101
|
Ellingson BM, Wen PY, van den Bent MJ, Cloughesy TF. Pros and cons of current brain tumor imaging. Neuro Oncol 2015; 16 Suppl 7:vii2-11. [PMID: 25313235 DOI: 10.1093/neuonc/nou224] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Over the past 20 years, very few agents have been approved for the treatment of brain tumors. Recent studies have highlighted some of the challenges in assessing activity in novel agents for the treatment of brain tumors. This paper reviews some of the key challenges related to assessment of tumor response to therapy in adult high-grade gliomas and discusses the strengths and limitations of imaging-based endpoints. Although overall survival is considered the "gold standard" endpoint in the field of oncology, progression-free survival and response rate are endpoints that hold great value in neuro-oncology. Particular focus is given to advancements made since the January 2006 Brain Tumor Endpoints Workshop, including the development of Response Assessment in Neuro-Oncology criteria, the value of T2/fluid-attenuated inversion recovery, use of objective response rates and progression-free survival in clinical trials, and the evaluation of pseudoprogression, pseudoresponse, and inflammatory response in radiographic images.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Patrick Y Wen
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Martin J van den Bent
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Timothy F Cloughesy
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| |
Collapse
|
102
|
Antiangiogenic Therapies and Extracranial Metastasis in Glioblastoma: A Case Report and Review of the Literature. Case Rep Oncol Med 2015. [PMID: 26199775 PMCID: PMC4493308 DOI: 10.1155/2015/431819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We present a case report of a patient with glioblastoma multiforme (GBM) complicated by extracranial metastasis (ECM) whose survival of nearly four years surpassed the anticipated life expectancy given numerous negative prognostic factors including EGFRvIII-mutation, unmethylated MGMT promoter status, and ECM. Interestingly, while this patient suffered from locally aggressive disease with multiple intracranial recurrences, the proximal cause of death was progressive extracranial disease and complications related to pulmonary metastases. Herein, we review potential mechanisms of ECM with an emphasis upon glioblastoma molecular and genetic profiles and the potential implications of targeted agents such as bevacizumab.
Collapse
|
103
|
Abstract
PURPOSE OF REVIEW A major recent clinical research focus for glioblastoma has been the therapeutic evaluation of antiangiogenic agents. Several vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitors and a soluble decoy VEGF receptor have demonstrated nominal benefit among patients. In contrast, bevacizumab, a humanized VEGF monoclonal antibody, exhibits evidence of apparent antitumor benefit, although these data remain controversial. In this review, we summarize how results of clinical trials evaluating bevacizumab to date influence the future of this therapeutic for recurrent and newly diagnosed glioblastoma patients. RECENT FINDINGS Recently reported, placebo-controlled phase III studies demonstrate a meaningful progression-free survival increment, but no overall survival benefit among newly diagnosed patients treated with bevacizumab. For unclear reasons, quality-of-life surveys from these studies revealed divergent results. Among recurrent patients, uncontrolled trials demonstrate improved overall radiographic response and progression-free survival rates, although the impact of bevacizumab on overall survival remains to be defined by an ongoing randomized phase III trial. SUMMARY The role of bevacizumab for glioblastoma remains uncertain but will likely be strongly influenced by results of a randomized phase III study among recurrent patients as well as further investigation of gene expression biomarker profiles to identify newly diagnosed patients more likely to derive survival benefit.
Collapse
|
104
|
Cavazos DA, Brenner AJ. Hypoxia in astrocytic tumors and implications for therapy. Neurobiol Dis 2015; 85:227-233. [PMID: 26094595 DOI: 10.1016/j.nbd.2015.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 06/01/2015] [Accepted: 06/12/2015] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma (GBM, Grade IV astrocytoma) is the most common and most aggressive of the primary malignant brain tumors in adults. Hypoxia is a distinct feature in GBM and plays a significant role in tumor progression, resistance to treatment and poor outcomes. This review considers the effects of hypoxia on astrocytic tumors and the mechanisms that contribute to tumor progression and therapeutic resistance, with a focus on the vascular changes, chemotaxic signaling pathways and metabolic alterations involved.
Collapse
Affiliation(s)
- David A Cavazos
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | - Andrew J Brenner
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA.
| |
Collapse
|
105
|
Abstract
Glioblastoma multiforme (GBM) tumor invasion is facilitated by cell migration and degradation of the extracellular matrix. Invadopodia are actin-rich structures that protrude from the plasma membrane in direct contact with the extracellular matrix and are proposed to participate in epithelial-mesenchymal transition. We characterized the invasiveness of 9 established GBM cell lines using an invadopodia assay and performed quantitative mass spectrometry-based proteomic analyses on enriched membrane fractions. All GBM cells produced invadopodia, with a 65% difference between the most invasive cell line (U87MG) and the least invasive cell line (LN229) (p = 0.0001). Overall, 1,141 proteins were identified in the GBM membrane proteome; the levels of 49 proteins correlated with cell invasiveness. Ingenuity Pathway Analysis predicted activation "cell movement" (z-score = 2.608, p = 3.94E(-04)) in more invasive cells and generated a network of invasion-associated proteins with direct links to key regulators of invadopodia formation. Gene expression data relating to the invasion-associated proteins ITGA5 (integrin α5), CD97, and ANXA1 (annexin A1) showed prognostic significance in independent GBM cohorts. Fluorescence microscopy demonstrated ITGA5, CD97, and ANXA1 localization in invadopodia assays, and small interfering RNA knockdown of ITGA5 reduced invadopodia formation in U87MG cells. Thus, invasion-associated proteins, including ITGA5, may prove to be useful anti-invasive targets; volociximab, a therapeutic antibody against integrin α5β1, may be useful for treatment of patients with GBM.
Collapse
|
106
|
Carvalho BF, Fernandes AC, Almeida DS, Sampaio LV, Costa A, Caeiro C, Osório L, Castro L, Linhares P, Damasceno M, Vaz RC. Second-Line Chemotherapy in Recurrent Glioblastoma: A 2-Cohort Study. Oncol Res Treat 2015; 38:348-54. [DOI: 10.1159/000431236] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 04/07/2015] [Indexed: 11/19/2022]
|
107
|
Prognostic implication of progression pattern after anti-VEGF bevacizumab treatment for recurrent malignant gliomas. J Neurooncol 2015; 124:101-10. [DOI: 10.1007/s11060-015-1808-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 05/09/2015] [Indexed: 12/22/2022]
|
108
|
Nagpal S, Recht CK, Bertrand S, Thomas RP, Ajlan A, Pena J, Gershon M, Coffey G, Kunz PL, Li G, Recht LD. Phase II pilot study of single-agent etirinotecan pegol (NKTR-102) in bevacizumab-resistant high grade glioma. J Neurooncol 2015; 123:277-82. [PMID: 25935109 PMCID: PMC4452613 DOI: 10.1007/s11060-015-1795-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/20/2015] [Indexed: 11/24/2022]
Abstract
Patients with recurrence of high-grade glioma (HGG) after bevacizumab (BEV) have an extremely poor prognosis. Etirinotecan pegol (EP) is the first long-acting topoisomerase-I inhibitor designed to concentrate in and provide continuous tumor exposure throughout the entire chemotherapy cycle. Here we report results of a Phase 2, single arm, open-label trial evaluating EP in HGG patients who progressed after BEV. Patients age >18 with histologically proven anaplastic astrocytoma or glioblastoma (GB) who previously received standard chemo-radiation and recurred after BEV were eligible. A predicted life expectancy >6 weeks and KPS ≥ 50 were required. The primary endpoint was PFS at 6-weeks. Secondary endpoint was overall survival from first EP infusion. Response was assessed by RANO criteria. Single agent EP was administered IV every 3 weeks at 145 mg/m2. Patients did not receive BEV while on EP. 20 patients (90 % GB) were enrolled with a median age of 50 and median KPS of 70. Three patients with GB (16.7 % of GB) had partial MRI responses. 6-week PFS was 55 %. Median and 6-month PFS were 2.2 months (95 % CI 1.4–3.4 months) and 11.2 % (95 % CI 1.9–28.9 %) respectively. Median overall survival from first EP infusion was 4.5 months (95 % CI 2.4–5.9). Only one patient had grade 3 toxicity (diarrhea with dehydration) attributable to EP. Hematologic toxicity was mild. Three patients had confirmed partial responses according to RANO criteria. These clinical data combined with a favorable safety profile warrant further clinical investigation of this agent in HGG.
Collapse
Affiliation(s)
- Seema Nagpal
- Division of Neuro-Oncology, Department of Neurology, Stanford University, 875 Blake Wilbur Drive CC2221, Stanford, CA, 94305, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Abstract
Glioblastoma (GBM) is the most common adult primary brain neoplasm. Despite advances in treatment, GBM continues to be associated with considerable morbidity and mortality as compared with other malignancies. Standard treatment for GBM results in survival of 12.9 months (95% CI: 12.3-13.7 months) with a median progression-free survival of 7.2 months (95% CI: 6.4-8.2 months) in a modern GBM cohort. These aggressive tumors recur and treatment for recurrent GBM continues to have very poor outcomes. Prior to the use of bevacizumab, monoclonal antibody to VEGF, 6-month progression-free survival in clinical trials for recurrent GBM ranged from 9 to 15%. Trials utilizing bevacizumab and its subsequent US FDA approval have given more hope to recurrent GBM and this concise review discusses bevacizumab in recurrent GBM. This review focuses on time-to-event outcomes (overall survival, progression-free survival and 6-month progression-free survival) in clinical trials utilizing bevacizumab for the treatment of recurrent GBM. For this review, we have chosen to focus primarily on Phase II clinical trials that have been published and available in the literature (PubMed). While we focused primarily on time-to-event variables, toxicity and safety of bevacizumab is very important and this agent can be associated with serious life-threatening toxicities. We have included a general section of toxicities but for a more lengthy review please see the excellent study by Odia and colleagues.
Collapse
Affiliation(s)
- Ashley Ghiaseddin
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, PO Box 3624, Durham, NC 27710, USA
| | | |
Collapse
|
110
|
Kegelman TP, Hu B, Emdad L, Das SK, Sarkar D, Fisher PB. In vivo modeling of malignant glioma: the road to effective therapy. Adv Cancer Res 2015; 121:261-330. [PMID: 24889534 DOI: 10.1016/b978-0-12-800249-0.00007-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite an increased emphasis on developing new therapies for malignant gliomas, they remain among the most intractable tumors faced today as they demonstrate a remarkable ability to evade current treatment strategies. Numerous candidate treatments fail at late stages, often after showing promising preclinical results. This disconnect highlights the continued need for improved animal models of glioma, which can be used to both screen potential targets and authentically recapitulate the human condition. This review examines recent developments in the animal modeling of glioma, from more established rat models to intriguing new systems using Drosophila and zebrafish that set the stage for higher throughput studies of potentially useful targets. It also addresses the versatility of mouse modeling using newly developed techniques recreating human protocols and sophisticated genetically engineered approaches that aim to characterize the biology of gliomagenesis. The use of these and future models will elucidate both new targets and effective combination therapies that will impact on disease management.
Collapse
Affiliation(s)
- Timothy P Kegelman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Bin Hu
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|
111
|
Abstract
Glioblastoma is the most common and malignant primary brain tumour in adults. Maximum feasible surgical resection, radiotherapy and temozolomide chemotherapy at initial diagnosis have improved prognosis but rapid recurrence is typical and survival remains brief. There is an urgent need for effective new treatments and approval of the antiangiogenic agent bevacizumab for recurrent glioblastoma by Health Canada in 2009 has been the most notable recent therapeutic advance for this disease. This review with illustrative case studies highlights how bevacizumab has been incorporated into the treatment of glioblastoma in Canada and describes the ongoing controversies surrounding its clinical application.
Collapse
|
112
|
Schmainda KM. Diffusion-weighted MRI as a biomarker for treatment response in glioma. CNS Oncol 2015; 1:169-80. [PMID: 23936625 DOI: 10.2217/cns.12.25] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Diffusion-weighted imaging (DWI) is a powerful MRI method, which probes abnormalities of tissue structure by detecting microscopic changes in water mobility at a cellular level beyond what is available with other imaging techniques. Accordingly, DWI has the potential to identify pathology before gross anatomic changes are evident on standard anatomical brain images. These features of tissue characterization and earlier detection are what make DWI particularly appealing for the evaluation of gliomas and the newer therapies where standard anatomical imaging is proving insufficient. This article focuses on the basic principles and applications of DWI, and its derived parameter, the apparent diffusion coefficient, for the purposes of diagnosis and evaluation of glioma, especially in the context of monitoring response to therapy.
Collapse
|
113
|
Schnegg CI, Yang MH, Ghosh SK, Hsu MY. Induction of Vasculogenic Mimicry Overrides VEGF-A Silencing and Enriches Stem-like Cancer Cells in Melanoma. Cancer Res 2015; 75:1682-90. [PMID: 25769726 DOI: 10.1158/0008-5472.can-14-1855] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 01/31/2015] [Indexed: 12/15/2022]
Abstract
The basis for resistance to VEGF inhibition is not fully understood despite its clinical importance. In this study, we examined the adaptive response to VEGF-A inhibition by a loss-of-function analysis using plasmid-based shRNA. Tumor xenografts that initially responded to VEGF-A inhibition underwent an adaptation in vivo, leading to acquired resistance. VEGF-A blockade in tumors was associated with HIF1α expression and an increase in CD144(+) vasculogenic mimicry (VM), leading to formation of channels displaying Tie-1 and MMP-2 upregulation. CD133(+) and CD271(+) melanoma stem-like cells (MSLC) accumulated in the perivascular niche. Tumor xenografts of melanoma cell populations that were intrinsically resistant to VEGF-A blockade did not exhibit any of these features, compared with nontarget control counterparts. Thus, melanomas that are initially sensitive to VEGF-A blockade acquire adaptive resistance by adopting VM as an alternate angiogenic strategy, thereby enriching for deposition of MSLC in the perivascular niche through an HIF1α-dependent process. Conversely, melanomas that are intrinsically resistant to VEGF-A blockade do not show any evidence of compensatory survival mechanisms that promote MSLC accumulation. Our work highlights the potential risk of anti-VEGF treatments owing to a selective pressure for an adaptive resistance mechanism that empowers the development of stem-like cancer cells, with implications for how to design combination therapies that can improve outcomes in patients.
Collapse
Affiliation(s)
- Caroline I Schnegg
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts
| | - Moon Hee Yang
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts
| | - Subrata K Ghosh
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts
| | - Mei-Yu Hsu
- Department of Dermatology, Boston University Medical Center, Boston, Massachusetts.
| |
Collapse
|
114
|
Brandes AA, Mason W, Pichler J, Nowak AK, Gil M, Saran F, Revil C, Lutiger B, Carpentier AF. Can bevacizumab prolong survival for glioblastoma patients through multiple lines of therapy? Future Oncol 2015; 10:1137-45. [PMID: 24947255 DOI: 10.2217/fon.14.75] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Glioblastoma has a poor prognosis accompanied by debilitating neurological symptoms and impaired quality of life. Effective treatment strategies are needed, beyond the current standard of care (SOC), to improve outcomes. Glioblastomas are highly vascularized with elevated levels of VEGF, representing an appropriate target for selective therapies. The role of the anti-VEGF agent bevacizumab in newly diagnosed and recurrent glioblastoma is not fully clear at this time. Although bevacizumab has demonstrated improvements in progression-free survival in newly diagnosed and recurrent glioblastoma, there remain challenges in assessing disease progression after antiangiogenic treatment. The bevacizumab mechanism of action suggests a rationale for continuing bevacizumab treatment through multiple lines of therapy, strengthened by longer progression-free and overall survival observed with bevacizumab continuation beyond progression in a Phase III study in metastatic colorectal cancer and in pooled analyses of Phase II trials in glioblastoma. A novel study (randomized, double-blind, Phase IIIb; TAMIGA [MO28347]) aims to evaluate whether continuing bevacizumab plus lomustine (as second-line therapy) and SOC (third line and beyond) improves survival compared with placebo plus lomustine and then placebo plus SOC in patients with glioblastoma who progressed after first-line bevacizumab plus SOC.
Collapse
Affiliation(s)
- Alba A Brandes
- Department of Medical Oncology, Azienda USL Bellaria-Maggiore Hospital, Via Altura 3, Bologna, 40139, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Lassman AB, Pugh SL, Gilbert MR, Aldape KD, Geinoz S, Beumer JH, Christner SM, Komaki R, DeAngelis LM, Gaur R, Youssef E, Wagner H, Won M, Mehta MP. Phase 2 trial of dasatinib in target-selected patients with recurrent glioblastoma (RTOG 0627). Neuro Oncol 2015; 17:992-8. [PMID: 25758746 DOI: 10.1093/neuonc/nov011] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/14/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND We conducted a phase II trial to evaluate the efficacy of dasatinib, a multitargeted tyrosine kinase inhibitor, for adults with recurrent glioblastoma (GBM). METHODS Eligibility requirements were Karnofsky performance status ≥ 60%; no concurrent hepatic enzyme-inducing anticonvulsants; prior treatment with surgery, radiotherapy, and temozolomide exclusively; and activation or overexpression of ≥ 2 putative dasatinib targets in GBM (ie, SRC, c-KIT, EPHA2, and PDGFR). Using a 2-stage design, 77 eligible participants (27 in stage 1, if favorable, and then 50 in stage 2) were needed to detect an absolute improvement in the proportion of patients either alive and progression-free patients at 6 months (6mPFS) or responding (any duration) from a historical 11% to 25%. RESULTS A high rate of ineligibility (27%) to stage 1 precluded a powered assessment of efficacy, but there was also infrequent treatment-related toxicity at 100 mg twice daily. Therefore, the study was redesigned to allow intrapatient escalation by 50 mg daily every cycle as tolerated (stage 1B) before determining whether to proceed to stage 2. Escalation was tolerable in 10 of 17 (59%) participants evaluable for that endpoint; however, among all eligible patients (stages 1 and 1B, n = 50), there were no radiographic responses, median overall survival was 7.9 months, median PFS was 1.7 months, and the 6mPFS rate was 6%. The clinical benefit was insufficient to correlate tested biomarkers with efficacy. The trial was closed without proceeding to stage 2. CONCLUSIONS Intraparticipant dose escalation was feasible, but dasatinib was ineffective in recurrent GBM. Clinical trials.gov identified. NCT00423735 (available at http://clinicaltrials.gov/ct2/show/NCT00423735).
Collapse
Affiliation(s)
- Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Stephanie L Pugh
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Mark R Gilbert
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Kenneth D Aldape
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Sandrine Geinoz
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Jan H Beumer
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Susan M Christner
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Ritsuko Komaki
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Lisa M DeAngelis
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Rakesh Gaur
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Emad Youssef
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Henry Wagner
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Minhee Won
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| | - Minesh P Mehta
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York (A.B.L. current); Memorial Sloan Kettering Cancer Center, New York, New York (A.B.L. during accrual, L.M.D.); NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania (S.L.P., S.G., M.W); The University of Texas MD Anderson Cancer Center, Houston, Texas (M.R.G., K.D.A. during accrual; R.K.); Neuro-Oncology Branch, National Cancer Institute/National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (M.R.G. current); University of Toronto and Princess Margaret Cancer Centre, Toronto, Canada (K.D.A. current); Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania (J.H.B., S.M.C.); Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy Pittsburgh, Pennsylvania (J.H.B.); NCI Community Oncology Research Program - Kansas City, Prairie Village, Kansas (R.G.); Arizona Oncology Services Foundation, Tucson, Arizona (E.Y.); Penn State University and The Milton S. Hershey Medical Center, Hershey, Pennsylvania (H.W.); University of Maryland Medical Systems, Baltimore, Maryland (M.P.M.)
| |
Collapse
|
116
|
Lu KV, Bergers G. Mechanisms of evasive resistance to anti-VEGF therapy in glioblastoma. CNS Oncol 2015; 2:49-65. [PMID: 23750318 DOI: 10.2217/cns.12.36] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Angiogenesis inhibitors targeting the VEGF signaling pathway have been US FDA approved for various cancers including glioblastoma (GBM), one of the most lethal and angiogenic tumors. This has led to the routine use of the anti-VEGF antibody bevacizumab in recurrent GBM, conveying substantial improvements in radiographic response, progression-free survival and quality of life. Despite these encouraging beneficial effects, patients inevitably develop resistance and frequently fail to demonstrate significantly better overall survival. Unlike chemotherapies, to which tumors exhibit resistance due to genetic mutation of drug targets, emerging evidence suggests that tumors bypass antiangiogenic therapy while VEGF signaling remains inhibited through a variety of mechanisms that are just beginning to be recognized. Because of the indirect nature of resistance to VEGF inhibitors there is promise that strategies combining angiogenesis inhibitors with drugs targeting such evasive resistance pathways will lead to more durable antiangiogenic efficacy and improved patient outcomes. Further identifying and understanding of evasive resistance mechanisms and their clinical importance in GBM relapse is therefore a timely and critical issue.
Collapse
|
117
|
Leu K, Pope WB, Cloughesy TF, Lai A, Nghiemphu PL, Chen W, Liau LM, Ellingson BM. Imaging biomarkers for antiangiogenic therapy in malignant gliomas. CNS Oncol 2015; 2:33-47. [PMID: 24570837 DOI: 10.2217/cns.12.29] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The discovery that malignant gliomas produce an excessive amount of VEGF, a key mediator of angiogenesis, has heightened interest in developing drugs that block angiogenic pathways. These antiangiogenic drugs tend to decrease vascular permeability, thereby diminishing tumor contrast enhancement independent of anti-tumor effects. This has made the determination of tumor response difficult, since contrast enhancement on post-contrast T1-weighted images is standard for assessing therapy effectiveness. In light of these unique challenges in assessing antiangiogenic therapy, new biomarkers have been proposed, based on advanced magnetic resonance techniques and PET. This article outlines the challenges associated with the evaluation of antiangiogenic therapy in malignant gliomas and describes how new imaging biomarkers can be used to better predict response.
Collapse
|
118
|
Wang W, Sivakumar W, Torres S, Jhaveri N, Vaikari VP, Gong A, Howard A, Golden EB, Louie SG, Schönthal AH, Hofman FM, Chen TC. Effects of convection-enhanced delivery of bevacizumab on survival of glioma-bearing animals. Neurosurg Focus 2015; 38:E8. [DOI: 10.3171/2015.1.focus14743] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECT
Bevacizumab (Avastin), an antibody to vascular endothelial growth factor (VEGF), alone or in combination with irinotecan (Camptosar [CPT-11]), is a promising treatment for recurrent glioblastoma. However, the intravenous (IV) administration of bevacizumab produces a number of systemic side effects, and the increase in survival it provides for patients with recurrent glioblastoma is still only a few months. Because bevacizumab is an antibody against VEGF, which is secreted into the extracellular milieu by glioma cells, the authors hypothesized that direct chronic intratumoral delivery techniques (i.e., convection-enhanced delivery [CED]) can be more effective than IV administration. To test this hypothesis, the authors compared outcomes for these routes of bevacizumab application with respect to animal survival, microvessel density (MVD), and inflammatory cell distribution.
METHODS
Two human glioma cell lines, U87 and U251, were used as sources of intracranial tumor cells. The glioma cell lines were implanted into the brains of mice in an orthotopic xenograft mouse tumor model. After 7 days, the mice were treated with one of the following: 1) vehicle, 2) CED bevacizumab, 3) IV bevacizumab, 4) intraperitoneal (IP) irinotecan, 5) CED bevacizumab plus IP irinotecan, or 6) IV bevacizumab plus IP irinotecan. Alzet micro-osmotic pumps were used to introduce bevacizumab directly into the tumor. Survival was monitored. Excised tumor tissue samples were immunostained to measure MVD and inflammatory cell and growth factor levels.
RESULTS
The results demonstrate that mice treated with CED of bevacizumab alone or in combination with irinotecan survived longer than those treated systemically; CED-treated animals survived 30% longer than IV-treated animals. In combination studies, CED bevacizumab plus CPT-11 increased survival by more than 90%, whereas IV bevacizumab plus CPT-11 increased survival by 40%. Furthermore, CED bevacizumab-treated tissues exhibited decreased MVD compared with that of IV-treated tissues. In additional studies, the infiltration of macrophages and dendritic cells into CED-treated animals were increased compared with those in IV-treated animals, suggesting a highly active inflammatory response taking place in CED-treated mice.
CONCLUSIONS
The administration of bevacizumab via CED increases survival over that of treatment with IV bevacizumab. Thus, CED of bevacizumab alone or in combination with chemotherapy can be an effective protocol for treating gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Stan G. Louie
- 4Department of Clinical Pharmacy and Pharmaceutical Economics and Policy, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Axel H. Schönthal
- 3Molecular Microbiology and Immunology, Keck School of Medicine; and
| | | | | |
Collapse
|
119
|
Rhun EL, Taillibert S, Chamberlain MC. The future of high-grade glioma: Where we are and where are we going. Surg Neurol Int 2015; 6:S9-S44. [PMID: 25722939 PMCID: PMC4338495 DOI: 10.4103/2152-7806.151331] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/15/2014] [Indexed: 01/12/2023] Open
Abstract
High-grade glioma (HGG) are optimally treated with maximum safe surgery, followed by radiotherapy (RT) and/or systemic chemotherapy (CT). Recently, the treatment of newly diagnosed anaplastic glioma (AG) has changed, particularly in patients with 1p19q codeleted tumors. Results of trials currenlty ongoing are likely to determine the best standard of care for patients with noncodeleted AG tumors. Trials in AG illustrate the importance of molecular characterization, which are germane to both prognosis and treatment. In contrast, efforts to improve the current standard of care of newly diagnosed glioblastoma (GB) with, for example, the addition of bevacizumab (BEV), have been largely disappointing and furthermore molecular characterization has not changed therapy except in elderly patients. Novel approaches, such as vaccine-based immunotherapy, for newly diagnosed GB are currently being pursued in multiple clinical trials. Recurrent disease, an event inevitable in nearly all patients with HGG, continues to be a challenge. Both recurrent GB and AG are managed in similar manner and when feasible re-resection is often suggested notwithstanding limited data to suggest benefit from repeat surgery. Occassional patients may be candidates for re-irradiation but again there is a paucity of data to commend this therapy and only a minority of selected patients are eligible for this approach. Consequently systemic therapy continues to be the most often utilized treatment in recurrent HGG. Choice of therapy, however, varies and revolves around re-challenge with temozolomide (TMZ), use of a nitrosourea (most often lomustine; CCNU) or BEV, the most frequently used angiogenic inhibitor. Nevertheless, no clear standard recommendation regarding the prefered agent or combination of agents is avaliable. Prognosis after progression of a HGG remains poor, with an unmet need to improve therapy.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Department of Neuro-oncology, Roger Salengro Hospital, University Hospital, Lille, and Neurology, Department of Medical Oncology, Oscar Lambret Center, Lille, France, Inserm U-1192, Laboratoire de Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM), Lille 1 University, Villeneuve D’Ascq, France
| | - Sophie Taillibert
- Neurology, Mazarin and Radiation Oncology, Pitié Salpétrière Hospital, University Pierre et Marie Curie, Paris VI, Paris, France
| | - Marc C. Chamberlain
- Department of Neurology and Neurological Surgery, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
120
|
Ellingson BM, Kim E, Woodworth DC, Marques H, Boxerman JL, Safriel Y, McKinstry RC, Bokstein F, Jain R, Chi TL, Sorensen AG, Gilbert MR, Barboriak DP. Diffusion MRI quality control and functional diffusion map results in ACRIN 6677/RTOG 0625: a multicenter, randomized, phase II trial of bevacizumab and chemotherapy in recurrent glioblastoma. Int J Oncol 2015; 46:1883-92. [PMID: 25672376 PMCID: PMC4383029 DOI: 10.3892/ijo.2015.2891] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 12/30/2014] [Indexed: 12/27/2022] Open
Abstract
Functional diffusion mapping (fDM) is a cancer imaging technique that quantifies voxelwise changes in apparent diffusion coefficient (ADC). Previous studies have shown value of fDMs in bevacizumab therapy for recurrent glioblastoma multiforme (GBM). The aim of the present study was to implement explicit criteria for diffusion MRI quality control and independently evaluate fDM performance in a multicenter clinical trial (RTOG 0625/ACRIN 6677). A total of 123 patients were enrolled in the current multicenter trial and signed institutional review board-approved informed consent at their respective institutions. MRI was acquired prior to and 8 weeks following therapy. A 5-point QC scoring system was used to evaluate DWI quality. fDM performance was evaluated according to the correlation of these metrics with PFS and OS at the first follow-up time-point. Results showed ADC variability of 7.3% in NAWM and 10.5% in CSF. A total of 68% of patients had usable DWI data and 47% of patients had high quality DWI data when also excluding patients that progressed before the first follow-up. fDM performance was improved by using only the highest quality DWI. High pre-treatment contrast enhancing tumor volume was associated with shorter PFS and OS. A high volume fraction of increasing ADC after therapy was associated with shorter PFS, while a high volume fraction of decreasing ADC was associated with shorter OS. In summary, DWI in multicenter trials are currently of limited value due to image quality. Improvements in consistency of image quality in multicenter trials are necessary for further advancement of DWI biomarkers.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Eunhee Kim
- Center for Statistical Sciences, Brown University, Providence, RI, USA
| | - Davis C Woodworth
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Helga Marques
- Center for Statistical Sciences, Brown University, Providence, RI, USA
| | - Jerrold L Boxerman
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Yair Safriel
- Radiology Associates of Clearwater, University of South Florida, Clearwater, FL, USA
| | - Robert C McKinstry
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Felix Bokstein
- Neuro-Oncology Service, Tel Aviv Sourasky Medical Center, Israel
| | - Rajan Jain
- Departments of Radiology and Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - T Linda Chi
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Gregory Sorensen
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Mark R Gilbert
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel P Barboriak
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
121
|
Pham K, Luo D, Siemann DW, Law BK, Reynolds BA, Hothi P, Foltz G, Harrison JK. VEGFR inhibitors upregulate CXCR4 in VEGF receptor-expressing glioblastoma in a TGFβR signaling-dependent manner. Cancer Lett 2015; 360:60-7. [PMID: 25676691 DOI: 10.1016/j.canlet.2015.02.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 11/16/2022]
Abstract
The failure of standard treatment for patients diagnosed with glioblastoma (GBM) coupled with the highly vascularized nature of this solid tumor has led to the consideration of agents targeting VEGF or VEGFRs, as alternative therapeutic strategies for this disease. Despite modest achievements in survival obtained with such treatments, failure to maintain an enduring survival benefit and more invasive relapsing tumors are evident. Our study suggests a potential mechanism by which anti-VEGF/VEGFR therapies regulate the enhanced invasive phenotype through a pathway that involves TGFβR and CXCR4. VEGFR signaling inhibitors (Cediranib and Vandetanib) elevated the expression of CXCR4 in VEGFR-expressing GBM cell lines and tumors, and enhanced the in vitro migration of these lines toward CXCL12. The combination of VEGFR inhibitor and CXCR4 antagonist provided a greater survival benefit to tumor-bearing animals. The upregulation of CXCR4 by VEGFR inhibitors was dependent on TGFβ/TGFβR, but not HGF/MET, signaling activity, suggesting a mechanism of crosstalk among VEGF/VEGFR, TGFβ/TGFβR, and CXCL12/CXCR4 pathways in the malignant phenotype of recurrent tumors after anti-VEGF/VEGFR therapies. Thus, the combination of VEGFR, CXCR4, and TGFβR inhibitors could provide an alternative strategy to halt GBM progression.
Collapse
Affiliation(s)
- Kien Pham
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Defang Luo
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Dietmar W Siemann
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brian K Law
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brent A Reynolds
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Parvinder Hothi
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | - Gregory Foltz
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | - Jeffrey K Harrison
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
122
|
Hochberg FH, Atai NA, Gonda D, Hughes MS, Mawejje B, Balaj L, Carter RS. Glioma diagnostics and biomarkers: an ongoing challenge in the field of medicine and science. Expert Rev Mol Diagn 2014; 14:439-52. [PMID: 24746164 DOI: 10.1586/14737159.2014.905202] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glioma is the most common brain tumor. For the more aggressive form, glioblastoma, standard treatment includes surgical resection, irradiation with adjuvant temozolomide and, on recurrence, experimental chemotherapy. However, the survival of patients remains poor. There is a critical need for minimally invasive biomarkers for diagnosis and as measures of response to therapeutic interventions. Glioma shed extracellular vesicles (EVs), which invade the surrounding tissue and circulate within both the cerebrospinal fluid and the systemic circulation. These tumor-derived EVs and their content serve as an attractive source of biomarkers. In this review, we discuss the current state of the art of biomarkers for glioma with emphasis on their EV derivation.
Collapse
Affiliation(s)
- Fred H Hochberg
- Department of Neurology and Program in Neuroscience, Massachusetts General Hospital and Harvard Medical School, Suite 340, 175 Cambridge Street, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
123
|
Batchelor TT, Reardon DA, de Groot JF, Wick W, Weller M. Antiangiogenic therapy for glioblastoma: current status and future prospects. Clin Cancer Res 2014; 20:5612-9. [PMID: 25398844 PMCID: PMC4234180 DOI: 10.1158/1078-0432.ccr-14-0834] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glioblastoma is characterized by high expression levels of proangiogenic cytokines and microvascular proliferation, highlighting the potential value of treatments targeting angiogenesis. Antiangiogenic treatment likely achieves a beneficial impact through multiple mechanisms of action. Ultimately, however, alternative proangiogenic signal transduction pathways are activated, leading to the development of resistance, even in tumors that initially respond. The identification of biomarkers or imaging parameters to predict response and to herald resistance is of high priority. Despite promising phase II clinical trial results and patient benefit in terms of clinical improvement and longer progression-free survival, an overall survival benefit has not been demonstrated in four randomized phase III trials of bevacizumab or cilengitide in newly diagnosed glioblastoma or cediranib or enzastaurin in recurrent glioblastoma. However, future studies are warranted. Predictive markers may allow appropriate patient enrichment, combination with chemotherapy may ultimately prove successful in improving overall survival, and novel agents targeting multiple proangiogenic pathways may prove effective.
Collapse
Affiliation(s)
- Tracy T Batchelor
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts.
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wolfgang Wick
- Neurooncology, University Clinic Heidelberg and German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Michael Weller
- Department of Neurology and Brain Tumor Center, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
124
|
Barone A, Sengupta R, Warrington NM, Smith E, Wen PY, Brekken RA, Romagnoli B, Douglas G, Chevalier E, Bauer MP, Dembowsky K, Piwnica-Worms D, Rubin JB. Combined VEGF and CXCR4 antagonism targets the GBM stem cell population and synergistically improves survival in an intracranial mouse model of glioblastoma. Oncotarget 2014; 5:9811-22. [PMID: 25238146 PMCID: PMC4259439 DOI: 10.18632/oncotarget.2443] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 09/08/2014] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma recurrence involves the persistence of a subpopulation of cells with enhanced tumor-initiating capacity (TIC) that reside within the perivascular space, or niche (PVN). Anti-angiogenic therapies may prevent the formation of new PVN but have not prevented recurrence in clinical trials, suggesting they cannot abrogate TIC activity. We hypothesized that combining anti-angiogenic therapy with blockade of PVN function would have superior anti-tumor activity. We tested this hypothesis in an established intracranial xenograft model of GBM using a monoclonal antibody specific for murine and human VEGF (mcr84) and a Protein Epitope Mimetic (PEM) CXCR4 antagonist, POL5551. When doses of POL5551 were increased to overcome an mcr84-induced improvement in vascular barrier function, combinatorial therapy significantly inhibited intracranial tumor growth and improved survival. Anti-tumor activity was associated with significant changes in tumor cell proliferation and apoptosis, and a reduction in the numbers of perivascular cells expressing the TIC marker nestin. A direct effect on TICs was demonstrated for POL5551, but not mcr84, in three primary patient-derived GBM isolates. These findings indicate that targeting the structure and function of the PVN has superior anti-tumor effect and provide a strong rationale for clinical evaluation of POL5551 and Avastin in patients with GBM.
Collapse
Affiliation(s)
- Amy Barone
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
| | - Rajarshi Sengupta
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
| | - Nicole M. Warrington
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
| | - Erin Smith
- BRIGHT Institute, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
| | - Patrick Y. Wen
- Center for Neuro-Oncology, Dana Farber/Brigham and Women’s Cancer Center, Brookline Ave, Boston, MA
- Division of Neuro-Oncology, Department of Neurology, Brigham and Women’s Hospital, Brookline Ave, Boston, MA
| | - Rolf A. Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Harry Hines Blvd. Dallas, TX
| | | | - Garry Douglas
- PolyPhor Ltd, Hegenheimermattweg 125 CH-4123 Allschwil, Switzerland
| | - Eric Chevalier
- PolyPhor Ltd, Hegenheimermattweg 125 CH-4123 Allschwil, Switzerland
| | - Michael P. Bauer
- PolyPhor Ltd, Hegenheimermattweg 125 CH-4123 Allschwil, Switzerland
| | - Klaus Dembowsky
- PolyPhor Ltd, Hegenheimermattweg 125 CH-4123 Allschwil, Switzerland
| | - David Piwnica-Worms
- BRIGHT Institute, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
- Department of Cell Biology & Physiology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Holcombe Dr., Houston, TX
| | - Joshua B. Rubin
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
- Department of Anatomy and Neurobiology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO
| |
Collapse
|
125
|
Omar AI. Tumor treating field therapy in combination with bevacizumab for the treatment of recurrent glioblastoma. J Vis Exp 2014:e51638. [PMID: 25407354 PMCID: PMC4541554 DOI: 10.3791/51638] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
A novel device that employs TTF therapy has recently been developed and is currently in use for the treatment of recurrent glioblastoma (rGBM). It was FDA approved in April 2011 for the treatment of patients 22 years or older with rGBM. The device delivers alternating electric fields and is programmed to ensure maximal tumor cell kill1. Glioblastoma is the most common type of glioma and has an estimated incidence of approximately 10,000 new cases per year in the United States alone2. This tumor is particularly resistant to treatment and is uniformly fatal especially in the recurrent setting3-5. Prior to the approval of the TTF System, the only FDA approved treatment for rGBM was bevacizumab6. Bevacizumab is a humanized monoclonal antibody targeted against the vascular endothelial growth factor (VEGF) protein that drives tumor angiogenesis7. By blocking the VEGF pathway, bevacizumab can result in a significant radiographic response (pseudoresponse), improve progression free survival and reduce corticosteroid requirements in rGBM patients8,9. Bevacizumab however failed to prolong overall survival in a recent phase III trial26. A pivotal phase III trial (EF-11) demonstrated comparable overall survival between physicians’ choice chemotherapy and TTF Therapy but better quality of life were observed in the TTF arm10. There is currently an unmet need to develop novel approaches designed to prolong overall survival and/or improve quality of life in this unfortunate patient population. One appealing approach would be to combine the two currently approved treatment modalities namely bevacizumab and TTF Therapy. These two treatments are currently approved as monotherapy11,12, but their combination has never been evaluated in a clinical trial. We have developed an approach for combining those two treatment modalities and treated 2 rGBM patients. Here we describe a detailed methodology outlining this novel treatment protocol and present representative data from one of the treated patients.
Collapse
Affiliation(s)
- Ayman I Omar
- Southern Illinois University School of Medicine;
| |
Collapse
|
126
|
Poulsen HS, Urup T, Michaelsen SR, Staberg M, Villingshøj M, Lassen U. The impact of bevacizumab treatment on survival and quality of life in newly diagnosed glioblastoma patients. Cancer Manag Res 2014; 6:373-87. [PMID: 25298738 PMCID: PMC4186574 DOI: 10.2147/cmar.s39306] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) remains one of the most devastating tumors, and patients have a median survival of 15 months despite aggressive local and systemic therapy, including maximal surgical resection, radiation therapy, and concomitant and adjuvant temozolomide. The purpose of antineoplastic treatment is therefore to prolong life, with a maintenance or improvement of quality of life. GBM is a highly vascular tumor and overexpresses the vascular endothelial growth factor A, which promotes angiogenesis. Preclinical data have suggested that anti-angiogenic treatment efficiently inhibits tumor growth. Bevacizumab is a humanized monoclonal antibody against vascular endothelial growth factor A, and treatment has shown impressive response rates in recurrent GBM. In addition, it has been shown that response is correlated to prolonged survival and improved quality of life. Several investigations in newly diagnosed GBM patients have been performed during recent years to test the hypothesis that newly diagnosed GBM patients should be treated with standard multimodality treatment, in combination with bevacizumab, in order to prolong life and maintain or improve quality of life. The results of these studies along with relevant preclinical data will be described, and pitfalls in clinical and paraclinical endpoints will be discussed.
Collapse
Affiliation(s)
- Hans Skovgaard Poulsen
- Department of Radiation Biology, Copenhagen University Hospital, Copenhagen, Denmark ; Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas Urup
- Department of Radiation Biology, Copenhagen University Hospital, Copenhagen, Denmark ; Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Signe Regner Michaelsen
- Department of Radiation Biology, Copenhagen University Hospital, Copenhagen, Denmark ; Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mikkel Staberg
- Department of Radiation Biology, Copenhagen University Hospital, Copenhagen, Denmark ; Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mette Villingshøj
- Department of Radiation Biology, Copenhagen University Hospital, Copenhagen, Denmark ; Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ulrik Lassen
- Department of Radiation Biology, Copenhagen University Hospital, Copenhagen, Denmark ; Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark ; Phase I Unit, The Finsencenter, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
127
|
Epidermal growth factor-like module containing mucin-like hormone receptor 2 expression in gliomas. J Neurooncol 2014; 121:53-61. [PMID: 25200831 DOI: 10.1007/s11060-014-1606-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 08/25/2014] [Indexed: 12/11/2022]
Abstract
Epidermal growth factor (EGF) module-containing mucin-like receptor 2 (EMR2) is a member of the seven span transmembrane adhesion G-protein coupled receptor subclass. This protein is expressed in a subset of glioblastoma (GBM) cells and associated with an invasive phenotype. The expression pattern and functional significance of EMR2 in low grade or anaplastic astrocytomas is unknown and our goal was to expand and further define EMR2's role in gliomas with an aggressive invasive phenotype. Using the TCGA survival data we describe EMR2 expression patterns across histologic grades of gliomas and demonstrate an association between increased EMR2 expression and poor survival (p < 0.05). This data supports prior functional data depicting that EMR2-positive neoplasms possess a greater capacity for infiltrative and metastatic spread. Genomic analysis suggests that EMR2 overexpression is associated with the mesenchymal GBM subtype (p < 0.0001). We also demonstrate that immunohistorchemistry is a feasible method for screening GBM patients for EMR2 expression. Protein and mRNA analysis demonstrated variable expression of all isoforms of EMR2 in all glioma grades, however GBM displayed the most diverse isoforms expression pattern as well as the highest expression of the EGF1-5 isoform of EMR2. Finally, a correlation of an increased EMR2 expression after bevacizumab treatment in glioma cells lines is identified. This observation should serve as the impetus for future studies to determine if this up-regulation of EMR2 plays a role in the observation of the diffuse and increasingly invasive recurrence patterns witnessed in a subset of GBM patients after bevacizumab treatment.
Collapse
|
128
|
Hamza MA, Mandel JJ, Conrad CA, Gilbert MR, Yung WKA, Puduvalli VK, DeGroot JF. Survival outcome of early versus delayed bevacizumab treatment in patients with recurrent glioblastoma. J Neurooncol 2014; 119:135-40. [PMID: 24803001 PMCID: PMC4297475 DOI: 10.1007/s11060-014-1460-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 04/23/2014] [Indexed: 12/12/2022]
Abstract
Bevacizumab (BEV) is widely used for treatment of patients with recurrent glioblastoma. It is not known if there are differences in outcome between early versus delayed BEV treatment of recurrent glioblastoma. We examined the relationship between the time of starting BEV treatment and outcomes in patients with recurrent glioblastoma. In this retrospective chart review, we identified patients with recurrent glioblastoma diagnosed between 2005 and 2011 who were treated with BEV alone or BEV-containing regimens. Data was analyzed to determine overall survival (OS) from time of diagnosis and progression free survival (PFS) from time of starting BEV. A total of 298 patients were identified, 112 patients received early BEV, 133 patients received delayed BEV, and 53 patients were excluded because they either progressed within 3 months of radiation or received BEV at the time of diagnosis. There was no significant difference in PFS between patients that received early BEV and those that received delayed BEV (5.2 vs. 4.3 months, p = 0.2). Patients treated with delayed BEV had longer OS when compared to those treated with early BEV (25.9 vs. 20.8 months, p = 0.005). In patients with recurrent glioblastoma, there was no significant difference in PFS from the time of starting BEV between early and delayed BEV. Although patients treated with delayed BEV seemed to have longer OS, a conclusion regarding OS outcome requires further prospective trials. These results may indicate that delaying treatment with BEV is not detrimental for survival of patients with recurrent glioblastoma.
Collapse
Affiliation(s)
- Mohamed A Hamza
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA,
| | | | | | | | | | | | | |
Collapse
|
129
|
Seyfried TN, Flores R, Poff AM, D'Agostino DP, Mukherjee P. Metabolic therapy: a new paradigm for managing malignant brain cancer. Cancer Lett 2014; 356:289-300. [PMID: 25069036 DOI: 10.1016/j.canlet.2014.07.015] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 01/18/2023]
Abstract
Little progress has been made in the long-term management of glioblastoma multiforme (GBM), considered among the most lethal of brain cancers. Cytotoxic chemotherapy, steroids, and high-dose radiation are generally used as the standard of care for GBM. These procedures can create a tumor microenvironment rich in glucose and glutamine. Glucose and glutamine are suggested to facilitate tumor progression. Recent evidence suggests that many GBMs are infected with cytomegalovirus, which could further enhance glucose and glutamine metabolism in the tumor cells. Emerging evidence also suggests that neoplastic macrophages/microglia, arising through possible fusion hybridization, can comprise an invasive cell subpopulation within GBM. Glucose and glutamine are major fuels for myeloid cells, as well as for the more rapidly proliferating cancer stem cells. Therapies that increase inflammation and energy metabolites in the GBM microenvironment can enhance tumor progression. In contrast to current GBM therapies, metabolic therapy is designed to target the metabolic malady common to all tumor cells (aerobic fermentation), while enhancing the health and vitality of normal brain cells and the entire body. The calorie restricted ketogenic diet (KD-R) is an anti-angiogenic, anti-inflammatory and pro-apoptotic metabolic therapy that also reduces fermentable fuels in the tumor microenvironment. Metabolic therapy, as an alternative to the standard of care, has the potential to improve outcome for patients with GBM and other malignant brain cancers.
Collapse
Affiliation(s)
| | | | - Angela M Poff
- Department of Molecular Pharmacology and Physiology, University of South Florida, 33612 Tampa, FL, USA
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, 33612 Tampa, FL, USA
| | | |
Collapse
|
130
|
LaViolette PS, Mickevicius NJ, Cochran EJ, Rand SD, Connelly J, Bovi JA, Malkin MG, Mueller WM, Schmainda KM. Precise ex vivo histological validation of heightened cellularity and diffusion-restricted necrosis in regions of dark apparent diffusion coefficient in 7 cases of high-grade glioma. Neuro Oncol 2014; 16:1599-606. [PMID: 25059209 DOI: 10.1093/neuonc/nou142] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Recent conflicting reports have found both brain tumor hypercellularity and necrosis in regions of restricted diffusion on MRI-derived apparent diffusion coefficient (ADC) images. This study precisely compares ADC and cell density voxel by voxel using postmortem human whole brain samples. METHODS Patients with meningioma were evaluated to determine a normative ADC distribution within benign fluid attenuated inversion recovery (FLAIR) T2/hyperintensity surrounding tumor. This distribution was used to calculate a minimum ADC threshold to define regions of ADC-FLAIR mismatch (AFMM), where restricted diffusion presented in conjunction with T2/FLAIR hyperintensity. Contrast-enhancing voxels were excluded from this analysis. AFMM maps were generated using imaging acquired prior to death in 7 patients with high-grade glioma who eventually donated their brains upon death. Histological samples were taken from numerous regions of abnormal FLAIR and AFMM. Each sample was computationally processed to determine cell density. Custom software was then used to downsample coregistered microscopic histology to the more coarse MRI resolution. A voxel-by-voxel evaluation comparing ADC and cellularity was then performed. RESULTS An ADC threshold of 0.929 × 10(-3) mm(2)/s was calculated from meningioma-induced edema and was used to define AFMM. Regions of AFMM showed significantly greater cell density in 6 of 7 high-grade glioma cases compared with regions of hyperintense FLAIR alone (P < .0001). Two patients had small regions of diffusion-restricted necrosis that had significantly lower ADC than nearby hypercellularity. CONCLUSIONS Regions of AFMM contain hypercellularity except for regions with extremely restricted diffusion, where necrosis is present.
Collapse
Affiliation(s)
- Peter S LaViolette
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nikolai J Mickevicius
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Elizabeth J Cochran
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Scott D Rand
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jennifer Connelly
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Joseph A Bovi
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mark G Malkin
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Wade M Mueller
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kathleen M Schmainda
- Department of Radiology (P.S.L., S.D.R., K.M.S.), Department of Biophysics (N.J.M., K.M.S.), Department of Pathology (E.J.C.), Department of Neurology (J.C., M.G.M.), Department of Radiation Oncology (J.A.B.), and Department of Neurosurgery (M.G.M., W.M.M.), Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
131
|
Woodworth GF, Dunn GP, Nance EA, Hanes J, Brem H. Emerging insights into barriers to effective brain tumor therapeutics. Front Oncol 2014; 4:126. [PMID: 25101239 PMCID: PMC4104487 DOI: 10.3389/fonc.2014.00126] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/13/2014] [Indexed: 12/27/2022] Open
Abstract
There is great promise that ongoing advances in the delivery of therapeutics to the central nervous system (CNS) combined with rapidly expanding knowledge of brain tumor patho-biology will provide new, more effective therapies. Brain tumors that form from brain cells, as opposed to those that come from other parts of the body, rarely metastasize outside of the CNS. Instead, the tumor cells invade deep into the brain itself, causing disruption in brain circuits, blood vessel and blood flow changes, and tissue swelling. Patients with the most common and deadly form, glioblastoma (GBM) rarely live more than 2 years even with the most aggressive treatments and often with devastating neurological consequences. Current treatments include maximal safe surgical removal or biopsy followed by radiation and chemotherapy to address the residual tumor mass and invading tumor cells. However, delivering effective and sustained treatments to these invading cells without damaging healthy brain tissue is a major challenge and focus of the emerging fields of nanomedicine and viral and cell-based therapies. New treatment strategies, particularly those directed against the invasive component of this devastating CNS disease, are sorely needed. In this review, we (1) discuss the history and evolution of treatments for GBM, (2) define and explore three critical barriers to improving therapeutic delivery to invasive brain tumors, specifically, the neuro-vascular unit as it relates to the blood brain barrier, the extra-cellular space in regard to the brain penetration barrier, and the tumor genetic heterogeneity and instability in association with the treatment efficacy barrier, and (3) identify promising new therapeutic delivery approaches that have the potential to address these barriers and create sustained, meaningful efficacy against GBM.
Collapse
Affiliation(s)
- Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine , Baltimore, MD , USA ; Department of Anatomy and Neurobiology, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Gavin P Dunn
- Department of Neurosurgery, Pathology and Immunology, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine , St. Louis, MO , USA
| | - Elizabeth A Nance
- Center for Nanomedicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Justin Hanes
- Center for Nanomedicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Ophthalmology, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Neurosurgery, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| |
Collapse
|
132
|
Ajaz M, Jefferies S, Brazil L, Watts C, Chalmers A. Current and investigational drug strategies for glioblastoma. Clin Oncol (R Coll Radiol) 2014; 26:419-30. [PMID: 24768122 DOI: 10.1016/j.clon.2014.03.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 11/21/2022]
Abstract
Medical treatments for glioblastoma face several challenges. Lipophilic alkylators remain the mainstay of treatment, emphasising the primacy of good blood-brain barrier penetration. Temozolomide has emerged as a major contributor to improved patient survival. The roles of procarbazine and vincristine in the procarbazine, lomustine and vincristine (PCV) schedule have attracted scrutiny and several lines of evidence now support the use of lomustine as effective single-agent therapy. Bevacizumab has had a convoluted development history, but clearly now has no major role in first-line treatment, and may even be detrimental to quality of life in this setting. In later disease, clinically meaningful benefits are achievable in some patients, but more impressively the combination of bevacizumab and lomustine shows early promise. Over the last decade, investigational strategies in glioblastoma have largely subscribed to the targeted kinase inhibitor paradigm and have mostly failed. Low prevalence dominant driver lesions such as the FGFR-TACC fusion may represent a niche role for this agent class. Immunological, metabolic and radiosensitising approaches are being pursued and offer more generalised efficacy. Finally, trial design is a crucial consideration. Progress in clinical glioblastoma research would be greatly facilitated by improved methodologies incorporating: (i) routine pharmacokinetic and pharmacodynamic assessments by preoperative dosing; and (ii) multi-stage, multi-arm protocols incorporating new therapy approaches and high-resolution biology in order to guide necessary improvements in science.
Collapse
Affiliation(s)
- M Ajaz
- Surrey Cancer Research Institute, University of Surrey, Guildford, UK.
| | - S Jefferies
- Oncology Centre, Addenbrooke's Hospital, Cambridge, UK
| | - L Brazil
- Guy's, St Thomas' and King's College Hospitals, London, UK
| | - C Watts
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - A Chalmers
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
133
|
Increased survival using delayed gamma knife radiosurgery for recurrent high-grade glioma: a feasibility study. World Neurosurg 2014; 82:e623-32. [PMID: 24930898 DOI: 10.1016/j.wneu.2014.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 03/04/2014] [Accepted: 06/09/2014] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The current study retrospectively assessed delayed gamma knife radiosurgery (GKRS) in the management of high-grade glioma recurrences. METHODS A total of 55 consecutive patients with high-grade glioma comprising 68 World Health Organization (WHO) III and WHO IV were treated with GKRS for local recurrences between 2001 and 2007. All patients had undergone microsurgery and radiochemotherapy, considered as standard therapy for high-grade glioma. Complete follow-up was available in all patients; median follow-up was 17.2 months (2.5-114.2 months). Median tumor volume was 5.2 mL, prescription dose was 20 Gy (14-22 Gy), and median max dose was 45 Gy (30-77.3 Gy). RESULTS The patients with WHO III tumors showed a median survival of 49.6 months with and a 2-year survival of 90%. After GKRS of the recurrences, these patients showed a median survival of 24.2 months and a 2-year survival of 50%. The patients with WHO IV tumors had a median survival of 24.5 months with a 2-year survival of 51.4%. After the recurrence was treated with GKRS, the median survival was 11.3 months and a 2-year survival: 22.9% for the WHO IV patients. CONCLUSION The current study shows a survival benefit for high-grade glioma recurrences when GKRS was administered after standard therapy. This is a relevant improvement compared with earlier studies that had had not been able to provide a beneficial effect timing radiosurgery in close vicinity to EBRT.
Collapse
|
134
|
Lacouture ME, Elizabeth Davis M, Elzinga G, Butowski N, Tran D, Villano JL, DiMeglio L, Davies AM, Wong ET. Characterization and Management of Dermatologic Adverse Events With the NovoTTF-100A System, a Novel Anti-mitotic Electric Field Device for the Treatment of Recurrent Glioblastoma. Semin Oncol 2014; 41 Suppl 4:S1-14. [DOI: 10.1053/j.seminoncol.2014.03.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
135
|
Kaloshi G, Roji A, Seferi A, Cakani B, Bushati T, Roci E, Petrela M. Spinal Dissemination of Intracranial Glioblastoma in Bevacizumab Era: a Potential Bevacizumab-induced Mechanism. Acta Inform Med 2014; 22:142-4. [PMID: 24825943 PMCID: PMC4008035 DOI: 10.5455/aim.2014.22.142-144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Spinal metastasis, a devastating neurologic complication of intracranial glioblastomas is not as uncommon as initially thought. It varies from 25% in supratentorial glioblastomas to 60% in infratentorial glioblastomas. The underlying pathogenesis spinal spread of high-grade gliomas is still unclear. To date, no causal responsibility of Bevacizumab (BEV) was noted. Here, we report for the first time, a case of thoracic intramedullary metastases from a cerebral glioblastoma pre-treated with BEV. A critical and exhaustive review is provided.
Collapse
Affiliation(s)
- Gentian Kaloshi
- Department of neurosurgery, University Hospital Center "Mother Theresa", Tirana, Albania
| | - Arben Roji
- Department of neurosurgery, University Hospital Center "Mother Theresa", Tirana, Albania
| | - Arsen Seferi
- Department of neurosurgery, University Hospital Center "Mother Theresa", Tirana, Albania
| | - Bujar Cakani
- Department of neurosurgery, University Hospital Center "Mother Theresa", Tirana, Albania
| | - Teona Bushati
- Department of neurosurgery, University Hospital Center "Mother Theresa", Tirana, Albania
| | - Ermir Roci
- Department of neurosurgery, University Hospital Center "Mother Theresa", Tirana, Albania
| | - Mentor Petrela
- Department of neurosurgery, University Hospital Center "Mother Theresa", Tirana, Albania
| |
Collapse
|
136
|
Boer JC, Walenkamp AME, den Dunnen WFA. Recruitment of bone marrow derived cells during anti-angiogenic therapy in GBM: the potential of combination strategies. Crit Rev Oncol Hematol 2014; 92:38-48. [PMID: 24933160 DOI: 10.1016/j.critrevonc.2014.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/10/2014] [Accepted: 05/02/2014] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) is a highly vascular tumor characterized by rapid and invasive tumor growth, followed by oxygen depletion, hypoxia and neovascularization, which generate a network of disorganized, tortuous and permeable vessels. Recruitment of bone marrow derived cells (BMDC) is crucial for vasculogenesis. These cells may act as vascular progenitors by integrating into the newly formed blood vessels or as vascular modulators by releasing pro-angiogenic factors. In patients with recurrent GBM, anti-vascular endothelial growth factor (VEGF) therapy has been evaluated in combination with chemotherapy, yielding improvements in progression-free survival (PFS). However, benefits are temporary as vascular tumors acquire angiogenic pathways independently of VEGF. Specifically, acute hypoxia following prolonged VEGF depletion induces the recruitment of certain myeloid cell subpopulations, which highly contribute to treatment refractoriness. Here we review the molecular mechanisms of neovascularization in relation to bevacizumab therapy with special emphasis on the recruitment of BMDCs and possible combination therapies for GBM patients.
Collapse
Affiliation(s)
- Jennifer C Boer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Annemiek M E Walenkamp
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
137
|
Khasraw M, Ameratunga M, Grommes C. Bevacizumab for the treatment of high-grade glioma: an update after phase III trials. Expert Opin Biol Ther 2014; 14:729-40. [PMID: 24655021 DOI: 10.1517/14712598.2014.898060] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Gliomas are highly vascular and rich in VEGF, which promotes angiogenesis. Bevacizumab is a monoclonal antibody against VEGF, inhibiting angiogenesis by preventing receptor activation. Early Phase II clinical trials using bevacizumab in both newly diagnosed and recurrent high-grade gliomas (HGG) showed promising results, but these have not been confirmed in recent Phase III trials. This review is an update including recently reported Phase II and III study results. AREAS COVERED This is a review of clinical trials investigating bevacizumab in newly diagnosed and recurrent HGG with a focus on outcome results. A future perspective about the expected future role of bevacizumab is given. Bevacizumab efficacy, safety and tolerability, the combination of radiation and bevacizumab, as well as the use of bevacizumab to treat pseudoprogression are discussed. Further criteria of response evaluation needed to be adjusted in the age of antiangiogenic therapy are also discussed. EXPERT OPINION Bevacizumab has been shown to be safe and tolerable in HGG. In the recurrent disease setting, bevacizumab might offer clinical benefits and is currently approved as a single agent for this indication. Although clinical trials demonstrate a prolonged progression-free survival (PFS) in bevacizumab-treated HGG, a benefit on overall survival has not been demonstrated. Research so far shows that bevacizumab appears to prolong PFS in newly diagnosed glioblastoma. Available data do not demonstrate a survival benefit in newly diagnosed patients. In the recurrent setting, there is no adequately powered randomized clinical trial to address whether there is a PFS or survival benefit with bevacizumab. Bevacizumab has also been introduced into other settings in neuro-oncology, including concurrent administration with re-irradiation for recurrent HGG.
Collapse
Affiliation(s)
- Mustafa Khasraw
- Geelong Hospital, Andrew Love Cancer Centre , 70 Swanston Street, Geelong, VIC 3220 , Australia +61 3 42152700 ; +61 3 42152836 ;
| | | | | |
Collapse
|
138
|
Mazaris P, Hong X, Altshuler D, Schultz L, Poisson LM, Jain R, Mikkelsen T, Rosenblum M, Kalkanis S. Key determinants of short-term and long-term glioblastoma survival: a 14-year retrospective study of patients from the Hermelin Brain Tumor Center at Henry Ford Hospital. Clin Neurol Neurosurg 2014; 120:103-12. [PMID: 24731587 DOI: 10.1016/j.clineuro.2014.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 02/06/2014] [Accepted: 03/01/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Glioblastoma (GBM) is a heterogeneous neoplasm with a small percentage of long-term survivors. Despite aggressive surgical resection and advances in radiotherapy and chemotherapy, the median survival for patients with GBM is 12-14 months. Factors associated with a favorable prognosis include young age, high performance status, gross resection >98%, non-eloquent tumor location and O6-methylguanine methyltransferase (MGMT) promoter methylation. We retrospectively analyzed the relationship of clinical, epidemiologic, genetic and molecular characteristics with survival in patients with GBM. METHODS This retrospective analysis of overall survival looked at the outcomes of 480 patients diagnosed with GBM over 14 years at a single institution. Multivariate analysis was performed examining multiple patient characteristics. RESULTS Median survival time improved from 11.8 months in patients diagnosed from 1995 to 1999 to 15.9 months in those diagnosed from 2005 to 2008. Factors associated with survivor groups were age, KPS, tumor resection, treatment received and early progression. 18 cancer-related genes were upregulated in short-term survivors and five genes were downregulated in short-term survivors. CONCLUSIONS Epidemiologic, clinical, and molecular characteristics all contribute to GBM prognosis. Identifying factors associated with survival is important for treatment strategies as well as research for novel therapeutics and technologies. This study demonstrated improved survival for patients over time as well as significant differences among survivor groups.
Collapse
Affiliation(s)
- Paul Mazaris
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Xin Hong
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - David Altshuler
- Wayne State University School of Medicine, 1313 Scott Hall, Detroit 48201, USA
| | - Lonni Schultz
- Public Health Sciences, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Laila M Poisson
- Public Health Sciences, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Rajan Jain
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA; Radiology, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Tom Mikkelsen
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Mark Rosenblum
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Steven Kalkanis
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA.
| |
Collapse
|
139
|
Anderson MD, Hamza MA, Hess KR, Puduvalli VK. Implications of bevacizumab discontinuation in adults with recurrent glioblastoma. Neuro Oncol 2014; 16:823-8. [PMID: 24596117 DOI: 10.1093/neuonc/nou021] [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: 12/24/2022] Open
Abstract
BACKGROUND Patients with recurrent glioblastoma benefiting from bevacizumab are often treated indefinitely due to concerns regarding rebound tumor recurrence upon discontinuation. However, treatment is discontinued for reasons other than disease progression in a subset of these patients, the characteristics and outcomes of which are poorly defined. METHODS Of 342 adults with recurrent glioblastoma in our database treated with bevacizumab, 82 received treatment for ≥ 6 months; of these, bevacizumab was discontinued for reasons other than tumor progression in 18 patients (Bev-D) and for disease progression in the remainder (Bev-S). The impact of discontinuation on outcome was assessed with discontinuation as a time-dependent covariate in a Cox hazards model for progression-free survival. RESULTS There was no difference in hazard rates for progression between Bev-D and Bev-S groups; the adjusted hazard ratio for progression using discontinuation as a time-dependent covariate was 0.91 (95% CI:0.47, 1.78). The median PFS after bevacizumab-discontinuation was 27 weeks (95% CI:15-NR). At progression, a higher proportion of Bev-D patients had local progression compared with the Bev-S patients. Salvage therapy in Bev-D patients yielded a PFS-26 weeks of 47% (95% CI:23%-94%) with a median PFS of 23 weeks (95% CI:12-NR), vs. 5% (95% CI: 1%-21%) and 9 weeks (95% CI: 6-11) in Bev-S patients (HR:0.3;CI, 0.1-0.6) (P = .0007). CONCLUSIONS Bevacizumab discontinuation unrelated to disease progression does not appear to cause rebound recurrence or worsen PFS in patients who benefit from bevacizumab. Additionally, Bev-D patients had an improved response to salvage therapy, findings which provide a strong basis for a prospective study.
Collapse
Affiliation(s)
- Mark D Anderson
- Department of Neuro-Oncology (M.A., M.H., V.P.); Department of Biostatistics (K.H.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mohamed A Hamza
- Department of Neuro-Oncology (M.A., M.H., V.P.); Department of Biostatistics (K.H.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kenneth R Hess
- Department of Neuro-Oncology (M.A., M.H., V.P.); Department of Biostatistics (K.H.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vinay K Puduvalli
- Department of Neuro-Oncology (M.A., M.H., V.P.); Department of Biostatistics (K.H.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
140
|
Wong ET, Lok E, Swanson KD, Gautam S, Engelhard HH, Lieberman F, Taillibert S, Ram Z, Villano JL. Response assessment of NovoTTF-100A versus best physician's choice chemotherapy in recurrent glioblastoma. Cancer Med 2014; 3:592-602. [PMID: 24574359 PMCID: PMC4101750 DOI: 10.1002/cam4.210] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 01/16/2014] [Accepted: 01/17/2014] [Indexed: 12/24/2022] Open
Abstract
The NovoTTF-100A device emits frequency-tuned alternating electric fields that interfere with tumor cell mitosis. In phase III trial for recurrent glioblastomas, NovoTTF-100A was shown to have equivalent efficacy and less toxicity when compared to Best Physician's Choice (BPC) chemotherapy. We analyzed the characteristics of responders and nonresponders in both cohorts to determine the characteristics of response and potential predictive factors. Tumor response and progression were determined by Macdonald criteria. Time to response, response duration, progression-free survival (PFS) ± Simon–Makuch correction, overall survival (OS), prognostic factors, and relative hazard rates were compared between responders and nonresponders. Median response duration was 7.3 versus 5.6 months for NovoTTF-100A and BPC chemotherapy, respectively (P = 0.0009). Five of 14 NovoTTF-100A responders but none of seven BPC responders had prior low-grade histology. Mean cumulative dexamethasone dose was 35.9 mg for responders versus 485.6 mg for nonresponders in the NovoTTF-100A cohort (P < 0.0001). Hazard analysis showed delayed tumor progression in responders compared to nonresponders. Simon–Makuch-adjusted PFS was longer in responders than in nonresponders treated with NovoTTF-100A (P = 0.0007) or BPC chemotherapy (P = 0.0222). Median OS was longer for responders than nonresponders treated with NovoTTF-100A (P < 0.0001) and BPC chemotherapy (P = 0.0235). Pearson analysis showed strong correlation between response and OS in NovoTTF-100A (P = 0.0002) but not in BPC cohort (P = 0.2900). Our results indicate that the response characteristics favor NovoTTF-100A and data on prior low-grade histology and dexamethasone suggest potential genetic and epigenetic determinants of NovoTTF-100A response.
Collapse
Affiliation(s)
- Eric T Wong
- Brain Tumor Center and Neuro-Oncology Unit, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Dowson N, Thomas P, Fay M, Jeffree RL, Gal Y, Bourgeat P, Smith J, Winter C, Coulthard A, Salvado O, Crozier S, Rose S. Early prediction of treatment response in advanced gliomas with (18)F-dopa positron-emission tomography. ACTA ACUST UNITED AC 2014; 21:e172-8. [PMID: 24523617 DOI: 10.3747/co.21.1772] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Imaging markers that enable prediction of survival are of interest for aiding clinical decision-making for patients with advanced glioma. [...]
Collapse
Affiliation(s)
- Nicholas Dowson
- The Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, Royal Brisbane and Women's Hospital, Brisbane, Australia,
| | - Paul Thomas
- Specialised pet Services Queensland, Royal Brisbane and Women's Hospital, School of Medicine, The University of Queensland, Brisbane, Australia
| | - Michael Fay
- Department of Radiation Oncology, Royal Brisbane and Women's Hospital, School of Medicine, The University of Queensland, Brisbane, Australia
| | - Rosalind L Jeffree
- Department of Neurosurgery, Royal Brisbane and Women's Hospital, School of Medicine, The University of Queensland, Brisbane, Australia
| | - Yaniv Gal
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia
| | - Pierrick Bourgeat
- The Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, Brisbane, Australia
| | - Jye Smith
- Specialised pet Services Queensland, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Craig Winter
- Department of Neurosurgery, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Alan Coulthard
- Department of Radiology, Royal Brisbane and Women's Hospital, School of Medicine, The University of Queensland, Brisbane, Australia
| | - Olivier Salvado
- The Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, Brisbane, Australia
| | - Stuart Crozier
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia
| | - Stephen Rose
- The Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, The University of Queensland, Brisbane, Australia
| |
Collapse
|
142
|
Soffietti R, Trevisan E, Bertero L, Cassoni P, Morra I, Fabrini MG, Pasqualetti F, Lolli I, Castiglione A, Ciccone G, Rudà R. Bevacizumab and fotemustine for recurrent glioblastoma: a phase II study of AINO (Italian Association of Neuro-Oncology). J Neurooncol 2014; 116:533-41. [PMID: 24293233 PMCID: PMC3905193 DOI: 10.1007/s11060-013-1317-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/17/2013] [Indexed: 01/05/2023]
Abstract
The optimal combination of bevacizumab with cytotoxic or cytostatic drugs in recurrent glioblastoma is unknown. We performed a phase 2 trial of combined bevacizumab and fotemustine for patients with glioblastoma at first relapse after radiotherapy and temozolomide. The primary endpoint was 6-month progression-free survival (PFS), while secondary endpoints were overall survival (OS), response rate based on RANO criteria and toxicity. Fifty-four patients with recurrent GBM were enrolled. The authors observed a 6-month PFS rate of 42.6% (95% CI 29.3-55.2) and a median PFS of 5.2 months (95% CI 3.8-6.6). The median OS was 9.1 months (95% CI 7.3-10.3). Twenty-eight patients (52%) had a radiographic response, and a significant neurological improvement with steroid reduction was observed in 25/42 symptomatic patients (60%). MGMT promoter methylation was significantly associated with improved PFS in univariate analysis. Most unifocal tumors at baseline had a focal enhancing progression (76%), while the diffuse non-enhancing progression accounted for 9.5%. Response or survival were not associated with any pattern of progression. Survival after failure of treatment was short. Twelve out of 54 patients (22%) discontinued fotemustine for grade 3/4 myelotoxicity, while 4/54 (7.4%) discontinued bevacizumab. This study failed to demonstrate a superiority of the combination of bevacizumab and fotemustine over either bevacizumab or fotemustine alone as historical controls. Future studies should explore alternative regimens of combination of the two drugs.
Collapse
Affiliation(s)
- Riccardo Soffietti
- Dept. Neuro-Oncology, University and City of Health and Science Hospital of Turin, Via Cherasco 15, 10126, Turin, Italy,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Magnuson W, Ian Robins H, Mohindra P, Howard S. Large volume reirradiation as salvage therapy for glioblastoma after progression on bevacizumab. J Neurooncol 2014; 117:133-9. [PMID: 24469853 DOI: 10.1007/s11060-014-1363-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 01/06/2014] [Indexed: 10/25/2022]
Abstract
Outcomes after bevacizumab failure for recurrent glioblastoma (GBM) are poor. Our analysis of 16 phase II trials (n = 995) revealed a median overall survival (OS) of 3.8 months (±1.0 month SD) after bevacizumab failure with no discernible activity of salvage chemotherapy. Thus, the optimal treatment for disease progression after bevacizumab has yet to be elucidated. This study evaluated the efficacy of reirradiation for patients with GBM after progression on bevacizumab. An IRB approved retrospective (2/2008-5/2013) analysis was performed of 23 patients with recurrent GBM (after standard radiotherapy/temozolomide) treated with bevacizumab (10 mg/kg) every 2 weeks until progression (median age 53 years; median KPS 80; median progression free survival on bevacizumab 3.7 months). Within 7-14 days of progression on bevacizumab, patients initiated reirradiation to a dose of 54 Gy in 27 fractions using pulsed-reduced dose rate (PRDR) radiotherapy. The median planning target volume was 424 cm(3). At the start of reirradiation, bevacizumab (10 mg/kg) was given every 4 weeks for two additional cycles. The median OS and 6 month OS after bevacizumab failure was 6.9 months and 65 %, respectively. Reirradiation was well tolerated with no symptomatic grade 3-4 toxicities. Favorable outcomes of reirradiation after bevacizumab failure in patients with recurrent GBM suggest its role as a treatment option for large volume recurrences not amenable to stereotactic radiosurgery. As PRDR is easily accomplished from a technological standpoint, we are in the process of expanding this approach to a multi-institutional cooperative group trial.
Collapse
Affiliation(s)
- William Magnuson
- Department of Radiation Oncology, University of Wisconsin, Madison, WI, USA
| | | | | | | |
Collapse
|
144
|
Wiestler B, Radbruch A, Osswald M, Combs SE, Jungk C, Winkler F, Bendszus M, Unterberg A, Platten M, Wick W, Wick A. Towards optimizing the sequence of bevacizumab and nitrosoureas in recurrent malignant glioma. J Neurooncol 2014; 117:85-92. [PMID: 24458956 DOI: 10.1007/s11060-013-1356-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 12/31/2013] [Indexed: 12/24/2022]
Abstract
Studies on the monoclonal VEGF-A antibody bevacizumab gave raise to questions regarding the lack of an overall survival benefit, the optimal timing in the disease course and potential combination and salvage therapies. We retrospectively assessed survival, radiological progression type on bevacizumab and efficacy of salvage therapies in 42 patients with recurrent malignant gliomas who received bevacizumab and nitrosourea sequentially. 15 patients received bevacizumab followed by nitrosourea at progression and 27 patients vice versa. Time to treatment failure, defined as time from initiation of one to failure of the other treatment, was similar in both groups (9.6 vs. 9.2 months, log rank p = 0.19). Progression-free survival on nitrosoureas was comparable in both groups, while progression-free survival on bevacizumab was longer in the group receiving bevacizumab first (5.3 vs. 4.1 months, log rank p = 0.03). Survival times were similar for patients with grade III (n = 9) and grade IV (n = 33) tumors. Progression-free survival on bevacizumab for patients developing contrast-enhancing T1 progression was longer than for patients who displayed a non-enhancing T2 progression. However, post-progression survival times after bevacizumab failure were not different. Earlier treatment with bevacizumab was not associated with better outcome in this series. The fact that earlier as compared to later bevacizumab treatment does not result in a different time to treatment failure highlights the challenge for first-line or recurrence trials with bevacizumab to demonstrate an overall survival benefit if crossover of bevacizumab-naïve patients after progression occurs.
Collapse
Affiliation(s)
- Benedikt Wiestler
- Department of Neurooncology, University Hospital Heidelberg and National Center for Tumor Diseases, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Trevisan E, Bertero L, Bosa C, Magistrello M, Pellerino A, Rudà R, Soffietti R. Antiangiogenic therapy of brain tumors: the role of bevacizumab. Neurol Sci 2014; 35:507-14. [DOI: 10.1007/s10072-014-1627-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 01/03/2014] [Indexed: 12/18/2022]
|
146
|
Gillies RJ, Flowers CI, Drukteinis JS, Gatenby RA. A unifying theory of carcinogenesis, and why targeted therapy doesn't work. Eur J Radiol 2013; 81 Suppl 1:S48-50. [PMID: 23083599 DOI: 10.1016/s0720-048x(12)70018-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Robert J Gillies
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33602, USA.
| | | | | | | |
Collapse
|
147
|
Nagane M, Nishikawa R. Bevacizumab for glioblastoma-a promising drug or not? Cancers (Basel) 2013; 5:1456-68. [PMID: 24213559 PMCID: PMC3875948 DOI: 10.3390/cancers5041456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/24/2013] [Indexed: 12/31/2022] Open
Abstract
Two double blind, placebo-controlled, and randomized phase III studies were conducted, and the results including OS’s were reported at the ASCO Meeting in June 2013, which was the beginning of confusion surrounding this topic. This is a review article not only summarizing the previous evidence, but also looking beyond.
Collapse
Affiliation(s)
- Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan; E-Mail:
| | - Ryo Nishikawa
- Department of Neuro-Oncology/Neurosurgery, International Medical Center, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama-ken 350-1298, Japan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-42-984-4111; Fax: +81-42-984-4741
| |
Collapse
|
148
|
Ogura K, Mizowaki T, Arakawa Y, Sakanaka K, Miyamoto S, Hiraoka M. Efficacy of salvage stereotactic radiotherapy for recurrent glioma: impact of tumor morphology and method of target delineation on local control. Cancer Med 2013; 2:942-9. [PMID: 24403268 PMCID: PMC3892399 DOI: 10.1002/cam4.154] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 09/30/2013] [Accepted: 10/01/2013] [Indexed: 01/17/2023] Open
Abstract
In this study, we assessed the efficacy of salvage stereotactic radiotherapy (SRT) for recurrent glioma. From August 2008 to December 2012, 30 patients with recurrent glioma underwent salvage SRT. The initial histological diagnoses were World Health Organization (WHO) grades II, III, and IV in 6, 9, and 15 patients, respectively. Morphologically, the type of recurrence was classified as diffuse or other. Two methods of clinical target delineation were used: A, a contrast-enhancing tumor; or B, a contrast-enhancing tumor with a 3–10-mm margin and/or surrounding fluid attenuation inversion recovery (FLAIR) high-intensity areas. The prescribed dose was 22.5–35 Gy delivered in five fractions at an isocenter using a dynamic conformal arc technique. The overall survival (OS) and local control probability (LCP) after SRT were calculated using the Kaplan–Meier method. A univariate analysis was used to test the effect of clinical variables on OS/LCP. The median follow-up period was 272 days after SRT. The OS and LCP were 83% and 56% at 6 months after SRT, respectively. Morphologically, the tumor type correlated significantly with both OS and LCP (P = 0.006 and <0.001, respectively). The method of target delineation also had a significant influence on LCP (P = 0.016). Grade 3 radiation necrosis was observed in two patients according to Common Terminology Criteria for Adverse Events, version 3. Salvage SRT was safe and effective for recurrent glioma, especially non-diffuse recurrences. Improved local control might be obtained by adding a margin to contrast-enhancing tumors or including increased FLAIR high-intensity areas.
Collapse
Affiliation(s)
- Kengo Ogura
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | | | | | | | | | | |
Collapse
|
149
|
Lu-Emerson C, Snuderl M, Kirkpatrick ND, Goveia J, Davidson C, Huang Y, Riedemann L, Taylor J, Ivy P, Duda DG, Ancukiewicz M, Plotkin SR, Chi AS, Gerstner ER, Eichler AF, Dietrich J, Stemmer-Rachamimov AO, Batchelor TT, Jain RK. Increase in tumor-associated macrophages after antiangiogenic therapy is associated with poor survival among patients with recurrent glioblastoma. Neuro Oncol 2013; 15:1079-87. [PMID: 23828240 DOI: 10.1093/neuonc/not082] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Antiangiogenic therapy is associated with increased radiographic responses in glioblastomas, but tumors invariably recur. Because tumor-associated macrophages have been shown to mediate escape from antiangiogenic therapy in preclinical models, we examined the role of macrophages in patients with recurrent glioblastoma. We compared autopsy brain specimens from 20 patients with recurrent glioblastoma who received antiangiogenic treatment and chemoradiation with 8 patients who received chemotherapy and/or radiotherapy without antiangiogenic therapy or no treatment. Tumor-associated macrophages were morphologically and phenotypically analyzed using flow cytometry and immunohistochemistry for CD68, CD14, CD163, and CD11b expression. Flow cytometry showed an increase in macrophages in the antiangiogenic-treated patients. Immunohistochemical analysis demonstrated an increase in CD68+ macrophages in the tumor bulk (P < .01) and infiltrative areas (P = .02) in antiangiogenic-treated patients. We also observed an increase in CD11b+ cells in the tumor bulk (P < .01) and an increase in CD163+ macrophages in infiltrative tumor (P = .02). Of note, an increased number of CD11b+ cells in bulk and infiltrative tumors (P = .05 and P = .05, respectively) correlated with poor overall survival among patients who first received antiangiogenic therapy at recurrence. In summary, recurrent glioblastomas showed an increased infiltration in myeloid populations in the tumor bulk and in the infiltrative regions after antiangiogenic therapy. Higher numbers of CD11b+ cells correlated with poor survival among these patients. These data suggest that tumor-associated macrophages may participate in escape from antiangiogenic therapy and may represent a potential biomarker of resistance and a potential therapeutic target in recurrent glioblastoma.
Collapse
Affiliation(s)
- Christine Lu-Emerson
- Department of Neurology, Radiation Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Schulman KL, Berenson K, Tina Shih YC, Foley KA, Ganguli A, de Souza J, Yaghmour NA, Shteynshlyuger A. A checklist for ascertaining study cohorts in oncology health services research using secondary data: report of the ISPOR oncology good outcomes research practices working group. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2013; 16:655-669. [PMID: 23796301 DOI: 10.1016/j.jval.2013.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
OBJECTIVES The ISPOR Oncology Special Interest Group formed a working group at the end of 2010 to develop standards for conducting oncology health services research using secondary data. The first mission of the group was to develop a checklist focused on issues specific to selection of a sample of oncology patients using a secondary data source. METHODS A systematic review of the published literature from 2006 to 2010 was conducted to characterize the use of secondary data sources in oncology and inform the leadership of the working group prior to the construction of the checklist. A draft checklist was subsequently presented to the ISPOR membership in 2011 with subsequent feedback from the larger Oncology Special Interest Group also incorporated into the final checklist. RESULTS The checklist includes six elements: identification of the cancer to be studied, selection of an appropriate data source, evaluation of the applicability of published algorithms, development of custom algorithms (if needed), validation of the custom algorithm, and reporting and discussions of the ascertainment criteria. The checklist was intended to be applicable to various types of secondary data sources, including cancer registries, claims databases, electronic medical records, and others. CONCLUSIONS This checklist makes two important contributions to oncology health services research. First, it can assist decision makers and reviewers in evaluating the quality of studies using secondary data. Second, it highlights methodological issues to be considered when researchers are constructing a study cohort from a secondary data source.
Collapse
|