101
|
Hou J, Deng Q, Zhou J, Zou J, Zhang Y, Tan P, Zhang W, Cui H. CSN6 controls the proliferation and metastasis of glioblastoma by CHIP-mediated degradation of EGFR. Oncogene 2016; 36:1134-1144. [PMID: 27546621 DOI: 10.1038/onc.2016.280] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/04/2016] [Accepted: 06/27/2016] [Indexed: 12/31/2022]
Abstract
CSN6, a critical subunit of the constitutive photomorphogenesis 9 (COP9) signalosome (CSN), has received attention as a regulator of the degradation of cancer-related proteins such as p53, c-myc and c-Jun, through the ubiquitin-proteasome system, suggesting its importance in cancerogenesis. However, the biological functions and molecular mechanisms of CSN6 in glioblastoma (GBM) remain poorly understood. Here, we report that GBM tumors overexpressed CSN6 compared with normal brain tissues and that CSN6 promoted GBM cell proliferation, migration, invasion and tumorigenesis. Erlotinib, a small-molecule epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, was used to reveal that the proliferative and metastatic effects of CSN6 on GBM cells were EGFR dependent. We also found that CSN6 positively regulated EGFR stability via reduced levels of EGFR ubiquitination, thereby elevating steady expression of EGFR. In addition, this study is the first description of a novel role for the CSN6-interacting E3 ligase, CHIP (carboxyl terminus of heat-shock protein 70-interacting protein), regulating EGFR ubiquitination in cancer cells. We showed that CSN6 associated with CHIP and led to CHIP destabilization by increasing CHIP self-ubiquitination. Moreover, CSN6 decreased CHIP expression and increased EGFR expression in the tumor samples. Deregulation of this axis promoted GBM cell's proliferation and metastasis. Thus, our study provides insights into the applicability of using the CSN6-CHIP-EGFR axis as a potential therapeutic target in cancer.
Collapse
Affiliation(s)
- J Hou
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Q Deng
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - J Zhou
- Department of Neurosurgery, Rocket Force General Hospital, Chinese People's Liberation Army, Beijing, China
| | - J Zou
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Y Zhang
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - P Tan
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - W Zhang
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - H Cui
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| |
Collapse
|
102
|
ROS1 amplification mediates resistance to gefitinib in glioblastoma cells. Oncotarget 2016; 6:20388-95. [PMID: 25978031 PMCID: PMC4653012 DOI: 10.18632/oncotarget.3981] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 04/15/2015] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor in adults and remains incurable despite multimodal intensive treatment regimens. The majority of GBM tumors show a mutated or overexpressed EGFR, however, tumors treated with tyrosine kinase inhibitors (TKIs) will inevitably recur highlighting the need to identify signalling pathways involved in GBM resistance to these drugs. To this end, we treated GBM cells that overexpress EGFR with increasing concentrations of gefitinib and isolated resistant clones. These resistant clones were subject to RNAseq and the expression of several genes was found to be upregulated. These genes are mainly tyrosine kinase receptors and include ROS1, DDR1 and PDGFRA and are known to control several downstream targets of EGFR. The upregulation of ROS1 and DDR1 was confirmed at the protein level by western blot. Treatment with a potent and highly specific pyrazole ROS1 inhibitor in ROS1 overexpressing clones led to a sensitization of these cells to low concentrations of gefitinib. Combined treatment with gefitinib and ROS1 inhibitor induces massive cell death by apoptosis following a prolonged S phase cell cycle arrest. Our current study led to the discovery of alternative pathways used by GBM cells to evade cell death following treatment with gefitinib and identifies new therapeutic targets to prevent GBM cell resistance to the drug.
Collapse
|
103
|
Endovascular therapies for malignant gliomas: Challenges and the future. J Clin Neurosci 2016; 26:26-32. [DOI: 10.1016/j.jocn.2015.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 10/25/2015] [Indexed: 12/17/2022]
|
104
|
Xue M, Wei W, Su Y, Johnson D, Heath JR. Supramolecular Probes for Assessing Glutamine Uptake Enable Semi-Quantitative Metabolic Models in Single Cells. J Am Chem Soc 2016; 138:3085-93. [PMID: 26916347 PMCID: PMC4887079 DOI: 10.1021/jacs.5b12187] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We describe a supramolecular surface competition assay for quantifying glutamine uptake from single cells. Cy3-labeled cyclodextrins were immobilized on a glass surface as a supramolecular host/FRET donor, and adamantane-BHQ2 conjugates were employed as the guest/quencher. An adamantane-labeled glutamine analog was selected through screening a library of compounds and validated by cell uptake experiments. When integrated onto a single cell barcode chip with a multiplex panel of 15 other metabolites, associated metabolic enzymes, and phosphoproteins, the resultant data provided input for a steady-state model that describes energy potential in single cells and correlates that potential with receptor tyrosine kinase signaling. We utilize this integrated assay to interrogate a dose-dependent response of model brain cancer cells to EGFR inhibition. We find that low-dose (1 μM erlotinib) drugging actually increases cellular energy potential even as glucose uptake and phosphoprotein signaling is repressed. We also identify new interactions between phosphoprotein signaling and cellular energy processes that may help explain the facile resistance exhibited by certain cancer patients to EGFR inhibitors.
Collapse
Affiliation(s)
- Min Xue
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Wei Wei
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Yapeng Su
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Dazy Johnson
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - James R. Heath
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
105
|
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
|
106
|
Abstract
Glioblastoma, the most aggressive of the gliomas, has a high recurrence and mortality rate. The nature of this poor prognosis resides in the molecular heterogeneity and phenotypic features of this tumor. Despite research advances in understanding the molecular biology, it has been difficult to translate this knowledge into effective treatment. Nearly all will have tumor recurrence, yet to date very few therapies have established efficacy as salvage regimens. This challenge is further complicated by imaging confounders and to an even greater degree by the ever increasing molecular heterogeneity that is thought to be both sporadic and treatment-induced. The development of novel clinical trial designs to support the development and testing of novel treatment regimens and drug delivery strategies underscore the need for more precise techniques in imaging and better surrogate markers to help determine treatment response. This review summarizes recent approaches to treat patients with recurrent glioblastoma and considers future perspectives.
Collapse
Affiliation(s)
- Carlos Kamiya-Matsuoka
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
107
|
Kast RE. Erlotinib augmentation with dapsone for rash mitigation and increased anti-cancer effectiveness. SPRINGERPLUS 2015; 4:638. [PMID: 26543772 PMCID: PMC4628020 DOI: 10.1186/s40064-015-1441-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 10/14/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND The epidermal growth factor receptor tyrosine kinase inhibitor erlotinib has failed in many ways to be as potent in the anti-cancer role as pre-clinical studies would have suggested. This paper traces some aspects of this failure to a compensatory erlotinib-mediated increase in interleukin-8. Many other-but not all- cancer chemotherapeutic cytotoxic drugs also provoke a compensatory increase in a malignant clone's interleukin-8 synthesis. Untreated glioblastoma and other cancer cells themselves natively synthesize interleukin-8. Interleukin-8 has tumor growth promoting, mobility and metastasis formation enhancing, effects as well as pro-angiogenesis effects. FINDINGS The old sulfone antibiotic dapsone- one of the very first antibiotics in clinical use- has demonstrated several interleukin-8 system inhibiting actions. Review of these indicates dapsone has potential to augment erlotinib effectiveness. Erlotinib typically gives a rash that has recently been proven to come about via an erlotinib triggered up-regulated keratinocyte interleukin-8 synthesis. The erlotinib rash shares histological features reminiscent of typical neutrophilic dermatoses. Dapsone has an established therapeutic role in current treatment of other neutrophilic dermatoses. CONCLUSION Thus, dapsone has potential to both improve the quality of life in erlotinib treated patients by amelioration of rash as well as to short-circuit a growth-enhancing aspect of erlotinib when used in the anti-cancer role.
Collapse
Affiliation(s)
- R E Kast
- IIAIGC Study Center, 22 Church Street, Burlington, VT 05401 USA
| |
Collapse
|
108
|
Whittle JR, Lickliter JD, Gan HK, Scott AM, Simes J, Solomon BJ, MacDiarmid JA, Brahmbhatt H, Rosenthal MA. First in human nanotechnology doxorubicin delivery system to target epidermal growth factor receptors in recurrent glioblastoma. J Clin Neurosci 2015; 22:1889-94. [PMID: 26279503 DOI: 10.1016/j.jocn.2015.06.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/08/2015] [Indexed: 01/27/2023]
Abstract
There are limited treatment options for patients with recurrent glioblastoma (GBM). The EnGeneIC delivery vehicle (EDV) is a novel nanocellular (minicell) compound which packages theoretically effective concentrations of chemotherapeutic drugs that are designed to target tumors via minicell-surface attached bispecific proteins (EnGeneIC, Lane Cove West, NSW, Australia). Epidermal growth factor receptor (EGFR) is overexpressed in 40-50% of patients with GBM and is a promising target for new therapeutics. (V)EDVDox contains doxorubicin (Dox) within the minicells and targets EGFR through Vectibix (V; Amgen Biologicals, Thousand Oaks, CA, USA). We conducted a first in human Phase I study of (V)EDVDox in adults with recurrent GBM expressing EGFR on immunohistochemistry, following standard therapy including radiation and temozolomide, to establish a safe maximum tolerated dose and determine a recommended Phase II dose (RPTD). (V)EDVDox was administered weekly in an 8week cycle, with dose escalation in successive cohorts of patients using a standard 3+3 design. In total, 14 patients were treated at three dose levels, and the RPTD was identified as 5×10(9)(V)EDVDox. Overall (V)EDVDox was well tolerated, with no dose limiting toxicity and no withdrawals from the study due to adverse events. The most common adverse events were nausea, fever, and chills or rigors, experienced in seven, five and five patients, respectively. Transient uncomplicated hypophosphatemia was seen in seven patients and was not dose-related. Our results demonstrate that (V)EDVDox, up to a dose of 5×10(9)(V)EDVDox weekly, is well tolerated in patients with recurrent GBM.
Collapse
Affiliation(s)
- James R Whittle
- Department of Medical Oncology, Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia.
| | | | - Hui K Gan
- Olivia Newton John Cancer and Wellness Centre, Austin Hospital, Heidelberg, VIC, Australia; Ludwig Institute for Cancer Research, Austin Hospital, Heidelberg, VIC, Australia
| | - Andrew M Scott
- Olivia Newton John Cancer and Wellness Centre, Austin Hospital, Heidelberg, VIC, Australia; Ludwig Institute for Cancer Research, Austin Hospital, Heidelberg, VIC, Australia
| | - John Simes
- Royal Prince Alfred Hospital, Camperdown, Sydney, NSW, Australia
| | | | | | | | - Mark A Rosenthal
- Department of Medical Oncology, Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia
| |
Collapse
|
109
|
Flanigan PM, Aghi MK. Adaptation to antiangiogenic therapy in neurological tumors. Cell Mol Life Sci 2015; 72:3069-82. [PMID: 25943307 PMCID: PMC4506875 DOI: 10.1007/s00018-015-1916-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 01/15/2023]
Abstract
Because tumors require a vascular supply for their survival and growth, angiogenesis is considered an important therapeutic target in most human cancers including cancer of the central nervous system. Antiangiogenic therapy has focused on inhibitors of the vascular endothelial growth factor (VEGF) signaling pathway. VEGF pathway-targeted drugs have shown therapeutic efficacy in several CNS tumors and have been tried most frequently in glioblastoma. These therapies, however, have been less effective than anticipated as some patients do not respond to therapy and some receive only modest benefit. Underlying this suboptimal response are multiple mechanisms of drug resistance involving changes in both tumor cells and their microenvironment. In this review, we discuss the multiple proposed mechanisms by which neurological tumors evolve to become resistant to antiangiogenic therapies. A better understanding of these mechanisms, their context, and their interplay will likely facilitate improvements in pharmacological strategies for the targeted treatment of neurological tumors.
Collapse
Affiliation(s)
| | - Manish K. Aghi
- Department of Neurological Surgery, California Center for Pituitary Disorders, University of California, San Francisco, USA
| |
Collapse
|
110
|
Abstract
Standard treatment for glioblastoma multiforme is surgery followed by radiotherapy and chemotherapy, generally with temozolomide. However, disease recurs in almost all patients. Diagnosis of progression is complex given the possibility of pseudoprogression. The Response Assessment in Neuro-Oncology criteria increase the sensitivity for detecting progression. Most patients will not be candidates for new surgery or re-irradiation, and anticancer drugs are the most common approach for second-line treatment, if the patient's condition allows. Antiangiogenics, inhibitors of the epidermal growth factor receptor, nitrosoureas, and re-treatment with temozolomide have been studied in the second line, but a standard therapy has not yet been established. This review considers currently available medical treatment options for patients with glioblastoma recurrence.
Collapse
Affiliation(s)
- O. Gallego
- Medical Oncology, Hospital Santa Creu i Sant Pau, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
111
|
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor and is notorious for its poor prognosis. The highly invasive nature of GBM and its inherent resistance to therapy lead to very high rates of recurrence. Recently, a small cohort of tumor cells, called cancer stem cells (CSCs), has been recognized as a subset of tumor cells with self-renewal ability and multilineage capacity. These properties, along with the remarkable tumorigenicity of CSCs, are thought to account for the high rates of tumor recurrence after treatment. Recent research has been geared toward understanding the unique biological characteristics of CSCs to enable development of targeted therapy. Strategies include inhibition of CSC-specific pathways and receptors; agents that increase sensitivity of CSCs to chemotherapy and radiotherapy; CSC differentiation agents; and CSC-specific immunotherapy, virotherapy, and gene therapy. These approaches could inform the development of newer therapeutics for GBM.
Collapse
|
112
|
Lau D, Magill ST, Aghi MK. Molecularly targeted therapies for recurrent glioblastoma: current and future targets. Neurosurg Focus 2015; 37:E15. [PMID: 25434384 DOI: 10.3171/2014.9.focus14519] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECT Glioblastoma is the most aggressive and diffusely infiltrative primary brain tumor. Recurrence is expected and is extremely difficult to treat. Over the past decade, the accumulation of knowledge regarding the molecular and genetic profile of glioblastoma has led to numerous molecularly targeted therapies. This article aims to review the literature and highlight the mechanisms and efficacies of molecularly targeted therapies for recurrent glioblastoma. METHODS A systematic search was performed with the phrase "(name of particular agent) and glioblastoma" as a search term in PubMed to identify all articles published up until 2014 that included this phrase in the title and/or abstract. The references of systematic reviews were also reviewed for additional sources. The review included clinical studies that comprised at least 20 patients and reported results for the treatment of recurrent glioblastoma with molecular targeted therapies. RESULTS A total of 42 articles were included in this review. In the treatment of recurrent glioblastoma, various targeted therapies have been tested over the past 10-15 years. The targets of interest include epidermal growth factor receptor, vascular endothelial growth factor receptor, platelet-derived growth factor receptor, Ras pathway, protein kinase C, mammalian target of rapamycin, histone acetylation, and integrins. Unfortunately, the clinical responses to most available targeted therapies are modest at best. Radiographic responses generally range in the realm of 5%-20%. Progression-free survival at 6 months and overall survival were also modest with the majority of studies reporting a 10%-20% 6-month progression-free survival and 5- to 8-month overall survival. There have been several clinical trials evaluating the use of combination therapy for molecularly targeted treatments. In general, the outcomes for combination therapy tend to be superior to single-agent therapy, regardless of the specific agent studied. CONCLUSIONS Recurrent glioblastoma remains very difficult to treat, even with molecular targeted therapies and anticancer agents. The currently available targeted therapy regimens have poor to modest activity against recurrent glioblastoma. As newer agents are actively being developed, combination regimens have provided the most promising results for improving outcomes. Targeted therapies matched to molecular profiles of individual tumors are predicted to be a critical component necessary for improving efficacy in future trials.
Collapse
Affiliation(s)
- Darryl Lau
- Department of Neurological Surgery, University of California, San Francisco, California
| | | | | |
Collapse
|
113
|
Gilbert MR, Armstrong TS, Pope WB, van den Bent MJ, Wen PY. Facing the future of brain tumor clinical research. Clin Cancer Res 2015; 20:5591-600. [PMID: 25398842 DOI: 10.1158/1078-0432.ccr-14-0835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This edition of CCR Focus provides critical reviews of several important areas in the field, including the application of findings from genomic investigations of brain tumors to improve diagnosis, clinical trial design, and ultimately optimizing individual patient treatment. Another article is a critical review provided by experts in the field that discusses the recent clinical trials using angiogenesis inhibitors, possible explanations for the results, and how to move forward. There is a concise discussion of the application of immunotherapy to brain tumors by key investigators in this field, reflecting the potential opportunities as well as the disease-specific challenges. Finally, leading pediatric brain tumor investigators provide an overview of the field and insights about the recent seminal discoveries in two pediatric brain tumors, supporting the paradigm that laboratory investigations lead to more precise diagnosis, prognosis, and ultimately better treatment. Herein, an overview of the recent advances and challenges in the area of clinical and translational brain tumor research is provided to set the stage for the contributions that follow.
Collapse
Affiliation(s)
- Mark R Gilbert
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Terri S Armstrong
- University of Texas Health Science Center School of Nursing and Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Whitney B Pope
- Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | | |
Collapse
|
114
|
Compound 331 selectively induces glioma cell death by upregulating miR-494 and downregulating CDC20. Sci Rep 2015; 5:12003. [PMID: 26153143 PMCID: PMC4495416 DOI: 10.1038/srep12003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 06/12/2015] [Indexed: 12/27/2022] Open
Abstract
Malignant gliomas are the most common malignant tumors in the central nervous system (CNS). Up to date, the prognosis of glioma is still very poor, effective therapy with less side-effect is very necessary. Herein, we identify a compound named as "331" selectively induced cell death in glioma cells but not in astrocytes. Compound 331 upregulated miR-494 and downregulated CDC20 in glioma cells but not in astrocytes. These results suggest that compound 331 could be a potential drug selectively targeting glioma cells through upregulating miR-494 and downregulating CDC20.
Collapse
|
115
|
Reardon DA, Wen PY, Mellinghoff IK. Targeted molecular therapies against epidermal growth factor receptor: past experiences and challenges. Neuro Oncol 2015; 16 Suppl 8:viii7-13. [PMID: 25342602 DOI: 10.1093/neuonc/nou232] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) has emerged as a highly attractive therapeutic target in glioblastoma (GBM) based on its high frequency of gene amplification and mutation and its identification as an upstream trigger of dysregulated cell signaling cascades that drive GBM pathophysiology. Extensive investment has been committed in an attempt to exploit EGFR therapeutically to improve outcome for GBM patients, including the development of a variety of EGFR-targeting therapeutics as well as the participation of hundreds of participants in multiple, carefully constructed clinical trials. In this review, we summarize the design and results of clinical trials evaluating EGFR tyrosine kinase inhibitors in recurrent and newly diagnosed GBM patients. While overall results thus far have been disappointing, it is premature to discount EGFR as a therapeutic target in GBM on the basis of these studies given the limitations in study design and the pharmacology of first-generation EGFR kinase inhibitors. Although important lessons have been learned, critical questions remain unanswered and warrant further study.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ingo K Mellinghoff
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
116
|
Hasan S, Chen E, Lanciano R, Yang J, Hanlon A, Lamond J, Arrigo S, Ding W, Mikhail M, Ghaneie A, Brady L. Salvage Fractionated Stereotactic Radiotherapy with or without Chemotherapy and Immunotherapy for Recurrent Glioblastoma Multiforme: A Single Institution Experience. Front Oncol 2015; 5:106. [PMID: 26029663 PMCID: PMC4432688 DOI: 10.3389/fonc.2015.00106] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/21/2015] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The current standard of care for salvage treatment of glioblastoma multiforme (GBM) is gross total resection and adjuvant chemoradiation for operable patients. Limited evidence exists to suggest that any particular treatment modality improves survival for recurrent GBM, especially if inoperable. We report our experience with fractionated stereotactic radiotherapy (fSRT) with and without chemo/immunotherapy, identifying prognostic factors associated with prolonged survival. METHODS From 2007 to 2014, 19 patients between 29 and 78 years old (median 55) with recurrent GBM following resection and chemoradiation for their initial tumor, received 18-35 Gy (median 25) in three to five fractions via CyberKnife fSRT. Clinical target volume (CTV) ranged from 0.9 to 152 cc. Sixteen patients received adjuvant systemic therapy with bevacizumab (BEV), temozolomide (TMZ), anti-epidermal growth factor receptor (125)I-mAb 425, or some combination thereof. RESULTS The median overall survival (OS) from date of recurrence was 8 months (2.5-61) and 5.3 months (0.6-58) from the end of fSRT. The OS at 6 and 12 months was 47 and 32%, respectively. Three of 19 patients were alive at the time of this review at 20, 49, and 58 months from completion of fSRT. Hazard ratios for survival indicated that patients with a frontal lobe tumor, adjuvant treatment with either BEV or TMZ, time to first recurrence >16 months, CTV <36 cc, recursive partitioning analysis <5, and Eastern Cooperative Oncology Group performance status <2 were all associated with improved survival (P < 0.05). There was no evidence of radionecrosis for any patient. CONCLUSION Radiation Therapy Oncology Group (RTOG) 1205 will establish the role of re-irradiation for recurrent GBM, however our study suggests that CyberKnife with chemotherapy can be safely delivered, and is most effective in patients with smaller frontal lobe tumors, good performance status, or long interval from diagnosis.
Collapse
Affiliation(s)
- Shaakir Hasan
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA
| | - Eda Chen
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA
| | - Rachelle Lanciano
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - Jun Yang
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - Alex Hanlon
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Nursing, University of Pennsylvania , Philadelphia, PA , USA
| | - John Lamond
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - Stephen Arrigo
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - William Ding
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| | - Michael Mikhail
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA
| | - Arezoo Ghaneie
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA
| | - Luther Brady
- Philadelphia CyberKnife/Crozer Keystone Healthcare System , Philadelphia, PA , USA ; School of Medicine, Drexel University , Philadelphia, PA , USA
| |
Collapse
|
117
|
Zahonero C, Aguilera P, Ramírez-Castillejo C, Pajares M, Bolós MV, Cantero D, Perez-Nuñez A, Hernández-Laín A, Sánchez-Gómez P, Sepúlveda JM. Preclinical Test of Dacomitinib, an Irreversible EGFR Inhibitor, Confirms Its Effectiveness for Glioblastoma. Mol Cancer Ther 2015; 14:1548-58. [PMID: 25939761 DOI: 10.1158/1535-7163.mct-14-0736] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 04/27/2015] [Indexed: 11/16/2022]
Abstract
Glioblastomas (GBM) are devastating tumors in which there has been little clinical improvement in the last decades. New molecularly directed therapies are under development. EGFR is one of the most promising targets, as this receptor is mutated and/or overexpressed in nearly half of the GBMs. However, the results obtained with first-generation tyrosine-kinase inhibitors have been disappointing with no clear predictive markers of tumor response. Here, we have tested the antitumoral efficacy of a second-generation inhibitor, dacomitinib (PF299804, Pfizer), that binds in an irreversible way to the receptor. Our results confirm that dacomitinib has an effect on cell viability, self-renewal, and proliferation in EGFR-amplified ± EGFRvIII GBM cells. Moreover, systemic administration of dacomitinib strongly impaired the in vivo tumor growth rate of these EGFR-amplified cell lines, with a decrease in the expression of stem cell-related markers. However, continuous administration of the compound was required to maintain the antitumor effect. The data presented here confirm that dacomitinib clearly affects receptor signaling in vivo and that its strong antitumoral effect is independent of the presence of mutant receptor isoforms although it could be affected by the PTEN status (as it is less effective in a PTEN-deleted GBM line). Dacomitinib is being tested in second line for EGFR-amplified GBMs. We hope that our results could help to select retrospectively molecular determinants of this response and to implement future trials with dacomitinib (alone or in combination with other inhibitors) in newly diagnosed GBMs.
Collapse
Affiliation(s)
- Cristina Zahonero
- Neuro-Oncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
| | - Pilar Aguilera
- Neuro-Oncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
| | | | - Marta Pajares
- Neuro-Oncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
| | | | - Diana Cantero
- Unidad Multidisciplinar de Neurooncología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Angel Perez-Nuñez
- Unidad Multidisciplinar de Neurooncología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Aurelio Hernández-Laín
- Unidad Multidisciplinar de Neurooncología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Juan Manuel Sepúlveda
- Unidad Multidisciplinar de Neurooncología, Hospital Universitario 12 de Octubre, Madrid, Spain.
| |
Collapse
|
118
|
Ilkhanizadeh S, Lau J, Huang M, Foster DJ, Wong R, Frantz A, Wang S, Weiss WA, Persson AI. Glial progenitors as targets for transformation in glioma. Adv Cancer Res 2015; 121:1-65. [PMID: 24889528 DOI: 10.1016/b978-0-12-800249-0.00001-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioma is the most common primary malignant brain tumor and arises throughout the central nervous system. Recent focus on stem-like glioma cells has implicated neural stem cells (NSCs), a minor precursor population restricted to germinal zones, as a potential source of gliomas. In this review, we focus on the relationship between oligodendrocyte progenitor cells (OPCs), the largest population of cycling glial progenitors in the postnatal brain, and gliomagenesis. OPCs can give rise to gliomas, with signaling pathways associated with NSCs also playing key roles during OPC lineage development. Gliomas can also undergo a switch from progenitor- to stem-like phenotype after therapy, consistent with an OPC-origin even for stem-like gliomas. Future in-depth studies of OPC biology may shed light on the etiology of OPC-derived gliomas and reveal new therapeutic avenues.
Collapse
Affiliation(s)
- Shirin Ilkhanizadeh
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Jasmine Lau
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Miller Huang
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Daniel J Foster
- Department of Neurology, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Sandler Neurosciences Center, University of California, San Francisco, California, USA
| | - Robyn Wong
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Aaron Frantz
- Department of Neurology, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Sandler Neurosciences Center, University of California, San Francisco, California, USA
| | - Susan Wang
- Department of Neurology, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Sandler Neurosciences Center, University of California, San Francisco, California, USA
| | - William A Weiss
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Department of Neurology, University of California, San Francisco, California, USA
| | - Anders I Persson
- Department of Neurology, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Sandler Neurosciences Center, University of California, San Francisco, California, USA.
| |
Collapse
|
119
|
Grant R, Kolb L, Moliterno J. Molecular and genetic pathways in gliomas: the future of personalized therapeutics. CNS Oncol 2015; 3:123-36. [PMID: 25055018 DOI: 10.2217/cns.14.7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, we have seen significant advances in brain imaging, which have resulted in more detailed anatomic and functional localization of gliomas in relation to the eloquent cortex, as well as improvements in microsurgical techniques and enhanced delivery of adjuvant stereotactic radiation. While these advancements have led to a relatively modest improvement in clinical outcomes for patients with malignant gliomas, much more work remains to be done. As with other types of cancer, we are now rapidly moving past the era of histopathology dictating treatment for brain tumors and into the realm of molecular diagnostics and associated targeted therapies, specifically based on the genomic architecture of individual gliomas. In this review, we discuss the current era of molecular glioma characterization and how these profiles will allow for individualized, patient-specific targeted treatments.
Collapse
Affiliation(s)
- Ryan Grant
- Department of Neurosurgery, Yale University School of Medicine, Yale-New Haven Hospital, 333 Cedar Street, TMP4, New Haven, CT 06510, USA
| | | | | |
Collapse
|
120
|
|
121
|
AXL as a modulator of sunitinib response in glioblastoma cell lines. Exp Cell Res 2015; 332:1-10. [PMID: 25637219 DOI: 10.1016/j.yexcr.2015.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 01/15/2015] [Accepted: 01/19/2015] [Indexed: 02/01/2023]
Abstract
Receptor tyrosine kinase (RTK) targeted therapy has been explored for glioblastoma treatment. However, it is unclear which RTK inhibitors are the most effective and there are no predictive biomarkers available. We recently identified the RTK AXL as a putative target for the pan-RTK inhibitors cediranib and sunitinib, which are under clinical trials for glioblastoma patients. Here, we provide evidence that AXL activity can modulate sunitinib response in glioblastoma cell lines. We found that AXL knockdown conferred lower sensitivity to sunitinib by rescuing migratory defects and inhibiting apoptosis in cells expressing high AXL basal levels. Accordingly, overactivation of AXL by its ligand GAS6 rendered AXL positive glioblastoma cells more sensitive to sunitinib. AXL knockdown induced a cellular rewiring of several growth signaling pathways through activation of RTKs, such as EGFR, as well as intracellular pathways such as MAPK and AKT. The combination of sunitinib with a specific AKT inhibitor reverted the resistance of AXL-silenced cells to sunitinib. Together, our results suggest that sunitinib inhibits AXL and AXL activation status modulates therapy response of glioblastoma cells to sunitinib. Moreover, it indicates that combining sunitinib therapy with AKT pathway inhibitors could overcome sunitinib resistance.
Collapse
|
122
|
Individualized dosing of tyrosine kinase inhibitors: are we there yet? Drug Discov Today 2015; 20:18-36. [DOI: 10.1016/j.drudis.2014.09.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/25/2014] [Accepted: 09/12/2014] [Indexed: 12/11/2022]
|
123
|
Emerging Strategies for the Treatment of Tumor Stem Cells in Central Nervous System Malignancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 853:167-87. [DOI: 10.1007/978-3-319-16537-0_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
124
|
Incorporation of biomarkers in phase II studies of recurrent glioblastoma. Tumour Biol 2014; 36:153-62. [PMID: 25534238 DOI: 10.1007/s13277-014-2960-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/05/2014] [Indexed: 01/15/2023] Open
Abstract
The survival trends for glioblastoma (GBM) patients have remained largely static, reflecting a lack of improvement in the therapeutic options for patients. Less than 5 % of newly diagnosed GBM survives more than 5 years. Tumor relapse is nearly universal and the majority of patients do not respond to further systemic therapy. The results from phase II studies conducted with recurrent GBM patients have not translated to successful confirmatory studies and thus we have reached a significant roadblock in the development of new treatments for patients with recurrent GBM. The development of new, active, and potentially targeted drugs for the treatment of recurrent GBM represents a major unmet need. The incorporation of diagnostic/companion biomarker combinations into the phase II studies and appropriate stratification of the patients is lagging significantly behind other larger cancer groups such as breast, non-small cell lung cancer, and melanoma. We herein carried out a systematic review of the phase II clinical studies conducted in patients with recurrent GBM (2010-2013 inclusive) to assess the degree of biomarker incorporation within the clinical trial design.
Collapse
|
125
|
Atkins RJ, Ng W, Stylli SS, Hovens CM, Kaye AH. Repair mechanisms help glioblastoma resist treatment. J Clin Neurosci 2014; 22:14-20. [PMID: 25444993 DOI: 10.1016/j.jocn.2014.09.003] [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: 12/23/2013] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 12/28/2022]
Abstract
Glioblastoma multiforme (GBM) is a malignant and incurable glial brain tumour. The current best treatment for GBM includes maximal safe surgical resection followed by concomitant radiotherapy and adjuvant temozolomide. Despite this, median survival is still only 14-16 months. Mechanisms that lead to chemo- and radio-resistance underpin treatment failure. Insights into the DNA repair mechanisms that permit resistance to chemoradiotherapy in GBM may help improve patient responses to currently available therapies.
Collapse
Affiliation(s)
- Ryan J Atkins
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia.
| | - Wayne Ng
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Christopher M Hovens
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia; Australian Prostate Cancer Research Centre at Epworth, Richmond, VIC, Australia
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, Australia
| |
Collapse
|
126
|
Yoshida Y, Ozawa T, Yao TW, Shen W, Brown D, Parsa AT, Raizer JJ, Cheng SY, Stegh AH, Mazar AP, Giles FJ, Sarkaria JN, Butowski N, Nicolaides T, James CD. NT113, a pan-ERBB inhibitor with high brain penetrance, inhibits the growth of glioblastoma xenografts with EGFR amplification. Mol Cancer Ther 2014; 13:2919-29. [PMID: 25313012 DOI: 10.1158/1535-7163.mct-14-0306] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This report describes results from our analysis of the activity and biodistribution of a novel pan-ERBB inhibitor, NT113, when used in treating mice with intracranial glioblastoma (GBM) xenografts. Approaches used in this investigation include: bioluminescence imaging (BLI) for monitoring intracranial tumor growth and response to therapy; determination of survival benefit from treatment; analysis of tumor IHC reactivity for indication of treatment effect on proliferation and apoptotic response; Western blot analysis for determination of effects of treatment on ERBB and ERBB signaling mediator activation; and high-performance liquid chromatography for determination of NT113 concentration in tissue extracts from animals receiving oral administration of inhibitor. Our results show that NT113 is active against GBM xenografts in which wild-type EGFR or EGFRvIII is highly expressed. In experiments including lapatinib and/or erlotinib, NT113 treatment was associated with the most substantial improvement in survival, as well as the most substantial tumor growth inhibition, as indicated by BLI and IHC results. Western blot analysis results indicated that NT113 has inhibitory activity, both in vivo and in vitro, on ERBB family member phosphorylation, as well as on the phosphorylation of downstream signaling mediator Akt. Results from the analysis of animal tissues revealed significantly higher NT113 normal brain-to-plasma and intracranial tumor-to-plasma ratios for NT113, relative to erlotinib, indicating superior NT113 partitioning to intracranial tissue compartments. These data provide a strong rationale for the clinical investigation of NT113, a novel ERBB inhibitor, in treating patients with GBM.
Collapse
Affiliation(s)
- Yasuyuki Yoshida
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Tomoko Ozawa
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Tsun-Wen Yao
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California. Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Wang Shen
- NewGen Therapeutics, Inc., Menlo Park, California
| | - Dennis Brown
- NewGen Therapeutics, Inc., Menlo Park, California
| | - Andrew T Parsa
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jeffrey J Raizer
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Shi-Yuan Cheng
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Alexander H Stegh
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Andrew P Mazar
- Northwestern Medicine Developmental Therapeutics Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Francis J Giles
- Northwestern Medicine Developmental Therapeutics Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Theodore Nicolaides
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California. Department of Pediatrics, University of California San Francisco, San Francisco, California.
| | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois. Northwestern Medicine Developmental Therapeutics Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| |
Collapse
|
127
|
Abstract
The survival outcome of patients with malignant gliomas is still poor, despite advances in surgical techniques, radiation therapy and the development of novel chemotherapeutic agents. The heterogeneity of molecular alterations in signaling pathways involved in the pathogenesis of these tumors contributes significantly to their resistance to treatment. Several molecular targets for therapy have been discovered over the last several years. Therapeutic agents targeting these signaling pathways may provide more effective treatments and may improve survival. This review summarizes the important molecular therapeutic targets and the outcome of published clinical trials involving targeted therapeutic agents in glioma patients.
Collapse
|
128
|
Bastien JIL, McNeill KA, Fine HA. Molecular characterizations of glioblastoma, targeted therapy, and clinical results to date. Cancer 2014; 121:502-16. [PMID: 25250735 DOI: 10.1002/cncr.28968] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 06/18/2014] [Accepted: 06/26/2014] [Indexed: 12/22/2022]
Abstract
During the last decade, extensive multiplatform genome-wide analysis has yielded a wealth of knowledge regarding the genetic and molecular makeup of glioblastoma multiforme (GBM). These profiling studies support the emerging view that GBM comprises a group of highly heterogeneous tumor types, each with its own distinct molecular and genetic signatures. This heterogeneity complicates the process of defining reliable intertumor/intratumor biological states, which will ultimately be needed for classifying tumors and for designing effective customized therapies that target resultant disease pathways. The increased understanding of the molecular pathogenesis of GBM has brought the hope and expectation that such knowledge will lead to better and more rational therapies directed toward specific molecular targets. To date, however, these expectations have largely been unrealized. This review discusses some of the principal genetic and epigenetic aberrations found in GBM that appear promising for targeted therapies now and in the near future, and it offers suggestions for future directions concerning the rather disappointing results of clinical trials to date.
Collapse
Affiliation(s)
- Jayson I L Bastien
- Laura & Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | | | | |
Collapse
|
129
|
Hottinger AF, Stupp R, Homicsko K. Standards of care and novel approaches in the management of glioblastoma multiforme. CHINESE JOURNAL OF CANCER 2014; 33:32-9. [PMID: 24384238 PMCID: PMC3905088 DOI: 10.5732/cjc.013.10207] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor in adults. Standard therapeutic approaches provide modest improvement in the progression-free and overall survival, necessitating the investigation of novel therapies. We review the standard treatment options for GBM and evaluate the results obtained in clinical trials for promising novel approaches, including the inhibition of angiogenesis, targeted approaches against molecular pathways, immunotherapies, and local treatment with low voltage electric fields.
Collapse
Affiliation(s)
- Andreas F Hottinger
- Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne 1011, Switzerland.
| | | | | |
Collapse
|
130
|
Zahonero C, Sánchez-Gómez P. EGFR-dependent mechanisms in glioblastoma: towards a better therapeutic strategy. Cell Mol Life Sci 2014; 71:3465-88. [PMID: 24671641 PMCID: PMC11113227 DOI: 10.1007/s00018-014-1608-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 02/06/2014] [Accepted: 03/11/2014] [Indexed: 12/11/2022]
Abstract
Glioblastoma is a particularly resilient cancer, and while therapies may be able to reach the brain by crossing the blood-brain barrier, they then have to deal with a highly invasive tumor that is very resistant to DNA damage. It seems clear that in order to kill aggressive glioma cells more efficiently and with fewer side effects on normal tissue, there must be a shift from classical cytotoxic chemotherapy to more targeted therapies. Since the epidermal growth factor receptor (EGFR) is altered in almost 50% of glioblastomas, it currently represents one of the most promising therapeutic targets. In fact, it has been associated with several distinct steps in tumorigenesis, from tumor initiation to tumor growth and survival, and also with the regulation of cell migration and angiogenesis. However, inhibitors of the EGFR kinase have produced poor results with this type of cancer in clinical trials, with no clear explanation for the tumor resistance observed. Here we will review what we know about the expression and function of EGFR in cancer and in particular in gliomas. We will also evaluate which are the possible molecular and cellular escape mechanisms. As a result, we hope that this review will help improve the design of future EGFR-targeted therapies for glioblastomas.
Collapse
Affiliation(s)
- Cristina Zahonero
- Neuro-Oncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
| | | |
Collapse
|
131
|
Reardon DA, Nabors LB, Mason WP, Perry JR, Shapiro W, Kavan P, Mathieu D, Phuphanich S, Cseh A, Fu Y, Cong J, Wind S, Eisenstat DD. Phase I/randomized phase II study of afatinib, an irreversible ErbB family blocker, with or without protracted temozolomide in adults with recurrent glioblastoma. Neuro Oncol 2014; 17:430-9. [PMID: 25140039 DOI: 10.1093/neuonc/nou160] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/07/2014] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND This phase I/II trial evaluated the maximum tolerated dose (MTD) and pharmacokinetics of afatinib plus temozolomide as well as the efficacy and safety of afatinib as monotherapy (A) or with temozolomide (AT) vs temozolomide monotherapy (T) in patients with recurrent glioblastoma (GBM). METHODS Phase I followed a traditional 3 + 3 dose-escalation design to determine MTD. Treatment cohorts were: afatinib 20, 40, and 50 mg/day (plus temozolomide 75 mg/m(2)/day for 21 days per 28-day cycle). In phase II, participants were randomized (stratified by age and KPS) to receive A, T or AT; A was dosed at 40 mg/day and T at 75 mg/m(2) for 21 of 28 days. Primary endpoint was progression-free survival rate at 6 months (PFS-6). Participants were treated until intolerable adverse events (AEs) or disease progression. RESULTS Recommended phase II dose was 40 mg/day (A) + T based on safety data from phase I (n = 32). Most frequent AEs in phase II (n = 119) were diarrhea (71% [A], 82% [AT]) and rash (71% [A] and 69% [AT]). Afatinib and temozolomide pharmacokinetics were unaffected by coadministration. Independently assessed PFS-6 rate was 3% (A), 10% (AT), and 23% (T). Median PFS was longer in afatinib-treated participants with epidermal growth factor receptor (EFGR) vIII-positive tumors versus EGFRvIII-negative tumors. Best overall response included partial response in 1 (A), 2 (AT), and 4 (T) participants and stable disease in 14 (A), 14 (AT), and 21 (T) participants. CONCLUSIONS Afatinib has a manageable safety profile but limited single-agent activity in unselected recurrent GBM patients.
Collapse
Affiliation(s)
- David A Reardon
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - Louis B Nabors
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - Warren P Mason
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - James R Perry
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - William Shapiro
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - Petr Kavan
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - David Mathieu
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - Surasak Phuphanich
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - Agnieszka Cseh
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - Yali Fu
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - Julie Cong
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - Sven Wind
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | - David D Eisenstat
- Dana-Farber Cancer Institute, Boston, Massachusetts (D.A.R.); University of Alabama, Birmingham, Alabama (L.B.N.); Princess Margaret Hospital, Toronto, Ontario, Canada (W.P.M.); Odette Cancer Centre, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada (J.R.P.); Barrow Neurological Institute, Phoenix, Arizona (W.S.); Department of Medical Oncology, McGill University, Montréal, Quebec, Canada (P.K.); Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada (D.M.); Johnnie Cochran Brain Tumor Center, Cedars-Sinai Medical Center, Los Angeles, California (S.P., A.C.); Boehringer Ingelheim R.C.V GmbH & Co KG, 1120 Vienna, Austria (A.C.); Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut (Y.F., J.C.); Boehringer Ingelheim Pharma GmbH & Co. K.G., 88400 Biberach, Germany (S.S.W.); CancerCare Manitoba, Winnipeg, Manitoba, Canada (D.D.E.)
| | | |
Collapse
|
132
|
Xie Q, Mittal S, Berens ME. Targeting adaptive glioblastoma: an overview of proliferation and invasion. Neuro Oncol 2014; 16:1575-84. [PMID: 25082799 DOI: 10.1093/neuonc/nou147] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma is one of the most devastating cancers, in which tumor cell infiltration into surrounding normal brain tissue confounds clinical management. This review describes basic and translational research into glioma proliferation and invasion, in particular the phenotypic switch underlying a stochastic "go or grow" model of tumor cell behavior. We include recent progress in system genomics, cancer stem cell theory, and tumor-microenvironment interaction, from which novel therapeutic strategies may emerge for managing this malignant disease. We suggest that an effective therapeutic strategy should target both adaptive glioblastoma cells and the stroma-tumor interaction.
Collapse
Affiliation(s)
- Qian Xie
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan (Q.X.); Department of Neurosurgery (S.M.); Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan (S.M.); Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona (M.E.B.)
| | - Sandeep Mittal
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan (Q.X.); Department of Neurosurgery (S.M.); Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan (S.M.); Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona (M.E.B.)
| | - Michael E Berens
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan (Q.X.); Department of Neurosurgery (S.M.); Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan (S.M.); Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona (M.E.B.)
| |
Collapse
|
133
|
Microenvironmental stiffness enhances glioma cell proliferation by stimulating epidermal growth factor receptor signaling. PLoS One 2014; 9:e101771. [PMID: 25000176 PMCID: PMC4084995 DOI: 10.1371/journal.pone.0101771] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/10/2014] [Indexed: 01/13/2023] Open
Abstract
The aggressive and rapidly lethal brain tumor glioblastoma (GBM) is associated with profound tissue stiffening and genomic lesions in key members of the epidermal growth factor receptor (EGFR) pathway. Previous studies from our laboratory have shown that increasing microenvironmental stiffness in culture can strongly enhance glioma cell behaviors relevant to tumor progression, including proliferation, yet it has remained unclear whether stiffness and EGFR regulate proliferation through common or independent signaling mechanisms. Here we test the hypothesis that microenvironmental stiffness regulates cell cycle progression and proliferation in GBM tumor cells by altering EGFR-dependent signaling. We began by performing an unbiased reverse phase protein array screen, which revealed that stiffness modulates expression and phosphorylation of a broad range of signals relevant to proliferation, including members of the EGFR pathway. We subsequently found that culturing human GBM tumor cells on progressively stiffer culture substrates both dramatically increases proliferation and facilitates passage through the G1/S checkpoint of the cell cycle, consistent with an EGFR-dependent process. Western Blots showed that increasing microenvironmental stiffness enhances the expression and phosphorylation of EGFR and its downstream effector Akt. Pharmacological loss-of-function studies revealed that the stiffness-sensitivity of proliferation is strongly blunted by inhibition of EGFR, Akt, or PI3 kinase. Finally, we observed that stiffness strongly regulates EGFR clustering, with phosphorylated EGFR condensing into vinculin-positive focal adhesions on stiff substrates and dispersing as microenvironmental stiffness falls to physiological levels. Our findings collectively support a model in which tissue stiffening promotes GBM proliferation by spatially and biochemically amplifying EGFR signaling.
Collapse
|
134
|
Abstract
Glioblastoma is the most common and most aggressive primary brain tumor in adults. Optimized standard treatment only confers a modest improvement in progression and overall survival, underscoring the pressing need for the development of novel therapies. Our understanding of glioblastoma (a molecularly heterogeneous disorder) has been accelerated in the setting of large scale genomic analyses, lending insight into potential actionable targets. Antiangiogenic therapies have been used in the treatment of glioblastoma, and our understanding of the means to optimize the role of these agents is continuing to evolve. Recently, immunotherapy has garnered increasing attention as a therapeutic approach in the treatment of gliomas. Promising novel approaches are under active development in the treatment of glioblastoma.
Collapse
|
135
|
Addeo R, Zappavigna S, Parlato C, Caraglia M. Erlotinib: early clinical development in brain cancer. Expert Opin Investig Drugs 2014; 23:1027-37. [PMID: 24836441 DOI: 10.1517/13543784.2014.918950] [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] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common brain cancer in adults. It is also, unfortunately, the most aggressive type and the least responsive to therapy. Overexpression of EGFR and/or EGFRvIII is frequently found in GBM and is frequently associated with the more malignant phenotype of the disease and a poor clinical outcome. EGFR-targeted therapy represents a promising anti-GBM therapy. Two EGFR kinase inhibitors, gefitinib and erlotinib have been tested in clinical trials for malignant gliomas. However, the clinical efficacy of EGFR-targeted therapy has been only modest in GBM patients. AREAS COVERED The authors provide an evaluation of erlotinib as a potential therapy for GBM. The authors highlight experiences drawn from clinical trials and discuss the challenges, which include the insufficient penetration through the blood-brain barrier (BBB) and chemoresistance. EXPERT OPINION Malignant brain tumours have a very complex signalling network that is not only driven by EGFR. This complexity dictates tumour sensitivity to EGFR-targeted therapies. Alternative kinase signalling pathways may be involved in parallel with the inhibited target, so that a single target's inactivation is not sufficient to block downstream oncogenic signalling. The use of nanocarriers offers many opportunities, such as the release of the drug to specific cells or tissues, together with the ability to overcome different biological barriers, like the BBB.
Collapse
|
136
|
Intratumor heterogeneity and its impact on drug distribution and sensitivity. Clin Pharmacol Ther 2014; 96:224-38. [PMID: 24827540 DOI: 10.1038/clpt.2014.105] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/07/2014] [Indexed: 01/04/2023]
Abstract
We provide an overview of the available information on the distribution of chemotherapeutics in human tumors, highlighting the progress made to assess the heterogeneity of drug concentrations in relation to the complex neoplastic tissue using novel analytical methods, e.g., mass spectrometry imaging. The increase in interstitial fluid pressure due to abnormal vascularization and stiffness of tumor stroma explains the variable and heterogeneous drug concentrations. Therapeutic strategies to enhance tumor drug distribution, thus possibly increasing efficacy, are discussed.
Collapse
|
137
|
Wilson TA, Karajannis MA, Harter DH. Glioblastoma multiforme: State of the art and future therapeutics. Surg Neurol Int 2014; 5:64. [PMID: 24991467 PMCID: PMC4078454 DOI: 10.4103/2152-7806.132138] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 03/13/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common and lethal primary malignancy of the central nervous system (CNS). Despite the proven benefit of surgical resection and aggressive treatment with chemo- and radiotherapy, the prognosis remains very poor. Recent advances of our understanding of the biology and pathophysiology of GBM have allowed the development of a wide array of novel therapeutic approaches, which have been developed. These novel approaches include molecularly targeted therapies, immunotherapies, and gene therapy. METHODS We offer a brief review of the current standard of care, and a survey of novel therapeutic approaches for treatment of GBM. RESULTS Despite promising results in preclinical trials, many of these therapies have demonstrated limited therapeutic efficacy in human clinical trials. Thus, although survival of patients with GBM continues to slowly improve, treatment of GBM remains extremely challenging. CONCLUSION Continued research and development of targeted therapies, based on a detailed understanding of molecular pathogenesis can reasonably be expected to yield improved outcomes for patients with GBM.
Collapse
Affiliation(s)
- Taylor A Wilson
- Department of Neurosurgery, Division of Oncology, New York University School of Medicine, NY, USA
| | - Matthias A Karajannis
- Department of Pediatrics, Division of Oncology, New York University School of Medicine, NY, USA
| | - David H Harter
- Department of Neurosurgery, Division of Oncology, New York University School of Medicine, NY, USA
| |
Collapse
|
138
|
The role of targeted therapies in the management of progressive glioblastoma. J Neurooncol 2014; 118:557-99. [DOI: 10.1007/s11060-013-1339-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 12/28/2013] [Indexed: 12/28/2022]
|
139
|
Wen PY, Chang SM, Lamborn KR, Kuhn JG, Norden AD, Cloughesy TF, Robins HI, Lieberman FS, Gilbert MR, Mehta MP, Drappatz J, Groves MD, Santagata S, Ligon AH, Yung WKA, Wright JJ, Dancey J, Aldape KD, Prados MD, Ligon KL. Phase I/II study of erlotinib and temsirolimus for patients with recurrent malignant gliomas: North American Brain Tumor Consortium trial 04-02. Neuro Oncol 2014; 16:567-78. [PMID: 24470557 DOI: 10.1093/neuonc/not247] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Inhibition of epidermal growth factor receptor (EGFR) and the mechanistic target of rapamycin (mTOR) may have synergistic antitumor effects in high-grade glioma patients. METHODS We conducted a phase I/II study of the EGFR inhibitor erlotinib (150 mg/day) and the mTOR inhibitor temsirolimus. Patients initially received temsirolimus 50 mg weekly, and the dose adjusted based on toxicities. In the phase II component, the primary endpoint was 6-month progression-free survival (PFS6) among glioblastoma patients. RESULTS Twenty-two patients enrolled in phase I, 47 in phase II. Twelve phase I patients treated at the maximum tolerated dosage were included in the phase II cohort for analysis. The maximum tolerated dosage was 15 mg temsirolimus weekly with erlotinib 150 mg daily. Dose-limiting toxicities were rash and mucositis. Among 42 evaluable glioblastoma patients, 12 (29%) achieved stable disease, but there were no responses, and PFS6 was 13%. Among 16 anaplastic glioma patients, 1 (6%) achieved complete response, 1 (6%) partial response, and 2 (12.5%) stable disease, with PFS6 of 8%. Tumor levels of both drugs were low, and posttreatment tissue in 3 patients showed no reduction in the mTOR target phosphorylated (phospho-)S6(S235/236) but possible compensatory increase in phospho-Akt(S473). Presence of EGFR variant III, phospho-EGFR, and EGFR amplification did not correlate with survival, but patients with elevated phospho-extracellular signal-regulated kinase or reduced phosphatase and tensin homolog protein expression had decreased progression-free survival at 4 months. CONCLUSION Because of increased toxicity, the maximum tolerated dosage of temsirolimus in combination with erlotinib proved lower than expected. Insufficient tumor drug levels and redundant signaling pathways may partly explain the minimal antitumor activity noted.
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology, Dana Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (P.Y.W., A.D.N., J.D.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (S.M.C., K.R.L., M.D.P.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Division of Neuro-Oncology, Department of Neurology, University of California, Los Angeles, Los Angeles, California (T.F.C.); University of Wisconsin, Madison Wisconsin (H.I.R., M.P.M.); Neurooncology Program, Division of Hematology/Oncology, University of Pittsburgh Medical Center Cancer Pavilion, Pittsburgh, Pennsylvania (F.S.L.); Division of Neuro-Oncology, MD Anderson Cancer Center, Houston, Texas (M.R.G., M.D.G., W.K.A.Y., K.D.A.); Center for Molecular Oncologic Pathology, Dana Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (S.S., A.H.L.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.S., A.H.L., K.L.L.); Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland (J.D.*, J.J.W.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Ahluwalia MS, Patel M, Peereboom DM. Role of tyrosine kinase inhibitors in the management of high-grade gliomas. Expert Rev Anticancer Ther 2014; 11:1739-48. [DOI: 10.1586/era.11.166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
141
|
Nakada M, Kita D, Watanabe T, Hayashi Y, Hamada JI. The mechanism of chemoresistance against tyrosine kinase inhibitors in malignant glioma. Brain Tumor Pathol 2014; 31:198-207. [DOI: 10.1007/s10014-013-0174-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/27/2013] [Indexed: 11/24/2022]
|
142
|
Olar A, Aldape KD. Using the molecular classification of glioblastoma to inform personalized treatment. J Pathol 2014; 232:165-77. [PMID: 24114756 PMCID: PMC4138801 DOI: 10.1002/path.4282] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 08/23/2013] [Accepted: 09/24/2013] [Indexed: 12/19/2022]
Abstract
Glioblastoma is the most common and most aggressive diffuse glioma, associated with short survival and uniformly fatal outcome, irrespective of treatment. It is characterized by morphological, genetic and gene-expression heterogeneity. The current standard of treatment is maximal surgical resection, followed by radiation, with concurrent and adjuvant chemotherapy. Due to the heterogeneity, most tumours develop resistance to treatment and shortly recur. Following recurrence, glioblastoma is quickly fatal in the majority of cases. Recent genetic molecular advances have contributed to a better understanding of glioblastoma pathophysiology and disease stratification. In this paper we review basic glioblastoma pathophysiology, with emphasis on clinically relevant genetic molecular alterations and potential targets for further drug development.
Collapse
Affiliation(s)
- Adriana Olar
- Department of Pathology, University of Texas MD Anderson Cancer Centre, Houston, TX, USA
| | | |
Collapse
|
143
|
Gallego O, Cuatrecasas M, Benavides M, Segura PP, Berrocal A, Erill N, Colomer A, Quintana MJ, Balaña C, Gil M, Gallardo A, Murata P, Barnadas A. Efficacy of erlotinib in patients with relapsed gliobastoma multiforme who expressed EGFRVIII and PTEN determined by immunohistochemistry. J Neurooncol 2013; 116:413-9. [PMID: 24352766 PMCID: PMC3890043 DOI: 10.1007/s11060-013-1316-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 11/17/2013] [Indexed: 11/26/2022]
Abstract
Epidermal growth factor receptor gene (EGFR) alteration is a common feature in most of glioblastoma multiforme (GBM). Robust response of anti-EGFR treatments has been mostly associated with the EGFR deletion mutant variant III (EGFRvIII) and expression of PTEN. We have performed a prospective trial in order to confirm the efficacy of erlotinib treatment in patients with relapsed GBM who expressed EGFRvIII and PTEN. All patients included in the trial were required to be PTEN (+++), EGFR (+++) and EGFRvIII (+++) positives by immunohistochemistry. This new phase II trial enrolled 40 patients and was design to be stopped in case of fewer than two responses in the first 13 patients. Patient eligibility included histopathology criteria, radiological progression, more than 18 years old, Karnofsky performed status, KPS > 50, and adequate bone marrow and organ function. There was no limit to the number of prior treatments for relapses. No enzyme-inducing antiepileptic drugs were allowed. The primary endpoints were response and progression-free survival at 6 months (PFS6). Thirteen patients (6 men, 7 women) with recurrent GBM received erlotinib 150 mg/day. Median age was 53 years, median KPS was 80, and median prior treatments for relapses were 2. There was one partial response and three stable diseases (one at 18 months). PFS at 6 months was 20 %. Dose reduction for toxicity was not needed in any patient. Dermatitis was the main treatment-related toxicity, grade 1 in 8 patients and grade 2 in 5 patients. No grade 3 toxicity was observed. Median survival was 7 months (95 % IC 1.41–4.7). As conclusion, monotherapy with erlotinib in GBM relapses patients with high protein expression for PTEN (+++), EGFR (+++), and EGFRvlII (+++) showed low toxicity but minimal efficacy and the trial stopped.
Collapse
Affiliation(s)
- Oscar Gallego
- Medical Oncology Service, Santa Creu i Sant Pau Hospital, Sant Antoni Mº Claret nº 167, 08025, Barcelona, Spain,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Bruna J, Miró J, Velasco R. Epilepsy in glioblastoma patients: basic mechanisms and current problems in treatment. Expert Rev Clin Pharmacol 2013; 6:333-44. [PMID: 23656344 DOI: 10.1586/ecp.13.12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Glioblastoma-related epilepsy requires paying careful attention to a combination of factors with an integrated approach. Major interrelated issues must be considered in the seizure care of glioblastoma patients. Seizure control frequently requires the administration of antiepileptic drugs simultaneously with other treatments, including surgery, radiotherapy and chemotherapy, with complete seizure relief often being difficult to achieve. The pharmacological interactions between antiepileptic drugs and antineoplastic agents can modify the activity of both treatments, compromising their efficacy and increasing the probability of developing adverse events related to both therapies. This review summarizes the new pathophysiological pathways involved in the epileptogenesis of glioblastoma-related seizures and the interactions between antiepileptic drugs and oncological treatment, paying special attention to its impact on survival and the current evidence of the antiepileptic treatment efficacy, including the potential usefulness of new third-generation compounds.
Collapse
Affiliation(s)
- Jordi Bruna
- Unit of Neuro-Oncology, Hospital Universitari de Bellvitge-ICO Duran i Reynals, Barcelona, Spain
| | | | | |
Collapse
|
145
|
Tanase CP, Enciu AM, Mihai S, Neagu AI, Calenic B, Cruceru ML. Anti-cancer Therapies in High Grade Gliomas. CURR PROTEOMICS 2013; 10:246-260. [PMID: 24228024 PMCID: PMC3821381 DOI: 10.2174/1570164611310030007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/10/2013] [Accepted: 06/11/2013] [Indexed: 12/28/2022]
Abstract
High grade gliomas represent one of the most aggressive and treatment-resistant types of human cancer, with only 1–2 years median survival rate for patients with grade IV glioma. The treatment of glioblastoma is a considerable therapeutic challenge; combination therapy targeting multiple pathways is becoming a fast growing area of research. This review offers an up-to-date perspective of the literature about current molecular therapy targets in high grade glioma, that include angiogenic signals, tyrosine kinase receptors, nodal signaling proteins and cancer stem cells related approaches. Simultaneous identification of proteomic signatures could provide biomarker panels for diagnostic and personalized treatment of different subsets of glioblastoma. Personalized medicine is starting to gain importance in clinical care, already having recorded a series of successes in several types of cancer; nonetheless, in brain tumors it is still at an early stage.
Collapse
Affiliation(s)
- Cristiana Pistol Tanase
- Victor Babes National Institute of Pathology, Department of Biochemistry-Proteomics, no 99-101 Splaiul Inde-pendentei, 050096 sect 5 Bucharest, Romania
| | | | | | | | | | | |
Collapse
|
146
|
Alexander BM, Lee EQ, Reardon DA, Wen PY. Current and future directions for Phase II trials in high-grade glioma. Expert Rev Neurother 2013; 13:369-87. [PMID: 23545053 DOI: 10.1586/ern.12.158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite surgery, radiation and chemotherapy, the prognosis for high-grade glioma (HGG) is poor. Our understanding of the molecular pathways involved in gliomagenesis and progression has increased in recent years, leading to the development of novel agents that specifically target these pathways. Results from most single-agent trials have been modest at best, however. Despite the initial success of antiangiogenesis agents in HGG, the clinical benefit is short-lived and most patients eventually progress. Several novel agents, multi-targeted agents and combination therapies are now in clinical trials for HGG and several more strategies are being pursued.
Collapse
Affiliation(s)
- Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, 75 Francis Street, ASB1-L2, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
147
|
Cloughesy TF, Cavenee WK, Mischel PS. Glioblastoma: from molecular pathology to targeted treatment. ANNUAL REVIEW OF PATHOLOGY 2013; 9:1-25. [PMID: 23937436 DOI: 10.1146/annurev-pathol-011110-130324] [Citation(s) in RCA: 400] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glioblastoma (GBM) is one of the most lethal human cancers. Genomic analyses are defining the molecular architecture of GBM, uncovering relevant subsets of patients whose disease may require different treatments. Many pharmacological targets have been revealed, promising to transform patient care through targeted therapies. However, for most patients, clinical responses to targeted inhibitors are either not apparent or not durable. In this review, we address the challenge of developing more effective, molecularly guided approaches for the treatment of GBM patients. We summarize the current state of knowledge regarding molecular classifiers and examine their benefit for stratifying patients for treatment. We survey the molecular landscape of the disease, discussing the challenges raised by acquired drug resistance. Furthermore, we analyze the biochemical features of GBM, suggesting a next generation of drug targets, and we examine the contribution of tumor heterogeneity and its implications. We conclude with an analysis of the experimental approaches and their potential benefit to patients.
Collapse
Affiliation(s)
- Timothy F Cloughesy
- Department of Neurology and Neuro-Oncology Program, University of California, Los Angeles, California 90095;
| | | | | |
Collapse
|
148
|
Pinel S, Mriouah J, Vandamme M, Chateau A, Plénat F, Guérin E, Taillandier L, Bernier-Chastagner V, Merlin JL, Chastagner P. Synergistic antitumor effect between gefitinib and fractionated irradiation in anaplastic oligodendrogliomas cannot be predicted by the Egfr signaling activity. PLoS One 2013; 8:e68333. [PMID: 23874590 PMCID: PMC3715478 DOI: 10.1371/journal.pone.0068333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 05/28/2013] [Indexed: 11/18/2022] Open
Abstract
In high-grade gliomas, the identification of patients that could benefit from EGFR inhibitors remains a challenge, hindering the use of these agents. Using xenografts models, we evaluated the antitumor effect of the combined treatment "gefitinib + radiotherapy" and aimed to identify the profile of responsive tumors. Expression of phosphorylated proteins involved in the EGFR-dependent signaling pathways was analyzed in 10 glioma models. We focused on three models of anaplastic oligodendrogliomas (TCG2, TCG3 and TCG4) harboring high levels of phospho-EGFR, phospho-AKT and phospho-MEK1. They were treated with gefitinib (GEF 75 mg/kg/day x 5 days/week, for 2 weeks) and/or fractionated radiotherapy (RT: 5x2Gy/week for 2 weeks). Our results showed that GEF and/or RT induced significant tumor growth delays. However, only the TCG3 xenografts were highly responsive to the combination GEF+RT, with ∼50% of tumor cure. Phosphoproteins analysis five days after treatment onset demonstrated in TCG3 xenografts, but not in TCG2 model, that the EGFR-dependent pathways were inhibited after GEF treatment. Moreover, TCG3-bearing mice receiving GEF monotherapy exhibited a transient beneficial therapeutic response, rapidly followed by tumor regrowth, along with a major vascular remodeling. Taken together, our data evoked an "EGFR-addictive" behavior for TCG3 tumors. This study confirms that combination of gefitinib with fractionated irradiation could be a potent therapeutic strategy for anaplastic oligodendrogliomas harboring EGFR abnormalities but this treatment seems mainly beneficial for "EGFR-addictive" tumors. Unfortunately, neither the usual molecular markers (EGFR amplification, PTEN loss) nor the basal overexpression of phosphoproteins were useful to distinguish this responsive tumor. Evaluating the impact of TKIs on the EGFR-dependent pathways during the treatment might be more relevant, and requires further validation.
Collapse
Affiliation(s)
- Sophie Pinel
- Université de Lorraine, CRAN, UMR 7039, Campus Science, Vandoeuvre-les-Nancy, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Solomón MT, Selva JC, Figueredo J, Vaquer J, Toledo C, Quintanal N, Salva S, Domíngez R, Alert J, Marinello JJ, Catalá M, Griego MG, Martell JA, Luaces PL, Ballesteros J, de-Castro N, Bach F, Crombet T. Radiotherapy plus nimotuzumab or placebo in the treatment of high grade glioma patients: results from a randomized, double blind trial. BMC Cancer 2013; 13:299. [PMID: 23782513 PMCID: PMC3691625 DOI: 10.1186/1471-2407-13-299] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 06/14/2013] [Indexed: 01/23/2023] Open
Abstract
Background The prognosis of patients bearing high grade glioma remains dismal. Epidermal Growth Factor Receptor (EGFR) is well validated as a primary contributor of glioma initiation and progression. Nimotuzumab is a humanized monoclonal antibody that recognizes the EGFR extracellular domain and reaches Central Nervous System tumors, in nonclinical and clinical setting. While it has similar activity when compared to other anti-EGFR antibodies, it does not induce skin toxicity or hypomagnesemia. Methods A randomized, double blind, multicentric clinical trial was conducted in high grade glioma patients (41 anaplastic astrocytoma and 29 glioblastoma multiforme) that received radiotherapy plus nimotuzumab or placebo. Treatment and placebo groups were well-balanced for the most important prognostic variables. Patients received 6 weekly doses of 200 mg nimotuzumab or placebo together with irradiation as induction therapy. Maintenance treatment was given for 1 year with subsequent doses administered every 3 weeks. The objectives of this study were to assess the comparative overall survival, progression free survival, response rate, immunogenicity and safety. Results The median cumulative dose was 3200 mg of nimotuzumab given over a median number of 16 doses. The combination of nimotuzumab and RT was well-tolerated. The most prevalent related adverse reactions included nausea, fever, tremors, anorexia and hepatic test alteration. No anti-idiotypic response was detected, confirming the antibody low immunogenicity. The mean and median survival time for subjects treated with nimotuzumab was 31.06 and 17.76 vs. 21.07 and 12.63 months for the control group. Conclusions In this randomized trial, nimotuzumab showed an excellent safety profile and significant survival benefit in combination with irradiation. Trial registration Cuban National Register for clinical trials (No. 1745) (http://registroclinico.sld.cu/ensayos).
Collapse
|
150
|
Abstract
Angiogenesis, recruitment of new blood vessels, is an essential component of the metastatic pathway. These vessels provide the principal route by which tumor cells exit the primary tumor site and enter the circulation. For many tumors, the vascular density can provide a prognostic indicator of metastatic potential, with the highly vascular primary tumors having a higher incidence of metastasis than poorly vascular tumors. The discovery and characterization of tumor-derived angiogenesis modulators greatly contributed to our understanding of how tumors regulate angiogenesis. However, although angiogenesis appears to be a rate-limiting event in tumor growth and metastatic dissemination, a direct connection between the induction of angiogenesis and the progression to tumor malignancy is less well understood. In this review, we discuss the observations concerning the modulation of angiogenesis and their implications in various neurological disorders, as well as their potential impact on cancer therapy.
Collapse
Affiliation(s)
- Göksemin Acar
- Department of Neurology, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| | | | | |
Collapse
|