
59
|
Zanca C, Villa GR, Benitez JA, Thorne AH, Koga T, D'Antonio M, Ikegami S, Ma J, Boyer AD, Banisadr A, Jameson NM, Parisian AD, Eliseeva OV, Barnabe GF, Liu F, Wu S, Yang H, Wykosky J, Frazer KA, Verkhusha VV, Isaguliants MG, Weiss WA, Gahman TC, Shiau AK, Chen CC, Mischel PS, Cavenee WK, Furnari FB. Glioblastoma cellular cross-talk converges on NF-κB to attenuate EGFR inhibitor sensitivity. Genes Dev 2017; 31:1212-1227. [PMID: 28724615 PMCID: PMC5558924 DOI: 10.1101/gad.300079.117] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/16/2017] [Indexed: 01/31/2023]
Abstract
Zanca et al. show that heterogeneous expression of the wild-type EGFR receptor and its constitutively active mutant form, EGFRvIII, limits sensitivity to EGFR-directed therapies through an interclonal communication mechanism mediated by IL-6 cytokine secreted from EGFRvIII-positive tumor cells. In glioblastoma (GBM), heterogeneous expression of amplified and mutated epidermal growth factor receptor (EGFR) presents a substantial challenge for the effective use of EGFR-directed therapeutics. Here we demonstrate that heterogeneous expression of the wild-type receptor and its constitutively active mutant form, EGFRvIII, limits sensitivity to these therapies through an interclonal communication mechanism mediated by interleukin-6 (IL-6) cytokine secreted from EGFRvIII-positive tumor cells. IL-6 activates a NF-κB signaling axis in a paracrine and autocrine manner, leading to bromodomain protein 4 (BRD4)-dependent expression of the prosurvival protein survivin (BIRC5) and attenuation of sensitivity to EGFR tyrosine kinase inhibitors (TKIs). NF-κB and survivin are coordinately up-regulated in GBM patient tumors, and functional inhibition of either protein or BRD4 in in vitro and in vivo models restores sensitivity to EGFR TKIs. These results provide a rationale for improving anti-EGFR therapeutic efficacy through pharmacological uncoupling of a convergence point of NF-κB-mediated survival that is leveraged by an interclonal circuitry mechanism established by intratumoral mutational heterogeneity.
Collapse
Affiliation(s)
- Ciro Zanca
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Genaro R Villa
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA.,Department of Molecular and Medical Pharmacology, School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA.,Medical Scientist Training Program, School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Jorge A Benitez
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | | | - Tomoyuki Koga
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Matteo D'Antonio
- Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| | - Shiro Ikegami
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Jianhui Ma
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Antonia D Boyer
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Afsheen Banisadr
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Nathan M Jameson
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Alison D Parisian
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Olesja V Eliseeva
- Gamaleya Research Center of Epidemiology and Microbiology, Moscow 123098, Russian Federation
| | | | - Feng Liu
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA.,National Research Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sihan Wu
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Huijun Yang
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Jill Wykosky
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Kelly A Frazer
- Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA.,Institute for Genomic Medicine, University of California at San Diego, La Jolla, California 92093, USA.,Department of Pediatrics, Rady Children's Hospital, Division of Genome Information Sciences, University of California at San Diego, La Jolla, California 92093, USA
| | - Vladislav V Verkhusha
- Department of Anatomy and Structural Biology, Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Maria G Isaguliants
- Gamaleya Research Center of Epidemiology and Microbiology, Moscow 123098, Russian Federation.,Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm 17177, Sweden.,Department of Research, Riga Stradins University, Riga LV-1007, Latvia
| | - William A Weiss
- Department of Neurology, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94159, USA.,Department of Pediatrics, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94159, USA.,Department of Neurosurgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94159, USA
| | - Timothy C Gahman
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Andrew K Shiau
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA
| | - Clark C Chen
- Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA.,Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA.,Department of Pathology, School of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA.,Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA.,Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Frank B Furnari
- Ludwig Institute for Cancer Research, La Jolla, California 92093, USA.,Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA.,Department of Pathology, School of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
60
|
Morgan E, Mason W. What are the prospects for combination therapy for glioblastoma? Expert Rev Neurother 2017; 17:947-949. [PMID: 28678557 DOI: 10.1080/14737175.2017.1351300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
61
|
Touat M, Idbaih A, Sanson M, Ligon KL. Glioblastoma targeted therapy: updated approaches from recent biological insights. Ann Oncol 2017; 28:1457-1472. [PMID: 28863449 PMCID: PMC5834086 DOI: 10.1093/annonc/mdx106] [Citation(s) in RCA: 311] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Indexed: 12/29/2022] Open
Abstract
Glioblastoma (WHO grade IV astrocytoma) is the most frequent primary brain tumor in adults, representing a highly heterogeneous group of neoplasms that are among the most aggressive and challenging cancers to treat. An improved understanding of the molecular pathways that drive malignancy in glioblastoma has led to the development of various biomarkers and the evaluation of several agents specifically targeting tumor cells and the tumor microenvironment. A number of rational approaches are being investigated, including therapies targeting tumor growth factor receptors and downstream pathways, cell cycle and epigenetic regulation, angiogenesis and antitumor immune response. Moreover, recent identification and validation of prognostic and predictive biomarkers have allowed implementation of modern trial designs based on matching molecular features of tumors to targeted therapeutics. However, while occasional targeted therapy responses have been documented in patients, to date no targeted therapy has been formally validated as effective in clinical trials. The lack of knowledge about relevant molecular drivers in vivo combined with a lack of highly bioactive and brain penetrant-targeted therapies remain significant challenges. In this article, we review the most promising biological insights that have opened the way for the development of targeted therapies in glioblastoma, and examine recent data from clinical trials evaluating targeted therapies and immunotherapies. We discuss challenges and opportunities for the development of these agents in glioblastoma.
Collapse
Affiliation(s)
- M. Touat
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris
- Gustave Roussy, Université Paris-Saclay, Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Villejuif
| | - A. Idbaih
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris
- AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - M. Sanson
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris
- AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - K. L. Ligon
- Department of Oncologic Pathology, Dana-Farber/Brigham and Women's Cancer Center, Boston, USA
| |
Collapse
|
62
|
Yue Q, Gao X, Yu Y, Li Y, Hua W, Fan K, Zhang R, Qian J, Chen L, Li C, Mao Y. An EGFRvIII targeted dual-modal gold nanoprobe for imaging-guided brain tumor surgery. NANOSCALE 2017; 9:7930-7940. [PMID: 28569328 DOI: 10.1039/c7nr01077j] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Surgery is a mainstay to treat malignant brain tumors. However, due to the infiltrative nature of these tumors, it is a great challenge for surgeons to accurately identify and excise all the tumor foci. EGFRvIII, a variant of epidermal growth factor receptor (EGFR), is found in 20% of glioblastoma cases, which is the brain tumor with the highest malignancy. In this study, we developed an EGFRvIII-targeted nanoprobe to guide glioblastoma surgery by pre-operatively defining the tumor boundary via magnetic resonance imaging (MRI) and intra-operatively guiding resection by surface-enhanced resonance Raman scattering (SERRS) imaging. In vivo MRI studies show that this nanoprobe delineates an orthotopic EGFRvIII+ U87MG glioblastoma xenograft with a higher target to background ratio than the control nanoprobe without targeting specificity. With the assistance of a handheld Raman scanner, this nanoprobe successfully guided EGFRvIII+ glioblastoma resection by tracking its characteristic SERRS signal peaks. Ex vivo Raman microscopy and histological images verified that this nanoprobe precisely demarcated the glioblastoma boundary and no residual neoplastic foci were observed in the tumor bed. This dual-modal nanoprobe not only precisely guided glioblastoma resection, but also overcame the brain shift induced false-positive signal by real-timely co-registering pre-operative and intra-operative images. This nanoprobe is promising for the improvement in diagnostic accuracy and surgical outcome of EGFRvIII+ glioblastoma.
Collapse
Affiliation(s)
- Qi Yue
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Döbber A, Phoa AF, Abbassi RH, Stringer BW, Day BW, Johns TG, Abadleh M, Peifer C, Munoz L. Development and Biological Evaluation of a Photoactivatable Small Molecule Microtubule-Targeting Agent. ACS Med Chem Lett 2017; 8:395-400. [PMID: 28435525 DOI: 10.1021/acsmedchemlett.6b00483] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/15/2017] [Indexed: 01/21/2023] Open
Abstract
Photoremovable protecting groups added to bioactive molecules provide spatial and temporal control of the biological effects. We present synthesis and characterization of the first photoactivatable small-molecule tubulin inhibitor. By blocking the pharmacophoric OH group on compound 1 with photoremovable 4,5-dimethoxy-2-nitrobenzyl moiety we developed the photocaged prodrug 2 that had no effect in biological assays. Short UV light exposure of the derivative 2 or UV-irradiation of cells treated with 2 resulted in fast and potent inhibition of tubulin polymerization, attenuation of cell viability, and apoptotic cell death, implicating release of the parent active compound. This study validates for the first time the photoactivatable prodrug concept in the field of small molecule tubulin inhibitors. The caged derivative 2 represents a novel tool in antitubulin approaches.
Collapse
Affiliation(s)
- Alexander Döbber
- School of Medical
Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße
76, 24118 Kiel, Germany
| | - Athena F. Phoa
- School of Medical
Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ramzi H. Abbassi
- School of Medical
Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Brett W. Stringer
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Bryan W. Day
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Terrance G. Johns
- Oncogenic Signalling Laboratory and Brain
Cancer Discovery Collaborative, Centre for Cancer Research, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
- Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Mohammed Abadleh
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße
76, 24118 Kiel, Germany
| | - Christian Peifer
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße
76, 24118 Kiel, Germany
| | - Lenka Munoz
- School of Medical
Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
64
|
Zhang J, Liu H, Tong H, Wang S, Yang Y, Liu G, Zhang W. Clinical Applications of Contrast-Enhanced Perfusion MRI Techniques in Gliomas: Recent Advances and Current Challenges. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:7064120. [PMID: 29097933 PMCID: PMC5612612 DOI: 10.1155/2017/7064120] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/23/2017] [Indexed: 01/12/2023]
Abstract
Gliomas possess complex and heterogeneous vasculatures with abnormal hemodynamics. Despite considerable advances in diagnostic and therapeutic techniques for improving tumor management and patient care in recent years, the prognosis of malignant gliomas remains dismal. Perfusion-weighted magnetic resonance imaging techniques that could noninvasively provide superior information on vascular functionality have attracted much attention for evaluating brain tumors. However, nonconsensus imaging protocols and postprocessing analysis among different institutions impede their integration into standard-of-care imaging in clinic. And there have been very few studies providing a comprehensive evidence-based and systematic summary. This review first outlines the status of glioma theranostics and tumor-associated vascular pathology and then presents an overview of the principles of dynamic contrast-enhanced MRI (DCE-MRI) and dynamic susceptibility contrast-MRI (DSC-MRI), with emphasis on their recent clinical applications in gliomas including tumor grading, identification of molecular characteristics, differentiation of glioma from other brain tumors, treatment response assessment, and predicting prognosis. Current challenges and future perspectives are also highlighted.
Collapse
Affiliation(s)
- Junfeng Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Heng Liu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Haipeng Tong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Sumei Wang
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Yizeng Yang
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Weiguo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing 400042, China
| |
Collapse
|
65
|
Mohan S, Bonni A, Jahani-Asl A. Targeting OSMR in glioma stem cells. Oncotarget 2017; 8:16103-16104. [PMID: 28177884 PMCID: PMC5369948 DOI: 10.18632/oncotarget.15066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 02/01/2017] [Indexed: 01/31/2023] Open
Affiliation(s)
- Sushmetha Mohan
- Department of Oncology, Integrated Program in Neuroscience, McGill University, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Azad Bonni
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Arezu Jahani-Asl
- Department of Oncology, Integrated Program in Neuroscience, McGill University, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| |
Collapse
|
66
|
Kambach DM, Halim AS, Cauer A, Sun Q, Tristan CA, Celiku O, Kesarwala AH, Shankavaram U, Batchelor E, Stommel JM. Disabled cell density sensing leads to dysregulated cholesterol synthesis in glioblastoma. Oncotarget 2017; 8:14860-14875. [PMID: 28118603 PMCID: PMC5362450 DOI: 10.18632/oncotarget.14740] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/10/2017] [Indexed: 01/09/2023] Open
Abstract
A hallmark of cellular transformation is the evasion of contact-dependent inhibition of growth. To find new therapeutic targets for glioblastoma, we looked for pathways that are inhibited by high cell density in astrocytes but not in glioma cells. Here we report that glioma cells have disabled the normal controls on cholesterol synthesis. At high cell density, astrocytes turn off cholesterol synthesis genes and have low cholesterol levels, but glioma cells keep this pathway on and maintain high cholesterol. Correspondingly, cholesterol pathway upregulation is associated with poor prognosis in glioblastoma patients. Densely-plated glioma cells increase oxygen consumption, aerobic glycolysis, and the pentose phosphate pathway to synthesize cholesterol, resulting in a decrease in reactive oxygen species, TCA cycle intermediates, and ATP. This constitutive cholesterol synthesis is controlled by the cell cycle, as it can be turned off by cyclin-dependent kinase inhibitors and it correlates with disabled cell cycle control though loss of p53 and RB. Finally, glioma cells, but not astrocytes, are sensitive to cholesterol synthesis inhibition downstream of the mevalonate pathway, suggesting that specifically targeting cholesterol synthesis might be an effective treatment for glioblastoma.
Collapse
Affiliation(s)
- Diane M. Kambach
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alan S. Halim
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - A.Gesine Cauer
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qian Sun
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carlos A. Tristan
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Orieta Celiku
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aparna H. Kesarwala
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eric Batchelor
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jayne M. Stommel
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
67
|
A monoclonal antibody targeting the dimer interface of epidermal growth factor receptor (EGFR). Immunol Lett 2016; 180:39-45. [DOI: 10.1016/j.imlet.2016.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/14/2016] [Accepted: 10/31/2016] [Indexed: 01/19/2023]
|
68
|
Abstract
INTRODUCTION Gliomas are the most common malignant primary brain tumors in adults. Despite aggressive treatment with surgery, radiation and chemotherapy, these tumors are incurable and invariably recur. Molecular characterization of these tumors in recent years has advanced our understanding of gliomagenesis and offered an array of pathways that can be specifically targeted. Areas covered: The most commonly dysregulated signaling pathways found in gliomas will be discussed, as well as the biologic importance of these disrupted pathways and how each may contribute to tumor development. Our knowledge regarding these pathways are most relevant to Grade IV glioma/glioblastoma, but we will also discuss genomic categorization of low grade glioma. Further, drugs targeting single pathways, which have undergone early phase clinical trials will be reviewed, followed by an in depth discussion of emerging treatments on the horizon, which will include inhibitors of Epidermal Growth Factor Receptor (EGFR) and receptor tyrosine kinases, Phosphoinositide-3-Kinase (PI3K), angiogenesis, cell cycle and mutant Isocitrate Dehydrogenase (IDH) mutations. Expert opinion: Results from single agent targeted therapy trials have been modest. Lack of efficacy may stem from a combination of poor blood brain barrier penetration, the genetically heterogeneous make-up of the tumors and the emergence of resistance mechanisms. These factors can be overcome by rational drug design that capitalizes on ways to target critical pathways and limits upregulation of redundant pathways.
Collapse
Affiliation(s)
- Julie J Miller
- a Neuro-Oncology Fellow, Dana-Farber Cancer Institute , Massachusetts General Hospital , Boston , USA
| | - Patrick Y Wen
- b Center for Neuro-Oncology, Dana-Farber/Brigham Cancer Center, Division of Neuro-Oncology, Department of Neurology, and Harvard Medical School , Boston , USA
| |
Collapse
|
69
|
Kaur A, Denisova OV, Qiao X, Jumppanen M, Peuhu E, Ahmed SU, Raheem O, Haapasalo H, Eriksson J, Chalmers AJ, Laakkonen P, Westermarck J. PP2A Inhibitor PME-1 Drives Kinase Inhibitor Resistance in Glioma Cells. Cancer Res 2016; 76:7001-7011. [DOI: 10.1158/0008-5472.can-16-1134] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/16/2016] [Accepted: 09/08/2016] [Indexed: 11/16/2022]
|
70
|
Jeong KH, Song YJ, Han JY, Kim KU. Relationship Between Cytogenetic Complexity and Peritumoral Edema in High-Grade Astrocytoma. Ann Lab Med 2016; 36:583-9. [PMID: 27578512 PMCID: PMC5011112 DOI: 10.3343/alm.2016.36.6.583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/09/2016] [Accepted: 07/27/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The purpose of the study is to reveal the association of cytogenetic compltyexi and peritumoral edema volume (PTEV) and its prognostic significance in high-grade astrocytoma patients by culturing patient tumor cells. METHODS Twenty-seven high-grade astrocytoma patients were divided into three groups according to karyotype complexity: normal, non-complex karyotype (NCK), and complex karyotype (CK). Endothelial growth factor receptor (EGFR) amplification was detected by FISH, and its association with chromosome 7 abnormalities was analyzed. Mean PTEV of each group was compared by ANOVA to evaluate the relationship between PTEV and cytogenetic complexity. RESULTS The PTEV of patients in normal (n=6), NCK (n=8), and CK (n=13) groups were 24.52±17.73, 34.26±35.04, and 86.31±48.7 cm³, respectively (P=0.005). Ten out of 11 patients with EGFR amplification showed abnormalities in chromosome 7. The mean PTEV of EGFR-amplified and non-amplified groups were 80.4±53.7 and 41.3±37.9 cm³, respectively (P=0.035). The average survival of patients with PTEV less than 90 cm³ was 30.52±26.11 months, while in patients with PTEVs over or equal to 90 cm³, it was 10.83±5.53 months (P=0.007). CONCLUSIONS The results show an association of complex karyotype with the PTEV of high-grade astrocytoma. EGFR amplification plays a significant role in the formation of peritumoral edema, causing PTEV to increase, which is related with survival. This implies that cytogenetic karyotype can be applied as a prognostic factor.
Collapse
Affiliation(s)
- Kyung Ho Jeong
- Department of Neurosurgery, Medical Science Research Center, College of Medicine, Dong-A University, Busan, Korea
| | - Young Jin Song
- Department of Neurosurgery, Medical Science Research Center, College of Medicine, Dong-A University, Busan, Korea.,Brain Tumor Institute, Medical Science Research Center, College of Medicine, Dong-A University, Busan, Korea
| | - Jin Yeong Han
- Department of Laboratory Medicine, College of Medicine, Dong-A University, Busan, Korea
| | - Ki Uk Kim
- Department of Neurosurgery, Medical Science Research Center, College of Medicine, Dong-A University, Busan, Korea.,Brain Tumor Institute, Medical Science Research Center, College of Medicine, Dong-A University, Busan, Korea.
| |
Collapse
|
71
|
Affiliation(s)
- Sameer A Greenall
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3800, Australia
| | - Terrance G Johns
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
72
|
Chen R, Cohen AL, Colman H. Targeted Therapeutics in Patients With High-Grade Gliomas: Past, Present, and Future. Curr Treat Options Oncol 2016; 17:42. [DOI: 10.1007/s11864-016-0418-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
73
|
Tanboon J, Williams EA, Louis DN. The Diagnostic Use of Immunohistochemical Surrogates for Signature Molecular Genetic Alterations in Gliomas. J Neuropathol Exp Neurol 2016; 75:4-18. [PMID: 26671986 DOI: 10.1093/jnen/nlv009] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A number of key mutations that affect treatment and prognosis have been identified in human gliomas. Two major ways to identify these mutations in a tumor sample are direct interrogation of the mutated DNA itself and immunohistochemistry to assess the effects of the mutated genes on proteins. Immunohistochemistry is an affordable, robust, and widely available technology that has been in place for decades. For this reason, the use of immunohistochemical approaches to assess molecular genetic changes has become an essential component of state-of-the-art practice. In contrast, even though DNA sequencing technologies are undergoing rapid development, many medical centers do not have access to such methodologies and may be thwarted by the relatively high costs of sending out such tests to reference laboratories. This review summarizes the current experience using immunohistochemistry of glioma samples to identify mutations in IDH1, TP53, ATRX, histone H3 genes, BRAF, EGFR, MGMT, CIC, and FUBP1 as well as guidelines for prudent use of DNA sequencing as a supplemental method.
Collapse
|
74
|
Touat M, Duran-Peña A, Alentorn A, Lacroix L, Massard C, Idbaih A. Emerging circulating biomarkers in glioblastoma: promises and challenges. Expert Rev Mol Diagn 2016; 15:1311-23. [PMID: 26394701 DOI: 10.1586/14737159.2015.1087315] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glioblastoma (GBM) is the most common and devastating primary malignant brain tumor in adults. The past few years have seen major progress in our understanding of the molecular basis of GBM. These advances, which have contributed to the development of novel targeted therapies, will change the paradigms in GBM therapy from disease-based to individually tailored molecular target-based treatment. No validated circulating biomarkers have yet been integrated into clinical practice for GBM. There is thus a critical need to implement minimally invasive clinical tests enabling molecular stratification and prognosis assessment, as well as the prediction and monitoring of treatment response. After examination of data from recent studies exploring several categories of tumor-associated biomarkers (circulating tumor cells, extracellular vesicles, nucleic acids and oncometabolites) identified in the blood, cerebrospinal fluid and urine, this article discusses the challenges and prospects for the development of circulating biomarkers in GBM.
Collapse
Affiliation(s)
- Mehdi Touat
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,b 2 Département d'innovations thérapeutiques précoces, Gustave Roussy, F-94805 Villejuif, France
| | - Alberto Duran-Peña
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France
| | - Agusti Alentorn
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France.,d 4 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Ludovic Lacroix
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,e 5 Département de biologie médicale et de pathologie, Gustave Roussy, F-94805 Villejuif, France.,f 6 Laboratoire de recherche translationnelle et centre de ressources biologiques, Gustave Roussy, F-94805 Villejuif, France
| | - Christophe Massard
- a 1 Inserm U981, Université Paris Sud, Gustave Roussy, F-94805 Villejuif, France.,b 2 Département d'innovations thérapeutiques précoces, Gustave Roussy, F-94805 Villejuif, France
| | - Ahmed Idbaih
- c 3 AP-HP, Hôpital Universitaire la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France.,d 4 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| |
Collapse
|
75
|
Staberg M, Michaelsen SR, Olsen LS, Nedergaard MK, Villingshøj M, Stockhausen MT, Hamerlik P, Poulsen HS. Combined EGFR- and notch inhibition display additive inhibitory effect on glioblastoma cell viability and glioblastoma-induced endothelial cell sprouting in vitro. Cancer Cell Int 2016; 16:34. [PMID: 27118928 PMCID: PMC4845362 DOI: 10.1186/s12935-016-0309-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/18/2016] [Indexed: 12/11/2022] Open
Abstract
Background For Glioblastoma (GBM) patients, a number of anti-neoplastic strategies using specifically targeting drugs have been tested; however, the effects on survival have been limited. One explanation could be treatment resistance due to redundant signaling pathways, which substantiates the need for combination therapies. In GBM, both the epidermal growth factor receptor (EGFR) and the notch signaling pathways are often deregulated and linked to cellular growth, invasion and angiogenesis. Several studies have confirmed cross-talk and co-dependence of these pathways. Therefore, this study aimed at testing a combination treatment strategy using inhibitors targeting the notch and EGFR pathways. Methods For evaluation of cell viability a standard MTT assay was used. Western blotting (WB) and Q-RT-PCR were employed in order to assess the protein- and mRNA expression levels, respectively. In order to determine angiogenic processes, we used an endothelial spheroid sprouting assay. For assessment of secreted VEGF from GBM cells we performed a VEGF-quantikine ELISA. Results GBM cells were confirmed to express EGFR and Notch and to have the capacity to induce endothelial cell sprouting. Inhibition of EGFR and Notch signaling was achieved using either Iressa (gefitinib) or the gamma-secretase inhibitor DAPT. Our data showed that DAPT combined with Iressa treatment displayed increased inhibitory effect on cell viability and abrogated expression and activation of major pro-survival pathways. Similarly, the combinational treatment significantly increased abrogation of GBM-induced endothelial cell sprouting suggesting reduced GBM angiogenesis. Conclusion This study finds that simultaneous targeting of notch and EGFR signaling leads to enhanced inhibitory effects on GBM-induced angiogenesis and cell viability, thereby stressing the importance of further evaluation of this targeting approach in a clinical setting. Electronic supplementary material The online version of this article (doi:10.1186/s12935-016-0309-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mikkel Staberg
- Department of Radiation Biology, The Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Signe Regner Michaelsen
- Department of Radiation Biology, The Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Louise Stobbe Olsen
- Department of Radiation Biology, The Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mette Kjølhede Nedergaard
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Copenhagen University Hospital and University of Copenhagen, Copenhagen, Denmark
| | - Mette Villingshøj
- Department of Radiation Biology, The Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie-Thérése Stockhausen
- Department of Radiation Biology, The Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Petra Hamerlik
- Department of Radiation Biology, The Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark.,Brain Tumor Biology Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Hans Skovgaard Poulsen
- Department of Radiation Biology, The Finsen Center, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
76
|
Venkatesan S, Lamfers MLM, Dirven CMF, Leenstra S. Genetic biomarkers of drug response for small-molecule therapeutics targeting the RTK/Ras/PI3K, p53 or Rb pathway in glioblastoma. CNS Oncol 2016; 5:77-90. [PMID: 26986934 DOI: 10.2217/cns-2015-0005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Glioblastoma is the most deadly and frequently occurring primary malignant tumor of the central nervous system. Genomic studies have shown that mutated oncogenes and tumor suppressor genes in glioblastoma mainly occur in three pathways: the RTK/Ras/PI3K signaling, the p53 and the Rb pathways. In this review, we summarize the modulatory effects of genetic aberrations in these three pathways to drugs targeting these specific pathways. We also provide an overview of the preclinical efforts made to identify genetic biomarkers of response and resistance. Knowledge of biomarkers will finally promote patient stratification in clinical trials, a prerequisite for trial design in the era of precision medicine.
Collapse
Affiliation(s)
- Subramanian Venkatesan
- Department of Neurosurgery, Brain Tumor Center of the Erasmus Medical Center, Rotterdam, The Netherlands.,UCL Cancer Institute, Paul O'Gorman Building, London, UK
| | - Martine L M Lamfers
- Department of Neurosurgery, Brain Tumor Center of the Erasmus Medical Center, Rotterdam, The Netherlands
| | - Clemens M F Dirven
- Department of Neurosurgery, Brain Tumor Center of the Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sieger Leenstra
- Department of Neurosurgery, Brain Tumor Center of the Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Neurosurgery, Elisabeth Hospital, Tilburg, The Netherlands
| |
Collapse
|
77
|
Zorzan M, Giordan E, Redaelli M, Caretta A, Mucignat-Caretta C. Molecular targets in glioblastoma. Future Oncol 2016; 11:1407-20. [PMID: 25952786 DOI: 10.2217/fon.15.22] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma is the most lethal brain tumor. The poor prognosis results from lack of defined tumor margins, critical location of the tumor mass and presence of chemo- and radio-resistant tumor stem cells. The current treatment for glioblastoma consists of neurosurgery, followed by radiotherapy and temozolomide chemotherapy. A better understanding of the role of molecular and genetic heterogeneity in glioblastoma pathogenesis allowed the design of novel targeted therapies. New targets include different key-role signaling molecules and specifically altered pathways. The new approaches include interference through small molecules or monoclonal antibodies and RNA-based strategies mediated by siRNA, antisense oligonucleotides and ribozymes. Most of these treatments are still being tested yet they stay as solid promises for a clinically relevant success.
Collapse
Affiliation(s)
- Maira Zorzan
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | | | | | | |
Collapse
|
78
|
Thorne AH, Zanca C, Furnari F. Epidermal growth factor receptor targeting and challenges in glioblastoma. Neuro Oncol 2016; 18:914-8. [PMID: 26755074 DOI: 10.1093/neuonc/nov319] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/13/2015] [Indexed: 12/23/2022] Open
Abstract
With the evolution of technology, there is now a deeper understanding of glioblastoma as an inter- and intraheterogeneous disease comprising a multitude of genetically and epigenetically different cancer cells. Greater characterization of glioblastoma at the molecular level has improved its initial pathophysiological staging and classification. With this knowledge comes the hope that more efficacious therapies to combat this highly lethal disease are on the horizon. One possibility for intervention is represented by the targeting of epidermal growth factor receptor (EGFR), which is amplified and mutated in a large subset of patients. In this review, we provide a brief overview of EGFR and its mutated form, EGFR variant III, describing the downstream cellular pathways activated by each receptor, available animal models, therapeutic strategies to inhibit the receptor, and possible intervention routes to efficiently target this receptor and prevent the emergence of resistant mechanisms which to date have hampered a successful therapeutic outcome.
Collapse
Affiliation(s)
- Amy Haseley Thorne
- Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, California (A.H.T., C.Z., F.F.); Moores Cancer Center, University of California at San Diego, La Jolla, California (F.F.); Department of Pathology, University of California at San Diego, La Jolla, California (F.F.)
| | - Ciro Zanca
- Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, California (A.H.T., C.Z., F.F.); Moores Cancer Center, University of California at San Diego, La Jolla, California (F.F.); Department of Pathology, University of California at San Diego, La Jolla, California (F.F.)
| | - Frank Furnari
- Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, California (A.H.T., C.Z., F.F.); Moores Cancer Center, University of California at San Diego, La Jolla, California (F.F.); Department of Pathology, University of California at San Diego, La Jolla, California (F.F.)
| |
Collapse
|
79
|
Gatson NTN, Weathers SPS, de Groot JF. ReACT Phase II trial: a critical evaluation of the use of rindopepimut plus bevacizumab to treat EGFRvIII-positive recurrent glioblastoma. CNS Oncol 2015; 5:11-26. [PMID: 26670466 DOI: 10.2217/cns.15.38] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma is the most deadly primary brain tumor in adults and has long represented a therapeutic challenge. Disease recurrence is inevitable, and the management of recurrent disease is complicated by spontaneous or induced tumor heterogeneity which confers resistance to therapy and increased oncogenicity. EGFR and the tumor-specific mutation EGFRvIII is commonly altered in glioblastoma making it an appealing therapeutic target. Immunotherapy is an emerging and promising therapeutic approach to glioma and the EGFRvIII vaccine, rindopepimut, is the first immunotherapeutic drug to enter Phase III clinical trials for glioblastoma. Rindopepimut activates a specific immune response against tumor cells harboring the EGFRvIII protein. This review evaluates the recently completed ReACT Phase II trial using rindopepimut plus bevacizumab in the setting of EGFRvIII-positive recurrent glioblastoma (Clinical Trials identifier: NCT01498328).
Collapse
Affiliation(s)
- Na Tosha N Gatson
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0431, Houston, TX 77054, USA
| | - Shiao-Pei S Weathers
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0431, Houston, TX 77054, USA
| | - John F de Groot
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0431, Houston, TX 77054, USA
| |
Collapse
|
80
|
Ma Y, Tang N, Thompson RC, Mobley BC, Clark SW, Sarkaria JN, Wang J. InsR/IGF1R Pathway Mediates Resistance to EGFR Inhibitors in Glioblastoma. Clin Cancer Res 2015; 22:1767-76. [PMID: 26561558 DOI: 10.1158/1078-0432.ccr-15-1677] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/23/2015] [Indexed: 01/09/2023]
Abstract
PURPOSE Aberrant activation of EGFR is a hallmark of glioblastoma. However, EGFR inhibitors exhibit at best modest efficacy in glioblastoma. This is in sharp contrast with the observations in EGFR-mutant lung cancer. We examined whether activation of functionally redundant receptor tyrosine kinases (RTKs) conferred resistance to EGFR inhibitors in glioblastoma. EXPERIMENTAL DESIGN We collected a panel of patient-derived glioblastoma xenograft (PDX) lines that maintained expression of wild-type or mutant EGFR in serial xenotransplantation and tissue cultures. Using this physiologically relevant platform, we tested the abilities of several RTK ligands to protect glioblastoma cells against an EGFR inhibitor, gefitinib. Based on the screening results, we further developed a combination therapy cotargeting EGFR and insulin receptor (InsR)/insulin-like growth factor 1 receptor (IGF1R). RESULTS Insulin and IGF1 induced significant protection against gefitinib in the majority of EGFR-dependent PDX lines with one exception that did not express InsR or IGF1R. Blockade of the InsR/IGF1R pathway synergistically improved sensitivity to gefitinib or dacomitinib. Gefitinib alone effectively attenuated EGFR activities and the downstream MEK/ERK pathway. However, repression of AKT and induction of apoptosis required concurrent inhibition of both EGFR and InsR/IGF1R. A combination of gefitinib and OSI-906, a dual InsR/IGF1R inhibitor, was more effective than either agent alone to treat subcutaneous glioblastoma xenograft tumors. CONCLUSIONS Our results suggest that activation of the InsR/IGF1R pathway confers resistance to EGFR inhibitors in EGFR-dependent glioblastoma through AKT regulation. Concurrent blockade of these two pathways holds promise to treat EGFR-dependent glioblastoma.
Collapse
Affiliation(s)
- Yufang Ma
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nan Tang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Reid C Thompson
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bret C Mobley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Steven W Clark
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jialiang Wang
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee. Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee. Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
81
|
Phoa AF, Browne S, Gurgis FMS, Åkerfeldt MC, Döbber A, Renn C, Peifer C, Stringer BW, Day BW, Wong C, Chircop M, Johns TG, Kassiou M, Munoz L. Pharmacology of novel small-molecule tubulin inhibitors in glioblastoma cells with enhanced EGFR signalling. Biochem Pharmacol 2015; 98:587-601. [PMID: 26519552 DOI: 10.1016/j.bcp.2015.10.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/20/2015] [Indexed: 01/19/2023]
Abstract
We recently reported that CMPD1, originally developed as an inhibitor of MK2 activation, primarily inhibits tubulin polymerisation and induces apoptosis in glioblastoma cells. In the present study we provide detailed pharmacological investigation of CMPD1 analogues with improved molecular properties. We determined their anti-cancer efficacy in glioblastoma cells with enhanced EGFR signalling, as deregulated EGFR often leads to chemoresistance. Eight analogues of CMPD1 with varying lipophilicity and basicity were synthesised and tested for efficacy in the cell viability assay using established glioblastoma cell lines and patient-derived primary glioblastoma cells. The mechanism of action for the most potent analogue 15 was determined using MK2 activation and tubulin polymerisation assays, together with the immunofluorescence analysis of the mitotic spindle formation. Apoptosis was analysed by Annexin V staining, immunoblotting analysis of bcl-2 proteins and PARP cleavage. The apoptotic activity of CMPD1 and analogue 15 was comparable across glioblastoma cell lines regardless of the EGFR status. Primary glioblastoma cells of the classical subtype that are characterized by enhanced EGFR activity were most sensitive to the treatment with CMPD1 and 15. In summary, we present mechanism of action for a novel small molecule tubulin inhibitor, compound 15 that inhibits tubulin polymerisation and mitotic spindle formation, induces degradation of anti-apoptotic bcl-2 proteins and leads to apoptosis of glioblastoma cells. We also demonstrate that the enhanced EGFR activity does not decrease the efficacy of tubulin inhibitors developed in this study.
Collapse
Affiliation(s)
- Athena F Phoa
- School of Medical Sciences, The University of Sydney, NSW 2006, Australia
| | - Stephen Browne
- School of Medical Sciences, The University of Sydney, NSW 2006, Australia
| | - Fadi M S Gurgis
- School of Medical Sciences, The University of Sydney, NSW 2006, Australia
| | - Mia C Åkerfeldt
- School of Medical Sciences, The University of Sydney, NSW 2006, Australia
| | - Alexander Döbber
- School of Medical Sciences, The University of Sydney, NSW 2006, Australia; Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Christian Renn
- School of Medical Sciences, The University of Sydney, NSW 2006, Australia; Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Christian Peifer
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Brett W Stringer
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Bryan W Day
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Chin Wong
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2154, Australia
| | - Megan Chircop
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, NSW 2154, Australia
| | - Terrance G Johns
- Oncogenic Signalling Laboratory and Brain Cancer Discovery Collaborative, Centre for Cancer Research, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia; Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Michael Kassiou
- School of Chemistry and Faculty of Health Sciences, The University of Sydney, NSW 2006, Australia
| | - Lenka Munoz
- School of Medical Sciences, The University of Sydney, NSW 2006, Australia.
| |
Collapse
|
82
|
Wang K, Johnson A, Ali SM, Klempner SJ, Bekaii-Saab T, Vacirca JL, Khaira D, Yelensky R, Chmielecki J, Elvin JA, Lipson D, Miller VA, Stephens PJ, Ross JS. Comprehensive Genomic Profiling of Advanced Esophageal Squamous Cell Carcinomas and Esophageal Adenocarcinomas Reveals Similarities and Differences. Oncologist 2015; 20:1132-9. [PMID: 26336083 DOI: 10.1634/theoncologist.2015-0156] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/25/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinomas (ESCCs) and esophageal adenocarcinomas (EACs) account for >95% of esophageal malignancies and represent a major global health burden. ESCC is the dominant histology globally but represents a minority of U.S. cases, with EAC accounting for the majority of U.S. CASES The patient outcomes for advanced ESCC and EAC are poor, and new therapeutic options are needed. Using a sensitive sequencing assay, we compared the genomic profiles of ESCC and EAC with attention to identification of therapeutically relevant genomic alterations. METHODS Next-generation sequencing-based comprehensive genomic profiling was performed on hybridization-captured, adaptor ligation-based libraries to a median coverage depth of >650× for all coding exons of 315 cancer-related genes plus selected introns from 28 genes frequently rearranged in cancer. Results from a single sample were evaluated for all classes of genomic alterations (GAs) including point mutations, short insertions and deletions, gene amplifications, homozygous deletions, and fusions/rearrangements. Clinically relevant genomic alterations (CRGAs) were defined as alterations linked to approved drugs and those under evaluation in mechanism-driven clinical trials. RESULTS There were no significant differences by sex for either tumor type, and the median age for all patients was 63 years. All ESCCs and EACs were at an advanced stage at the time of sequencing. All 71 ESCCs and 231 EACs featured GAs on profiling, with 522 GAs in ESCC (7.4 per sample) and 1,303 GAs in EAC (5.6 per sample). The frequency of clinically relevant GAs in ESCC was 94% (2.6 per sample) and 93% in EAC (2.7 per sample). CRGAs occurring more frequently in EAC included KRAS (23% EAC vs. 6% ESCC) and ERBB2 (23% EAC vs. 3% ESCC). ESCC samples were enriched for CRGA in PIK3CA (24% ESCC vs. 10% EAC), PTEN (11% ESCC vs. 4% EAC), and NOTCH1 (17% ESCC vs. 3% EAC). Other GAs that differed significantly between histologic tumor types included SMAD4 (14% EAC vs. 1% ESCC), RB1 (14% ESCC vs. 2% EAC), SOX2 (18% ESCC vs. 1% EAC), and NFE2L2 (24% ESCC vs. 1% EAC). CONCLUSION ESCC and EAC share similarly high frequencies of overall and clinically relevant genomic alterations; however, the profiles of genomic alterations in the two diseases differ widely, with KRAS and ERBB2 far more frequently altered in EAC compared with ESCC and with mammalian target of rapamycin (MTOR) pathway genes (PIK3CA and PTEN) and NOTCH1 more frequently altered in ESCC compared with EAC. Comprehensive genomic profiling highlights the promise of identifying clinically relevant genomic alterations in both ESCC and EAC and suggests new avenues for molecularly directed therapies in esophageal cancer.
Collapse
Affiliation(s)
- Kai Wang
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Adrienne Johnson
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Siraj M Ali
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Samuel J Klempner
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Tanios Bekaii-Saab
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Jeffrey L Vacirca
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Depinder Khaira
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Roman Yelensky
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Juliann Chmielecki
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Julia A Elvin
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Doron Lipson
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Vincent A Miller
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Philip J Stephens
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| |
Collapse
|
83
|
Krůpa P, Řehák S, Diaz-Garcia D, Filip S. NANOTECHNOLOGY - NEW TRENDS IN THE TREATMENT OF BRAIN TUMOURS. ACTA MEDICA (HRADEC KRÁLOVÉ) 2015; 57:142-50. [PMID: 25938897 DOI: 10.14712/18059694.2015.79] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
High grade gliomas are some of the deadliest human tumours. Conventional treatments such as surgery, radiotherapy and chemotherapy have only a limited effect. Nowadays, resection is the common treatment of choice and although new approaches, such as perioperative magnetic resonance imaging or fluorescent microscopy have been developed, the survival rate of diagnosed patients is still very low. The inefficacy of conventional methods has led to the development of new strategies and the significant progress of nanotechnology in recent years. These platforms can be used either as novel imaging tools or to improve anticancer drug delivery into tumours while minimizing its distribution and toxicity in healthy tissues. Amongst the new nanotechnology platforms used for delivery into the brain tissue are: polymeric nanoparticles, liposomes, dendrimers, nanoshells, carbon nanotubes, superparamagnetic nanoparticles and nucleic acid based nanoparticles (DNA, RNA interference [RNAi] and antisense oligonucleotides [ASO]). These nanoparticles have been applied in the delivery of small molecular weight drugs as well as macromolecules - proteins, peptides and genes. The unique properties of these nanoparticles, such as surface charge, particle size, composition and ability to modify their surface with tissue recognition ligands and antibodies, improve their biodistribution and pharmacokinetics. All of the above mentioned characteristics make of nanoplatforms a very suitable tool for its use in targeted, personalized medicine, where they could possibly carry large doses of therapeutic agents specifically into malignant cells while avoiding healthy cells. This review poses new possibilities in the large field of nanotechnology with special interest in the treatment of high grade brain tumours.
Collapse
Affiliation(s)
- Petr Krůpa
- Charles University in Prague, Department of Neurosurgery, Faculty of Medicine in Hradec Králové, and University Hospital Hradec Králové, Czech Republic.
| | - Svatopluk Řehák
- Charles University in Prague, Department of Neurosurgery, Faculty of Medicine in Hradec Králové, and University Hospital Hradec Králové, Czech Republic
| | - Daniel Diaz-Garcia
- Charles University in Prague, Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, and University Hospital Hradec Králové, Czech Republic
| | - Stanislav Filip
- Charles University in Prague, Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Králové, and University Hospital Hradec Králové, Czech Republic
| |
Collapse
|
84
|
Alentorn A, Duran-Peña A, Pingle SC, Piccioni DE, Idbaih A, Kesari S. Molecular profiling of gliomas: potential therapeutic implications. Expert Rev Anticancer Ther 2015; 15:955-62. [PMID: 26118895 DOI: 10.1586/14737140.2015.1062368] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gliomas are the most common primary malignant brain tumor. Over the last decade, significant advances have been made in the molecular characterization of this tumor group, identifying predictive biomarkers or molecular actionable targets, and paving the way to molecular-based targeted therapies. This personalized therapeutic approach is effective and illustrated in the present review. Among many molecular abnormalities, BRAF mutation and mTOR activation in pilocytic astrocytomas and subependymal giant cell astrocytomas are actionable targets sensitive to vemurafenib and everolimus, respectively. Chromosome arms 1p/19q co-deletion and IDH mutational status are pivotal in driving delivery of early procarbazine, lomustine and vincristine chemotherapy in anaplastic oligodendroglial tumors. Although consensus to assess MGMT promoter methylation is not reached yet, it may be useful in predicting resistance to temozolomide in elderly patients.
Collapse
Affiliation(s)
- Agusti Alentorn
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de neurologie 2-Mazarin, Paris, France
| | | | | | | | | | | |
Collapse
|
85
|
Glioblastoma antigen discovery--foundations for immunotherapy. J Neurooncol 2015; 123:347-58. [PMID: 26045361 DOI: 10.1007/s11060-015-1836-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 05/30/2015] [Indexed: 01/07/2023]
Abstract
Prognosis for patients with glioblastoma (GBM), the most common high-grade primary central nervous system (CNS) tumor, remains discouraging despite multiple discoveries and clinical advances. Immunotherapy has emerged as a promising approach to GBM therapy as the idea the human CNS is immunoprivileged is being challenged. Early clinical studies of vaccine-based approaches have been encouraging, but further investigation is required before these therapies become clinically meaningful. A key challenge in immunotherapy involves identification of target antigens that are specific and sensitive for GBM. Here we discuss tumor-associated antigens that have been targeted for GBM therapy, strategies for discovery of novel antigens, and the theory of epitope spreading as it applies to GBM immunotherapy.
Collapse
|