1
|
Sung KS, Cho WH, Cha SH, Kim YW, Choi SH, Kim HJ, Yun MS. Saturated Fatty Acid Emulsions Open the Blood-Brain Barrier and Promote Drug Delivery in Rat Brains. Pharmaceutics 2024; 16:246. [PMID: 38399300 PMCID: PMC10893510 DOI: 10.3390/pharmaceutics16020246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/29/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
We performed this study to evaluate whether saturated fatty acid (SFA) emulsions affect the BBB and determine the duration of BBB opening, thereby promoting drug delivery to the brain. Butyric, valeric, caproic, enanthic, and caprylic acid emulsions were infused into the carotid artery of the rat model. We evaluated the BBB opening and drug delivery over time. The trypan blue and doxorubicin delivery studies were repeated from 30 min to 6 h. In the 1 h rats in each group, transmission electron microscopy (TEM) was performed to morphologically evaluate tight junctions, and the delivery of temozolomide was assessed by desorption electrospray ionization mass spectrometry. The ipsilateral hemisphere was positive for trypan blue staining in all the five SFA emulsion groups. In the valeric, enanthic, and caprylic acid emulsion groups, RGB ratios were significantly higher at 30 min and decreased thereafter. Doxorubicin delivery increased in all emulsion groups at all time points. Tight junctions were observed to be open in all groups. TMZ delivery was significantly higher in the ipsilateral hemisphere. In conclusion, intra-arterially infused SFA emulsions opened the BBB and promoted drug delivery within 30 min, which decreased thereafter. Therefore, SFA emulsions may aid BBB research and promote drug delivery to the brain.
Collapse
Affiliation(s)
- Kyoung Su Sung
- Department of Neurosurgery, Dong-A University Hospital, Dong-A University College of Medicine, Busan 49201, Republic of Korea;
- Department of Medicine, The Graduate School of Medicine, Pusan National University, Busan 49241, Republic of Korea
| | - Won Ho Cho
- Department of Neurosurgery, Pusan National University Hospital, Biomedical Institute of Pusan National University Hospital, School of Medicine, Pusan National University, Busan 49241, Republic of Korea; (W.H.C.); (S.H.C.)
| | - Seung Heon Cha
- Department of Neurosurgery, Pusan National University Hospital, Biomedical Institute of Pusan National University Hospital, School of Medicine, Pusan National University, Busan 49241, Republic of Korea; (W.H.C.); (S.H.C.)
| | - Yong-Woo Kim
- Department of Radiology, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Seon Hee Choi
- Institute for Research and Industry Cooperation, Pusan National University, Busan 49241, Republic of Korea;
| | - Hak Jin Kim
- Department of Radiology, Pusan National University Hospital, Biomedical Institute of Pusan National University Hospital, School of Medicine, Pusan National University, Busan 49241, Republic of Korea
| | - Mi Sook Yun
- Division of Biostatistics, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| |
Collapse
|
2
|
Hargrave DR, Terashima K, Hara J, Kordes UR, Upadhyaya SA, Sahm F, Bouffet E, Packer RJ, Witt O, Sandalic L, Kieloch A, Russo M, Cohen KJ. Phase II Trial of Dabrafenib Plus Trametinib in Relapsed/Refractory BRAF V600-Mutant Pediatric High-Grade Glioma. J Clin Oncol 2023; 41:5174-5183. [PMID: 37643378 PMCID: PMC10666989 DOI: 10.1200/jco.23.00558] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/16/2023] [Accepted: 06/28/2023] [Indexed: 08/31/2023] Open
Abstract
PURPOSE BRAF V600 mutation is detected in 5%-10% of pediatric high-grade gliomas (pHGGs), and effective treatments are limited. In previous trials, dabrafenib as monotherapy or in combination with trametinib demonstrated activity in children and adults with relapsed/refractory BRAF V600-mutant HGG. METHODS This phase II study evaluated dabrafenib plus trametinib in patients with relapsed/refractory BRAF V600-mutant pHGG. The primary objective was overall response rate (ORR) by independent review by Response Assessment in Neuro-Oncology criteria. Secondary objectives included ORR by investigator determination, duration of response (DOR), progression-free survival, overall survival (OS), and safety. RESULTS A total of 41 pediatric patients with previously treated BRAF V600-mutant HGG were enrolled. At primary analysis, median follow-up was 25.1 months, and 51% of patients remained on treatment. Sixteen of 20 discontinuations were due to progressive disease in this relapsed/refractory pHGG population. Independently assessed ORR was 56% (95% CI, 40 to 72). Median DOR was 22.2 months (95% CI, 7.6 months to not reached [NR]). Fourteen deaths were reported. Median OS was 32.8 months (95% CI, 19.2 months to NR). The most common all-cause adverse events (AEs) were pyrexia (51%), headache (34%), and dry skin (32%). Two patients (5%) had AEs (both rash) leading to discontinuation. CONCLUSION In relapsed/refractory BRAF V600-mutant pHGG, dabrafenib plus trametinib improved ORR versus previous trials of chemotherapy in molecularly unselected patients with pHGG and was associated with durable responses and encouraging survival. These findings suggest that dabrafenib plus trametinib is a promising targeted therapy option for children and adolescents with relapsed/refractory BRAF V600-mutant HGG.
Collapse
Affiliation(s)
- Darren R Hargrave
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Keita Terashima
- National Center for Child Health and Development, Tokyo, Japan
| | | | - Uwe R Kordes
- University Medical Center Eppendorf, Hamburg, Germany
| | | | - Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- CCU Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg University Hospital, Heidelberg, Germany
- Hopp Children's Cancer Center (KiTZ), German Cancer Research Center (DKFZ), Heidelberg University Hospital, Heidelberg, Germany
| | - Eric Bouffet
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | - Olaf Witt
- Hopp Children's Cancer Center (KiTZ), German Cancer Research Center (DKFZ), Heidelberg University Hospital, Heidelberg, Germany
| | | | | | - Mark Russo
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | - Kenneth J Cohen
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
3
|
Seung WB, Cha SH, Kim HJ, Choi SH, Lee J, Kwak D, Hyun Woo K, You JW, Kim YW, Kim SK, Lee DS. Triolein emulsion enhances temozolomide brain delivery: an experimental study in rats. Drug Deliv 2021; 28:2373-2382. [PMID: 34747271 PMCID: PMC8583762 DOI: 10.1080/10717544.2021.1998247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose To evaluate the enhancement of temozolomide (TMZ) delivery in the rat brain using a triolein emulsion. Materials and Methods Rats were divided into the five groups as following: group 1 (negative control), group 2 (treated with triolein emulsion and TMZ 20 mg/kg), and group 3 (TMZ 20 mg/kg treatment without triolein), group 4 (treated with triolein emulsion and TMZ 10 mg/kg), and group 5 (TMZ 10 mg/kg treatment without triolein). Triolein emulsion was infused into the right common carotid artery. One hour later, the TMZ concentration was evaluated quantitatively and qualitatively using high-performance liquid chromatography (HPLC-MS) and desorption electrospray ionization mass spectrometry (DESI-MS) imaging, respectively. The concentration ratios of the ipsilateral to contralateral hemisphere in each group were determined and the statistical analysis was conducted using an unpaired t-test. Results Quantitatively, the TMZ concentration ratio of the ipsilateral to the control hemisphere was 2.41 and 1.13 in groups 2 and 3, and were 2.49 and 1.14 in groups 4 and 5, respectively. Thus, the TMZ signal intensities of TMZ in group 2 and 4 were statistically high in the ipsilateral hemispheres. Qualitatively, the signal intensity of TMZ was remarkably high in the ipsilateral hemisphere in group 2 and 4. Conclusions The triolein emulsion efficiently opened the blood-brain barrier and could provide a potential new strategy to enhance the therapeutic effect of TMZ. HPLC-MS and DESI-MS imaging were shown to be suitable for analyses of enhancement of brain TMZ concentrations.
Collapse
Affiliation(s)
- Won-Bae Seung
- Department of Neurosurgery, Dongguk University College of Medicine, Dongguk University Gyeongju Hospital, Gyeongju, South Korea.,Department of Neurosurgery, SMG Yeonse Hospital, Changwon, South Korea
| | - Seung Heon Cha
- College of Medicine, Pusan National University, Busan, Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Hak Jin Kim
- College of Medicine, Pusan National University, Busan, Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Seon Hee Choi
- College of Medicine, Pusan National University, Busan, Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Juho Lee
- College of Pharmacy, Pusan National University, Busan, South Korea
| | - Dongmin Kwak
- College of Pharmacy, Pusan National University, Busan, South Korea
| | - Kim Hyun Woo
- College of Pharmacy, Pusan National University, Busan, South Korea
| | - Jin-Wook You
- College of Pharmacy, Pusan National University, Busan, South Korea
| | - Yong-Woo Kim
- Pusan National University Yangsan Hospital, College of Medicine, Pusan National University, Busan, South Korea
| | - Sang Kyoon Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Da-Sol Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| |
Collapse
|
4
|
Shi S, Lu S, Jing X, Liao J, Li Q. The Prognostic Impact of Radiotherapy in Conjunction with Temozolomide in Diffuse Intrinsic Pontine Glioma: A Systematic Review and Meta-Analysis. World Neurosurg 2021; 148:e565-e571. [PMID: 33476781 DOI: 10.1016/j.wneu.2021.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Diffuse intrinsic pontine glioma (DIPG) is a rare and devastating brainstem glioma that occurs predominately in children. To date, the prognostic impact of radiotherapy (RT) in conjunction with temozolomide (TMZ) in DIPG has not been thoroughly analyzed. The aim of this meta-analysis was to analyze the effectiveness of RT quantitatively and precisely in conjunction with TMZ in improving the prognosis of DIPG. METHODS A systematic search of 8 electronic databases was conducted. Articles mainly discussing the prognostic impact of RT in conjunction with TMZ in DIPG were selected. The pooled 1- and 2-year overall survival (OS) and progression-free survival (PFS) were calculated. RESULTS A total of 14 studies fulfilled our inclusion criteria, involving 283 cases of patients with DIPG who were treated with RT in conjunction with TMZ. The pooled 1- and 2-year OS of this treatment was 43% and 11%, respectively. The pooled 1- and 2-year PFS was 20% and 2%, respectively. Subgroup analysis revealed that the heterogeneity remained almost the same in all stratum. Egger's test demonstrated that the possibility of publication bias was low. CONCLUSIONS Requirements of up-to-date evidence on evaluating the prognostic impact of this therapy are urgent.
Collapse
Affiliation(s)
- Shuai Shi
- Department of Neurosurgery, Tianjin Medical University, Tianjin, PR China; Department of Neurology, Tianjin Huanhu Hospital, Tianjin, PR China
| | - Shan Lu
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, PR China
| | - Xiyue Jing
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, PR China
| | - Jianwen Liao
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, PR China
| | - Qingguo Li
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, PR China.
| |
Collapse
|
5
|
Miklja Z, Yadav VN, Cartaxo RT, Siada R, Thomas CC, Cummings JR, Mullan B, Stallard S, Paul A, Bruzek AK, Wierzbicki K, Yang T, Garcia T, Wolfe I, Leonard M, Robertson PL, Garton HJ, Wahl DR, Parmar H, Sarkaria JN, Kline C, Mueller S, Nicolaides T, Glasser C, Leary SE, Venneti S, Kumar-Sinha C, Chinnaiyan AM, Mody R, Pai MP, Phoenix TN, Marini BL, Koschmann C. Everolimus improves the efficacy of dasatinib in PDGFRα-driven glioma. J Clin Invest 2020; 130:5313-5325. [PMID: 32603316 PMCID: PMC7524471 DOI: 10.1172/jci133310] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 06/24/2020] [Indexed: 12/26/2022] Open
Abstract
Pediatric and adult high-grade gliomas (HGGs) frequently harbor PDGFRA alterations. We hypothesized that cotreatment with everolimus may improve the efficacy of dasatinib in PDGFRα-driven glioma through combinatorial synergism and increased tumor accumulation of dasatinib. We performed dose-response, synergism, P-glycoprotein inhibition, and pharmacokinetic studies in in vitro and in vivo human and mouse models of HGG. Six patients with recurrent PDGFRα-driven glioma were treated with dasatinib and everolimus. We found that dasatinib effectively inhibited the proliferation of mouse and human primary HGG cells with a variety of PDGFRA alterations. Dasatinib exhibited synergy with everolimus in the treatment of HGG cells at low nanomolar concentrations of both agents, with a reduction in mTOR signaling that persisted after dasatinib treatment alone. Prolonged exposure to everolimus significantly improved the CNS retention of dasatinib and extended the survival of PPK tumor-bearing mice (mutant TP53, mutant PDGFRA, H3K27M). Six pediatric patients with glioma tolerated this combination without significant adverse events, and 4 patients with recurrent disease (n = 4) had a median overall survival of 8.5 months. Our results show that the efficacy of dasatinib treatment of PDGFRα-driven HGG was enhanced with everolimus and suggest a promising route for improving targeted therapy for this patient population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Hemant Parmar
- Department of Radiology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Jann N. Sarkaria
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Cassie Kline
- Department of Pediatrics and
- Department of Neurology, UCSF, San Francisco, California, USA
| | - Sabine Mueller
- Department of Pediatrics and
- Department of Neurology, UCSF, San Francisco, California, USA
| | - Theodore Nicolaides
- Division of Pediatric Hematology/Oncology, NYU Langone Medical Center, New York, New York, USA
| | - Chana Glasser
- Division of Pediatric Hematology/Oncology, NYU Winthrop Hospital, Mineola, New York, USA
| | - Sarah E.S. Leary
- Seattle Children’s Hospital/University of Washington (UW), Seattle, Washington, USA
| | | | | | - Arul M. Chinnaiyan
- Department of Pathology
- Department of Urology
- Michigan Center for Translational Pathology
- Howard Hughes Medical Institute
- Rogel Cancer Center, and
| | | | - Manjunath P. Pai
- College of Pharmacy, Michigan Medicine, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
6
|
Ruggiero A, Ariano A, Triarico S, Capozza MA, Romano A, Maurizi P, Mastrangelo S, Attinà G. Temozolomide and oral etoposide in children with recurrent malignant brain tumors. Drugs Context 2020; 9:2020-3-1. [PMID: 32547627 PMCID: PMC7271709 DOI: 10.7573/dic.2020-3-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/01/2020] [Accepted: 05/08/2020] [Indexed: 11/26/2022] Open
Abstract
Despite advances in the treatment of brain tumors, the prognosis of children with recurrent malignant brain tumors remains poor. Etoposide (VP-16), an inhibitor of nuclear enzyme deoxyribonucleic acid (DNA)-topoisomerase II, has shown activity in brain tumors. Its efficacy appears schedule dependent but, to date, the most effective schedule of administration has not been well defined. Temozolomide (TMZ), like VP-16, penetrates the blood-brain barrier and has activity against malignant brain tumors. This novel alkylating agent is rapidly absorbed and is highly bioavailable after oral administration. The antitumor activity of TMZ has been shown to be schedule dependent. Based on the evidence of different mechanisms of cytotoxicity, TMZ and VP-16 have been utilized in combination in patients with malignant brain tumors. This review evaluates the results derived from the combination use of TMZ and oral VP-16. The reported data suggest potential activity of oral VP-16 and TMZ alone or in combination. Further clinical trials are needed to explore and confirm their promising activity in relapsed brain neoplasms.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Anna Ariano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Michele Antonio Capozza
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Zhao L, Koschmann C. Integrating subgroups with mixed-type endpoints in early phase oncology trials. Stat Methods Med Res 2019; 29:498-507. [PMID: 30945609 DOI: 10.1177/0962280219840050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Testing anti-cancer agents with multiple disease subtypes is challenging and it becomes more complicated when the subgroups have different types of endpoints (such as binary endpoints of tumor response and progression-free survival endpoints). When this occurs, one common approach in oncology is to conduct a series of small screening trials in specific patient subgroups, and these trials are typically run in parallel, independent of each other. However, this approach does not consider the possibility that some of the patient subpopulations respond similarly to therapy. In this article, we developed a simple approach to jointly model subgroups with mixed-type endpoints, which allows borrowing strength across subgroups for efficient estimation of treatment effects.
Collapse
Affiliation(s)
- Lili Zhao
- Biostatistics Department, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Development of the CNS TAP tool for the selection of precision medicine therapies in neuro-oncology. J Neurooncol 2017; 137:155-169. [PMID: 29235051 DOI: 10.1007/s11060-017-2708-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/04/2017] [Indexed: 01/08/2023]
Abstract
The number of targeted therapies utilized in precision medicine are rapidly increasing. Neuro-oncology offers a unique challenge due to the varying blood brain barrier (BBB) penetration of each agent. Neuro-oncologists face a difficult task weighing the growing number of potential targeted therapies and their likelihood of BBB penetration. We developed the CNS TAP Working Group and performed an extensive literature review for the evidence-based creation of the CNS TAP tool, which was retrospectively validated by analyzing brain tumor patients who underwent therapy targeted based on genomic results from an academic sequencing study (MiOncoseq, n = 17) or private molecular profiling (Foundation One, n = 7). The CNS TAP tool scores relevant targeted agents by applying multiple variables (i.e., pre-clinical data, clinical data, BBB permeability) to patient specific genomic information and clinical trial availability. In the Michigan cohort, the CNS TAP tool predicted the selected agent 85.7% of the time. The CNS TAP tool predicted the agent independently selected by pediatric neuro-oncologists in the Colorado cohort 50% of the time. Patients with recurrent brain tumors treated with agents predicted by the CNS TAP tool demonstrated a median progression-free survival of 4 months and four patients with recurrent high-grade glioma maintained ongoing partial responses of at least 6 months. The CNS TAP tool is a formalized algorithm to assist clinicians select the optimal targeted therapy for neuro-oncology patients. The CNS TAP tool has relatively high concordance with selected therapies and clinical outcomes in patients receiving targeted therapy in this heterogeneous retrospective cohort were promising.
Collapse
|
9
|
Sayour EJ, Mitchell DA. Immunotherapy for Pediatric Brain Tumors. Brain Sci 2017; 7:brainsci7100137. [PMID: 29065490 PMCID: PMC5664064 DOI: 10.3390/brainsci7100137] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/20/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022] Open
Abstract
Malignant brain tumors are the most common cause of solid cancer death in children. New targeted therapies are vital to improve treatment outcomes, but must be developed to enable trafficking across the blood brain barrier (BBB). Since activated T cells cross the BBB, cancer immunotherapy can be harnessed to unlock the cytotoxic potential of the immune system. However, standard of care treatments (i.e., chemotherapy and radiation) applied concomitant to pediatric brain tumor immunotherapy may abrogate induction of immunotherapeutic responses. This review will discuss the development of immunotherapies within this paradigm using emerging approaches being investigated in phase I/II trials in children with refractory brain tumors, including checkpoint inhibitors, vaccine immunotherapy, and adoptive cell therapy.
Collapse
Affiliation(s)
- Elias J Sayour
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lilian S. Wells Department of Neurosurgery, 1149 South Newell Drive, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| | - Duane A Mitchell
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lilian S. Wells Department of Neurosurgery, 1149 South Newell Drive, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
10
|
Jones C, Karajannis MA, Jones DTW, Kieran MW, Monje M, Baker SJ, Becher OJ, Cho YJ, Gupta N, Hawkins C, Hargrave D, Haas-Kogan DA, Jabado N, Li XN, Mueller S, Nicolaides T, Packer RJ, Persson AI, Phillips JJ, Simonds EF, Stafford JM, Tang Y, Pfister SM, Weiss WA. Pediatric high-grade glioma: biologically and clinically in need of new thinking. Neuro Oncol 2017; 19:153-161. [PMID: 27282398 PMCID: PMC5464243 DOI: 10.1093/neuonc/now101] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/14/2016] [Indexed: 12/14/2022] Open
Abstract
High-grade gliomas in children are different from those that arise in adults. Recent collaborative molecular analyses of these rare cancers have revealed previously unappreciated connections among chromatin regulation, developmental signaling, and tumorigenesis. As we begin to unravel the unique developmental origins and distinct biological drivers of this heterogeneous group of tumors, clinical trials need to keep pace. It is important to avoid therapeutic strategies developed purely using data obtained from studies on adult glioblastoma. This approach has resulted in repetitive trials and ineffective treatments being applied to these children, with limited improvement in clinical outcome. The authors of this perspective, comprising biology and clinical expertise in the disease, recently convened to discuss the most effective ways to translate the emerging molecular insights into patient benefit. This article reviews our current understanding of pediatric high-grade glioma and suggests approaches for innovative clinical management.
Collapse
Affiliation(s)
- Chris Jones
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Matthias A Karajannis
- Division of Pediatric Hematology/Oncology, NYU Langone Medical Center, New York, NY, USA
| | - David T W Jones
- Division of Pediatric Neurooncology, German Cancer Research Centre, Heidelberg, Germany
| | - Mark W Kieran
- Pediatric Medical Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michelle Monje
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, California, USA
| | - Suzanne J Baker
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Oren J Becher
- Departments of Pediatrics and Pathology, Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Yoon-Jae Cho
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, California, USA
| | - Nalin Gupta
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Cynthia Hawkins
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Darren Hargrave
- Neuro-oncology and Experimental Therapeutics, Great Ormond Street Hospital for Children, London, UK
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Nada Jabado
- Department of Pediatrics, McGill University, Montreal, Canada
| | - Xiao-Nan Li
- Brain Tumor Program, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Sabine Mueller
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Theo Nicolaides
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, District of Columbia, USA
| | - Anders I Persson
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Joanna J Phillips
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Erin F Simonds
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - James M Stafford
- Department of Biochemistry, NYU Langone Medical Center, New York, New York, USA
| | - Yujie Tang
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Centre, Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - William A Weiss
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
11
|
Agliano A, Balarajah G, Ciobota DM, Sidhu J, Clarke PA, Jones C, Workman P, Leach MO, Al-Saffar NMS. Pediatric and adult glioblastoma radiosensitization induced by PI3K/mTOR inhibition causes early metabolic alterations detected by nuclear magnetic resonance spectroscopy. Oncotarget 2017; 8:47969-47983. [PMID: 28624789 PMCID: PMC5564619 DOI: 10.18632/oncotarget.18206] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 04/29/2017] [Indexed: 11/25/2022] Open
Abstract
Poor outcome for patients with glioblastomas is often associated with radioresistance. PI3K/mTOR pathway deregulation has been correlated with radioresistance; therefore, PI3K/mTOR inhibition could render tumors radiosensitive. In this study, we show that NVP-BEZ235, a dual PI3K/mTOR inhibitor, potentiates the effects of irradiation in both adult and pediatric glioblastoma cell lines, resulting in early metabolic changes detected by nuclear magnetic resonance (NMR) spectroscopy. NVP-BEZ235 radiosensitises cells to X ray exposure, inducing cell death through the inhibition of CDC25A and the activation of p21cip1(CDKN1A). Lactate and phosphocholine levels, increased with radiation, are decreased after NVP-BEZ235 and combination treatment, suggesting that inhibiting the PI3K/mTOR pathway reverses radiation induced metabolic changes. Importantly, NVP-BEZ235 potentiates the effects of irradiation in a xenograft model of adult glioblastoma, where we observed a decrease in lactate and phosphocholine levels after seven days of combination treatment. Although tumor size was not affected due to the short length of the treatment, a significant increase in CASP3 mRNA was observed in the combination group. Taken together, our data suggest that NMR metabolites could be used as biomarkers to detect an early response to combination therapy with PI3K/mTOR inhibitors and radiotherapy in adult and pediatric glioblastoma patients.
Collapse
Affiliation(s)
- Alice Agliano
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Geetha Balarajah
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- The Centre for Molecular Pathology, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Daniela M Ciobota
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Jasmin Sidhu
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Paul A Clarke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Chris Jones
- Divisions of Cancer Therapeutics and Molecular Pathology, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Martin O Leach
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Nada M S Al-Saffar
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
12
|
Al-Saffar NMS, Agliano A, Marshall LV, Jackson LE, Balarajah G, Sidhu J, Clarke PA, Jones C, Workman P, Pearson ADJ, Leach MO. In vitro nuclear magnetic resonance spectroscopy metabolic biomarkers for the combination of temozolomide with PI3K inhibition in paediatric glioblastoma cells. PLoS One 2017; 12:e0180263. [PMID: 28704425 PMCID: PMC5509135 DOI: 10.1371/journal.pone.0180263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 06/13/2017] [Indexed: 11/18/2022] Open
Abstract
Recent experimental data showed that the PI3K pathway contributes to resistance to temozolomide (TMZ) in paediatric glioblastoma and that this effect is reversed by combination treatment of TMZ with a PI3K inhibitor. Our aim is to assess whether this combination results in metabolic changes that are detectable by nuclear magnetic resonance (NMR) spectroscopy, potentially providing metabolic biomarkers for PI3K inhibition and TMZ combination treatment. Using two genetically distinct paediatric glioblastoma cell lines, SF188 and KNS42, in vitro 1H-NMR analysis following treatment with the dual pan-Class I PI3K/mTOR inhibitor PI-103 resulted in a decrease in lactate and phosphocholine (PC) levels (P<0.02) relative to control. In contrast, treatment with TMZ caused an increase in glycerolphosphocholine (GPC) levels (P≤0.05). Combination of PI-103 with TMZ showed metabolic effects of both agents including a decrease in the levels of lactate and PC (P<0.02) while an increase in GPC (P<0.05). We also report a decrease in the protein expression levels of HK2, LDHA and CHKA providing likely mechanisms for the depletion of lactate and PC, respectively. Our results show that our in vitro NMR-detected changes in lactate and choline metabolites may have potential as non-invasive biomarkers for monitoring response to combination of PI3K/mTOR inhibitors with TMZ during clinical trials in children with glioblastoma, subject to further in vivo validation.
Collapse
Affiliation(s)
- Nada M. S. Al-Saffar
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Alice Agliano
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Lynley V. Marshall
- Divisions of Cancer Therapeutics and Molecular Pathology, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - L. Elizabeth Jackson
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Geetha Balarajah
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Jasmin Sidhu
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Paul A. Clarke
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Chris Jones
- Divisions of Cancer Therapeutics and Molecular Pathology, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Andrew D. J. Pearson
- Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Martin O. Leach
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
13
|
Rizzo D, Ruggiero A, Martini M, Rizzo V, Maurizi P, Riccardi R. Molecular Biology in Pediatric High-Grade Glioma: Impact on Prognosis and Treatment. BIOMED RESEARCH INTERNATIONAL 2015; 2015:215135. [PMID: 26448930 PMCID: PMC4584033 DOI: 10.1155/2015/215135] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/04/2014] [Indexed: 12/17/2022]
Abstract
High-grade gliomas are the main cause of death in children with brain tumours. Despite recent advances in cancer therapy, their prognosis remains poor and the treatment is still challenging. To date, surgery followed by radiotherapy and temozolomide is the standard therapy. However, increasing knowledge of glioma biology is starting to impact drug development towards targeted therapies. The identification of agents directed against molecular targets aims at going beyond the traditional therapeutic approach in order to develop a personalized therapy and improve the outcome of pediatric high-grade gliomas. In this paper, we critically review the literature regarding the genetic abnormalities implicated in the pathogenesis of pediatric malignant gliomas and the current development of molecularly targeted therapies. In particular, we analyse the impact of molecular biology on the prognosis and treatment of pediatric high-grade glioma, comparing it to that of adult gliomas.
Collapse
Affiliation(s)
- Daniela Rizzo
- Department of Pediatric Oncology, A. Gemelli Hospital, Catholic University of Rome, Largo A Gemelli, 1, 00168 Rome, Italy
| | - Antonio Ruggiero
- Department of Pediatric Oncology, A. Gemelli Hospital, Catholic University of Rome, Largo A Gemelli, 1, 00168 Rome, Italy
| | - Maurizio Martini
- Anatomic Pathology, Catholic University, “A. Gemelli” Hospital, 00168 Rome, Italy
| | - Valentina Rizzo
- Department of Pediatric Oncology, A. Gemelli Hospital, Catholic University of Rome, Largo A Gemelli, 1, 00168 Rome, Italy
| | - Palma Maurizi
- Department of Pediatric Oncology, A. Gemelli Hospital, Catholic University of Rome, Largo A Gemelli, 1, 00168 Rome, Italy
| | - Riccardo Riccardi
- Department of Pediatric Oncology, A. Gemelli Hospital, Catholic University of Rome, Largo A Gemelli, 1, 00168 Rome, Italy
| |
Collapse
|
14
|
Rizzo D, Scalzone M, Ruggiero A, Maurizi P, Attinà G, Mastrangelo S, Lazzareschi I, Ridola V, Colosimo C, Caldarelli M, Balducci M, Riccardi R. Temozolomide in the treatment of newly diagnosed diffuse brainstem glioma in children: a broken promise? J Chemother 2015; 27:106-110. [PMID: 25466729 DOI: 10.1179/1973947814y.0000000228] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
BACKGROUND The purpose of this study was to assess the efficacy and toxicity of radiotherapy (RT) with concurrent temozolomide (TMZ) chemotherapy followed by adjuvant TMZ in children with diffuse intrinsic pontine glioma (DIPG). METHODS Patients younger than 18 years with newly diagnosed DIPG were enrolled. Children were treated with focal RT along with concurrent daily TMZ. Four weeks after completing the initial RT-TMZ schedule, adjuvant TMZ was given every 28 days up to 12 cycles or progression disease. RESULTS Fifteen children with a median age of 9 years were enrolled. Fourteenth out of the 15 patients completed the chemoradiotherapy. The toxicity associated with TMZ was primarily haematopoietic. At a median follow-up of 15 months 13 children had died and 2 children were alive with progressive disease. No patient experienced complete response (CR). The median time to progression was 7.15 months. CONCLUSION Chemoradiotherapy with TMZ followed by adjuvant TMZ did not improve the poor prognosis associated with DIPG in children.
Collapse
|
15
|
Agnihotri S, Burrell K, Buczkowicz P, Remke M, Golbourn B, Chornenkyy Y, Gajadhar A, Fernandez NA, Clarke ID, Barszczyk MS, Pajovic S, Ternamian C, Head R, Sabha N, Sobol RW, Taylor MD, Rutka JT, Jones C, Dirks PB, Zadeh G, Hawkins C. ATM regulates 3-methylpurine-DNA glycosylase and promotes therapeutic resistance to alkylating agents. Cancer Discov 2014; 4:1198-213. [PMID: 25100205 DOI: 10.1158/2159-8290.cd-14-0157] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
UNLABELLED Alkylating agents are a first-line therapy for the treatment of several aggressive cancers, including pediatric glioblastoma, a lethal tumor in children. Unfortunately, many tumors are resistant to this therapy. We sought to identify ways of sensitizing tumor cells to alkylating agents while leaving normal cells unharmed, increasing therapeutic response while minimizing toxicity. Using an siRNA screen targeting over 240 DNA damage response genes, we identified novel sensitizers to alkylating agents. In particular, the base excision repair (BER) pathway, including 3-methylpurine-DNA glycosylase (MPG), as well as ataxia telangiectasia mutated (ATM), were identified in our screen. Interestingly, we identified MPG as a direct novel substrate of ATM. ATM-mediated phosphorylation of MPG was required for enhanced MPG function. Importantly, combined inhibition or loss of MPG and ATM resulted in increased alkylating agent-induced cytotoxicity in vitro and prolonged survival in vivo. The discovery of the ATM-MPG axis will lead to improved treatment of alkylating agent-resistant tumors. SIGNIFICANCE Inhibition of ATM and MPG-mediated BER cooperate to sensitize tumor cells to alkylating agents, impairing tumor growth in vitro and in vivo with no toxicity to normal cells, providing an ideal therapeutic window.
Collapse
Affiliation(s)
- Sameer Agnihotri
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Kelly Burrell
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Pawel Buczkowicz
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Marc Remke
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Brian Golbourn
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Yevgen Chornenkyy
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Aaron Gajadhar
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Nestor A Fernandez
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Ian D Clarke
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Mark S Barszczyk
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Sanja Pajovic
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Christian Ternamian
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Renee Head
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Nesrin Sabha
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Robert W Sobol
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, Pennsylvania. Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Michael D Taylor
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - James T Rutka
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Chris Jones
- The Institute of Cancer Research, London, United Kingdom
| | - Peter B Dirks
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Gelareh Zadeh
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada. Division of Neurosurgery, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | - Cynthia Hawkins
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada. Division of Pathology, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| |
Collapse
|
16
|
Al-Saffar NMS, Marshall LV, Jackson LE, Balarajah G, Eykyn TR, Agliano A, Clarke PA, Jones C, Workman P, Pearson ADJ, Leach MO. Lactate and choline metabolites detected in vitro by nuclear magnetic resonance spectroscopy are potential metabolic biomarkers for PI3K inhibition in pediatric glioblastoma. PLoS One 2014; 9:e103835. [PMID: 25084455 PMCID: PMC4118961 DOI: 10.1371/journal.pone.0103835] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 07/02/2014] [Indexed: 01/09/2023] Open
Abstract
The phosphoinositide 3-kinase (PI3K) pathway is believed to be of key importance in pediatric glioblastoma. Novel inhibitors of the PI3K pathway are being developed and are entering clinical trials. Our aim is to identify potential non-invasive biomarkers of PI3K signaling pathway inhibition in pediatric glioblastoma using in vitro nuclear magnetic resonance (NMR) spectroscopy, to aid identification of target inhibition and therapeutic response in early phase clinical trials of PI3K inhibitors in childhood cancer. Treatment of SF188 and KNS42 human pediatric glioblastoma cell lines with the dual pan-Class I PI3K/mTOR inhibitor PI-103, inhibited the PI3K signaling pathway and resulted in a decrease in phosphocholine (PC), total choline (tCho) and lactate levels (p<0.02) as detected by phosphorus (31P)- and proton (1H)-NMR. Similar changes were also detected using the pan-Class I PI3K inhibitor GDC-0941 which lacks significant mTOR activity and is entering Phase II clinical trials. In contrast, the DNA damaging agent temozolomide (TMZ), which is used as current frontline therapy in the treatment of glioblastoma postoperatively (in combination with radiotherapy), increased PC, glycerophosphocholine (GPC) and tCho levels (p<0.04). PI-103-induced NMR changes were associated with alterations in protein expression levels of regulatory enzymes involved in glucose and choline metabolism including GLUT1, HK2, LDHA and CHKA. Our results show that by using NMR we can detect distinct biomarkers following PI3K pathway inhibition compared to treatment with the DNA-damaging anti-cancer agent TMZ. This is the first study reporting that lactate and choline metabolites are potential non-invasive biomarkers for monitoring response to PI3K pathway inhibitors in pediatric glioblastoma.
Collapse
Affiliation(s)
- Nada M. S. Al-Saffar
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Lynley V. Marshall
- Division of Molecular Pathology, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies. The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - L. Elizabeth Jackson
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Geetha Balarajah
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Thomas R. Eykyn
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, St Thomas’ Hospital, London, United Kingdom
| | - Alice Agliano
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Paul A. Clarke
- Division of Cancer Therapeutics, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Chris Jones
- Division of Molecular Pathology, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Paul Workman
- Division of Cancer Therapeutics, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Andrew D. J. Pearson
- Division of Cancer Therapeutics, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies. The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Martin O. Leach
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
17
|
Cefalo G, Massimino M, Ruggiero A, Barone G, Ridola V, Spreafico F, Potepan P, Abate ME, Mascarin M, Garrè ML, Perilongo G, Madon E, Colosimo C, Riccardi R. Temozolomide is an active agent in children with recurrent medulloblastoma/primitive neuroectodermal tumor: an Italian multi-institutional phase II trial. Neuro Oncol 2014; 16:748-753. [PMID: 24482446 PMCID: PMC3984557 DOI: 10.1093/neuonc/not320] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 12/18/2013] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The aim of this study was to assess the objective response rate (ORR) of children and young adults with recurrent medulloblastoma/primitive neuroectodermal tumor (MB/PNET) treated with temozolomide (TMZ). The secondary purpose was to analyze the toxicity profile of TMZ when administered orally for 5 days in 3 divided daily doses every 28 days. METHODS Forty-two patients with recurrent MB/PNET, aged 21 years and younger, were recruited. Patients were treated with oral TMZ. Starting doses ranged from 120 to 200 mg/m(2)/day based on previous treatments. A craniospinal MRI was performed prior to the first cycle of TMZ and following every 2 cycles of treatment. RESULTS Median age was 10 years (range, 2-21 years). Forty of 42 patients were assessed for response and toxicity. The objective response rate was 42.5%: 6 patients achieved a complete response, 11 had a partial response, and 10 had stable disease. Progression-free survival rates for all patients at 6 and 12 months were 30% and 7.5%, respectively. Their median overall survival rates at 6 and 12 months were 42.5% and 17.5%, respectively. No major extrahematological effects or life-threatening events were reported. The most common grade 3/4 toxicity included thrombocytopenia (17.5%), neutropenia (7.5%), and anemia (2.5%). CONCLUSIONS TMZ proved to be an effective agent in children and young adults with MB/PNET, heavily pre-treated, with a tolerable toxicity profile.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Riccardo Riccardi
- Pediatrics Unit, Fondazione IRCCS, Istituto Nazionale Tumori, Milan, Italy, (G.C., M.M., F.S.); Pediatric Oncology, A. Gemelli Hospital, Catholic University, Rome, Italy, (A.R., G.B., V.R., R.R.); Department of Radiodiagnostics, Fondazione IRCCS, Istituto Nazionale Tumori, Milan, Italy, (P.P.); Department of Chemotherapy, Istituto Ortopedico Rizzoli, Bologna, Italy, (M.E.A.); Department of Radiotherapy Unit, CRO, Aviano, Italy, (M.M.); Department of Neurosurgery, Ospedale G. Gaslini, Genova, Italy, (M.L.G.); Department of Pediatrics, University of Padua, Padua, Italy, (G.P.); Department of Pediatrics, Regina Margherita Children's Hospital, University of Turin, Turin, Italy, (E.M.); Department of Bioimaging and Radiological Sciences, Catholic University, Rome, Italy, (C.C.)
| |
Collapse
|
18
|
Ruggiero A, Rizzo D, Attinà G, Lazzareschi I, Maurizi P, Ridola V, Mastrangelo S, Migliorati R, Bertolini P, Colosimo C, Riccardi R. Phase I study of temozolomide combined with oral etoposide in children with malignant glial tumors. J Neurooncol 2013; 113:513-518. [PMID: 23666235 DOI: 10.1007/s11060-013-1145-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 04/28/2013] [Indexed: 11/29/2022]
Abstract
The treatment of children with malignant glioma remains challenging. The aim of this multicenter phase I study is to establish the recommended dose (RD) of the combination therapy with temozolomide (TMZ) and oral etoposide (VP-16) in children with relapsed or refractory malignant glioma and brainstem glioma at diagnosis. A phase I trial was conducted to establish the maximum tolerated dose (MTD) of TMZ and oral VP-16. This orally administered combination was investigated by a classical 3 + 3 design. Cohorts of patients were enrolled at 4 different levels: (1) TMZ 120 mg/m(2) on days 1-5 and VP-16 50 mg/m(2) on days 1-8; (2) TMZ 150 mg/m(2) on days 1-5 and VP-16 50 mg/m(2) on days 1-8; (3) TMZ 150 mg/m(2) on days 1-5 and VP-16 50 mg/m(2) on days 1-10; (4) TMZ 150 mg/m(2) on days 1-5 and VP-16 50 mg/m(2) on days 1-12. Therapy was administered in 28-day courses. A total of 118 courses were administered to 18 patients with a median age of 11.2 years. At dose level 1, none displayed toxicity. Of the 6 patients at dose level 2, 1 patient had dose limiting toxicity (DLT). None of the 3 patients at dose level 3 had DLT. At dose level 4, grade III/IV thrombocytopenia and neutropenia were observed in 2 out of the 6 patients enrolled. Therefore, the MTD was established at dose level 3. The RD for phase II trial in children with malignant glial is TMZ 150 mg/m(2) for 5 days and VP-16 50 mg/m(2) for 10 days every 28 days.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Division, Department of Pediatric Oncology, A Gemelli Hospital, Catholic University of Rome, Largo A Gemelli 1, 00168 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lee PY, Chen CL, Lin ZZ, Cheng AL, Chen EIT, Whang-Peng J, Huang CYF. The Aurora kinases inhibitor VE-465 is a novel treatment for glioblastoma multiforme. Oncology 2013; 84:326-35. [PMID: 23636005 DOI: 10.1159/000347021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 01/02/2013] [Indexed: 11/19/2022]
Abstract
Glioblastoma multiforme (GBM) is one of the most common and aggressive types of primary brain tumor. After complete surgical resection combined with radiation and chemotherapy, approximately 10% of patients survive for more than 5 years. Therefore, a novel therapy for GBM is needed. Aurora-A (AURKA) plays important roles in cell cycle regulation, such as centrosome maturation, chromatic separation, bipolar spindle assembly, and mitotic entry. To investigate the effects of AURKA inhibition, three GBM cell lines, including GBM 8401, GBM 8901, and U87-MG cells, were treated with the AURKA inhibitor VE-465. Sensitivities to VE-465, as indicated by 50% inhibitory concentration values for GBM 8401, GBM 8901, and U87-MG cells, were 6, 25, and 19 nM, respectively. Additionally, colony formation of GBM 8401 and GBM 8901 cells was decreased after treatment with the VE-465. VE-465 treatment increased polyploidy and p53 protein expression, and inhibited cell growth in a caspase-independent manner. Taken together, these results suggest that the inhibition of AURKA by a small-molecule inhibitor may have potential to serve as a novel therapeutic approach for GBM.
Collapse
Affiliation(s)
- Pei-Ying Lee
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, ROC. lpyalexandra @ gmail.com
| | | | | | | | | | | | | |
Collapse
|
20
|
A phase II single-arm study of irinotecan in combination with temozolomide (TEMIRI) in children with newly diagnosed high grade glioma: a joint ITCC and SIOPE-brain tumour study. J Neurooncol 2013; 113:127-34. [DOI: 10.1007/s11060-013-1098-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 02/22/2013] [Indexed: 10/27/2022]
|
21
|
Jung TY, Kim CY, Kim DS, Ra YS, Kim SH, Baek HJ, Choi HS, Kim IA. Prognosis of pediatric high-grade gliomas with temozolomide treatment: a retrospective, multicenter study. Childs Nerv Syst 2012; 28:1033-9. [PMID: 22576807 DOI: 10.1007/s00381-012-1786-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 04/25/2012] [Indexed: 12/22/2022]
Abstract
PURPOSE We analyzed the usefulness of initial or recurrent treatment of temozolomide (TMZ) in pediatric high-grade gliomas (HGGs). METHODS Between 2002 and 2010, we performed surgery on 35 patients with 17 glioblastomas, 14 anaplastic astrocytomas, 3 anaplastic oligodendrogliomas, and 1 anaplastic oligoastrocytoma. The male-to-female ratio was 21:14, and the median age was 13 years (range, 3-18 years). The mean follow-up period was 15.9 (± 1.8) months. As the TMZ treatment, 22 patients received the initial treatment and 13 patients at recurrence. We analyzed the prognostic significance of TMZ treatment, tumor location, extent of removal, pathology, and recurrence pattern. RESULTS The median progression-free survival (PFS) and overall survival (OS) were 9.7 (± 1.4) and 17.8 (± 2.5) months, respectively. Based on univariate analysis, the median PFS was 9.9 (± 1.6) months in the tumors located in the cerebral hemisphere and 5.6 (± 1.3) months in the diencephalon (p = 0.03). Median PFS was 12.5 (± 1.7) months in the initial treatment and 6.8 (±0.8) months in the recurrent treatment (p = 0.03). The median OS was 14.9 (± 2.3) months in glioblastomas and 24.4 (± 4.1) months in tumors with an anaplastic pathology (p = 0.01). The median OS was 12.1 (± 3.7) months in patients with cerebrospinal fluid (CSF) dissemination and 18.2 (± 2.9) months in patients without CSF dissemination (p = 0.02). Grades 3 and 4 treatment-related toxicity occurred in 7.7-9 % of the patients. CONCLUSIONS Initial or recurrent TMZ treatment in pediatric HGGs was safe and tolerable. Initial treatment showed improved PFS compared to recurrent treatment, and both showed similar OS.
Collapse
Affiliation(s)
- Tae-Young Jung
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Gwangju, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Jones C, Perryman L, Hargrave D. Paediatric and adult malignant glioma: close relatives or distant cousins? Nat Rev Clin Oncol 2012; 9:400-13. [PMID: 22641364 DOI: 10.1038/nrclinonc.2012.87] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gliomas in children differ from their adult counterparts by their distribution of histological grade, site of presentation and rate of malignant transformation. Although rare in the paediatric population, patients with high-grade gliomas have, for the most part, a comparably dismal clinical outcome to older patients with morphologically similar lesions. Molecular profiling data have begun to reveal the major genetic alterations underpinning these malignant tumours in children. Indeed, the accumulation of large datasets on adult high-grade glioma has revealed key biological differences between the adult and paediatric disease. Furthermore, subclassifications within the childhood age group can be made depending on age at diagnosis and tumour site. However, challenges remain on how to reconcile clinical data from adult patients to tailor novel treatment strategies specifically for paediatric patients.
Collapse
Affiliation(s)
- Chris Jones
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK
| | | | | |
Collapse
|
23
|
Abdelwahab MG, Fenton KE, Preul MC, Rho JM, Lynch A, Stafford P, Scheck AC. The ketogenic diet is an effective adjuvant to radiation therapy for the treatment of malignant glioma. PLoS One 2012; 7:e36197. [PMID: 22563484 PMCID: PMC3341352 DOI: 10.1371/journal.pone.0036197] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 04/03/2012] [Indexed: 01/23/2023] Open
Abstract
Introduction The ketogenic diet (KD) is a high-fat, low-carbohydrate diet that alters metabolism by increasing the level of ketone bodies in the blood. KetoCal® (KC) is a nutritionally complete, commercially available 4∶1 (fat∶ carbohydrate+protein) ketogenic formula that is an effective non-pharmacologic treatment for the management of refractory pediatric epilepsy. Diet-induced ketosis causes changes to brain homeostasis that have potential for the treatment of other neurological diseases such as malignant gliomas. Methods We used an intracranial bioluminescent mouse model of malignant glioma. Following implantation animals were maintained on standard diet (SD) or KC. The mice received 2×4 Gy of whole brain radiation and tumor growth was followed by in vivo imaging. Results Animals fed KC had elevated levels of β-hydroxybutyrate (p = 0.0173) and an increased median survival of approximately 5 days relative to animals maintained on SD. KC plus radiation treatment were more than additive, and in 9 of 11 irradiated animals maintained on KC the bioluminescent signal from the tumor cells diminished below the level of detection (p<0.0001). Animals were switched to SD 101 days after implantation and no signs of tumor recurrence were seen for over 200 days. Conclusions KC significantly enhances the anti-tumor effect of radiation. This suggests that cellular metabolic alterations induced through KC may be useful as an adjuvant to the current standard of care for the treatment of human malignant gliomas.
Collapse
Affiliation(s)
- Mohammed G. Abdelwahab
- Neuro-Oncology Research, Barrow Neurological Institute® of St. Joseph's Hospital and Medical Center, Phoenix, Arizona, United States of America
| | - Kathryn E. Fenton
- Neuro-Oncology Research, Barrow Neurological Institute® of St. Joseph's Hospital and Medical Center, Phoenix, Arizona, United States of America
| | - Mark C. Preul
- Neurosurgery Research, Barrow Neurological Institute® of St. Joseph's Hospital and Medical Center, Phoenix, Arizona, United States of America
| | - Jong M. Rho
- Pediatric Epilepsy Research, Barrow Neurological Institute® of St. Joseph's Hospital and Medical Center, Phoenix, Arizona, United States of America
| | - Andrew Lynch
- Nutricia Advanced Medical Nutrition, Danone Research, Centre for Specialised Nutrition, Liverpool, United Kingdom
| | - Phillip Stafford
- AZ Biodesign, Center for Innovations in Medicine, Arizona State University School of Life Sciences, Tempe, Arizona, United States of America
| | - Adrienne C. Scheck
- Neuro-Oncology Research, Barrow Neurological Institute® of St. Joseph's Hospital and Medical Center, Phoenix, Arizona, United States of America
- Neurosurgery Research, Barrow Neurological Institute® of St. Joseph's Hospital and Medical Center, Phoenix, Arizona, United States of America
- * E-mail:
| |
Collapse
|
24
|
Akyüz C, Demir HA, Varan A, Yalçin B, Kutluk T, Büyükpamukçu M. Temozolomide in relapsed pediatric brain tumors: 14 cases from a single center. Childs Nerv Syst 2012; 28:111-5. [PMID: 21866330 DOI: 10.1007/s00381-011-1561-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 08/10/2011] [Indexed: 11/25/2022]
Abstract
AIM Temozolomide is an active drug against gliomas in adults. It also has some promising effects in pediatric patients with brain tumors. We have conducted a retrospective study to investigate the effectiveness of temozolomide in patients with relapsed brain tumors. PATIENTS AND METHODS The files of 14 children treated at our hospital between 2005 and 2010 with the diagnoses of relapsed brain tumors were reviewed for pathological characteristics, treatment results, and outcomes. RESULTS The median age at relapse was 8 years (range, 1.08-23; F/M, 5/9). Diagnoses included medulloblastoma (n = 5), atypical teratoid rhabdoid tumor (n = 2), ependymoma (n = 2), glioneuronal tumor (n = 1), malignant neoplasm (n = 1), pontine glioma (n = 1), astrocytoma grade III (n = 1), and glioblastoma multiforme (n = 1). All patients except the one with pontine glioma had undergone surgical resection, and all had prior adjuvant chemotherapy. Twelve out of 14 patients had received radiotherapy. The median number of temozolomide courses was 5.0 (range, 1-24). Objective response rate in our patients was 35.7% (three complete responses, one partial response, and one minor response). Stable disease achieved in 14.3% of patients and 50% had progressive disease. Median survival time was 8 months (range, 1-55). At the end of the study, three patients were alive. Hematological toxicity was seen in 30.8% of all courses. CONCLUSIONS Relapsed brain tumors in childhood have an unfavorable prognosis. These data suggest that temozolomide might be an active agent against recurrent medulloblastoma. Although overall objective response rate was low, further multicentric studies with temozolomide may be warranted in children with recurrent brain tumors.
Collapse
Affiliation(s)
- Canan Akyüz
- Institute of Oncology, Department of Pediatric Oncology, Hacettepe University, 06100 Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
25
|
Lee JY, Park CK, Park SH, Wang KC, Cho BK, Kim SK. MGMT promoter gene methylation in pediatric glioblastoma: analysis using MS-MLPA. Childs Nerv Syst 2011; 27:1877-83. [PMID: 21789683 DOI: 10.1007/s00381-011-1525-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 07/06/2011] [Indexed: 01/11/2023]
Abstract
PURPOSE Promoter methylation of the O⁶-methylguanine-DNA-methyltransferase (MGMT) gene is widely recognized as an important predictive factor in the treatment of glioblastoma (GBM) patients with temozolomide. However, data regarding the methylation status of the MGMT promoter in pediatric GBM are yet to be elucidated. METHODS Nineteen tissue samples of pediatric GBM were evaluated for the MGMT promoter methylation status using methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA). Methylation status was also evaluated using methylation-specific polymerase chain reaction (MSP) for 17 of the 19 patients. The correlation between MGMT promoter methylation and clinical outcome was assessed. RESULTS Three of the 19 patients (16%) showed methylation of the MGMT promoter, according to MS-MLPA, as did 1 of the 17 (6%), according to MSP. The methylation status did not seem to have a definite effect on clinical outcome. CONCLUSIONS Pediatric GBMs rarely have methylated MGMT promoters. With a better clinical outcome and lower methylation rate than their adult counterparts, it may be suggested that, for pediatric GBM, MGMT promoter methylation does not play a significant role as a prognostic factor.
Collapse
Affiliation(s)
- Ji Yeoun Lee
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, 110-744, Republic of Korea
| | | | | | | | | | | |
Collapse
|
26
|
Christmann M, Verbeek B, Roos WP, Kaina B. O(6)-Methylguanine-DNA methyltransferase (MGMT) in normal tissues and tumors: enzyme activity, promoter methylation and immunohistochemistry. Biochim Biophys Acta Rev Cancer 2011; 1816:179-90. [PMID: 21745538 DOI: 10.1016/j.bbcan.2011.06.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/22/2011] [Accepted: 06/24/2011] [Indexed: 12/29/2022]
Abstract
O(6)-Methylguanine-DNA methyltransferase (MGMT) is a suicide enzyme that repairs the pre-mutagenic, pre-carcinogenic and pre-toxic DNA damage O(6)-methylguanine. It also repairs larger adducts on the O(6)-position of guanine, such as O(6)-[4-oxo-4-(3-pyridyl)butyl]guanine and O(6)-chloroethylguanine. These adducts are formed in response to alkylating environmental pollutants, tobacco-specific carcinogens and methylating (procarbazine, dacarbazine, streptozotocine, and temozolomide) as well as chloroethylating (lomustine, nimustine, carmustine, and fotemustine) anticancer drugs. MGMT is therefore a key node in the defense against commonly found carcinogens, and a marker of resistance of normal and cancer cells exposed to alkylating therapeutics. MGMT also likely protects against therapy-related tumor formation caused by these highly mutagenic drugs. Since the amount of MGMT determines the level of repair of toxic DNA alkylation adducts, the MGMT expression level provides important information as to cancer susceptibility and the success of therapy. In this article, we describe the methods employed for detecting MGMT and review the literature with special focus on MGMT activity in normal and neoplastic tissues. The available data show that the expression of MGMT varies greatly in normal tissues and in some cases this has been related to cancer predisposition. MGMT silencing in tumors is mainly regulated epigenetically and in brain tumors this correlates with a better therapeutic response. Conversely, up-regulation of MGMT during cancer treatment limits the therapeutic response. In malignant melanoma, MGMT is not related to the therapeutic response, which is due to other mechanisms of inherent drug resistance. For most cancers, studies that relate MGMT activity to therapeutic outcome following O(6)-alkylating drugs are still lacking.
Collapse
Affiliation(s)
- Markus Christmann
- Insitute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | | | | | | |
Collapse
|
27
|
Ridola V, Barone G, Lazzareschi I, Ruggiero A, Rizzo D, Riccardi R. Feasibility study of 21-day-on/7-day-off temozolomide in children with brain tumors. J Neurooncol 2011; 103:147-153. [PMID: 20811928 DOI: 10.1007/s11060-010-0371-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 08/18/2010] [Indexed: 11/30/2022]
Abstract
Temozolomide (TMZ) is an oral alkylating agent with proven antitumoral activity in preclinical and clinical studies in adults with high-grade glioma (HGG). However, only limited efficacy has been reported in children with HGG using the 5-day schedule. This study investigated the safety of administering TMZ to children and adolescents with brain tumors over an extended period. Extended schedules have been proven to overcome chemoresistance without any major toxicity. The toxicity of TMZ, administered at 70 mg/m(2)/day orally for 21 consecutive days every 28 days, was assessed in children with brain tumors. A total of 156 courses of TMZ were given to 17 patients (median age 12.5 years, range 1-17 years), who were recruited into the study. Eleven patients had progressive or relapsing disease, and six patients were newly diagnosed. In this cohort no cases of toxic death or nonhematological toxicity were reported. In comparison with the 5-day schedule, thrombocytopenia and neutropenia were noted to be less frequent. Grades 3 and 4 lymphopenia occurred in 10.8 and 22.4% of courses, respectively; among the lymphopenic patients there was one case of disseminated zoster (meningoencephalitis and cutaneous involvement), one case of rotavirus gastroenteritis, and two cases of herpetic stomatitis reported. The objective response rate was 11.8%. Overall, 82.3% of patients showed stable disease. The prolonged TMZ schedule appeared to be well tolerated, with few cases of neutropenia or thrombocytopenia recorded. Nevertheless, prolonged exposure to TMZ was associated with lymphopenia and may lead to a higher rate of viral infections.
Collapse
Affiliation(s)
- Vita Ridola
- Department of Pediatric Oncology, A. Gemelli Hospital, Catholic University of Rome, Largo A Gemelli 1, 00168 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
28
|
Bartels U, Baruchel S, Carret A, Crooks B, Hukin J, Johnston D, Silva M, Strother D, Wilson B, Zelcer S, Eisenstat D, Sung L, Bouffet E. The use and effectiveness of temozolomide in children with central nervous system tumours: a survey from the Canadian Paediatric Brain Tumour Consortium. Curr Oncol 2011; 18:e19-24. [PMID: 21331268 PMCID: PMC3031361 DOI: 10.3747/co.v18i1.675] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To describe the use of temozolomide (tmz) in Canadian children treated for brain tumours and to evaluate survival and predictors of survival for children treated with this agent. METHODS A survey was conducted within the Canadian Paediatric Brain Tumour Consortium (cpbtc), a group of tertiary care centres in pediatric neuro-oncology (n = 16) in Canada that are involved in the treatment of children with central nervous system tumours. RESULTS In 10 of the 16 participating pediatric oncology centres of the cpbtc, 137 children with brain tumours were treated with tmz between January 2000 and March 2006. Although 33% of the children were enrolled into a clinical trial, 67% were treated outside open studies. Most patients (72%) received tmz treatment on recurrence of their brain tumour (first or subsequent). The most commonly administered regimen was single-agent tmz 150-200 mg/m(2) administered on 5 consecutive days every 28 days. The median duration of tmz treatment was 141 days (range: 4-1102 days). Response data were provided for 127 of the 137 patients, of whom 6 showed a complete response. Sixteen patients experienced a minor or partial response, 53 had stable disease, and 52 had progressive disease. Of 32 patients alive at last follow-up, 19 had a diagnosis of low-grade glioma. CONCLUSIONS Temozolomide is used in a variety of pediatric brain tumours, often at the time of recurrence. The lack of insight into clear indications for this agent in pediatric brain tumours-used either alone or in combination therapy-may be a result of suboptimal design of phase i and ii studies and a lack of phase iii trials in the pediatric brain tumour population.
Collapse
Affiliation(s)
| | | | | | | | - J. Hukin
- British Columbia’s Children’s Hospital, Vancouver, BC
| | - D. Johnston
- Children’s Hospital of Easter n Ontario, Ottawa, ON
| | - M. Silva
- Kingston General Hospital, Kingston, ON
| | - D. Strother
- University of Calgary, Alberta Hospital, Calgary, AB
| | - B. Wilson
- Stollery Children’s Hospital, Edmonton, AB
| | - S. Zelcer
- Children’s Hospital of Western Ontario, London, ON
| | | | - L. Sung
- Hospital for Sick Children, Toronto, ON
| | | |
Collapse
|
29
|
Gaspar N, Marshall L, Perryman L, Bax DA, Little SE, Viana-Pereira M, Sharp SY, Vassal G, Pearson AD, Reis RM, Hargrave D, Workman P, Jones C. MGMT-independent temozolomide resistance in pediatric glioblastoma cells associated with a PI3-kinase-mediated HOX/stem cell gene signature. Cancer Res 2010; 70:9243-52. [PMID: 20935218 PMCID: PMC3935452 DOI: 10.1158/0008-5472.can-10-1250] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Sensitivity to temozolomide is restricted to a subset of glioblastoma patients, with the major determinant of resistance being a lack of promoter methylation of the gene encoding the repair protein DNA methyltransferase MGMT, although other mechanisms are thought to be active. There are, however, limited preclinical data in model systems derived from pediatric glioma patients. We screened a series of cell lines for temozolomide efficacy in vitro, and investigated the differential mechanisms of resistance involved. In the majority of cell lines, a lack of MGMT promoter methylation and subsequent protein overexpression were linked to temozolomide resistance. An exception was the pediatric glioblastoma line KNS42. Expression profiling data revealed a coordinated upregulation of HOX gene expression in resistant lines, especially KNS42, which was reversed by phosphoinositide 3-kinase pathway inhibition. High levels of HOXA9/HOXA10 gene expression were associated with a shorter survival in pediatric high-grade glioma patient samples. Combination treatment in vitro of pathway inhibition and temozolomide resulted in a highly synergistic interaction in KNS42 cells. The resistance gene signature further included contiguous genes within the 12q13-q14 amplicon, including the Akt enhancer PIKE, significantly overexpressed in the KNS42 line. These cells were also highly enriched for CD133 and other stem cell markers. We have thus shown an in vitro link between phosphoinositide 3-kinase-mediated HOXA9/HOXA10 expression, and a drug-resistant, progenitor cell phenotype in MGMT-independent pediatric glioblastoma.
Collapse
Affiliation(s)
- Nathalie Gaspar
- Paediatric Oncology, The Institute of Cancer Research, Sutton, UK
- Cancer Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
- Pharmacology and New Treatments of Cancer, Institut de Cancérologie Gustav Roussy, Villejuif, France
| | - Lynley Marshall
- Paediatric Oncology, The Institute of Cancer Research, Sutton, UK
- Cancer Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
- Paediatric Oncology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Lara Perryman
- Paediatric Oncology, The Institute of Cancer Research, Sutton, UK
| | - Dorine A. Bax
- Paediatric Oncology, The Institute of Cancer Research, Sutton, UK
| | | | - Marta Viana-Pereira
- Paediatric Oncology, The Institute of Cancer Research, Sutton, UK
- Life and Health Science Research Institute (ICVS), Universidade do Minho, Braga, Portugal
| | - Swee Y. Sharp
- Cancer Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Gilles Vassal
- Pharmacology and New Treatments of Cancer, Institut de Cancérologie Gustav Roussy, Villejuif, France
| | - Andrew D.J. Pearson
- Paediatric Oncology, The Institute of Cancer Research, Sutton, UK
- Paediatric Oncology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Rui M. Reis
- Life and Health Science Research Institute (ICVS), Universidade do Minho, Braga, Portugal
| | - Darren Hargrave
- Paediatric Oncology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Paul Workman
- Cancer Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Chris Jones
- Paediatric Oncology, The Institute of Cancer Research, Sutton, UK
| |
Collapse
|
30
|
Parekh C, Jubran R, Erdreich-Epstein A, Panigrahy A, Bluml S, Finlay J, Dhall G. Treatment of children with recurrent high grade gliomas with a bevacizumab containing regimen. J Neurooncol 2010; 103:673-80. [PMID: 21038110 DOI: 10.1007/s11060-010-0444-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Accepted: 10/21/2010] [Indexed: 01/01/2023]
Abstract
Children with recurrent high grade gliomas (HGG) have a dismal outcome with a median progression free survival (PFS) of 12 weeks. Adults with recurrent HGG treated with irinotecan and bevacizumab reportedly have a 63% response rate and a median PFS of 23 weeks. There is a paucity of corresponding published pediatric data. We retrospectively reviewed the records of patients less than 21 years of age with recurrent or progressive WHO grade 3-4 gliomas who were treated with bevacizumab containing regimens at our institution between January 2006 and September 2008. We identified eight patients. Six out of eight patients received irinotecan, temozolomide and bevacizumab, one patient received irinotecan and bevacizumab, and one patient received CCNU and bevacizumab. Three patients had stable disease for 30-93 weeks. The remaining five patients developed progressive disease within 17 weeks. The median PFS was 15 weeks and the 6-month PFS was 38%. Contrast enhancing disease responded or remained stable in five out of seven patients whereas non-enhancing disease progressed in three out of four patients. New distant non-enhancing lesions developed in three patients. The most common side effects included diarrhea, vomiting, thrombocytopenia and neutropenia. Bevacizumab was well tolerated when used in combination with conventional chemotherapy (irinotecan in most cases). PFS in our cohort was much shorter and the response rate was inferior in this small cohort of patients when compared with published adult data. However, bevacizumab containing regimens might be effective in a subset of pediatric patients, especially those with predominantly contrast-enhancing disease.
Collapse
Affiliation(s)
- Chintan Parekh
- Division of Pediatric Hematology Oncology, Childrens Hospital Los Angeles, 4650 Sunset Boulevard, MS# 54, Los Angeles, CA 90027, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Stafford P, Abdelwahab MG, Kim DY, Preul MC, Rho JM, Scheck AC. The ketogenic diet reverses gene expression patterns and reduces reactive oxygen species levels when used as an adjuvant therapy for glioma. Nutr Metab (Lond) 2010; 7:74. [PMID: 20831808 PMCID: PMC2949862 DOI: 10.1186/1743-7075-7-74] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Accepted: 09/10/2010] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Malignant brain tumors affect people of all ages and are the second leading cause of cancer deaths in children. While current treatments are effective and improve survival, there remains a substantial need for more efficacious therapeutic modalities. The ketogenic diet (KD) - a high-fat, low-carbohydrate treatment for medically refractory epilepsy - has been suggested as an alternative strategy to inhibit tumor growth by altering intrinsic metabolism, especially by inducing glycopenia. METHODS Here, we examined the effects of an experimental KD on a mouse model of glioma, and compared patterns of gene expression in tumors vs. normal brain from animals fed either a KD or a standard diet. RESULTS Animals received intracranial injections of bioluminescent GL261-luc cells and tumor growth was followed in vivo. KD treatment significantly reduced the rate of tumor growth and prolonged survival. Further, the KD reduced reactive oxygen species (ROS) production in tumor cells. Gene expression profiling demonstrated that the KD induces an overall reversion to expression patterns seen in non-tumor specimens. Notably, genes involved in modulating ROS levels and oxidative stress were altered, including those encoding cyclooxygenase 2, glutathione peroxidases 3 and 7, and periredoxin 4. CONCLUSIONS Our data demonstrate that the KD improves survivability in our mouse model of glioma, and suggests that the mechanisms accounting for this protective effect likely involve complex alterations in cellular metabolism beyond simply a reduction in glucose.
Collapse
Affiliation(s)
- Phillip Stafford
- Neuro-Oncology Research, Barrow Neurological Institute7 of St, Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Initial experience involving treatment and retreatment with carmustine wafers in combination with oral temozolomide: long-term survival in a child with relapsed glioblastoma multiforme. J Pediatr Hematol Oncol 2010; 32:e202-6. [PMID: 20523247 DOI: 10.1097/mph.0b013e3181e0d16b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Glioblastomas occur infrequently in children, and the prognosis is better than for glioblastomas seen in adults. Aggressive treatment is justified in pediatric patients. OBSERVATIONS We present the case of a 6-year-old child with malignant posterior temporal glioma treated with surgery, radiotherapy, local chemotherapy with carmustine wafers, and oral therapy with temozolomide, both at initial diagnosis and at relapse 18 months later. After 6 years, the patient seems healthy with no focal neurologic signs, and imaging studies show no evidence of disease. CONCLUSION Multimodal therapy was found to have a very positive outcome for a child with malignant glioma.
Collapse
|
33
|
Affiliation(s)
- Norbert Graf
- Department of Pediatric Oncology and Hematology, University of the Saarland, Homburg, Germany.
| |
Collapse
|
34
|
Kim EK, Chung DS, Shin HJ, Hong YK. Inhibitory Effect of IFN-beta, on the Antitumor Activity of Celecoxib in U87 Glioma Model. J Korean Neurosurg Soc 2009; 46:552-7. [PMID: 20062571 DOI: 10.3340/jkns.2009.46.6.552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 09/04/2009] [Accepted: 11/01/2009] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Interferon-beta, (IFN-beta) has been used in the treatment of cancers. Inhibition of the enzyme cyclooxygenase (COX) with celecoxib had a significantly suppressive effect on tumor growth, angiogenesis, and metastasis in a variety of tumors. The aim of this study was to elucidate the antiglioma effect of combined treatment with IFN-beta and celecoxib in U87 glioma model. METHODS The in vitro effects of IFN-beta (50-1,000 IU/mL) and celecoxib (50-250 microM) alone or combination of both on the proliferation and apoptosis of U87 cells were tested using MTT assay, FACS analysis and DNA condensation. To determine the in vivo effect, nude mice bearing intracerebral U87 xenograft inoculation were treated with IFN-beta intraperitoneally (2x10(5) IU/day for 15 days), celecoxib orally (5, 10 mg/kg) or their combination. RESULTS IFN-beta or celecoxib showed an inhibitory effect on the proliferation of U87 cells. When U87 cells were treated with IFN-beta and celecoxib combination, it seemed that IFN-beta interrupted the antiproliferative and apoptotic activity of celecoxib. No additive effect was observed on the survival of the tumor bearing mice by the combination of IFN-beta and celecoxib. CONCLUSION These results suggest that IFN-beta seems to inhibit the antiglioma effect of celecoxib, therefore combination of IFN-beta and celecoxib may be undesirable in the treatment of glioma.
Collapse
Affiliation(s)
- Eun-Kyoung Kim
- The Catholic University of Korea College of Medicine, Kangnam St. Mary's Hospital, Seoul, Korea
| | | | | | | |
Collapse
|
35
|
Bax DA, Little SE, Gaspar N, Perryman L, Marshall L, Viana-Pereira M, Jones TA, Williams RD, Grigoriadis A, Vassal G, Workman P, Sheer D, Reis RM, Pearson ADJ, Hargrave D, Jones C. Molecular and phenotypic characterisation of paediatric glioma cell lines as models for preclinical drug development. PLoS One 2009; 4:e5209. [PMID: 19365568 PMCID: PMC2666263 DOI: 10.1371/journal.pone.0005209] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 03/19/2009] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Although paediatric high grade gliomas resemble their adult counterparts in many ways, there appear to be distinct clinical and biological differences. One important factor hampering the development of new targeted therapies is the relative lack of cell lines derived from childhood glioma patients, as it is unclear whether the well-established adult lines commonly used are representative of the underlying molecular genetics of childhood tumours. We have carried out a detailed molecular and phenotypic characterisation of a series of paediatric high grade glioma cell lines in comparison to routinely used adult lines. PRINCIPAL FINDINGS All lines proliferate as adherent monolayers and express glial markers. Copy number profiling revealed complex genomes including amplification and deletions of genes known to be pivotal in core glioblastoma signalling pathways. Expression profiling identified 93 differentially expressed genes which were able to distinguish between the adult and paediatric high grade cell lines, including a number of kinases and co-ordinated sets of genes associated with DNA integrity and the immune response. SIGNIFICANCE These data demonstrate that glioma cell lines derived from paediatric patients show key molecular differences to those from adults, some of which are well known, whilst others may provide novel targets for evaluation in primary tumours. We thus provide the rationale and demonstrate the practicability of using paediatric glioma cell lines for preclinical and mechanistic studies.
Collapse
Affiliation(s)
- Dorine A. Bax
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
| | - Suzanne E. Little
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
| | - Nathalie Gaspar
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
- Cancer Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, Sutton, United Kingdom
- Pharmacology and New Treatments of Cancer, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Lara Perryman
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
| | - Lynley Marshall
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
- Cancer Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, Sutton, United Kingdom
- Paediatric Oncology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Marta Viana-Pereira
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
- Life and Health Science Research Institute (ICVS), Universidade do Minho, Braga, Portugal
| | - Tania A. Jones
- Neuroscience Centre, Institute of Cell and Molecular Science, Bart's and The London School of Medicine & Dentistry, London, United Kingdom
| | - Richard D. Williams
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
| | - Anita Grigoriadis
- Breakthrough Breast Cancer Unit, Guy's Hospital, London, United Kingdom
| | - Gilles Vassal
- Pharmacology and New Treatments of Cancer, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Paul Workman
- Pharmacology and New Treatments of Cancer, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Denise Sheer
- Neuroscience Centre, Institute of Cell and Molecular Science, Bart's and The London School of Medicine & Dentistry, London, United Kingdom
| | - Rui M. Reis
- Life and Health Science Research Institute (ICVS), Universidade do Minho, Braga, Portugal
| | - Andrew D. J. Pearson
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
- Paediatric Oncology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Darren Hargrave
- Paediatric Oncology, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Chris Jones
- Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom
| |
Collapse
|
36
|
Broniscer A, Baker SJ, Stewart CF, Merchant TE, Laningham FH, Schaiquevich P, Kocak M, Morris EB, Endersby R, Ellison DW, Gajjar A. Phase I and pharmacokinetic studies of erlotinib administered concurrently with radiotherapy for children, adolescents, and young adults with high-grade glioma. Clin Cancer Res 2009; 15:701-7. [PMID: 19147777 PMCID: PMC2629527 DOI: 10.1158/1078-0432.ccr-08-1923] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To estimate the maximum-tolerated dose (MTD) of erlotinib administered during and after radiotherapy, and to describe the pharmacokinetics of erlotinib and its metabolite OSI-420 in patients between 3 and 25 years with newly diagnosed high-grade glioma who did not require enzyme-inducing anticonvulsants. EXPERIMENTAL DESIGN Five dosage levels (70, 90, 120, 160, and 200 mg/m(2) per day) were planned in this phase I study. Dose-limiting toxicities (DLT) were evaluated during first 8 weeks of therapy. Local radiotherapy (dose between 54 and 59.4 Gy) and erlotinib started preferentially on the same day. Erlotinib was administered once daily for a maximum of 3 years. Pharmacokinetic studies were obtained after first dose and on day 8 of therapy. Mutational analysis of EGFR kinase domain, PIK3CA, and PTEN was done in tumor tissue. RESULTS Median age at diagnosis of 23 patients was 10.7 years (range, 3.7-22.5 years). MTD of erlotinib was 120 mg/m(2) per day. Skin rash and diarrhea were generally well controlled with supportive care. Dose-limiting toxicities were diarrhea (n = 1), increase in serum lipase (n = 1), and rash with pruritus (n = 1). The pharmacokinetic variables of erlotinib and OSI-420 in children were similar to those described in adults. However, there was no relationship between erlotinib dosage and drug exposure. No EGFR kinase domain mutations were observed. Two patients with glioblastoma harbored mutations in PIK3CA (n = 1) or PTEN (n = 1). CONCLUSIONS Although the MTD of erlotinib in children with newly diagnosed high-grade glioma was 120 mg/m(2) per day, pharmacokinetic studies showed wide interpatient variability in drug exposure.
Collapse
Affiliation(s)
- Alberto Broniscer
- Department of Oncology, St. Jude Children's Research Hospital, Mail Stop 260, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Finlay JL, Dhall G, Boyett JM, Dunkel IJ, Gardner SL, Goldman S, Yates AJ, Rosenblum MK, Stanley P, Zimmerman RA, Wallace D, Pollack IF, Packer RJ. Myeloablative chemotherapy with autologous bone marrow rescue in children and adolescents with recurrent malignant astrocytoma: outcome compared with conventional chemotherapy: a report from the Children's Oncology Group. Pediatr Blood Cancer 2008; 51:806-11. [PMID: 18802947 PMCID: PMC2844080 DOI: 10.1002/pbc.21732] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE Children and adolescents with malignant astrocytomas recurring after initial treatment have a dismal prognosis, with only rare patients surviving 1-year beyond recurrence. The purpose of this study was to attempt to improve their survival. METHODS Twenty-seven children and adolescents with malignant astrocytomas [17 glioblastoma multiforme and 10 anaplastic astrocytoma (AA)] following initial tumor progression, received myeloablative chemotherapy followed by autologous marrow rescue with one of three thiotepa and etoposide-based chemotherapy regimens, administered alone (n = 11) or combined with carmustine (n = 5) or carboplatin (n = 11). Time to progression and death following myeloablative chemotherapy for these patients was compared non-randomly with outcome of a contemporaneously treated cohort of similar patients who received only conventional chemotherapy following initial tumor progression. The two cohorts were compared for age, histology, prior therapies, extent of surgical resection at progression, and time from initial diagnosis to progression. RESULTS Five of 27 children (two with glioblastoma multiforme and three with AA) survive event-free from 8.3 to 13.3 years (median of 11.1 years) following myeloablative chemotherapy. Of 56 children with recurrent malignant astrocytoma who received conventional chemotherapy following initial progression, no patient survives. Differences in distributions of survival were not significant when stratified by surgical debulking (P = 0.39). However, for patients who were surgically debulked, the survival distributions are significantly different (P = 0.017). CONCLUSIONS Myeloablative chemotherapy with autologous marrow rescue can produce durable remissions in children and young adults with recurrent malignant gliomas, in the setting of minimal residual tumor burden achieved surgically.
Collapse
Affiliation(s)
- Jonathan L Finlay
- The Children's Center for Cancer & Blood Diseases, Department of Pediatrics, Children's Hospital, Los Angeles, California 90027, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Broniscer A, Gururangan S, MacDonald TJ, Goldman S, Packer RJ, Stewart CF, Wallace D, Danks MK, Friedman HS, Poussaint TY, Kun LE, Boyett JM, Gajjar A. Phase I trial of single-dose temozolomide and continuous administration of o6-benzylguanine in children with brain tumors: a pediatric brain tumor consortium report. Clin Cancer Res 2008; 13:6712-8. [PMID: 18006772 DOI: 10.1158/1078-0432.ccr-07-1016] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To estimate the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of escalating doses of temozolomide combined with O(6)-benzylguanine in patients < or =21 years with recurrent brain tumors. EXPERIMENTAL DESIGN Treatment strata consisted of patients who had previously received no or local radiotherapy (Str1) and patients who had undergone craniospinal radiotherapy or myeloablative chemotherapy (Str2). One-hour i.v. administration of O(6)-benzylguanine at 120 mg/m(2) was followed by 48-h continuous infusion at 30 mg/m(2)/day. Single-dose temozolomide at five dosage levels (267, 355, 472, 628, and 835 mg/m(2)) was given at least 6 h after completion of O(6)-benzylguanine bolus. Treatment was repeated after recovery from toxicities at least 4 weeks apart for a maximum of 12 courses. Dose escalation followed the modified continual reassessment method. Pharmacokinetic analyses of temozolomide and 5-triazeno imidazole carboxamide (MTIC) were done in 28 patients. RESULTS A total of 44 and 26 eligible patients were enrolled on Str1 and Str2, respectively. Median age at study entry in each stratum was 8.6 and 11.3 years, respectively. Predominant diagnoses were high-grade/brainstem glioma in Str1 and medulloblastoma in Str2. Whereas the estimated MTDs of temozolomide for Str1 and Str2 were 562 and 407 mg/m(2), respectively, the doses recommended for phase II investigations are 472 and 355 mg/m(2), respectively. DLTs were predominantly neutropenia and thrombocytopenia. Three patients with gliomas experienced centrally confirmed partial responses to therapy. Four patients completed all planned therapy. Temozolomide and MTIC exposures were statistically associated with temozolomide dosage. CONCLUSIONS The current schedule of temozolomide and O(6)-benzylguanine is safe and showed modest activity against recurrent brain tumors in children.
Collapse
Affiliation(s)
- Alberto Broniscer
- Department of Oncology, St. Jude Children's Research Hospital, 332 North Lauderdale, Memphis, TN 38105, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Temozolomide (Temodal, Temodar), an imidazol derivative, is a second-generation alkylating agent. The orally available prodrug with the capacity of crossing the blood-brain barrier received accelerated US FDA approval in 1999. Three pivotal Phase II trials showed modest activity in the treatment of recurrent anaplastic astrocytoma glioblastoma. In 2005, the FDA and the European Agency for the Evaluation of Medicinal Products approved temozolomide for use in newly diagnosed glioblastoma, in conjunction with radiotherapy, based on an European Organisation for Research and Treatment of Cancer/National Cancer Institute of Canada Phase III trial. The adverse events associated with temozolomide are mild-to-moderate and generally predictable; the most serious are noncumulative and reversible myelosuppression and, in particular, thrombocytopenia, which occurs in less than 5% of patients. Continuous temozolomide administration is associated with profound CD4-selective lymphocytopenia. Molecular studies have suggested that the benefit of temozolomide chemotherapy is restricted to patients whose tumors have a methylated methylguanine methyltransferase gene promotor and are thus unable to repair some of the chemotherapy-induced DNA damage. Temozolomide is under investigation for other disease entities, in particular lower-grade glioma, brain metastases and melanoma.
Collapse
Affiliation(s)
- Nicole Mutter
- Multidisciplinary Oncology Center University of Lausanne Hospitals 46 Rue du Bugnon, 1011 Lausanne, Switzerland.
| | | |
Collapse
|