1
|
Bennebroek CA, Schouten CR, Montauban-van Swijndregt MC, Saeed P, Porro GL, Pott JWR, Dittrich ATM, Oostenbrink R, Schouten-van Meeteren AY, de Jong MC, de Graaf P. Treatment evaluation by volumetric segmentation in pediatric optic pathway glioma: evaluation of the effect of bevacizumab on intra-tumor components. J Neurooncol 2024; 166:79-87. [PMID: 38150061 DOI: 10.1007/s11060-023-04516-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/16/2023] [Indexed: 12/28/2023]
Abstract
PURPOSE Progressive pediatric optic pathway gliomas (OPGs) are treated by diverse systemic antitumor modalities. Refined insights on the course of intra-tumoral components are limited. METHODS We performed an exploratory study on the longitudinal volumetric course of different (intra-)tumor components by manual segmentation of MRI at the start and after 3, 6 and 12 months of bevacizumab (BVZ) treatment. RESULTS Thirty-one patients were treated with BVZ (median 12 months, range: 2-39 months). During treatment the total tumor volume decreased with median 19.9% (range: - 62.3 to + 29.7%; n = 30) within the first 3 months, decreased 19.0% (range: - 68.8 to + 96.1%; n = 28) between start and 6 months and 27.2% (range: -73.4 to + 36.0%; n = 21) between start and 12 months. Intra-tumoral cysts were present in 12 OPGs, all showed a decrease of volume during treatment. The relative contrast enhanced volume of NF1 associated OPG (n = 11) showed an significant reduction compared to OPG with a KIAA1549-BRAF fusion (p < 0.01). Three OPGs progressed during treatment, but were not preceded by an increase of relative contrast enhancement. CONCLUSION Treatment with BVZ of progressive pediatric OPGs leads to a decrease of both total tumor volume and cystic volume for the majority of OPGs with emphasis on the first three months. NF1 and KIAA1549-BRAF fusion related OPGs showed a different (early) treatment effect regarding the tumor enhancing component on MRI, which did not correlate with tumor volume changes. Future research is necessary to further evaluate these findings and its relevance to clinical outcome parameters.
Collapse
Affiliation(s)
- Carlien A Bennebroek
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1100DD, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, The Netherlands.
| | - Christiaan R Schouten
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Peerooz Saeed
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1100DD, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, The Netherlands
| | - Giorgio L Porro
- Department of Ophthalmology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Jan W R Pott
- Department of Ophthalmology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anne T M Dittrich
- Department of Pediatrics, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Rianne Oostenbrink
- ENCORE-NF1 Center, Department of General Pediatrics, Erasmus MC, Rotterdam, The Netherlands
| | | | - Marcus C de Jong
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Bennebroek CAM, van Zwol J, Porro GL, Oostenbrink R, Dittrich ATM, Groot ALW, Pott JW, Janssen EJM, Bauer NJ, van Genderen MM, Saeed P, Lequin MH, de Graaf P, Schouten-van Meeteren AYN. Impact of Bevacizumab on Visual Function, Tumor Size, and Toxicity in Pediatric Progressive Optic Pathway Glioma: A Retrospective Nationwide Multicentre Study. Cancers (Basel) 2022; 14:cancers14246087. [PMID: 36551572 PMCID: PMC9776082 DOI: 10.3390/cancers14246087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUNDS Bevacizumab (BVZ) is used as a subsequent line of treatment for pediatric optic pathway glioma (OPG) in the case of progression. Data on the treatment effect concerning tumor progression and visual function are scarce and nationwide studies are lacking. METHODS We performed a retrospective, nationwide, multicentre cohort study including all pediatric patients with OPG treated with BVZ in the Netherlands (2009-2021). Progression-free survival, change in visual acuity and visual field, MRI-based radiologic response, and toxicity were evaluated. RESULTS In total, 33 pediatric patients with OPG were treated with BVZ (median 12 months). Visual acuity improved in 20.5%, remained stable in 74.4%, and decreased in 5.1% of 39 of all analysed eyes. The monocular visual field improved in 73.1%, remained stable in 15.4%, and decreased in 7.7% of 25 analysed eyes. Radiologic response at the end of therapy showed a partial response in 7 patients (21.9%), minor response in 7 (21.9%), stable disease in 15 (46.9%), and progressive disease in 3 (9.3%). Progression-free survival at 18 and 36 months after the start of BVZ reduced from 70.9% to 38.0%. Toxicity (≥grade 3 CTCAE) during treatment was observed in five patients (15.2%). CONCLUSION Treatment of BVZ in pediatric patients with OPG revealed stabilisation in the majority of patients, but was followed by progression at a later time point in more than 60% of patients. This profile seems relatively acceptable given the benefits of visual field improvement in more than 70% of analysed eyes and visual acuity improvement in more than 20% of eyes at the cessation of BVZ.
Collapse
Affiliation(s)
- Carlien A. M. Bennebroek
- Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, 1053 VE Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, 1081 HV Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-20-5669111
| | - Judith van Zwol
- Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, 1053 VE Amsterdam, The Netherlands
| | - Giorgio L. Porro
- Department of Ophthalmology Utrecht UMC, 3584 CX Utrecht, The Netherlands
| | - Rianne Oostenbrink
- ENCORE-NF1 Center, Department of General Pediatrics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Anne T. M. Dittrich
- Department of Pediatrics, Radboud University Medical Center, Amalia Children’s Hospital, 6525 GA Nijmegen, The Netherlands
| | - Annabel L. W. Groot
- Department of Ophthalmology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jan W. Pott
- Department of Ophthalmology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Etienne J. M. Janssen
- Department of Neurology, Maastricht University Medical Center, 6221 CZ Maastricht, The Netherlands
| | - Noël J. Bauer
- Department of Ophthalmology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Maria M. van Genderen
- Department of Ophthalmology Utrecht UMC, 3584 CX Utrecht, The Netherlands
- Diagnostic Center for Complex Visual Disorders, Bartiméus, 3703 AJ Zeist, The Netherlands
| | - Peerooz Saeed
- Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, 1053 VE Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, 1081 HV Amsterdam, The Netherlands
| | - Maarten H. Lequin
- Department of Radiology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
| | | |
Collapse
|
3
|
de Marcellus C, Tauziède-Espariat A, Cuinet A, Pasqualini C, Robert MP, Beccaria K, Puget S, Boddaert N, Figarella-Branger D, De Carli E, Bourdeaut F, Leblond P, Fouyssac F, Andre N, Bertozzi AI, Butel T, Dufour C, Valteau-Couanet D, Varlet P, Grill J. The role of irinotecan-bevacizumab as rescue regimen in children with low-grade gliomas: a retrospective nationwide study in 72 patients. J Neurooncol 2022; 157:355-364. [PMID: 35239111 DOI: 10.1007/s11060-022-03970-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/15/2022] [Indexed: 10/19/2022]
Abstract
INTRODUCTION At least half of children with low-grade glioma (LGG) treated with first line chemotherapy experience a relapse/progression and may therefore need a second-line chemotherapy. Irinotecan-bevacizumab has been recommended in this setting in France after encouraging results of pilot studies. We performed a retrospective analysis to define the efficacy, toxicity and predictors for response to the combination on a larger cohort. METHODS We reviewed the files from children < 19 years of age with progressive or refractory LGG treated between 2009 and 2016 in 7 French centers with this combination. RESULTS 72 patients (median age 7.8 years [range 1-19]) received a median of 16 courses (range 3-30). The median duration of treatment was 9 months (range 1.4-16.2). 96% of patients experienced at least disease stabilization. The 6-month and 2-year progression-free survivals (PFS) were 91.7% [IC 95% 85.5-98.3] and 38.2% [IC 95% 28.2-51.8] respectively. No progression occurred after treatment in 18 patients with a median follow-up of 35.6 months (range 7.6-75.9 months). Younger patients had a worse PFS (p = 0.005). Prior chemoresistance, NF1 status, duration of treatment, histopathology or radiologic response did not predict response. The most frequent toxicities related to bevacizumab included grades 1-2 proteinuria in 21, epistaxis in 10, fatigue in 12 and hypertension in 8 while gastro-intestinal toxicity was the most frequent side effect related to irinotecan. CONCLUSIONS Bevacizumab-irinotecan has the potential of disease control clinically and radiographically in children with recurrent LGG whatever their previous characteristics; in many cases however these responses are not sustained, especially in younger children.
Collapse
Affiliation(s)
- Charles de Marcellus
- Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, University Paris-Saclay, Villejuif, France.
- Pediatric Intensive Care Unit, Necker-Enfants Malades University Hospital Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France.
| | | | - Aurélie Cuinet
- Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, University Paris-Saclay, Villejuif, France
| | - Claudia Pasqualini
- Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, University Paris-Saclay, Villejuif, France
| | - Matthieu P Robert
- Department of Ophthalmology, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, University Paris Descarte, Paris, France
- Borelli Centre UMR 9010, CNRS-SSA-ENS Paris Saclay-Paris University, Paris, France
| | - Kevin Beccaria
- Department of Pediatric Neurosurgery, Necker-Enfants Malades University Hospital, Assistance Publique- Hôpitaux de Paris, Paris, France
| | - Stéphanie Puget
- Department of Pediatric Neurosurgery, Necker-Enfants Malades University Hospital, Assistance Publique- Hôpitaux de Paris, Paris, France
| | - Nathalie Boddaert
- Paediatric Radiology Department, AP-HP, Hôpital Necker Enfants Malades, Université de Paris, 75015, Paris, France
- Université de Paris, Institut Imagine INSERM U1163, 75015, Paris, France
| | - Dominique Figarella-Branger
- APHM, CHU Timone, Department of Pathology and Neuropathology and Aix-Marseille Univ, CNRS, INP Inst Neurophysiopathol, Marseille, France
| | - Emilie De Carli
- Department of Pediatric Oncology, Angers University Hospital, Angers, France
| | - Franck Bourdeaut
- SIREDO Center (Pediatric, Adolescent and Young Adults Oncology), Institut Curie, University of Paris, Paris, France
| | - Pierre Leblond
- Pediatric Oncology Unit, Oscar Lambret Comprehensive Cancer Center, Lille, France
| | - Fanny Fouyssac
- Department of Pediatric Hematology, Nancy University Hospital, Vandoeuvre-les-Nancy, France
| | - Nicolas Andre
- Department of Pediatric Hematology and Oncology, Aix Marseille Universityy, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Anne I Bertozzi
- Department of Hematology-Oncology, Children University Hospital, Toulouse, France
| | - Thibaut Butel
- Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, University Paris-Saclay, Villejuif, France
| | - Christelle Dufour
- Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, University Paris-Saclay, Villejuif, France
| | - Dominique Valteau-Couanet
- Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, University Paris-Saclay, Villejuif, France
| | - Pascale Varlet
- Department of Neuropathology, GHU Psychiatrie et Neurosciences, site Sainte-Anne, Paris, France
| | - Jacques Grill
- Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, University Paris-Saclay, Villejuif, France
- CNRS Unit 8203, Gustave Roussy, University Paris-Saclay, Villejuif, France
| |
Collapse
|
4
|
Patel JP, Spiller SE, Barker ED. Drug penetration in pediatric brain tumors: Challenges and opportunities. Pediatr Blood Cancer 2021; 68:e28983. [PMID: 33719183 DOI: 10.1002/pbc.28983] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
Larger clinical trial enrollments and a greater understanding of biological heterogeneity have led to improved survival rates for children diagnosed with brain tumors in the last 50 years. However, reducing long-term morbidities and improving survival rates of high-risk tumors remain major challenges. Chemotherapy can reduce tumor burden, but effective drug penetration at the tumor site is limited by barriers in the route of drug administration and within the tumor microenvironment. Bioavailability of drugs is impeded by the blood-brain barrier, plasma protein binding, and structural components by the tumor including the matrix and vasculature contributing to increased interstitial fluid pressure, hypoxia, and acidity. Designing drug delivery systems to circumvent these barriers could lead to improved drug penetration at the tumor site and reduce adverse systemic side effects. In this review, we expand on how systemic and local barriers limit drug penetration and present potential methods to enhance drug penetration in pediatric brain tumors.
Collapse
Affiliation(s)
- Jenny P Patel
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| | - Susan E Spiller
- Pediatric Hematology/Oncology, East Tennessee Children's Hospital, Knoxville, Tennessee
| | - Elizabeth D Barker
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| |
Collapse
|
5
|
Attina G, Maurizi P, Triarico S, Capozza MA, Romano A, Mastrangelo S, Ruggiero A. Management of Children with Optic Gliomas and Neurofibromatosis Type 1. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2020; 13:1601-1606. [DOI: 10.13005/bpj/2035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Optic pathway gliomas (OPG) are a common cancer in children with neurofibromatosis type 1. OPGs can cause clinical symptoms such as reduction of visual acuity, alterations of the visual field, pallor of the optical papilla, strabismus, endocrinological alterations up to diencephalic syndrome.The current guidelines provide for wait and see as the main approach if the tumor is not causing visual deterioration and adopting treatment only in the event of significant impairment of the visual function. Therefore, it is essential to early detect the visual deterioration changes as well as the identification of children eligible for treatment.
Collapse
Affiliation(s)
- Giorgio Attina
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Michele Antonio Capozza
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
6
|
Abstract
Pilocytic astrocytomas (PA) are highly vascular tumors with vascular endothelial growth factor (VEGF)-vascular endothelial growth factor receptor-2 (VEGFR-2) signaling present in the tumor vasculature. PA may, therefore, be responsive to VEGF blockade with bevacizumab (BEV). Data regarding the use of BEV in refractory PA in adults are limited primarily to case reports and case series of patients with recurrent PA. We conducted a single-center, retrospective cohort study from 2009 to 2018. We screened 426 patients with pathologically confirmed PA. We identified 5 adult patients with PA who received BEV at our institution with sufficient clinical follow-up to derive evidence of the efficacy and toxicity. All 5 patients experienced tumor progression after initial therapies which included surgery, radiation, and chemotherapy. Four patients received BEV as monotherapy, whereas 1 received BEV with the continuation of previously initiated alkylating chemotherapy (temozolomide). The average duration of BEV therapy was 10.2 months (range, 1 to 20 mo) with an average follow-up of 47 months (range, 6 to 112 mo). One patient had a severe necrotizing rash in areas of skin contact and discontinued after 1 cycle of BEV. All patients had stabilization per RANO criteria, with 1 patient experiencing progression after 10 months on treatment. One patient had disease progression 5 years after completion of BEV, but the tumor responded to repeat treatment with BEV. Our institution's experience with the use of BEV in recurrent PA is in line with previous reports of therapeutic benefit in recurrent adult PA.
Collapse
|
7
|
Banerjee A, Jakacki RI, Onar-Thomas A, Wu S, Nicolaides T, Young Poussaint T, Fangusaro J, Phillips J, Perry A, Turner D, Prados M, Packer RJ, Qaddoumi I, Gururangan S, Pollack IF, Goldman S, Doyle LA, Stewart CF, Boyett JM, Kun LE, Fouladi M. A phase I trial of the MEK inhibitor selumetinib (AZD6244) in pediatric patients with recurrent or refractory low-grade glioma: a Pediatric Brain Tumor Consortium (PBTC) study. Neuro Oncol 2018; 19:1135-1144. [PMID: 28339824 DOI: 10.1093/neuonc/now282] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Activation of the mitogen-activated protein kinase pathway is important for growth of pediatric low-grade gliomas (LGGs). The aim of this study was to determine the recommended phase II dose (RP2D) and the dose-limiting toxicities (DLTs) of the MEK inhibitor selumetinib in children with progressive LGG. Methods Selumetinib was administered orally starting at 33 mg/m2/dose b.i.d., using the modified continual reassessment method. Pharmacokinetic analysis was performed during the first course. BRAF aberrations in tumor tissue were determined by real-time polymerase chain reaction and fluorescence in situ hybridization. Results Thirty-eight eligible subjects were enrolled. Dose levels 1 and 2 (33 and 43 mg/m2/dose b.i.d.) were excessively toxic. DLTs included grade 3 elevated amylase/lipase (n = 1), headache (n = 1), mucositis (n = 2), and grades 2-3 rash (n = 6). At dose level 0 (25 mg/m2/dose b.i.d, the RP2D), only 3 of 24 subjects experienced DLTs (elevated amylase/lipase, rash, and mucositis). At the R2PD, the median (range) area under the curve (AUC0-∞) and apparent oral clearance of selumetinib were 3855 ng*h/mL (1780 to 7250 ng × h/mL) and 6.5 L × h-1 × m-2 (3.4 to 14.0 L × h-1 × m-2), respectively. Thirteen of 19 tumors had BRAF abnormalities. Among the 5 (20%) of 25 subjects with sustained partial responses, all at the RP2D, 4 had BRAF aberrations, 1 had insufficient tissue. Subjects received a median of 13 cycles (range: 1-26). Fourteen (37%) completed all protocol treatment (26 cycles [n = 13], 13 cycles [n = 1]) with at least stable disease; 2-year progression-free survival at the RP2D was 69 ± SE 9.8%. Conclusion Selumetinib has promising antitumor activity in children with LGG. Rash and mucositis were the most common DLTs.
Collapse
Affiliation(s)
- Anuradha Banerjee
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Regina I Jakacki
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Arzu Onar-Thomas
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Shengjie Wu
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Theodore Nicolaides
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tina Young Poussaint
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jason Fangusaro
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joanna Phillips
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Arie Perry
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - David Turner
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael Prados
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Roger J Packer
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ibrahim Qaddoumi
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sridharan Gururangan
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ian F Pollack
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stewart Goldman
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lawrence A Doyle
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Clinton F Stewart
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - James M Boyett
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Larry E Kun
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Maryam Fouladi
- University of California San Francisco, San Francisco, California; Boston Children's Hospital, Boston, Massachusetts; Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania; St Jude Children's Research Hospital, Memphis, Tennessee; Lurie Children's Hospital, Chicago, Illinois; Children's National Medical Center, Washington, DC; Duke University Medical Center, Durham, North Carolina; Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
8
|
Khatua S, Gutmann DH, Packer RJ. Neurofibromatosis type 1 and optic pathway glioma: Molecular interplay and therapeutic insights. Pediatr Blood Cancer 2018; 65. [PMID: 29049847 DOI: 10.1002/pbc.26838] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/21/2017] [Accepted: 09/02/2017] [Indexed: 12/17/2022]
Abstract
Children with neurofibromatosis type 1 (NF1) are predisposed to develop central nervous system neoplasms, the most common of which are low-grade gliomas (LGGs). The absence of human NF1 associated LGG-derived cell lines, coupled with an inability to generate patient-derived xenograft models, represents barriers to profile molecularly targeted therapies for these tumors. Thus, genetically engineered mouse models have been identified to evaluate the interplay between Nf1-deficient tumor cells and nonneoplastic stromal cells to evaluate potential therapies for these neoplasms. Future treatments might also consider targeting the nonneoplastic cells in NF1-LGGs to reduce tumor growth and neurologic morbidity in affected children.
Collapse
Affiliation(s)
- Soumen Khatua
- Department of Pediatrics, MD Anderson Cancer Center, Houston, Texas
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, District of Columbia
| |
Collapse
|
9
|
Genetic Variations of Kinase Inserts Domain Receptor (KDR) Gene Are Associated with the Risk of Astrocytomas. Mol Neurobiol 2015; 53:2541-9. [PMID: 26081139 DOI: 10.1007/s12035-015-9239-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 05/26/2015] [Indexed: 02/08/2023]
Abstract
Astrocytomas is one of the most common central nervous system (CNS) tumors with high mortality rate. Kinase insert domain receptor (KDR) is involved in the regulation of tumor angiogenesis, migration, and vascular permeability. The aim of the study was to explore the relationship between KDR polymorphisms and risk of astrocytomas. Blood samples were collected from 157 astrocytomas patients and 160 healthy controls. Three tag-SNPs (rs2071559C/T, rs2305948T/C, and rs1870377A/T) were identified from the International HapMap Project Databases and genotyped using the method of polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). We evaluated the astrocytomas risk caused by individual SNPs and haplotype using odds ratios (ORs) and their 95 % confidence intervals (CIs). In the overall individual SNP analysis, the C allele of rs2071559 was correlated with an increased risk of astrocytomas. However, individuals with mutant allele A and genotype TA + AA of rs1870377 showed a protective effect against astrocytomas. Subgroup analysis based on WHO tumor grade revealed that the C allele of rs2071559 had more influence with the risk of astrocytomas in the grade III-IV (OR = 1.91) subgroup than the grade I-II (OR = 1.47) group. Genotype TT of rs2305948 was found to be significantly associated with susceptibility of astrocytomas only in the grade III-IV subgroup. The protective effect of rs1870377 did not reveal significant difference between the grade III-IV and grade I-II subgroups. Meanwhile, stratified analysis demonstrated that mutation of rs2071559 and rs2305948 could elevate the risk of astrocytomas more significantly in the subgroup of smokers than the nonsmokers. Interestingly, the protective effect of rs1870377 was more obvious in the nonsmokers than the smokers. Additionally, haplotype-specific analysis showed that haplotype CCT and CTT were related with an increased risk of astrocytomas. We found that individual with variants of rs2071559*C and rs2305948*T might significantly elevate the risk of astrocytomas, while mutants of rs1870377*A was associated with the decreased risk of astrocytomas. Further studies about ethnically diverse populations with larger sample size should be performed to confirm the correlation between KDR gene polymorphisms and risk of astrocytomas.
Collapse
|
10
|
Identification of Glioblastoma Phosphotyrosine-Containing Proteins with Two-Dimensional Western Blotting and Tandem Mass Spectrometry. BIOMED RESEARCH INTERNATIONAL 2015; 2015:134050. [PMID: 26090378 PMCID: PMC4450212 DOI: 10.1155/2015/134050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/25/2014] [Accepted: 08/25/2014] [Indexed: 12/24/2022]
Abstract
To investigate the presence of, and the potential biological roles of, protein tyrosine phosphorylation in the glioblastoma pathogenesis, two-dimensional gel electrophoresis- (2DGE-) based Western blotting coupled with liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) analysis was used to detect and identify the phosphotyrosine immunoreaction-positive proteins in a glioblastoma tissue. MS/MS and Mascot analyses were used to determine the phosphotyrosine sites of each phosphopeptide. Protein domain and motif analysis and systems pathway analysis were used to determine the protein domains/motifs that contained phosphotyrosine residue and signal pathway networks to clarify the potential biological functions of protein tyrosine phosphorylation. A total of 24 phosphotyrosine-containing proteins were identified. Each phosphotyrosine-containing protein contained at least one tyrosine kinase phosphorylation motif and a certain structural and functional domains. Those phosphotyrosine-containing proteins were involved in the multiple signal pathway systems such as oxidative stress, stress response, and cell migration. Those data show 2DGE-based Western blotting, MS/MS, and bioinformatics are a set of effective approaches to detect and identify glioblastoma tyrosine-phosphorylated proteome and to effectively rationalize the biological roles of tyrosine phosphorylation in the glioblastoma biological systems. It provides novel insights regarding tyrosine phosphorylation and its potential role in the molecular mechanism of a glioblastoma.
Collapse
|
11
|
Sie M, den Dunnen WFA, Lourens HJ, Meeuwsen-de Boer TGJ, Scherpen FJG, Zomerman WW, Kampen KR, Hoving EW, de Bont ESJM. Growth-factor-driven rescue to receptor tyrosine kinase (RTK) inhibitors through Akt and Erk phosphorylation in pediatric low grade astrocytoma and ependymoma. PLoS One 2015; 10:e0122555. [PMID: 25799134 PMCID: PMC4370756 DOI: 10.1371/journal.pone.0122555] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 02/23/2015] [Indexed: 01/20/2023] Open
Abstract
Up to now, several clinical studies have been started investigating the relevance of receptor tyrosine kinase (RTK) inhibitors upon progression free survival in various pediatric brain tumors. However, single targeted kinase inhibition failed, possibly due to tumor resistance mechanisms. The present study will extend our previous observations that vascular endothelial growth factor receptor (VEGFR)-2, platelet derived growth factor receptor (PDGFR)β, Src, the epidermal growth factor receptor (ErbB) family, and hepatocyte growth factor receptor (HGFR/cMet) are potentially drugable targets in pediatric low grade astrocytoma and ependymoma with investigations concerning growth-factor-driven rescue. This was investigated in pediatric low grade astrocytoma and ependymoma cell lines treated with receptor tyrosine kinase (RTK) inhibitors e.g. sorafenib, dasatinib, canertinib and crizotinib. Flow cytometry analyses showed high percentage of cells expressing VEGFR-1, fibroblast growth factor receptor (FGFR)-1, ErbB1/EGFR, HGFR and recepteur d’origine nantais (RON) (respectively 52-77%, 34-51%, 63-90%, 83-98%, 65-95%). Their respective inhibitors induced decrease of cell viability, measured with WST-1 cell viability assays. At least this was partially due to increased apoptotic levels measured by Annexin V/Propidium Iodide apoptosis assays. EGF, HGF and FGF, which are normally expressed in brain (tumor) tissue, showed to be effective rescue inducing growth factors resulting in increased cell survival especially during treatment with dasatinib (complete rescue) or sorafenib (partial rescue). Growth-factor-driven rescue was less prominent when canertinib or crizotinib were used. Rescue was underscored by significantly activating downstream Akt and/or Erk phosphorylation and increased tumor cell migration. Combination treatment showed to be able to overcome the growth-factor-driven rescue. In conclusion, our study highlights the extensive importance of environmentally present growth factors in developing tumor escape towards RTK inhibitors in pediatric low grade astrocytoma and ependymoma. It is of great interest to anticipate upon these results for the design of new therapeutic trials with RTK inhibitors in these pediatric brain tumors.
Collapse
Affiliation(s)
- Mariska Sie
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wilfred F. A. den Dunnen
- Department of Pathology and Medical Biology, Pathology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Harm Jan Lourens
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Tiny G. J. Meeuwsen-de Boer
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Frank J. G. Scherpen
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Walderik W. Zomerman
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kim R. Kampen
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eelco W. Hoving
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eveline S. J. M. de Bont
- Department of Pediatrics, Beatrix Children’s Hospital, Pediatric Oncology/Hematology division, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- * E-mail:
| |
Collapse
|
12
|
Brossier NM, Gutmann DH. Improving outcomes for neurofibromatosis 1-associated brain tumors. Expert Rev Anticancer Ther 2015; 15:415-23. [PMID: 25652347 DOI: 10.1586/14737140.2015.1009043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Children and adults with neurofibromatosis type 1 (NF1) are predisposed to developing CNS tumors, including optic pathway gliomas (OPGs), brainstem gliomas (BSGs) and high-grade gliomas. Although current first-line treatments for low-grade gliomas (OPGs and BSGs) may prevent further tumor growth, they rarely result in restoration of the associated visual or neurological deficits. The availability of accurate small-animal models of NF1-associated brain tumors has established tractable experimental platforms for the discovery and evaluation of promising therapeutic agents. On the basis of these preclinical studies, biologically targeted agents are now being evaluated in children with NF1-associated low-grade brain tumors. Collectively, these models have also begun to reveal potential neuroprotective and risk assessment strategies for this brain tumor-prone population.
Collapse
Affiliation(s)
- Nicole M Brossier
- Department of Pediatrics, St. Louis Children's Hospital, St. Louis, MO, USA
| | | |
Collapse
|
13
|
Sie M, den Dunnen WF, Hoving EW, de Bont ES. Anti-angiogenic therapy in pediatric brain tumors: An effective strategy? Crit Rev Oncol Hematol 2014; 89:418-32. [DOI: 10.1016/j.critrevonc.2013.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/10/2013] [Accepted: 09/27/2013] [Indexed: 12/15/2022] Open
|
14
|
Gururangan S, Fangusaro J, Poussaint TY, McLendon RE, Onar-Thomas A, Wu S, Packer RJ, Banerjee A, Gilbertson RJ, Fahey F, Vajapeyam S, Jakacki R, Gajjar A, Goldman S, Pollack IF, Friedman HS, Boyett JM, Fouladi M, Kun LE. Efficacy of bevacizumab plus irinotecan in children with recurrent low-grade gliomas--a Pediatric Brain Tumor Consortium study. Neuro Oncol 2013; 16:310-7. [PMID: 24311632 DOI: 10.1093/neuonc/not154] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND A phase II study of bevacizumab (BVZ) plus irinotecan (CPT-11) was conducted in children with recurrent low-grade glioma to measure sustained response and/or stable disease lasting ≥6 months and progression-free survival. METHODS Thirty-five evaluable patients received 2 doses (10 mg/kg each) of single-agent BVZ intravenously 2 weeks apart and then BVZ + CPT-11 every 2 weeks until progressive disease, unacceptable toxicity, or a maximum of 2 years of therapy. Correlative studies included neuroimaging and expression of tumor angiogenic markers (vascular endothelial growth factor [VEGF], VEGF receptor 2, hypoxia-inducible factor 2α, and carbonic anhydrase 9). RESULTS Thirty-five evaluable patients (median age 8.4 y [range, 0.6-17.6]) received a median of 12 courses of BVZ + CPT-11 (range, 2-26). Twenty-nine of 35 patients (83%) received treatment for at least 6 months. Eight patients progressed on treatment at a median time of 5.4 months (range, 1-17.8). Six patients (17.7%) still in follow-up have had stable disease without receiving additional treatment for a median of 40.1 months (range, 30.6-49.3) from initiating therapy. The 6-month and 2-year progression-free survivals were 85.4% (SE ± 5.96%) and 47.8% (SE ± 9.27%), respectively. The commonest toxicities related to BVZ included grades 1-2 hypertension in 24, grades 1-2 fatigue in 23, grades 1-2 epistaxis in 18, and grades 1-4 proteinuria in 15. The median volume of enhancement decreased significantly between baseline and day 15 (P < .0001) and over the duration of treatment (P < .037). CONCLUSION The combination of BVZ + CPT-11 appears to produce sustained disease control in some children with recurrent low-grade gliomas.
Collapse
Affiliation(s)
- Sridharan Gururangan
- Corresponding author: Sri Gururangan, MRCP, Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Hanes House, Room 307, 330 Trent Drive, Box 102382, Durham, NC 27710.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Langenkamp E, Kamps JAAM, Mrug M, Verpoorte E, Niyaz Y, Horvatovich P, Bischoff R, Struijker-Boudier H, Molema G. Innovations in studying in vivo cell behavior and pharmacology in complex tissues--microvascular endothelial cells in the spotlight. Cell Tissue Res 2013; 354:647-69. [PMID: 24072341 DOI: 10.1007/s00441-013-1714-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/18/2013] [Indexed: 02/06/2023]
Abstract
Many studies on the molecular control underlying normal cell behavior and cellular responses to disease stimuli and pharmacological intervention are conducted in single-cell culture systems, while the read-out of cellular engagement in disease and responsiveness to drugs in vivo is often based on overall tissue responses. As the majority of drugs under development aim to specifically interact with molecular targets in subsets of cells in complex tissues, this approach poses a major experimental discrepancy that prevents successful development of new therapeutics. In this review, we address the shortcomings of the use of artificial (single) cell systems and of whole tissue analyses in creating a better understanding of cell engagement in disease and of the true effects of drugs. We focus on microvascular endothelial cells that actively engage in a wide range of physiological and pathological processes. We propose a new strategy in which in vivo molecular control of cells is studied directly in the diseased endothelium instead of at a (far) distance from the site where drugs have to act, thereby accounting for tissue-controlled cell responses. The strategy uses laser microdissection-based enrichment of microvascular endothelium which, when combined with transcriptome and (phospho)proteome analyses, provides a factual view on their status in their complex microenvironment. Combining this with miniaturized sample handling using microfluidic devices enables handling the minute sample input that results from this strategy. The multidisciplinary approach proposed will enable compartmentalized analysis of cell behavior and drug effects in complex tissue to become widely implemented in daily biomedical research and drug development practice.
Collapse
Affiliation(s)
- Elise Langenkamp
- University Medical Center Groningen, Department of Pathology and Medical Biology, Medical Biology section, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
World Health Organization (WHO) grade I astrocytomas include pilocytic astrocytoma (PA) and subependymal giant cell astrocytoma (SEGA). As technologies in pharmacologic neo-adjuvant therapy continue to progress and as molecular characteristics are progressively recognized as potential markers of both clinically significant tumor subtypes and response to therapy, interest in the biology of these tumors has surged. An updated review of the current knowledge of the molecular biology of these tumors is needed. We conducted a Medline search to identify published literature discussing the molecular biology of grade I astrocytomas. We then summarized this literature and discuss it in a logical framework through which the complex biology of these tumors can be clearly understood. A comprehensive review of the molecular biology of WHO grade I astrocytomas is presented. The past several years have seen rapid progress in the level of understanding of PA in particular, but the molecular literature regarding both PA and SEGA remains nebulous, ambiguous, and occasionally contradictory. In this review we provide a comprehensive discussion of the current understanding of the chromosomal, genomic, and epigenomic features of both PA and SEGA and provide a logical framework in which these data can be more readily understood.
Collapse
Affiliation(s)
- Nicholas F Marko
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|
17
|
Plate KH, Scholz A, Dumont DJ. Tumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisited. Acta Neuropathol 2012; 124:763-75. [PMID: 23143192 PMCID: PMC3508273 DOI: 10.1007/s00401-012-1066-5] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 11/02/2012] [Accepted: 11/02/2012] [Indexed: 12/13/2022]
Abstract
The cellular and molecular mechanisms of tumor angiogenesis and its prospects for anti-angiogenic cancer therapy are major issues in almost all current concepts of both cancer biology and targeted cancer therapy. Currently, (1) sprouting angiogenesis, (2) vascular co-option, (3) vascular intussusception, (4) vasculogenic mimicry, (5) bone marrow-derived vasculogenesis, (6) cancer stem-like cell-derived vasculogenesis and (7) myeloid cell-driven angiogenesis are all considered to contribute to tumor angiogenesis. Many of these processes have been described in developmental angiogenesis; however, the relative contribution and relevance of these in human brain cancer remain unclear. Preclinical tumor models support a role for sprouting angiogenesis, vascular co-option and myeloid cell-derived angiogenesis in glioma vascularization, whereas a role for the other four mechanisms remains controversial and rather enigmatic. The anti-angiogenesis drug Avastin (Bevacizumab), which targets VEGF, has become one of the most popular cancer drugs in the world. Anti-angiogenic therapy may lead to vascular normalization and as such facilitate conventional cytotoxic chemotherapy. However, preclinical and clinical studies suggest that anti-VEGF therapy using bevacizumab may also lead to a pro-migratory phenotype in therapy resistant glioblastomas and thus actively promote tumor invasion and recurrent tumor growth. This review focusses on (1) mechanisms of tumor angiogenesis in human malignant glioma that are of particular relevance for targeted therapy and (2) controversial issues in tumor angiogenesis such as cancer stem-like cell-derived vasculogenesis and bone-marrow-derived vasculogenesis.
Collapse
Affiliation(s)
- Karl H Plate
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School, Frankfurt, Germany.
| | | | | |
Collapse
|
18
|
Sikkema AH, den Dunnen WF, Hulleman E, van Vuurden DG, Garcia-Manero G, Yang H, Scherpen FJ, Kampen KR, Hoving EW, Kamps WA, Diks SH, Peppelenbosch MP, de Bont ES. EphB2 activity plays a pivotal role in pediatric medulloblastoma cell adhesion and invasion. Neuro Oncol 2012; 14:1125-35. [PMID: 22723427 PMCID: PMC3424207 DOI: 10.1093/neuonc/nos130] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 04/25/2012] [Indexed: 01/05/2023] Open
Abstract
Eph/ephrin signaling has been implicated in various types of key cancer-enhancing processes, like migration, proliferation, and angiogenesis. In medulloblastoma, invading tumor cells characteristically lead to early recurrence and a decreased prognosis. Based on kinase-activity profiling data published recently, we hypothesized a key role for the Eph/ephrin signaling system in medulloblastoma invasion. In primary medulloblastoma samples, a significantly higher expression of EphB2 and the ligand ephrin-B1 was observed compared with normal cerebellum. Furthermore, medulloblastoma cell lines showed high expression of EphA2, EphB2, and EphB4. Stimulation of medulloblastoma cells with ephrin-B1 resulted in a marked decrease in in vitro cell adhesion and an increase in the invasion capacity of cells expressing high levels of EphB2. The cell lines that showed an ephrin-B1-induced phenotype possessed increased levels of phosphorylated EphB2 and, to a lesser extent, EphB4 after stimulation. Knockdown of EphB2 expression by short hairpin RNA completely abolished ephrin ligand-induced effects on adhesion and migration. Analysis of signal transduction identified p38, Erk, and mTOR as downstream signaling mediators potentially inducing the ephrin-B1 phenotype. In conclusion, the observed deregulation of Eph/ephrin expression in medulloblastoma enhances the invasive phenotype, suggesting a potential role in local tumor cell invasion and the formation of metastases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Eveline S.J.M. de Bont
- Department of Pediatric Oncology/Hematology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, the Netherlands (A.H.S., F.J.G.S., K.R.K., W.A.K., S.H.D., E.S.J.M.B.); Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, the Netherlands (W.F.A.D.); Neuro-Oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, Amsterdam, the Netherlands (E.H., D.G.V.); Department of Leukemia, Division of Cancer Medicine, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, USA (G.G.-M., H.Y.); Department of Neurosurgery, University Medical Center Groningen, Hanzeplein 1, Groningen, the Netherlands (E.W.H.); Department of Gastroenterology and Hepatology, Erasmus MC, L-459, 's-Gravendijkwal 230, Rotterdam, the Netherlands (M.P.P.)
| |
Collapse
|
19
|
Peppelenbosch MP. Kinome profiling. SCIENTIFICA 2012; 2012:306798. [PMID: 24278683 PMCID: PMC3820527 DOI: 10.6064/2012/306798] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 07/12/2012] [Indexed: 06/02/2023]
Abstract
The use of arrays in genomics has led to a fast and reliable way to screen the transcriptome of an organism. It can be automated and analysis tools have become available and hence the technique has become widely used within the past few years. Signal-transduction routes rely mainly on the phosphorylation status of already available proteins; therefore kinases are central players in signal-transduction routes. The array technology can now also be used for the analysis of the kinome. To enable array analysis, consensus peptides for kinases are spot on a solid support. After incubation with cell lysates and in the presence of radioactive ATP, radioactive peptides can be visualized and the kinases that are active in the cells can be determined. The present paper reviews comprehensively the different kinome array platforms available and results obtained hitherto using such platforms. It will appear that this technology does not disappoint its high expectations and is especially powerful because of its species independence. Nevertheless, improvements are still possible and I shall also sketch future possible directions.
Collapse
Affiliation(s)
- Maikel P. Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center Rotterdam, L-459, P.O. Box 2040, NL-3000 CA Rotterdam, The Netherlands
| |
Collapse
|
20
|
Sikkema AH, den Dunnen WFA, Diks SH, Peppelenbosch MP, de Bont ESJM. Optimizing targeted cancer therapy: towards clinical application of systems biology approaches. Crit Rev Oncol Hematol 2012; 82:171-86. [PMID: 21641230 DOI: 10.1016/j.critrevonc.2011.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 04/28/2011] [Accepted: 05/04/2011] [Indexed: 12/13/2022] Open
Abstract
In cancer, genetic and epigenetic alterations ultimately culminate in discordant activation of signal transduction pathways driving the malignant process. Pharmacological or biological inhibition of such pathways holds significant promise with respect to devising rational therapy for cancer. Thus, technical concepts pursuing robust characterization of kinase activity in tissue samples from cancer patients have been subject of investigation. In the present review we provide a comprehensive overview of these techniques and discuss their advantages and disadvantages for systems biology approaches to identify kinase targets in oncological disease. Recent advances in the development and application of array-based peptide-substrate kinase activity screens show great promise in overcoming the discrepancy between the evaluation of aberrant cell signaling in specific malignancies or even individual patients and the currently available ensemble of highly specific targeted treatment strategies. These developments have the potential to result in a more effective selection of kinase inhibitors and thus optimize mechanism-based patient-specific therapeutic strategies. Given the results from current research on the tumor kinome, generating network views on aberrant tumor cell signaling is critical to meet this challenge.
Collapse
Affiliation(s)
- Arend H Sikkema
- Beatrix Children's Hospital, Department of Pediatric Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
21
|
Wang T, Agarwal S, Elmquist WF. Brain distribution of cediranib is limited by active efflux at the blood-brain barrier. J Pharmacol Exp Ther 2012; 341:386-95. [PMID: 22323823 DOI: 10.1124/jpet.111.190488] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Cediranib is an orally active tyrosine kinase inhibitor that targets the vascular endothelial growth factor receptor family. Because of its potent antiangiogenic and antitumor activities, cediranib has been evaluated for therapy in glioma, a primary brain tumor. This study investigated the influence of two important efflux transporters at the blood-brain barrier, P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp), on the delivery of cediranib to the central nervous system. In vitro studies indicated that cediranib is a dual substrate for both P-gp and Bcrp. It is noteworthy that in spite of the in vitro data the in vivo mouse disposition studies conclusively showed that P-gp was the dominant transporter restricting the brain distribution of cediranib. The brain-to-plasma partitioning (AUC(brain)/AUC(plasma), where AUC is area under the curve) and the steady-state brain-to-plasma concentration ratio of cediranib were approximately 20-fold higher in Mdr1a/b⁻/⁻ and Mdr1a/b⁻/⁻Bcrp1⁻/⁻ mice compared with wild-type and Bcrp1⁻/⁻ mice. Moreover, there was no significant difference in brain distribution of cediranib between wild-type and Bcrp1⁻/⁻ mice and between Mdr1a/b⁻/⁻ and Mdr1a/b⁻/⁻Bcrp1⁻/⁻ mice. These results show that, unlike other tyrosine kinase inhibitors that are dual substrates for P-gp and Bcrp, Bcrp does not restrict the distribution of cediranib across the blood-brain barrier. We also show that inhibition of P-gp using specific or nonspecific inhibitors resulted in significantly enhanced delivery of cediranib to the brain. Concurrent administration of cediranib with chemical modulators of efflux transporters can be used as a strategy to enhance delivery and thus efficacy of cediranib in the brain. These findings are clinically relevant to the efficacy of cediranib chemotherapy in glioma.
Collapse
Affiliation(s)
- Tianli Wang
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|