1
|
Al-Jilaihawi S, Spreafico F, Mavinkurve-Groothuis A, Drost J, Perotti D, Koenig C, Brok J. Bevacizumab-containing treatment for relapsed or refractory Wilms tumor. Expert Rev Anticancer Ther 2024; 24:837-843. [PMID: 39016020 DOI: 10.1080/14737140.2024.2381537] [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: 04/08/2024] [Accepted: 07/15/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION Angiogenesis is critical for tumor growth and metastasis. Bevacizumab is an antiangiogenic drug used to treat various adult and childhood solid tumors. Its potential efficacy in Wilms tumor (WT) with poor prognosis is not established. AREAS COVERED The response to bevacizumab-containing regimens in relapsed or refractory WT was reviewed in available literature. Searches were conducted using PubMed, Scopus, and ClinicalTrials.gov databases. Eight papers were identified, published between 2007 and 2020, including six treatment regimens, predominantly vincristine, irinotecan, and bevacizumab (VIB) ± temozolomide (VITB). Among 16 evaluable patients, there were two complete responses, seven partial responses, five patients achieved stable disease (SD), and two patients had progressive disease. Objective responses (OR) were observed in 56% of all cases. OR or SD was observed in 89% (8/9) patients who received VIB/VITB. Bevacizumab was generally well tolerated. Related toxicities included hypertension, proteinuria, and delayed wound healing. EXPERT OPINION This review suggests potential effectiveness and good tolerability of bevacizumab in the setting of relapsed/refractory WT when used in combination with other drugs. Such combination therapies may serve as a bridging treatment option to other interventions and more personalized treatment options in the future; however, focused trials are needed to obtain additional evidence.
Collapse
Affiliation(s)
- Sarah Al-Jilaihawi
- Department of Paediatric Oncology, Bristol Royal Hospital for Children, Bristol, UK
| | - Filippo Spreafico
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | | | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Daniela Perotti
- Predictive Medicine: Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Christa Koenig
- Pediatric Hematology/Oncology, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jesper Brok
- Department of Pediatric Hematology and Oncology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
2
|
Özkan A, Yağcı Küpeli B, Küpeli S, Sezgin G, Bayram İ. Nimotuzumab-vinorelbine combination therapy versus other regimens in the treatment of pediatric diffuse intrinsic pontine glioma. Childs Nerv Syst 2024; 40:1671-1680. [PMID: 38478066 DOI: 10.1007/s00381-024-06329-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/21/2024] [Indexed: 05/23/2024]
Abstract
PURPOSE Pediatric diffuse intrinsic pontine glioma (DIPG) is a fatal disease associated with a median survival of < 1 year despite aggressive treatments. This retrospective study analyzed the treatment outcomes of patients aged < 18 years who were diagnosed with DIPG between 2012 and 2022 and who received different chemotherapy regimens. METHODS After radiotherapy, patients with DIPG received nimotuzumab-vinorelbine combination or temozolomide-containing therapy. When nimotuzumab was unavailable, it was replaced by vincristine, etoposide, and carboplatin/cyclophosphamide (VECC). Temozolomide was administered as a single agent or a part of the combination chemotherapy comprising temozolomide, irinotecan, and bevacizumab. Furthermore, 1- and 3-year overall survival (OS), progression-free survival (PFS), and median OS and PFS were analyzed. RESULTS The median age of 40 patients with DIPG was 97 ± 46.93 (23-213) months; the median follow-up time was 12 months. One and 3-year OS were 35.0% and 7.5%, respectively. Median OS was 12 months in all patients (n = 40), and it was 16, 10, and 11 months in those who received first-line nimotuzumab-vinorelbine combination (n = 13), temozolomide-based (n = 14), and VECC (n = 6) chemotherapy regimens, respectively (p = 0.360). One patient who received gefitinib survived for 16 months. Conversely, patients who never received radiotherapy and any antineoplastic medicamentous therapy (n = 6) had a median OS of 4 months. CONCLUSION Nimotuzumab-vinorelbine combination therapy prolonged OS by 6 months compared with temozolomide-containing chemotherapy, although the difference was not statistically significant.
Collapse
Affiliation(s)
- Ayşe Özkan
- Department of Pediatric Oncology and Pediatric Bone Marrow Transplantation Unit, Faculty of Medicine, Balcali Hospital, Çukurova University, Adana, Turkey.
| | - Begül Yağcı Küpeli
- Department of Pediatric Hematology and Oncology, Adana City Training and Research Hospital, University of Health Sciences, Adana, Turkey
| | - Serhan Küpeli
- Department of Pediatric Oncology and Pediatric Bone Marrow Transplantation Unit, Faculty of Medicine, Balcali Hospital, Çukurova University, Adana, Turkey
| | - Gülay Sezgin
- Department of Pediatric Oncology and Pediatric Bone Marrow Transplantation Unit, Faculty of Medicine, Balcali Hospital, Çukurova University, Adana, Turkey
| | - İbrahim Bayram
- Department of Pediatric Oncology and Pediatric Bone Marrow Transplantation Unit, Faculty of Medicine, Balcali Hospital, Çukurova University, Adana, Turkey
| |
Collapse
|
3
|
Yağcı-Küpeli B, Pehlivan D. Bevacizumab-containing regimens for children with relapsed or refractory tumors. Indian J Cancer 2024; 61:375-382. [PMID: 36861699 DOI: 10.4103/ijc.ijc_907_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 03/07/2021] [Indexed: 03/03/2023]
Abstract
BACKGROUND We aimed to evaluate the effect of bevacizumab-containing regimens (BCRs) on the survival of children with relapsed or refractory solid tumors. MATERIALS AND METHODS Files of children with relapsed or refractory solid tumors treated with BCR were retrospectively reviewed for age, gender, follow-up time, histopathological diagnosis, adverse events observed with BCR, number of chemotherapy protocols used before BCR, the best overall response obtained with BCR, time to progression, number of BCR courses given to patients, the status of patient at last visit, and outcome. RESULTS Thirty patients (16 boys, 14 girls) were treated with BCR. The median age at diagnosis was 8.5 (2 - 17) years and at the time of the study was 11 (3-21) years. The median follow-up time was 25.7 (5-79.4) months. The median follow-up time after the start of BCR was 3.2 (1-27) months. Histopathological diagnosis was central nervous system tumors in 25, Ewing sarcoma in two, osteosarcoma in two, and rhabdomyosarcoma in one patient. BCR was given as second-line in 21, third-line in six, and fourth-line protocol in three patients. No chemotherapy toxicity was observed in 22 (73.3%) patients. The best overall response was progressive disease in 17 (56.7%), partial response in seven (23.3%), and stable disease in 6 (20%) patients at first-response evaluation. The median time until progression was 77 (12-690) days. During the study period, 17 patients died of progressive disease. CONCLUSION Our study revealed that adding antiangiogenic agent bevacizumab to cytotoxic chemotherapy provided no survival benefit in children with relapsed or refractory solid tumors.
Collapse
Affiliation(s)
- Begül Yağcı-Küpeli
- Department of Pediatric Hematology/Oncology, Adana City Training and Research Hospital, University of Health Sciences, Adana, Turkey
| | - Duygu Pehlivan
- Department of Pediatrics, Adana City Training and Research Hospital, University of Health Sciences, Adana, Turkey
| |
Collapse
|
4
|
Acanda De La Rocha AM, Berlow NE, Fader M, Coats ER, Saghira C, Espinal PS, Galano J, Khatib Z, Abdella H, Maher OM, Vorontsova Y, Andrade-Feraud CM, Daccache A, Jacome A, Reis V, Holcomb B, Ghurani Y, Rimblas L, Guilarte TR, Hu N, Salyakina D, Azzam DJ. Feasibility of functional precision medicine for guiding treatment of relapsed or refractory pediatric cancers. Nat Med 2024; 30:990-1000. [PMID: 38605166 PMCID: PMC11031400 DOI: 10.1038/s41591-024-02848-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 01/31/2024] [Indexed: 04/13/2024]
Abstract
Children with rare, relapsed or refractory cancers often face limited treatment options, and few predictive biomarkers are available that can enable personalized treatment recommendations. The implementation of functional precision medicine (FPM), which combines genomic profiling with drug sensitivity testing (DST) of patient-derived tumor cells, has potential to identify treatment options when standard-of-care is exhausted. The goal of this prospective observational study was to generate FPM data for pediatric patients with relapsed or refractory cancer. The primary objective was to determine the feasibility of returning FPM-based treatment recommendations in real time to the FPM tumor board (FPMTB) within a clinically actionable timeframe (<4 weeks). The secondary objective was to assess clinical outcomes from patients enrolled in the study. Twenty-five patients with relapsed or refractory solid and hematological cancers were enrolled; 21 patients underwent DST and 20 also completed genomic profiling. Median turnaround times for DST and genomics were within 10 days and 27 days, respectively. Treatment recommendations were made for 19 patients (76%), of whom 14 received therapeutic interventions. Six patients received subsequent FPM-guided treatments. Among these patients, five (83%) experienced a greater than 1.3-fold improvement in progression-free survival associated with their FPM-guided therapy relative to their previous therapy, and demonstrated a significant increase in progression-free survival and objective response rate compared to those of eight non-guided patients. The findings from our proof-of-principle study illustrate the potential for FPM to positively impact clinical care for pediatric and adolescent patients with relapsed or refractory cancers and warrant further validation in large prospective studies. ClinicalTrials.gov registration: NCT03860376 .
Collapse
Affiliation(s)
- Arlet M Acanda De La Rocha
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | | | - Maggie Fader
- Division of Pediatric Hematology Oncology, Department of Pediatrics, Nicklaus Children's Hospital, Miami, FL, USA
| | - Ebony R Coats
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Cima Saghira
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Paula S Espinal
- Center for Precision Medicine, Nicklaus Children's Hospital, Miami, FL, USA
| | - Jeanette Galano
- Center for Precision Medicine, Nicklaus Children's Hospital, Miami, FL, USA
| | - Ziad Khatib
- Division of Pediatric Hematology Oncology, Department of Pediatrics, Nicklaus Children's Hospital, Miami, FL, USA
| | - Haneen Abdella
- Division of Pediatric Hematology Oncology, Department of Pediatrics, Nicklaus Children's Hospital, Miami, FL, USA
| | - Ossama M Maher
- Division of Pediatric Hematology Oncology, Department of Pediatrics, Nicklaus Children's Hospital, Miami, FL, USA
| | - Yana Vorontsova
- Center for Precision Medicine, Nicklaus Children's Hospital, Miami, FL, USA
| | - Cristina M Andrade-Feraud
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Aimee Daccache
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Alexa Jacome
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Victoria Reis
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Baylee Holcomb
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Yasmin Ghurani
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Lilliam Rimblas
- Division of Pediatric Hematology Oncology, Department of Pediatrics, Nicklaus Children's Hospital, Miami, FL, USA
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Nan Hu
- Department of Biostatistics, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Daria Salyakina
- Center for Precision Medicine, Nicklaus Children's Hospital, Miami, FL, USA
| | - Diana J Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA.
| |
Collapse
|
5
|
Dinikina YV, Zheludkova OG, Belogurova MB, Spelnikov DM, Osipov NN, Nikitina IL. Personalized treatment options of refractory and relapsed medulloblastoma in children: literature review. JOURNAL OF MODERN ONCOLOGY 2024; 25:454-465. [DOI: 10.26442/18151434.2023.4.202521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Medulloblastoma (MB) is the most common malignant tumor of the central nervous system in pediatric patients. Despite the complex anticancer therapy approach, refractory and relapsing forms of the disease remain fatal in most cases and account for approximately 30%. To date, repeated surgery, radiation, and chemotherapy can be used as life-prolonging treatment options; nevertheless, it should be emphasized that there are no standardized approaches based on existing data of molecular variants of MB. It is obvious that only a deep understanding of the biological mechanisms in association with clinical aspects in refractory and relapsing forms of MB would make it possible to personalize second- and subsequent-line therapy in order to achieve maximum efficiency and minimize early and long-term toxicity. The article presents the current understanding of prognostic factors in relapsed/refractory forms of MB, methods of modern diagnostics, as well as existing and perspective treatment options based on the biological and clinical aspects of the disease.
Collapse
|
6
|
Adamczewska-Wawrzynowicz K, Wiącek A, Kozłowska A, Mikosza K, Szefler L, Dudlik W, Dey S, Varghese N, Derwich K. Modern treatment strategies in pediatric oncology and hematology. Discov Oncol 2023; 14:98. [PMID: 37314524 PMCID: PMC10267092 DOI: 10.1007/s12672-023-00658-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/14/2023] [Indexed: 06/15/2023] Open
Abstract
Every year, approximately 400 00 children worldwide are diagnosed with cancer. Although treatment results in most types of childhood neoplasms are excellent with survival more than 80%, there are some with poor prognosis. Also recurrent and resistant to treatment childhood cancer remain a therapeutic challenge. Besides chemotherapy, which has been the basis of cancer therapy for years, molecular methods and precisely targeted therapies have recently found their usage. As a result of that, survival has improved and has positively impacted the rate of toxicities associated with chemotherapy (Butler et al. in CA Cancer J Clin 71:315-332, 2021). These achievements have contributed to better quality of patients' lives. Current methods of treatment and ongoing trials give hope for patients with relapses and resistance to conventional chemotherapy. This review focuses on the most recent progress in pediatric oncology treatments and discusses specific therapy methods for particular cancers types of cancer. Targeted therapies and molecular approaches have become more beneficial but research need to be continued in this field. Despite significant breakthroughs in pediatric oncology in the last few years, there is still a need to find new and more specific methods of treatment to increase the survival of children with cancer.
Collapse
Affiliation(s)
- Katarzyna Adamczewska-Wawrzynowicz
- Institute of Pediatrics, Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Szpitalna 27/33 street, 61-572, Poznan, Poland
| | - Anna Wiącek
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Klaudia Mikosza
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Lidia Szefler
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Weronika Dudlik
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Shreya Dey
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Noel Varghese
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Derwich
- Institute of Pediatrics, Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Szpitalna 27/33 street, 61-572, Poznan, Poland.
| |
Collapse
|
7
|
van Bree NFHN, Wilhelm M. The Tumor Microenvironment of Medulloblastoma: An Intricate Multicellular Network with Therapeutic Potential. Cancers (Basel) 2022; 14:5009. [PMID: 36291792 PMCID: PMC9599673 DOI: 10.3390/cancers14205009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Medulloblastoma (MB) is a heterogeneous disease in which survival is highly affected by the underlying subgroup-specific characteristics. Although the current treatment modalities have increased the overall survival rates of MB up to 70-80%, MB remains a major cause of cancer-related mortality among children. This indicates that novel therapeutic approaches against MB are needed. New promising treatment options comprise the targeting of cells and components of the tumor microenvironment (TME). The TME of MB consists of an intricate multicellular network of tumor cells, progenitor cells, astrocytes, neurons, supporting stromal cells, microglia, immune cells, extracellular matrix components, and vasculature systems. In this review, we will discuss all the different components of the MB TME and their role in MB initiation, progression, metastasis, and relapse. Additionally, we briefly introduce the effect that age plays on the TME of brain malignancies and discuss the MB subgroup-specific differences in TME components and how all of these variations could affect the progression of MB. Finally, we highlight the TME-directed treatments, in which we will focus on therapies that are being evaluated in clinical trials.
Collapse
Affiliation(s)
| | - Margareta Wilhelm
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, 17165 Stockholm, Sweden
| |
Collapse
|
8
|
Al Dahhan NZ, Cox E, Nieman BJ, Mabbott DJ. Cross-translational models of late-onset cognitive sequelae and their treatment in pediatric brain tumor survivors. Neuron 2022; 110:2215-2241. [PMID: 35523175 DOI: 10.1016/j.neuron.2022.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/21/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Pediatric brain tumor treatments have a high success rate, but survivors are at risk of cognitive sequelae that impact long-term quality of life. We summarize recent clinical and animal model research addressing pathogenesis or evaluating candidate interventions for treatment-induced cognitive sequelae. Assayed interventions encompass a broad range of approaches, including modifications to radiotherapy, modulation of immune response, prevention of treatment-induced cell loss or promotion of cell renewal, manipulation of neuronal signaling, and lifestyle/environmental adjustments. We further emphasize the potential of neuroimaging as a key component of cross-translation to contextualize laboratory research within broader clinical findings. This cross-translational approach has the potential to accelerate discovery to improve pediatric cancer survivors' long-term quality of life.
Collapse
Affiliation(s)
- Noor Z Al Dahhan
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Elizabeth Cox
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Brian J Nieman
- Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada; Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Donald J Mabbott
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Psychology, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
9
|
A phase I study of irinotecan and temozolomide with bevacizumab in children with recurrent/refractory central nervous system tumors. Childs Nerv Syst 2022; 38:919-928. [PMID: 35260913 DOI: 10.1007/s00381-022-05479-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/26/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Children with relapsed/refractory central nervous system (CNS) tumors require novel combinations of therapies. Irinotecan and temozolomide (IT) is a frequently used therapy with an established toxicity profile. Bevacizumab is an anti-VEGF monoclonal antibody with demonstrated activity in CNS tumors. Therefore, the combination of these agents has therapeutic potential in CNS tumors. The objective of this study was to determine the maximum tolerated dose (MTD) of escalating dose IT combined with a fixed dose of bevacizumab (BIT) in children with relapsed/refractory CNS tumors. METHODS A phase I trial was performed in a 3 + 3 design. Therapy toxicities and radiologic responses to treatment were described. RESULTS One hundred eighty cycles of therapy were administered to 26 patients. The MTD of BIT was dose level 1, (bevacizumab 10 mg/kg on days 1 and 15, irinotecan 125 mg/m2 on days 1 and 15, and temozolomide 125 mg/m2 on days 1-5 of 28-day cycles). The regimen was well tolerated with primarily hematologic toxicity, which was not dose limiting. Among 22 response-evaluable patients, there was 1 complete response (CR), 6 partial responses (PR), and 10 stable diseases (SD) with an overall response rate (ORR: CR + PR) of 31.8%. CONCLUSION At the MTD, BIT therapy was well tolerated, and prolonged treatment courses of up to 24 cycles were feasible, with radiographic responses observed. Further evaluation is needed for efficacy in a phase II trial (NCT00876993, registered April 7, 2009, www. CLINICALTRIALS gov ).
Collapse
|
10
|
Saker Z, Rizk M, Bahmad HF, Nabha SM. Targeting Angiogenic Factors for the Treatment of Medulloblastoma. Curr Treat Options Oncol 2022; 23:864-886. [PMID: 35412196 DOI: 10.1007/s11864-022-00981-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2022] [Indexed: 11/24/2022]
Abstract
OPINION STATEMENT Medulloblastoma (MB) is the most frequent pediatric brain tumor. Despite conventional therapy, MB patients have high mortality and morbidity rates mainly due to the incomplete understanding of the molecular and cellular processes involved in development of this cancer. Similar to other solid tumors, MB demonstrated high endothelial cell proliferation and angiogenic activity, wherein new blood vessels arise from the pre-existing vasculature, a process named angiogenesis. MB angiogenesis is considered a hallmark for MB development, progression, and metastasis emphasizing its potential target for antitumor therapy. However, angiogenesis is tightly regulated by a set of angiogenic factors making it a complex process to be targeted. Although agents targeting these factors and their receptors are early in development, the potential for their targeting may translate into improvement in the clinical care for MB patients. In this review, we focus on the most potent angiogenic factors and their corresponding receptors, highlighting their basic properties and expression in MB. We describe their contribution to MB tumorigenesis and angiogenesis and the potential therapeutic targeting of these factors.
Collapse
Affiliation(s)
- Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mahdi Rizk
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, 33140, USA.
| | - Sanaa M Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
11
|
The Current Landscape of Targeted Clinical Trials in Non-WNT/Non-SHH Medulloblastoma. Cancers (Basel) 2022; 14:cancers14030679. [PMID: 35158947 PMCID: PMC8833659 DOI: 10.3390/cancers14030679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Medulloblastoma is a form of malignant brain tumor that arises predominantly in infants and young children and can be divided into different groups based on molecular markers. The group of non-WNT/non-SHH medulloblastoma includes a spectrum of heterogeneous subgroups that differ in their biological characteristics, genetic underpinnings, and clinical course of disease. Non-WNT/non-SHH medulloblastoma is currently treated with surgery, chemotherapy, and radiotherapy; however, new drugs are needed to treat patients who are not yet curable and to reduce treatment-related toxicity and side effects. We here review which new treatment options for non-WNT/non-SHH medulloblastoma are currently clinically tested. Furthermore, we illustrate the challenges that have to be overcome to reach a new therapeutic standard for non-WNT/non-SHH medulloblastoma, for instance the current lack of good preclinical models, and the necessity to conduct trials in a comparably small patient collective. Abstract Medulloblastoma is an embryonal pediatric brain tumor and can be divided into at least four molecularly defined groups. The category non-WNT/non-SHH medulloblastoma summarizes medulloblastoma groups 3 and 4 and is characterized by considerable genetic and clinical heterogeneity. New therapeutic strategies are needed to increase survival rates and to reduce treatment-related toxicity. We performed a noncomprehensive targeted review of the current clinical trial landscape and literature to summarize innovative treatment options for non-WNT/non-SHH medulloblastoma. A multitude of new drugs is currently evaluated in trials for which non-WNT/non-SHH patients are eligible, for instance immunotherapy, kinase inhibitors, and drugs targeting the epigenome. However, the majority of these trials is not restricted to medulloblastoma and lacks molecular classification. Whereas many new molecular targets have been identified in the last decade, which are currently tested in clinical trials, several challenges remain on the way to reach a new therapeutic strategy for non-WNT/non-SHH medulloblastoma. These include the severe lack of faithful preclinical models and predictive biomarkers, the question on how to stratify patients for clinical trials, and the relative lack of studies that recruit large, homogeneous patient collectives. Innovative trial designs and international collaboration will be a key to eventually overcome these obstacles.
Collapse
|
12
|
Hill RM, Plasschaert SLA, Timmermann B, Dufour C, Aquilina K, Avula S, Donovan L, Lequin M, Pietsch T, Thomale U, Tippelt S, Wesseling P, Rutkowski S, Clifford SC, Pfister SM, Bailey S, Fleischhack G. Relapsed Medulloblastoma in Pre-Irradiated Patients: Current Practice for Diagnostics and Treatment. Cancers (Basel) 2021; 14:126. [PMID: 35008290 PMCID: PMC8750207 DOI: 10.3390/cancers14010126] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023] Open
Abstract
Relapsed medulloblastoma (rMB) accounts for a considerable, and disproportionate amount of childhood cancer deaths. Recent advances have gone someway to characterising disease biology at relapse including second malignancies that often cannot be distinguished from relapse on imaging alone. Furthermore, there are now multiple international early-phase trials exploring drug-target matches across a range of high-risk/relapsed paediatric tumours. Despite these advances, treatment at relapse in pre-irradiated patients is typically non-curative and focuses on providing life-prolonging and symptom-modifying care that is tailored to the needs and wishes of the individual and their family. Here, we describe the current understanding of prognostic factors at disease relapse such as principal molecular group, adverse molecular biology, and timing of relapse. We provide an overview of the clinical diagnostic process including signs and symptoms, staging investigations, and molecular pathology, followed by a summary of treatment modalities and considerations. Finally, we summarise future directions to progress understanding of treatment resistance and the biological mechanisms underpinning early therapy-refractory and relapsed disease. These initiatives include development of comprehensive and collaborative molecular profiling approaches at relapse, liquid biopsies such as cerebrospinal fluid (CSF) as a biomarker of minimal residual disease (MRD), modelling strategies, and the use of primary tumour material for real-time drug screening approaches.
Collapse
Affiliation(s)
- Rebecca M. Hill
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne NE1 7RU, UK; (S.C.C.); (S.B.)
| | - Sabine L. A. Plasschaert
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (S.L.A.P.); (M.L.); (P.W.)
| | - Beate Timmermann
- Department of Particle Therapy, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, 45147 Essen, Germany;
| | - Christelle Dufour
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, 94800 Villejuif, France;
| | - Kristian Aquilina
- Department of Neurosurgery, Great Ormond Street Hospital, London WC1N 3JH, UK;
| | - Shivaram Avula
- Department of Radiology, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK;
| | - Laura Donovan
- UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK;
| | - Maarten Lequin
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (S.L.A.P.); (M.L.); (P.W.)
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn, 53127 Bonn, Germany;
| | - Ulrich Thomale
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Stephan Tippelt
- Department of Pediatrics III, Center for Translational Neuro- and Behavioral Sciences (CTNBS), University Hospital of Essen, 45147 Essen, Germany;
| | - Pieter Wesseling
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (S.L.A.P.); (M.L.); (P.W.)
- Department of Pathology, Amsterdam University Medical Centers/VUmc, 1081 HV Amsterdam, The Netherlands
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Steven C. Clifford
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne NE1 7RU, UK; (S.C.C.); (S.B.)
| | - Stefan M. Pfister
- Hopp Children’s Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany;
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Pediatric Oncology and Hematology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Simon Bailey
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne NE1 7RU, UK; (S.C.C.); (S.B.)
| | - Gudrun Fleischhack
- Department of Pediatrics III, Center for Translational Neuro- and Behavioral Sciences (CTNBS), University Hospital of Essen, 45147 Essen, Germany;
| |
Collapse
|
13
|
Yamauchi T, Kitai R, Arai H, Kidoguchi M, Isozaki M, Kodera T, Yamauchi T, Kikuta KI. Bevacizumab, irinotecan, and temozolomide with re-irradiation in adult recurrent medulloblastoma: A first case report. INTERDISCIPLINARY NEUROSURGERY 2021. [DOI: 10.1016/j.inat.2021.101249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
14
|
Korshunov A, Okonechnikov K, Stichel D, Ryzhova M, Schrimpf D, Sahm F, Sievers P, Absalyamova O, Zheludkova O, Golanov A, Jones DTW, Pfister SM, von Deimling A, Kool M. Integrated molecular analysis of adult sonic hedgehog (SHH)-activated medulloblastomas reveals two clinically relevant tumor subsets with VEGFA as potent prognostic indicator. Neuro Oncol 2021; 23:1576-1585. [PMID: 33589929 PMCID: PMC8408884 DOI: 10.1093/neuonc/noab031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Up to now, adult medulloblastoma (MB) patients are treated according to the protocols elaborated for pediatric MB although these tumors are different in terms of clinical outcomes and biology. Approximately 70% of adult MB disclose a sonic hedgehog (SHH) molecular signature in contrast to about 30% in pediatric cohorts. In addition, adult SHH-MB (aSHH-MB) are clinically heterogeneous but there is consensus neither on their optimal treatment nor on risk stratification. Thus, the identification of clinically relevant molecular subsets of aSHH-MB and identification of potential treatment targets remains inconclusive. METHODS We analyzed 96 samples of institutionally diagnosed aSHH-MB through genome-wide DNA methylation profiling, targeted DNA sequencing, and RNA sequencing to identify molecular subcategories of these tumors and assess their prognostic significance. RESULTS We defined two aSHH-MB numerically comparable epigenetic subsets with clinical and molecular variability. The subset "aSHH-MBI" (46%/48%) was associated with PTCH1/SMO (54%/46%) mutations, "neuronal" transcriptional signatures, and favorable outcomes after combined radio-chemotherapy (5-year PFS = 80% and OS = 92%). The clinically unfavorable "aSHH-MBII" subset (50%/52%; 5-year PFS = 24% and OS = 45%) disclosed GLI2 amplifications (8%), loss of 10q (22%), and gene expression signatures associated with angiogenesis and embryonal development. aSHH-MBII tumors revealed strong and ubiquitous expression of VEGFA both at transcript and protein levels that was correlated with unfavorable outcome. CONCLUSIONS (1) The histologically uniform aSHH-MB cohort exhibits clear molecular heterogeneity separating these tumors into two molecular subsets (aSHH-MBI and aSHH-MBII), which are associated with different cytogenetics, mutational landscapes, gene expression signatures, and clinical course. (2) VEGFA appears to be a promising biomarker to predict clinical course, which needs further prospective validation as its potential role in the pathogenesis of this subset.
Collapse
Affiliation(s)
- Andrey Korshunov
- Clinical Cooperation Unit Neuropathology (B300), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University of Heidelberg, Heidelberg, Germany
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Konstantin Okonechnikov
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neuro-oncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Damian Stichel
- Clinical Cooperation Unit Neuropathology (B300), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University of Heidelberg, Heidelberg, Germany
| | - Marina Ryzhova
- N.N. Burdenko Neurosurgical Research Centre, Moscow, Russia
| | - Daniel Schrimpf
- Clinical Cooperation Unit Neuropathology (B300), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University of Heidelberg, Heidelberg, Germany
| | - Felix Sahm
- Clinical Cooperation Unit Neuropathology (B300), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University of Heidelberg, Heidelberg, Germany
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Philipp Sievers
- Clinical Cooperation Unit Neuropathology (B300), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University of Heidelberg, Heidelberg, Germany
| | | | - Olga Zheludkova
- Department of Neuro-Oncology, Russian Scientific Center of Radiology, Moscow, Russia
| | - Andrey Golanov
- N.N. Burdenko Neurosurgical Research Centre, Moscow, Russia
| | - David T W Jones
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Pediatric Glioma Research Group (B360), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neuro-oncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, University of Heidelberg, Heidelberg, Germany
| | - Andreas von Deimling
- Clinical Cooperation Unit Neuropathology (B300), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University of Heidelberg, Heidelberg, Germany
| | - Marcel Kool
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neuro-oncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
15
|
Tsakatikas S, Papageorgiou G, Fioretzaki R, Kosmas C. An overview of current results with the vincristine-irinotecan-temozolomide combination with or without bevacizumab in pediatric, adolescence and adult solid tumors. Crit Rev Oncol Hematol 2021; 166:103457. [PMID: 34428555 DOI: 10.1016/j.critrevonc.2021.103457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/02/2021] [Accepted: 08/19/2021] [Indexed: 11/29/2022] Open
Abstract
Malignant tumors in young patients present a significant therapeutic challenge for physicians, partially due to their rarity and a relative lack of data, at least compared to adult tumors. As a result, there is an urgent need to explore new possible therapeutic regimens, either by introducing novel agents or by exploring combinations of existing agents. Vincristine, Temozolomide and Irinotecan are chemotherapeutic drugs which have emerged over the last six decades as monotherapy or as part of therapeutic regimens in various solid tumors. Combining these agents can yield strong synergistic effects, as suggested by preclinical data and results from clinical trials. Furthermore, adding novel molecules, such as anti-VEGF factor Bevacizumab to the aforementioned regimens, has shown efficacy in a limited number of trials, which are thoroughly analyzed throughout this review. Data presented throughout this paper suggest that VIT(b) regimen should be further explored in solid tumors in pediatric and adolescent patients.
Collapse
Affiliation(s)
- Sergios Tsakatikas
- Department of Medical Oncology & Hematopoietic Cell Transplant Unit, "Metaxa" Memorial Cancer Hospital, 18537, Piraeus, Greece.
| | - George Papageorgiou
- Department of Medical Oncology & Hematopoietic Cell Transplant Unit, "Metaxa" Memorial Cancer Hospital, 18537, Piraeus, Greece.
| | - Rodanthi Fioretzaki
- Department of Medical Oncology & Hematopoietic Cell Transplant Unit, "Metaxa" Memorial Cancer Hospital, 18537, Piraeus, Greece.
| | - Christos Kosmas
- Department of Medical Oncology & Hematopoietic Cell Transplant Unit, "Metaxa" Memorial Cancer Hospital, 18537, Piraeus, Greece.
| |
Collapse
|
16
|
Levy AS, Krailo M, Chi S, Villaluna D, Springer L, Williams-Hughes C, Fouladi M, Gajjar A. Temozolomide with irinotecan versus temozolomide, irinotecan plus bevacizumab for recurrent medulloblastoma of childhood: Report of a COG randomized Phase II screening trial. Pediatr Blood Cancer 2021; 68:e29031. [PMID: 33844469 PMCID: PMC8764558 DOI: 10.1002/pbc.29031] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/17/2021] [Accepted: 03/08/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Approximately 30% of children with medulloblastoma (MB) experience recurrence, which is usually incurable. This study compared the overall survival (OS) of patients receiving temozolomide (TMZ) and irinotecan with that of patients receiving TMZ, irinotecan, and bevacizumab for recurrent MB/central nervous system (CNS) primitive neuroectodermal tumor (PNET). METHODS Patients with relapsed/refractory MB or CNS PNET were randomly assigned to receive TMZ (150 mg/m2 /day PO on days 1-5) and irinotecan (50 mg/m2 /day IV on days 1-5) with or without bevacizumab (10 mg/kg IV on days 1 and 15). RESULTS One hundred five patients were eligible and treated on study. Median OS was 13 months in the standard arm and 19 months with the addition of bevacizumab; median event-free survival (EFS) was 6 months in the standard arm and 9 months with the addition of bevacizumab. The hazard ratio for death from the stratified relative-risk regression model is 0.63. Overall, 23 patients completed 12 courses of planned protocol therapy, 23% (12/52) in the experimental arm with bevacizumab versus 21% (11/53) in the standard arm. Toxicity profiles were comparable in both treatment arms. The estimate of the incidence of feasibility events associated with the bevacizumab arm is three of 52 (5.8%) (95% CI 1.2-16%). Events included myelosuppression, electrolyte abnormalities, diarrhea, and elevated transaminases. One intracranial hemorrhage event was observed in each arm. CONCLUSION The addition of bevacizumab to TMZ/irinotecan significantly reduced the risk of death in children with recurrent MB. The combination was relatively well tolerated in this heavily pretreated cohort. The three-drug regimen demonstrated a sufficient risk reduction to warrant further investigation.
Collapse
Affiliation(s)
| | - Mark Krailo
- Department of Preventive Medicine, University of Southern California, Los Angeles CA
| | - Susan Chi
- Dana-Farber/Harvard Cancer Center, Boston, MA
| | | | | | - Chris Williams-Hughes
- Department of Preventive Medicine, University of Southern California, Los Angeles CA
| | - Maryam Fouladi
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Amar Gajjar
- Saint Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
17
|
Shrestha S, Morcavallo A, Gorrini C, Chesler L. Biological Role of MYCN in Medulloblastoma: Novel Therapeutic Opportunities and Challenges Ahead. Front Oncol 2021; 11:694320. [PMID: 34195095 PMCID: PMC8236857 DOI: 10.3389/fonc.2021.694320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022] Open
Abstract
The constitutive and dysregulated expression of the transcription factor MYCN has a central role in the pathogenesis of the paediatric brain tumour medulloblastoma, with an increased expression of this oncogene correlating with a worse prognosis. Consequently, the genomic and functional alterations of MYCN represent a major therapeutic target to attenuate tumour growth in medulloblastoma. This review will provide a comprehensive synopsis of the biological role of MYCN and its family components, their interaction with distinct signalling pathways, and the implications of this network in medulloblastoma development. We will then summarise the current toolbox for targeting MYCN and highlight novel therapeutic avenues that have the potential to results in better-tailored clinical treatments.
Collapse
Affiliation(s)
- Sumana Shrestha
- Division of Clinical Studies, Institute of Cancer Research (ICR), London and Royal Marsden NHS Trust, Sutton, United Kingdom
| | - Alaide Morcavallo
- Division of Clinical Studies, Institute of Cancer Research (ICR), London and Royal Marsden NHS Trust, Sutton, United Kingdom
| | - Chiara Gorrini
- Division of Clinical Studies, Institute of Cancer Research (ICR), London and Royal Marsden NHS Trust, Sutton, United Kingdom
| | - Louis Chesler
- Division of Clinical Studies, Institute of Cancer Research (ICR), London and Royal Marsden NHS Trust, Sutton, United Kingdom.,Division of Cancer Therapeutics, The Institute of Cancer Research (ICR), and The Royal Marsden NHS Trust, Sutton, United Kingdom
| |
Collapse
|
18
|
Papageorgiou GI, Tsakatikas SA, Fioretzaki RG, Kosmas C. Notable response of a young adult with recurrent glioblastoma multiforme to vincristine-irinotecan-temozolomide and bevacizumab. Anticancer Drugs 2021; 32:330-336. [PMID: 33229903 DOI: 10.1097/cad.0000000000001021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Glioblastoma multiforme is a malignant central nervous system (CNS) disease with dismal prognosis. Current treatment modalities only offer modest activity and usually of short duration, so there is an urgent need for the conduct of clinical trials exploring new treatment options and modalities. The vincristine-irinotecan-temozolomide and bevacizumab (VITb) regimen is an option of special interest, as it has produced encouraging results in young patients with various relapsed/refractory childhood and adolescence solid tumors, with an acceptable toxicity profile. With the current report, we present the case of a young male patient who was treated for GBM in second relapse at out institution, after previous surgical attempts and two radiotherapy sessions in conjunction with temozolomide and experienced a major and long-lasting response, weaned off steroids, to the VITb regimen followed by bevacizumab maintenance. The above case is discussed in the context of the existing literature regarding available evidence of synergy between the drugs used and the activity of certain components of the combination (i.e. combination of temozolomide-irinotecan ± vincristine, or bevacizumab-irinotecan in GBM) or the complete VITb regimen in other pediatric/adolescence solid tumors and the few cases reported with GBM.
Collapse
Affiliation(s)
- Georgios I Papageorgiou
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, Metaxa Cancer Hospital, Piraeus, Greece
| | | | | | | |
Collapse
|
19
|
孙 艳, 刘 晶, 杜 淑, 武 万, 孙 黎. [Survival of children with recurrent medulloblastoma undergoing sequential therapy: an analysis of 101 cases]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:164-168. [PMID: 33627212 PMCID: PMC7921531 DOI: 10.7499/j.issn.1008-8830.2010044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/18/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To study the clinical features of children with recurrent medulloblastoma (MB) and treatment regimens. METHODS A retrospective analysis was performed on 101 children with recurrent MB who were admitted to the hospital from August 1, 2011 to July 31, 2017. The children were followed up to July 31, 2020. The Kaplan-Meier method was used for survival analysis. The Cox regression model was used for multivariate regression analysis. RESULTS Of the 101 children, 95 underwent remission induction therapy, among whom 51 had response, resulting in a response rate of 54%. The median overall survival (OS) time after recurrence was 13 months, and the 1-, 3-, and 5-year OS rates were 50.5%±5.0%, 19.8%±4.0%, and 10%±3.3% respectively. There was no significant difference in the 5-year OS rate between the children with different ages (< 3 years or 3-18 years), sexes, pathological types, or Change stages, between the children with or without radiotherapy before recurrence or re-irradiation after recurrence, and between the children with different times to recurrence (< 12 months or ≥ 12 months after surgery) (P > 0.05). There were significant differences in the 5-year OS rate between the children with or without reoperation after recurrence and between the children with different recurrence sites (P < 0.05). The children with reoperation after recurrence had a significantly longer survival time than those without reoperation (P=0.007), and the risk of death in children undergoing reoperation after recurrence was 0.389 times (95% confidence interval:0.196-0.774) that in children who did not undergo such reoperation. CONCLUSIONS As for the recurrence of MB, although remission induction therapy again can achieve remission, such children still have a short survival time. Only reoperation can significantly prolong survival time, and therefore, early reoperation can be considered to improve the outcome of children with recurrent MB.
Collapse
Affiliation(s)
- 艳玲 孙
- />首都医科大学附属北京世纪坛医院儿科, 北京 100038Department of Pediatrics, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| | - 晶晶 刘
- />首都医科大学附属北京世纪坛医院儿科, 北京 100038Department of Pediatrics, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| | - 淑旭 杜
- />首都医科大学附属北京世纪坛医院儿科, 北京 100038Department of Pediatrics, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| | - 万水 武
- />首都医科大学附属北京世纪坛医院儿科, 北京 100038Department of Pediatrics, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| | - 黎明 孙
- />首都医科大学附属北京世纪坛医院儿科, 北京 100038Department of Pediatrics, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China
| |
Collapse
|
20
|
Eibl T, Hammer A, Yakubov E, Blechschmidt C, Kalisch A, Steiner HH. Medulloblastoma in adults - reviewing the literature from a surgeon's point of view. Aging (Albany NY) 2021; 13:3146-3160. [PMID: 33497354 PMCID: PMC7880386 DOI: 10.18632/aging.202568] [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: 08/03/2020] [Accepted: 01/04/2021] [Indexed: 06/12/2023]
Abstract
Medulloblastoma is a common primary brain tumor in children but it is a rare cancer in adult patients. We reviewed the literature, searching PubMed for articles on this rare tumor entity, with a focus on tumor biology, advanced neurosurgical opportunities for safe tumor resection, and multimodal treatment options. Adult medulloblastoma occurs at a rate of 0.6 per one million people per year. There is a slight disparity between male and female patients, and patients with a fair skin tone are more likely to have a medulloblastoma. Patients present with cerebellar signs and signs of elevated intracranial pressure. Diagnostic efforts should consist of cerebral MRI and MRI of the spinal axis. Cerebrospinal fluid should be investigated to look for tumor dissemination. Medulloblastoma tumors can be classified as classic, desmoplastic, anaplastic, and large cell, according to the WHO tumor classification. Molecular subgroups include WNT, SHH, group 3, and group 4 tumors. Further molecular analyses suggest that there are several subgroups within the four existing subgroups, with significant differences in patient age, frequency of metastatic spread, and patient survival. As molecular markers have started to play an increasing role in determining treatment strategies and prognosis, their importance has increased rapidly. Treatment options include microsurgical tumor resection and radiotherapy and, in addition, chemotherapy that respects the tumor biology of individual patients offers targeted therapeutic approaches. For neurosurgeons, intraoperative imaging and tumor fluorescence may improve resection rates. Disseminated disease, residual tumor after surgery, lower radiation dose, and low Karnofsky performance status are all suggestive of a poor outcome. Extraneural spread occurs only in very few cases. The reported 5-year-survival rates range between 60% and 80% for all adult medulloblastoma patients.
Collapse
Affiliation(s)
- Thomas Eibl
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg 90471, Bavaria, Germany
| | - Alexander Hammer
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg 90471, Bavaria, Germany
| | - Eduard Yakubov
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg 90471, Bavaria, Germany
| | - Cristiane Blechschmidt
- Department of Neuropathology, Paracelsus Medical University, Nuremberg 90471, Bavaria, Germany
| | - Alexander Kalisch
- Department of Oncology, Paracelsus Medical University, Nuremberg 90471, Bavaria, Germany
| | - Hans-Herbert Steiner
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg 90471, Bavaria, Germany
| |
Collapse
|
21
|
Ollauri-Ibáñez C, Astigarraga I. Use of Antiangiogenic Therapies in Pediatric Solid Tumors. Cancers (Basel) 2021; 13:E253. [PMID: 33445470 PMCID: PMC7827326 DOI: 10.3390/cancers13020253] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 12/23/2022] Open
Abstract
Cancer is an important cause of death in childhood. In recent years, scientists have made an important effort to achieve greater precision and more personalized treatments against cancer. But since only a few pediatric patients have identifiable therapeutic targets, other ways to stop the neoplastic cell proliferation and dissemination are needed. Therefore, the inhibition of general processes involved in the growth and behavior of tumors can be a relevant strategy for the development of new cancer therapies. In the case of solid tumors, one of these processes is angiogenesis, essential for tumor growth and generation of metastases. This review summarizes the results obtained with the use of antiangiogenic drugs in the main pediatric malignant solid tumors and also an overview of clinical trials currently underway. It should be noted that due to the rarity and heterogeneity of the different types of pediatric cancer, most studies on antiangiogenic drugs include only a small number of patients or isolated clinical cases, so they are not conclusive and further studies are needed.
Collapse
Affiliation(s)
- Claudia Ollauri-Ibáñez
- Pediatric Oncology Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
| | - Itziar Astigarraga
- Pediatric Oncology Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
- Pediatrics Department, Hospital Universitario Cruces, 48903 Barakaldo, Spain
- Pediatrics Department, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| |
Collapse
|
22
|
Hill RM, Richardson S, Schwalbe EC, Hicks D, Lindsey JC, Crosier S, Rafiee G, Grabovska Y, Wharton SB, Jacques TS, Michalski A, Joshi A, Pizer B, Williamson D, Bailey S, Clifford SC. Time, pattern, and outcome of medulloblastoma relapse and their association with tumour biology at diagnosis and therapy: a multicentre cohort study. THE LANCET CHILD & ADOLESCENT HEALTH 2020; 4:865-874. [PMID: 33222802 PMCID: PMC7671998 DOI: 10.1016/s2352-4642(20)30246-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 01/08/2023]
Abstract
Background Disease relapse occurs in around 30% of children with medulloblastoma, and is almost universally fatal. We aimed to establish whether the clinical and molecular characteristics of the disease at diagnosis are associated with the nature of relapse and subsequent disease course, and whether these associations could inform clinical management. Methods In this multicentre cohort study we comprehensively surveyed the clinical features of medulloblastoma relapse (time to relapse, pattern of relapse, time from relapse to death, and overall outcome) in centrally reviewed patients who relapsed following standard upfront therapies, from 16 UK Children's Cancer and Leukaemia Group institutions and four collaborating centres. We compared these relapse-associated features with clinical and molecular features at diagnosis, including established and recently described molecular features, prognostic factors, and treatment at diagnosis and relapse. Findings 247 patients (175 [71%] boys and 72 [29%] girls) with medulloblastoma relapse (median year of diagnosis 2000 [IQR 1995–2006]) were included in this study. 17 patients were later excluded from further analyses because they did not meet the age and treatment criteria for inclusion. Patients who received upfront craniospinal irradiation (irradiated group; 178 [72%] patients) had a more prolonged time to relapse compared with patients who did not receive upfront craniospinal irradiation (non-irradiated group; 52 [21%] patients; p<0·0001). In the non-irradiated group, craniospinal irradiation at relapse (hazard ratio [HR] 0·27, 95% CI 0·11–0·68) and desmoplastic/nodular histology (0·23, 0·07–0·77) were associated with prolonged time to death after relapse, MYC amplification was associated with a reduced overall survival (23·52, 4·85–114·05), and re-resection at relapse was associated with longer overall survival (0·17, 0·05–0·57). In the irradiated group, patients with MBGroup3 tumours relapsed significantly more quickly than did patients with MBGroup4 tumours (median 1·34 [0·99–1·89] years vs 2·04 [1·39–3·42 years; p=0·0043). Distant disease was prevalent in patients with MBGroup3 (23 [92%] of 25 patients) and MBGroup4 (56 [90%] of 62 patients) tumour relapses. Patients with distantly-relapsed MBGroup3 and MBGroup4 displayed both nodular and diffuse patterns of disease whereas isolated nodular relapses were rare in distantly-relapsed MBSHH (1 [8%] of 12 distantly-relapsed MBSHH were nodular alone compared with 26 [34%] of 77 distantly-relapsed MBGroup3 and MBGroup4). In MBGroup3 and MBGroup4, nodular disease was associated with a prolonged survival after relapse (HR 0·42, 0·21–0·81). Investigation of second-generation MBGroup3 and MBGroup4 molecular subtypes refined our understanding of heterogeneous relapse characteristics. Subtype VIII had prolonged time to relapse and subtype II had a rapid time from relapse to death. Subtypes II, III, and VIII developed a significantly higher incidence of distant disease at relapse whereas subtypes V and VII did not (equivalent rates to diagnosis). Interpretation This study suggests that the nature and outcome of medulloblastoma relapse are biology and therapy-dependent, providing translational opportunities for improved disease management through biology-directed disease surveillance, post-relapse prognostication, and risk-stratified selection of second-line treatment strategies. Funding Cancer Research UK, Action Medical Research, The Tom Grahame Trust, The JGW Patterson Foundation, Star for Harris, The Institute of Child Health - Newcastle University - Institute of Child Health High-Risk Childhood Brain Tumour Network (co-funded by The Brain Tumour Charity, Great Ormond Street Children's Charity, and Children with Cancer UK).
Collapse
Affiliation(s)
- Rebecca M Hill
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Stacey Richardson
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Edward C Schwalbe
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK; Department of Applied Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Debbie Hicks
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Janet C Lindsey
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Stephen Crosier
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Gholamreza Rafiee
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK; School of Electronics, Electrical Engineering and Computer Science, Queen's University Belfast, Centre for Cancer Research & Cell Biology, UK
| | - Yura Grabovska
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Thomas S Jacques
- Neural Development Unit, UCL Institute of Child Health, London, UK
| | - Antony Michalski
- Neural Development Unit, UCL Institute of Child Health, London, UK
| | - Abhijit Joshi
- Department of Neuropathology, Royal Victoria Infirmary, Newcastle University Teaching Hospitals, NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Barry Pizer
- Institute of Translational Research, University of Liverpool, Liverpool, UK
| | - Daniel Williamson
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Simon Bailey
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Steven C Clifford
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK.
| |
Collapse
|
23
|
Lim M, Shulman DS, Roberts H, Li A, Clymer J, Bona K, Al-Sayegh H, Ma C, DuBois SG. Off-label prescribing of targeted anticancer therapy at a large pediatric cancer center. Cancer Med 2020; 9:6658-6666. [PMID: 32750219 PMCID: PMC7520353 DOI: 10.1002/cam4.3349] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/25/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Background Off‐label drug prescribing is common in pediatric clinical medicine, though the extent and impact of this practice in pediatric oncology has not yet been characterized. Methods We completed a retrospective single‐institution cohort study evaluating prevalence, characteristics, and clinical outcomes of off‐label prescribing of 108 FDA‐approved targeted anticancer drugs in patients < 30 years old treated for cancer from 2007 to 2017. Dosing strategies were adjusted for body size and compared to FDA‐approved adult dosing regimen. A composite toxicity endpoint was defined as a patient having unplanned clinic visits, emergency department visits, or unplanned hospital admissions that were at least possibly related to the off‐label treatment. Results The overall prevalence of off‐label use of targeted therapies was 9.2% (n = 374 patients). The prevalence increased significantly over the study period (P < .0001). Patients treated off‐label were more likely to have neuro‐oncology diagnoses compared to patients not treated off‐label (46% vs 29%; P < .0001). Of the 108 potential agents, 38 (35%) were used by at least one patient. The median starting dose was below the FDA‐approved normalized dose for 44.4% of agents. Fifteen percent of patients had a complete response while receiving off‐label therapy, 38% experienced toxicity as defined, and 13% discontinued off‐label therapy due to toxicity. Conclusions In this real‐world evaluation of prescribing at a large pediatric cancer center, off‐label prescribing of FDA‐approved targeted therapies was common, increasing in prevalence, encompassed a broad sample of targeted agents, and was tolerable. Clinicians commonly start dosing below the equivalent FDA‐approved dose.
Collapse
Affiliation(s)
- Mir Lim
- Boston University School of Medicine, Boston, MA, USA.,Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - David S Shulman
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Holly Roberts
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Anran Li
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Jessica Clymer
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Kira Bona
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Hasan Al-Sayegh
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Clement Ma
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Xu Y, Li Q, Ma H, Sun T, Xiang R, Di F. Therapeutic effect and side effects of Bevacizumab combined with Irinotecan in the treatment of paediatric intracranial tumours: Meta-analysis and Systematic Review. J Clin Pharm Ther 2020; 45:1363-1371. [PMID: 32598559 PMCID: PMC7689821 DOI: 10.1111/jcpt.13228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/19/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Bevacizumab (BVZ) is an angiogenesis inhibitor that often works well with chemotherapeutic drugs for the treatment of solid intracranial tumours in children. This meta-analysis discusses the efficacy and side effects of BVZ combined with irinotecan in the treatment of patients (younger than 21 years of age) with recurrent, progressive or refractory intracranial tumours. METHODS We searched for articles published before 31 October 2018 in PubMed, EMBASE, Cochrane library and Web of Science. We selected relevant literature on the combination of BVZ and irinotecan in the treatment of children with intracranial tumours. Objective response was evaluated by combining partial response (PR), complete response (CR), stable disease (SD) and progressive disease (PD), and survival time was evaluated by combining overall survival (OS) and progression-free survival (PFS); common side effects were also analysed. All data included were obtained from single-arm data, with no control groups. RESULTS AND DISCUSSION A total of 13 studies involving 272 patients were included. We found that out of 41% patients who showed an objective response following the BVZ therapy combined with irinotecan, 28% achieved a PR, 13% achieved a CR, 32% showed a SD, and 43% had a PD; PFS and OS were 6.47 and 11.9 months, respectively; gastrointestinal dysfunction, leukopenia and hypertension were the three most common adverse events, accounting for 36.7%, 33.6% and 22.1%, respectively, whereas musculoskeletal disorders had the lowest occurrence, accounting for 3.9%. WHAT IS NEW AND CONCLUSION BVZ combined with irinotecan-based chemotherapy had a better response and prolonged survival in the treatment of paediatric intracranial tumours than radiation therapy or chemotherapy. Gastrointestinal dysfunction, leukopenia and hypertension were the toxic side effects with the highest incidence.
Collapse
Affiliation(s)
- Yan Xu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Qiang Li
- Department of NeurosurgeryXinle City HospitalXinleHebeiChina
| | - Hai‐Yang Ma
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Tao Sun
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Ruo‐Lan Xiang
- Department of Physiology and PathophysiologyPeking University School of Basic Medical SciencesBeijingChina
| | - Fei Di
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of NeurosurgeryZhangJiakou First HospitalZhangjiakouHebeiChina
| |
Collapse
|
25
|
Luzzi S, Giotta Lucifero A, Brambilla I, Semeria Mantelli S, Mosconi M, Foiadelli T, Savasta S. Targeting the medulloblastoma: a molecular-based approach. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:79-100. [PMID: 32608377 PMCID: PMC7975825 DOI: 10.23750/abm.v91i7-s.9958] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The lack of success of standard therapies for medulloblastoma has highlighted the need to plan a new therapeutic approach. The purpose of this article is to provide an overview of the novel treatment strategies based on the molecular characterization and risk categories of the medulloblastoma, also focusing on up-to-date relevant clinical trials and the challenges in translating tailored approaches into clinical practice. METHODS An online search of the literature was carried out on the PubMed/MEDLINE and ClinicalTrials.gov websites about molecular classification of medulloblastomas, ongoing clinical trials and new treatment strategies. Only articles in the English language and published in the last five years were selected. The research was refined based on the best match and relevance. RESULTS A total 58 articles and 51 clinical trials were analyzed. Trials were of phase I, II, and I/II in 55%, 33% and 12% of the cases, respectively. Target and adoptive immunotherapies were the treatment strategies for newly diagnosed and recurrent medulloblastoma in 71% and 29% of the cases, respectively. CONCLUSION Efforts are focused on the fine-tuning of target therapies and immunotherapies, including agents directed to specific pathways, engineered T-cells and oncoviruses. The blood-brain barrier, chemoresistance, the tumor microenvironment and cancer stem cells are the main translational challenges to be overcome in order to optimize medulloblastoma treatment, reduce the long-term morbidity and increase the overall survival.
Collapse
Affiliation(s)
- Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - Ilaria Brambilla
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Simona Semeria Mantelli
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Mario Mosconi
- Orthopaedic and Traumatology Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - Thomas Foiadelli
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Salvatore Savasta
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| |
Collapse
|
26
|
Franceschi E, Hofer S, Brandes AA, Frappaz D, Kortmann RD, Bromberg J, Dangouloff-Ros V, Boddaert N, Hattingen E, Wiestler B, Clifford SC, Figarella-Branger D, Giangaspero F, Haberler C, Pietsch T, Pajtler KW, Pfister SM, Guzman R, Stummer W, Combs SE, Seidel C, Beier D, McCabe MG, Grotzer M, Laigle-Donadey F, Stücklin ASG, Idbaih A, Preusser M, van den Bent M, Weller M, Hau P. EANO-EURACAN clinical practice guideline for diagnosis, treatment, and follow-up of post-pubertal and adult patients with medulloblastoma. Lancet Oncol 2020; 20:e715-e728. [PMID: 31797797 DOI: 10.1016/s1470-2045(19)30669-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/13/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022]
Abstract
The European Association of Neuro-Oncology (EANO) and EUropean RAre CANcer (EURACAN) guideline provides recommendations for the diagnosis, treatment, and follow-up of post-pubertal and adult patients with medulloblastoma. The guideline is based on the 2016 WHO classification of tumours of the CNS and on scientific developments published since 1980. It aims to provide direction for diagnostic and management decisions, and for limiting unnecessary treatments and cost. In view of the scarcity of data in adults with medulloblastoma, we base our recommendations on adult data when possible, but also include recommendations derived from paediatric data if justified. Our recommendations are a resource for professionals involved in the management of post-pubertal and adult patients with medulloblastoma, for patients and caregivers, and for health-care providers in Europe. The implementation of this guideline requires multidisciplinary structures of care, and defined processes of diagnosis and treatment.
Collapse
Affiliation(s)
- Enrico Franceschi
- Department of Medical Oncology, Azienda USL, Bologna, Italy; IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Silvia Hofer
- Division of Medical Oncology, Luzerner Kantonsspital, Luzern, Switzerland
| | - Alba A Brandes
- Department of Medical Oncology, Azienda USL, Bologna, Italy; IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Didier Frappaz
- Department of Neuro-Oncology and Institut d'Hématologie et d'Oncologie Pédiatrique, Centre Léon Bérard, Lyon, France
| | | | - Jacoline Bromberg
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, Netherlands
| | - Volodia Dangouloff-Ros
- Paediatric Radiology Department, Hôpital Necker Enfants Malades, Paris, France; UMR 1163, Imagine Institute, Paris, France
| | - Nathalie Boddaert
- Paediatric Radiology Department, Hôpital Necker Enfants Malades, Paris, France; UMR 1163, Imagine Institute, Paris, France
| | - Elke Hattingen
- Department of Neuroradiology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Benedikt Wiestler
- Department of Neuroradiology, Technical University of Munich Hospital, Munich, Germany
| | - Steven C Clifford
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Dominique Figarella-Branger
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Felice Giangaspero
- Department of Radiological, Oncological and Anatomopathological Sciences, Policlinico Umberto I, Sapienza University, Rome, Italy; IRCCS Neuromed, Mediterranean Neurological Institute, Pozzilli, Italy
| | - Christine Haberler
- Institute of Neurology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Torsten Pietsch
- Department of Neuropathology, DGNN Brain Tumour Reference Center, University of Bonn Medical Center, Bonn, Germany
| | - Kristian W Pajtler
- KiTZ Hopp Children's Cancer Center Heidelberg, Division of Pediatric Neurooncology, DKFZ German Cancer Research Center, DKTK German Cancer Consortium, and Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan M Pfister
- KiTZ Hopp Children's Cancer Center Heidelberg, Division of Pediatric Neurooncology, DKFZ German Cancer Research Center, DKTK German Cancer Consortium, and Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Raphael Guzman
- Department of Neurosurgery, Division of Pediatric Neurosurgery, University Hospital and University Children's Hospital, Basel, Switzerland
| | - Walter Stummer
- Department of Neurosurgery, University Hospital Muenster, Muenster, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Technical University of Munich, Munich, Germany; Institute of Radiation Medicine, Department of Radiation Sciences, Helmholtz Zentrum München, Munich, Germany
| | - Clemens Seidel
- Department of Radiation Oncology, University Hospital Leipzig, Leipzig, Germany
| | - Dagmar Beier
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Martin G McCabe
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Michael Grotzer
- Department of Oncology, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Florence Laigle-Donadey
- Service de Neurologie 2-Mazarin, Hôpitaux Universitaires La Pitié-Salpêtrière-Charles Foix, Paris, France
| | - Ana S Guerreiro Stücklin
- Department of Oncology, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Ahmed Idbaih
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - Matthias Preusser
- Division of Oncology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Martin van den Bent
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, Netherlands
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Peter Hau
- Wilhelm Sander-NeuroOncology Unit and Department of Neurology, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
27
|
Franceschi E, Frappaz D, Rudà R, Hau P, Preusser M, Houillier C, Lombardi G, Asioli S, Dehais C, Bielle F, Di Nunno V, van den Bent M, Brandes AA, Idbaih A. Rare Primary Central Nervous System Tumors in Adults: An Overview. Front Oncol 2020; 10:996. [PMID: 32676456 PMCID: PMC7333775 DOI: 10.3389/fonc.2020.00996] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
Overall, tumors of primary central nervous system (CNS) are quite common in adults with an incidence rate close to 30 new cases/100,000 inhabitants per year. Significant clinical and biological advances have been accomplished in the most common adult primary CNS tumors (i.e., diffuse gliomas). However, most CNS tumor subtypes are rare with an incidence rate below the threshold defining rare disease of 6.0 new cases/100,000 inhabitants per year. Close to 150 entities of primary CNS tumors have now been identified by the novel integrated histomolecular classification published by the World Health Organization (WHO) and its updates by the c-IMPACT NOW consortium (the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy). While these entities can be better classified into smaller groups either by their histomolecular features and/or by their location, assessing their treatment by clinical trials and improving the survival of patients remain challenging. Despite these tumors are rare, research, and advances remain slower compared to diffuse gliomas for instance. In some cases (i.e., ependymoma, medulloblastoma) the understanding is high because single or few driver mutations have been defined. The European Union has launched European Reference Networks (ERNs) dedicated to support advances on the clinical side of rare diseases including rare cancers. The ERN for rare solid adult tumors is termed EURACAN. Within EURACAN, Domain 10 brings together the European patient advocacy groups (ePAGs) and physicians dedicated to improving outcomes in rare primary CNS tumors and also aims at supporting research, care and teaching in the field. In this review, we discuss the relevant biological and clinical characteristics, clinical management of patients, and research directions for the following types of rare primary CNS tumors: medulloblastoma, pineal region tumors, glioneuronal and rare glial tumors, ependymal tumors, grade III meningioma and mesenchymal tumors, primary central nervous system lymphoma, germ cell tumors, spinal cord tumors and rare pituitary tumors.
Collapse
Affiliation(s)
- Enrico Franceschi
- Department of Medical Oncology, Azienda USL/IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Didier Frappaz
- Department of Neuro-Oncology and Institut d'Hématologie et d'Oncologie Pédiatrique, Centre Léon Bérard, Lyon, France
| | - Roberta Rudà
- Department of Neuro-Oncology, City of Health and Science and University of Turin, Turin, Italy
| | - Peter Hau
- Wilhelm Sander NeuroOncology-Unit, Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Caroline Houillier
- Sorbonne Université, IHU, ICM, Service de Neurologie 2-Mazarin, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Giuseppe Lombardi
- Department of Oncology, Veneto Institute of Oncology-IRCCS, Padua, Italy
| | - Sofia Asioli
- Section of Anatomic Pathology "M. Malpighi", Department of Biomedical and Neuromotor Sciences, Bellaria Hospital, Bologna, Italy
| | - Caroline Dehais
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - Franck Bielle
- Department of Neuropathology, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, AP-HP, Sorbonne Université, SIRIC Curamus, Paris, France
| | - Vincenzo Di Nunno
- Department of Medical Oncology, Azienda USL/IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Alba A Brandes
- Department of Medical Oncology, Azienda USL/IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Ahmed Idbaih
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | | |
Collapse
|
28
|
Adile AA, Kameda-Smith MM, Bakhshinyan D, Banfield L, Salim SK, Farrokhyar F, Fleming AJ. Salvage therapy for progressive, treatment-refractory or recurrent pediatric medulloblastoma: a systematic review protocol. Syst Rev 2020; 9:47. [PMID: 32127049 PMCID: PMC7055028 DOI: 10.1186/s13643-020-01307-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/20/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Central nervous system tumors remain the leading cause of cancer-related mortality amongst children with solid tumors, with medulloblastoma (MB) representing the most common pediatric brain malignancy. Despite best current therapies, patients with recurrent MB experience have an alarmingly high mortality rate and often have limited therapeutic options beyond inadequate chemotherapy or experimental clinical trials. Therefore, a systematic review of the literature regarding treatment strategies employed in recurrent pediatric MB will evaluate previous salvage therapies in order to guide future clinical trials. The aim of this systematic review will be to investigate the efficacy and safety of salvage therapies for the management of children with progressive, treatment-refractory, or recurrent MB. METHODS We will conduct literature searches (from 1995 onwards) in MEDLINE, EMBASE, ClinicalTrials.gov, WHO International Clinical Trials Registry Platform, and Cochrane Central Register of Controlled Trials. Studies examining the survival and toxicity of therapies administered to treatment-refractory pediatric MB patients will be included. Two reviewers will independently assess the search results based on predefined selection criteria, complete data abstraction, and quality assessment. The primary outcomes of this review will be overall and progression-free survival. Secondary outcomes will include safety and toxicity of each therapy administered. The study methodological quality (or bias) will be appraised using an appropriate tool. Due to the nature of the research question and published literature, we expect large inter-study heterogeneity and therefore will use random effects regression analysis to extract the combined effect. In additional analyses, we will investigate the role of re-irradiation and mono- vs. poly-therapy in recurrent disease, and whether molecular subgrouping of MB influences salvage therapy. DISCUSSION This systematic review will provide an overview of the current literature regarding salvage therapies for relapsed MB patients. Investigation of clinically tested therapies for children with recurrent MB has significant implications for clinical practice. By reviewing the efficacy and toxicity of MB salvage therapies, this study will identify effective therapeutic strategies administered to recurrent MB patients and can inform future clinical trials aimed to improve patient survivorship and quality of life. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42020167421.
Collapse
Affiliation(s)
- Ashley A. Adile
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Michelle M. Kameda-Smith
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON Canada
- Department of Surgery, Division of Neurosurgery, McMaster University, Hamilton, ON Canada
| | - David Bakhshinyan
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Laura Banfield
- Health Sciences Library, McMaster University, Hamilton, Ontario Canada
| | - Sabra K. Salim
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Forough Farrokhyar
- Department of Surgery, Division of Neurosurgery, McMaster University, Hamilton, ON Canada
- Department of Health, Evidence and Impact, McMaster University, Hamilton, Ontario Canada
| | - Adam J. Fleming
- Department of Pediatrics, Division of Hematology-Oncology, McMaster University, Hamilton, Canada
| |
Collapse
|
29
|
Wu WS, Liu JJ, Sun YL, DU SX, Li CD, Li M, Ren SQ, Zhang J, Gong XJ, Sun LM. [Effect of bevacizumab in treatment of children with optic pathway glioma]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:1193-1197. [PMID: 31874658 PMCID: PMC7389006 DOI: 10.7499/j.issn.1008-8830.2019.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/27/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To investigate the effect of bevacizumab in the treatment of children with optic pathway glioma (OPG). METHODS A retrospective analysis was performed for the clinical data of 30 children with OPG who underwent chemotherapy. According to whether bevacizumab was used, they were divided into conventional chemotherapy (carboplatin, vincristine and etoposide) group with 12 children and combined chemotherapy (bevacizumab, carboplatin, vincristine and etoposide) group with 18 children. The children were followed up to 6 months after chemotherapy, and the two groups were compared in terms of visual acuity and tumor size before and after chemotherapy and adverse reactions during chemotherapy. RESULTS The combined chemotherapy group had a significantly higher proportion of children achieving tumor regression than the conventional chemotherapy group (P<0.05), while there were no significant differences between the two groups in the proportion of children with improved visual acuity or adverse reactions (P>0.05). No chemotherapy-related death was observed in either group. CONCLUSIONS Bevacizumab combined with conventional chemotherapy can effectively reduce tumor size. Compared with conventional chemotherapy, such combination does not increase adverse reactions and can thus become a new direction for the treatment of OPG in children.
Collapse
Affiliation(s)
- Wan-Shui Wu
- Department of Pediatrics, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cañedo G, Solis I, González-San Segundo C, Madero L, Lassaletta A. Treatment of radiation-induced myelopathy with bevacizumab. Clin Transl Oncol 2019; 22:957-960. [PMID: 31571152 DOI: 10.1007/s12094-019-02209-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 09/13/2019] [Indexed: 10/25/2022]
Affiliation(s)
- G Cañedo
- Department of Pediatric Hematology-Oncology, Hospital Infantil Universitario del Niño Jesús, Avenida Menendez Pelayo 65, 28009, Madrid, Spain
| | - I Solis
- Department of Radiology, Hospital Infantil Universitario del Niño Jesús, Madrid, Spain
| | - C González-San Segundo
- Department of Radiation Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - L Madero
- Department of Pediatric Hematology-Oncology, Hospital Infantil Universitario del Niño Jesús, Avenida Menendez Pelayo 65, 28009, Madrid, Spain
| | - A Lassaletta
- Department of Pediatric Hematology-Oncology, Hospital Infantil Universitario del Niño Jesús, Avenida Menendez Pelayo 65, 28009, Madrid, Spain.
| |
Collapse
|
31
|
Casein kinase 2 inhibition sensitizes medulloblastoma to temozolomide. Oncogene 2019; 38:6867-6879. [PMID: 31406250 PMCID: PMC6800621 DOI: 10.1038/s41388-019-0927-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/21/2019] [Accepted: 05/26/2019] [Indexed: 11/20/2022]
Abstract
Medulloblastoma (MB) is the most common malignant pediatric brain tumor. Since surviving patients experience severe neurocognitive disabilities, better and more effective treatments are needed to enhance their quality of life. Casein Kinase 2 (CK2) is known to regulate cell growth and survival in multiple cancers; however, the role of CK2 in MB is currently being studied. In this study we verified the importance of CK2 in MB tumorigenesis and discovered that inhibition of CK2 using the small molecule inhibitor, CX-4945, can sensitize MB cells to a well-known and tolerated chemotherapeutic, temozolomide (TMZ). To study the role of CK2 in MB we modulated CK2 expression in multiple MB cell. Exogenous expression of CK2 enhanced cell growth and tumor growth in mice, while depletion or inhibition of CK2 expression decreased MB tumorigenesis. Treatment with CX-4945 reduced MB growth and increased apoptosis. We conducted a high-throughput screen where 4,000 small molecule compounds were analyzed to identify compounds that increased the anti-tumorigenic properties of CX-4945. TMZ was found to work synergistically with CX-4945 to decrease cell survival and increase apoptosis in MB cells. O-6-methylguanine-DNA methyltransferase (MGMT) activity is directly correlated to TMZ sensitivity. We found that loss of CK2 activity reduced β-catenin expression, a known MGMT regulator, which in turn led to a decrease in MGMT expression and an increased sensitivity to TMZ. Our findings show that CK2 is important for MB maintenance and that treatment with CX-4945 can sensitize MB cells to TMZ treatment.
Collapse
|
32
|
Bevacizumab-containing regimen in relapsed/progressed brain tumors: a single-institution experience. Childs Nerv Syst 2019; 35:1007-1012. [PMID: 30903281 DOI: 10.1007/s00381-019-04117-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/08/2019] [Indexed: 01/01/2023]
Abstract
AIM The aim of the study is to assess tumor response, treatment-related toxicities, progression-free survival (PFS), and overall survival (OS) in patients with relapsed/refractory brain tumors treated with bevacizumab-containing regimen. METHODS Patients that had received I and II line treatments with or without megatherapy were included. Doses and schedule were as follows: bevacizumab (BVZ) 10 mg/kg i.v. with irinotecan (IRI) 150 mg/m2 i.v. every 2 weeks ± temozolamide (TMZ) 200 mg/m2 p.o. daily for 5 days every 4 weeks. TMZ was omitted in heavily pretreated cases. RESULTS Between 2013 and 2018, 12 patients (3F/9M), median age 161 months (range 66-348), affected with medulloblastoma (n 7), or low-grade glioma (n 2), or high-grade glioma (n 3), received BVZ/IRI association (median courses 20, range 4-67); 3 of them continued single-agent BVZ (median courses 23, range 8-39). TMZ (median courses 8, range 2-26) was administered in eight patients and then stopped in three of them because of myelotoxicity or lack of compliance. Treatment was well tolerated. After 3 months, two complete responses, two partial responses, seven stable diseases, and one progressive disease were observed. Nine cases experienced an improvement in neurological symptoms. Median time to progression was 11 months (95% confidence interval, 4-18 months). Six-month and 2-year PFS were 75% and 42%, respectively. The OS is 33%; interestingly, two cases (one medulloblastoma and one high-grade glioma) are progression-free off-therapy since 30 and 48 months, respectively. CONCLUSIONS BVZ/IRI association ± TMZ showed encouraging therapeutic activity and low toxicity in this series of relapsed/refractory brain tumors.
Collapse
|
33
|
Vargas López AJ, Sola Vendrell E, Fernández Carballal C. Late recurrence of medulloblastoma after 17 years of complete remission. J Clin Neurosci 2019; 64:33-35. [PMID: 30905661 DOI: 10.1016/j.jocn.2019.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/11/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND To describe an exceptional case of late recurrence of medulloblastoma after 17 years of complete remission. CASE DESCRIPTION A 42-year-old male consulted in ER for 10-day occipital headache. He had a previous history of cerebellar medulloblastoma 17 years ago treated with gross total resection, chemotherapy and radiotherapy. During his yearly follow-up he had maintained complete remission. MRi showed a cerebellar mass suggestive of medulloblastoma recurrence vs radio-induced tumor. Craniotomy and complete resection of the tumor was performed. The anatomopathological analysis confirmed the recurrence of medulloblastoma. The patient received high dose of adjuvant chemotherapy and he maintains complete remission after 18 months. CONCLUSION Recurrence of medulloblastoma may occur despite more than 15 years of complete remission. Because of this fact it is mandatory to continue the follow-up of these patients. Aggressive management of recurrence is recommended in absence of disease dissemination.
Collapse
Affiliation(s)
- Antonio Jose Vargas López
- Neurological Surgery Department, Hospital General Universitario Gregorio Marañón, C/ Doctor Esquerdo, 46 28007 Madrid, Spain.
| | - Emma Sola Vendrell
- Pathology Department, Hospital General Universitario Gregorio Marañón, C/ Doctor Esquerdo, 46 28007 Madrid, Spain
| | - Carlos Fernández Carballal
- Neurological Surgery Department, Hospital General Universitario Gregorio Marañón, C/ Doctor Esquerdo, 46 28007 Madrid, Spain
| |
Collapse
|
34
|
Lu G, Rao M, Zhu P, Liang B, El-Nazer RT, Fonkem E, Bhattacharjee MB, Zhu JJ. Triple-drug Therapy With Bevacizumab, Irinotecan, and Temozolomide Plus Tumor Treating Fields for Recurrent Glioblastoma: A Retrospective Study. Front Neurol 2019; 10:42. [PMID: 30766509 PMCID: PMC6366009 DOI: 10.3389/fneur.2019.00042] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Clinical studies treating pediatric and adult solid tumors, such as glioblastoma (GBM), with a triple-drug regimen of temozolomide (TMZ), bevacizumab (BEV), and irinotecan (IRI) [TBI] have demonstrated various efficacies, but with no unexpected toxicities. The TBI regimen has never been studied in recurrent GBM (rGBM) patients. In this retrospective study, we investigated the outcomes and side effects of rGBM patients who had received the TBI regimen. We identified 48 adult rGBM patients with a median age of 56 years (range: 26-76), who received Tumor Treating Fields (TTFields) treatment for 30 days or longer, and concurrent salvage chemotherapies. The patients were classified into two groups based on chemotherapies received: TBI with TTFields (TBI+T, N = 18) vs. bevacizumab (BEV)-based chemotherapies with TTFields (BBC+T, N = 30). BBC regimens were either BEV monotherapy, BEV+IRI or BEV+CCNU. Patients in TBI+T group received on average 19 cycles of TMZ, 26 and 21 times infusions with BEV and IRI, respectively. Median overall survival (OS) and progression-free survival (PFS) for rGBM (OS-R and PFS-R) patients who received TBI+T were 18.9 and 10.7 months, respectively. In comparison, patients who received BBC+T treatment had OS-R and PFS-R of 11.8 (P > 0.05) and 4.7 (P < 0.05) months, respectively. Although the median PFS results were significantly different by 1.5 months (6.6 vs. 5.1) between TBI+T and BBC+T groups, the median OS difference of 14.7 months (32.5 vs. 17.8) was more pronounced, P < 0.05. Patients tolerated TBI+T or BBC+T treatments well and there were no unexpected toxicities. The most common side effects from TBI+T treatment included grade III hypertension (38.9%) and leukopenia (22.2%). In conclusion, the TBI regimen might play a role in the improvement of PFS-R and OS-R among rGBM patients. Prospective studies with a larger sample size are warranted to study the efficacy and toxicity of TBI+T regimen for rGBM.
Collapse
Affiliation(s)
- Guangrong Lu
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
| | - Mayank Rao
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
| | - Ping Zhu
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
- Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, UTHealth School of Public Health, Houston, TX, United States
| | - Buqing Liang
- Baylor Scott and White Health, Temple, TX, United States
| | | | - Ekokobe Fonkem
- Baylor Scott and White Health, Temple, TX, United States
| | - Meenakshi B. Bhattacharjee
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
| | - Jay-Jiguang Zhu
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, United States
| |
Collapse
|
35
|
Clinical Characteristics and Outcome of Children With Relapsed Medulloblastoma: A Retrospective Study at a Single Center in China. J Pediatr Hematol Oncol 2018; 40:598-604. [PMID: 29927794 DOI: 10.1097/mph.0000000000001241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Relapsed medulloblastoma (MB) has a dire prognosis, and chemotherapy remains the main therapeutic option. We retrospectively analyzed the clinical characteristics and survival rates of 60 Chinese children with relapsed MB. The patients received 11 cycles of chemotherapy in sequence, followed by 12 cycles of oral temozolomide and etoposide. Thirty patients were simultaneously administered intrathecal methotrexate (IT-MTX). The Kaplan-Meier method was used to determine survival rates; the patients' median survival time after relapse was 2.8 years, 5-year progression-free survival (PFS) and overall survival (OS) rates were 26.7%±5.7% and 31.6%±6.9%, respectively. There was no significant difference between these rates according to histology or molecular subgroup. Tumor cells were detected in the cerebrospinal fluid of over 40% of patients; such patients had significantly shorter OS and PFS rates. Patients who received IT-MTX showed significantly longer survival than those who did not (3.73 vs. 2.06 y, respectively, P=0.000); the corresponding 5-year PFS and OS rates were 43.3%±9.0% versus 10.0%±5.5% and 49.5%±11.1% versus 14.6%±6.9%, respectively (P=0.000). In addition, tumor cell-positive cerebrospinal fluid and IT-MTX use significantly influenced PFS and OS in relapsed patients. Taken together, our data show that IT-MTX improves the survival of patients with relapsed MB.
Collapse
|
36
|
Khatua S, Song A, Citla Sridhar D, Mack SC. Childhood Medulloblastoma: Current Therapies, Emerging Molecular Landscape and Newer Therapeutic Insights. Curr Neuropharmacol 2018; 16:1045-1058. [PMID: 29189165 PMCID: PMC6120114 DOI: 10.2174/1570159x15666171129111324] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 11/06/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022] Open
Abstract
Background: Medulloblastoma is the most common malignant brain tumor in children, currently treated uniformly based on histopathology and clinico-radiological risk stratification leading to unpredictable relapses and therapeutic failures. Identification of molecular subgroups have thrown light on the reasons for these and now reveals clues to profile molecularly based personalized therapy against these tumors. Methods: Research and online contents were evaluated for pediatric medulloblastoma which included latest information on the molecular subgroups and their clinical relevance and update on efforts to translate them into clinics. Results: Scientific endeavors over the last decade have clearly identified four molecular variants (WNT, SHH, Group 3, and Group 4) and their demographic, genomic, and epigenetic profile. Latest revelations include significant heterogeneity within these subgroups and 12 different subtypes of MB are now identified with disparate outcomes and biology. These findings have important implications for stratification and profiling future clinical trials against these formidable tumors. Conclusion: With the continued outpouring of genomic/epigenomic data of these molecular subgroups and evolution of further subtypes in each subgroup, the challenge lies in comprehensive evaluation of these informations. Current and future endeavors are now needed to profile personalized therapy for each child based on the molecular risk stratification of medulloblastoma, with a hope to improve survival outcome and reduce relapses.
Collapse
Affiliation(s)
- Soumen Khatua
- Department of Pediatrics, MD Anderson Cancer Center, The University of Texas Health Science Center at Houston, Pediatrics Houston, Texas, United States
| | - Anne Song
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, The University of Texas Health Science Center at Houston, Pediatrics Houston, Texas, United States
| | - Divyaswathi Citla Sridhar
- Department of Pediatrics, The University of Texas Health Science Center at Houston, Pediatrics Houston, Texas, United States
| | - Stephen C Mack
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, The University of Texas Health Science Center at Houston, Pediatrics Houston, Texas, United States
| |
Collapse
|
37
|
Thompson EM, Keir ST, Venkatraman T, Lascola C, Yeom KW, Nixon AB, Liu Y, Picard D, Remke M, Bigner DD, Ramaswamy V, Taylor MD. The role of angiogenesis in Group 3 medulloblastoma pathogenesis and survival. Neuro Oncol 2018; 19:1217-1227. [PMID: 28379574 DOI: 10.1093/neuonc/nox033] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Of the 4 medulloblastoma subgroups, Group 3 is the most aggressive but the importance of angiogenesis is unknown. This study sought to determine the role of angiogenesis and identify clinically relevant biomarkers of tumor vascularity and survival in Group 3 medulloblastoma. Methods VEGFA mRNA expression and survival from several patient cohorts were analyzed. Group 3 xenografts were implanted intracranially in nude rats. Dynamic susceptibility weighted (DSC) MRI and susceptibility weighted imaging (SWI) were obtained. DSC MRI was used to calculate relative cerebral blood volume (rCBV) and flow (rCBF). Tumor vessel density and rat vascular endothelial growth factor alpha (VEGFA) expression were determined. Results Patient VEGFA mRNA levels were significantly elevated in Group 3 compared with the other subgroups (P < 0.001) and associated with survival. Xenografts D283, D341, and D425 were identified as Group 3 by RNA hierarchical clustering and MYC amplification. The D283 group had the lowest rCBV and rCBF, followed by D341 and D425 (P < 0.05). These values corresponded to histological vessel density (P < 0.05), rat VEGFA expression (P < 0.05), and survival (P = 0.002). Gene set enrichment analysis identified 5 putative genes with expression profiles corresponding with these findings: RNH1, SCG2, VEGFA, AGGF1, and PROK2. SWI identified 3 xenograft-independent categories of intratumoral vascular architecture with distinct survival (P = 0.004): organized, diffuse microvascular, and heterogeneous. Conclusions Angiogenesis plays an important role in Group 3 medulloblastoma pathogenesis and survival. DSC MRI and SWI are clinically relevant biomarkers for tumor vascularity and overall survival and can be used to direct the use of antivascular therapies for patients with Group 3 medulloblastoma.
Collapse
Affiliation(s)
- Eric M Thompson
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Stephen T Keir
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Talaignair Venkatraman
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Christopher Lascola
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Kristen W Yeom
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Andrew B Nixon
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Yingmiao Liu
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Picard
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Marc Remke
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Darell D Bigner
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Vijay Ramaswamy
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Michael D Taylor
- Department of Neurosurgery, Duke University, Durham, North Carolina; Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina; Brain Imaging and Analysis Center, Duke University, Durham, North Carolina; Department of Radiology, Duke University, Durham, North Carolina; Department of Radiology, Stanford University, Palo Alto, California; Department of Medicine, Duke University, Durham, North Carolina; Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany; Department of Pathology, Duke University, Durham, North Carolina; Division of Haematology/Oncology, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumour Research Centre, Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Craveiro RB, Ehrhardt M, Velz J, Olschewski M, Goetz B, Pietsch T, Dilloo D. The anti-neoplastic activity of Vandetanib against high-risk medulloblastoma variants is profoundly enhanced by additional PI3K inhibition. Oncotarget 2017; 8:46915-46927. [PMID: 28159923 PMCID: PMC5564532 DOI: 10.18632/oncotarget.14911] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/26/2016] [Indexed: 12/26/2022] Open
Abstract
Medulloblastoma is comprised of at least four molecular subgroups with distinct clinical outcome (WHO classification 2016). SHH-TP53-mutated as well as MYC-amplified Non-WNT/Non-SHH medulloblastoma show the worst prognosis.Here we present evidence that single application of the multi-kinase inhibitor Vandetanib displays anti-neoplastic efficacy against cell lines derived from high-risk SHH-TP53-mutated and MYC-amplified Non-WNT/Non-SHH medulloblastoma. The narrow target spectrum of Vandetanib along with a favourable toxicity profile renders this drug ideal for multimodal treatment approaches. In this context our investigation documents that Vandetanib in combination with the clinically available PI3K inhibitor GDC-0941 leads to enhanced cytotoxicity against MYC-amplified and SHH-TP53-mutated medulloblastoma. In line with these findings we show for MYC-amplified medulloblastoma a profound reduction in activity of the oncogenes STAT3 and AKT. Furthermore, we document that Vandetanib and the standard chemotherapeutic Etoposide display additive anti-neoplastic efficacy in the investigated medulloblastoma cell lines that could be further enhanced by PI3K inhibition. Of note, the combination of Vandetanib, GDC-0941 and Etoposide results in MYC-amplified and SHH-TP53-mutated cell lines in complete loss of cell viability. Our findings therefore provide a rational to further evaluate Vandetanib in combination with PI3K inhibitors as well as standard chemotherapeutics in vivo for the treatment of most aggressive medulloblastoma variants.
Collapse
Affiliation(s)
- Rogerio B Craveiro
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, D-53113 Bonn, Germany
| | - Michael Ehrhardt
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, D-53113 Bonn, Germany
| | - Julia Velz
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, D-53113 Bonn, Germany
| | - Martin Olschewski
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, D-53113 Bonn, Germany
| | - Barbara Goetz
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, D-53113 Bonn, Germany
| | - Torsten Pietsch
- Department of Neuropathology, University of Bonn, D-53105 Bonn, Germany
| | - Dagmar Dilloo
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, University of Bonn Medical Center, D-53113 Bonn, Germany
| |
Collapse
|
39
|
Gauvain K, Ponisio MR, Barone A, Grimaldi M, Parent E, Leeds H, Goyal M, Rubin J, McConathy J. 18F-FDOPA PET/MRI for monitoring early response to bevacizumab in children with recurrent brain tumors. Neurooncol Pract 2017; 5:28-36. [PMID: 29692922 DOI: 10.1093/nop/npx008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Noninvasively predicting early response to therapy in recurrent pediatric brain tumors provides a challenge. 3,4-dihydroxy-6-[18F]fluoro-L-phenylalanine (18F-FDOPA) PET/MRI has not been previously studied as a tool to evaluate early response to antiangiogenic therapy in children. The purpose of this study was to evaluate the safety and feasibility of using 18F-FDOPA PET/MRI to assess response to bevacizumab in children with relapsed brain tumors. Materials and Methods Six patients with recurrent gliomas (5 low-grade, 1 high-grade) planned to undergo treatment with bevacizumab were enrolled. 18F-FDOPA PET/MRI scans were obtained prior to and 4 weeks following the start of treatment, and these were compared with the clinical response determined at the 3-month MRI. The primary PET measure was metabolic tumor volume (MTV) at 10 to 15 min after 18F-FDOPA injection. For each tumor, the MTV was determined by manually defining initial tumor volumes of interest (VOI) and then applying a 1.5-fold threshold relative to the mean standardized uptake value (SUV) of a VOI in the frontal lobe contralateral to the tumor. Results 18F-FDOPA PET/MRI was well tolerated by all patients. All tumors were well visualized with 18F-FDOPA on the initial study, with peak tumor uptake occurring approximately 10 min after injection. Maximum and mean SUVs as well as tumor-to-brain ratios were not predictors of response at 3 months. Changes in MTVs after therapy ranged from 23% to 98% (n = 5). There is a trend towards the percent MTV change seen on the 4-week scan correlating with progression-free survival. Conclusion 18F-FDOPA PET/MRI was well tolerated in pediatric patients and merits further investigation as an early predictor of response to therapy.
Collapse
Affiliation(s)
- Karen Gauvain
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO
| | - Maria Rosana Ponisio
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Amy Barone
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO
| | - Michael Grimaldi
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Ephraim Parent
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Hayden Leeds
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Manu Goyal
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Joshua Rubin
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO
| | - Jonathan McConathy
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,University of Alabama at Birmingham, Department of Radiology, Birmingham, AL
| |
Collapse
|
40
|
Srinivasan VM, Ghali MGZ, North RY, Boghani Z, Hansen D, Lam S. Modern management of medulloblastoma: Molecular classification, outcomes, and the role of surgery. Surg Neurol Int 2016; 7:S1135-S1141. [PMID: 28194300 PMCID: PMC5299153 DOI: 10.4103/2152-7806.196922] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/14/2016] [Indexed: 12/20/2022] Open
Affiliation(s)
- Visish M Srinivasan
- Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Texas, USA
| | - Michael G Z Ghali
- Department of Neurobiology, Drexel University College of Medicine, Philadelphia, USA
| | - Robert Y North
- Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Texas, USA
| | - Zain Boghani
- Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Texas, USA
| | - Daniel Hansen
- Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Texas, USA
| | - Sandi Lam
- Department of Neurosurgery, Baylor College of Medicine, Texas Children's Hospital, Texas, USA
| |
Collapse
|
41
|
Abstract
Medulloblastoma accounts for nearly 10% of all childhood brain tumors. These tumors occur exclusively in the posterior fossa and have the potential for leptomeningeal spread. Treatment includes a combination of surgery, radiation therapy (in patients >3 years old). Patients >3 years old are stratified based on the volume of postoperative residual tumor and the presence or absence of metastases into "standard risk" and "high risk" categories with long-term survival rates of approximately 85% and 70%, respectively. Outcomes are inferior in infants and children younger than 3 years with exception of those patients with the medulloblastoma with extensive nodularity histologic subtype. Treatment for medulloblastoma is associated with significant morbidity, especially in the youngest patients. Recent molecular subclassification of medulloblastoma has potential prognostic and therapeutic implications. Future incorporation of molecular subgroups into treatment protocols will hopefully improve both survival outcomes and posttreatment quality of life.
Collapse
Affiliation(s)
- Nathan E Millard
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin C De Braganca
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
42
|
Hepatotoxicity by combination treatment of temozolomide, artesunate and Chinese herbs in a glioblastoma multiforme patient: case report review of the literature. Arch Toxicol 2016; 91:1833-1846. [DOI: 10.1007/s00204-016-1810-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 08/04/2016] [Indexed: 10/21/2022]
|
43
|
Bonney PA, Santucci JA, Maurer AJ, Sughrue ME, McNall-Knapp RY, Battiste JD. Dramatic response to temozolomide, irinotecan, and bevacizumab for recurrent medulloblastoma with widespread osseous metastases. J Clin Neurosci 2016; 26:161-3. [DOI: 10.1016/j.jocn.2015.10.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/25/2015] [Indexed: 01/23/2023]
|
44
|
Cellular and Antibody Based Approaches for Pediatric Cancer Immunotherapy. J Immunol Res 2015; 2015:675269. [PMID: 26587548 PMCID: PMC4637498 DOI: 10.1155/2015/675269] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/31/2015] [Indexed: 11/17/2022] Open
Abstract
Progress in the use of traditional chemotherapy and radiation-based strategies for the treatment of pediatric malignancies has plateaued in the past decade, particularly for patients with relapsing or therapy refractory disease. As a result, cellular and humoral immunotherapy approaches have been investigated for several childhood cancers. Several monoclonal antibodies are now FDA approved and commercially available, some of which are currently considered standard of practice. There are also several new cellular immunotherapy approaches under investigation, including chimeric antigen receptor (CAR) modified T cells, cancer vaccines and adjuvants, and natural killer (NK) cell therapies. In this review, we will discuss previous studies on pediatric cancer immunotherapy and new approaches that are currently being investigated in clinical trials.
Collapse
|
45
|
Novo J, Bitterman P, Guirguis A. Central-type primitive neuroectodermal tumor of the uterus: Case report of remission of stage IV disease using adjuvant cisplatin/etoposide/bevacizumab chemotherapy and review of the literature. Gynecol Oncol Rep 2015; 14:26-30. [PMID: 26793768 PMCID: PMC4688884 DOI: 10.1016/j.gore.2015.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/30/2015] [Accepted: 09/13/2015] [Indexed: 12/21/2022] Open
Abstract
Bevacizumab was an effective agent in one case of advanced uterine PNET. VEGF was expressed by the tumor, supporting a mechanism for effectiveness. Cisplatin/etoposide/bevacizumab should be further studied in clinical trials. Patient remains disease-free forty-eight months following intervention.
Collapse
Affiliation(s)
- Jorge Novo
- Rush University Medical Center, Department of Pathology, 1653 West Congress Parkway, 570 Jelke, Chicago, IL 60612, United States
| | - Pincas Bitterman
- Rush University Medical Center, Department of Pathology, 1653 West Congress Parkway, 570 Jelke, Chicago, IL 60612, United States
| | - Alfred Guirguis
- Gynecologic Cancer Institute of Chicago, 5716 W 95th Street, Oak Lawn, IL 60453, United States
| |
Collapse
|
46
|
Millan NC, Poveda MJ, Cruz O, Mora J. Safety of bevacizumab in patients younger than 4 years of age. Clin Transl Oncol 2015; 18:464-8. [PMID: 26318053 DOI: 10.1007/s12094-015-1389-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/17/2015] [Indexed: 12/28/2022]
Abstract
PURPOSE Limited data exist regarding the safety and efficacy of bevacizumab in pediatric patients under the age of 4 years. Here, we report a large cohort of pediatric patients under 4 years of age treated with bevacizumab. METHODS The primary objective was to document adverse events with a possible relationship to bevacizumab. Patients (n = 16) were identified through retrospective chart review and harbored a variety of conditions (44% central nervous system (CNS) tumors, 31% vascular anomalies, 13% neuroblastoma, 12% other). RESULTS The median age was 34.3 months (range 4.9-47.3), including five patients <2 years of age. Patients received bevacizumab for a median duration of 6.2 months, alone or with chemotherapy, and a median dose of 9.25 mg/kg (range 7.0-11.8). Partial responses were seen in 19% of patients, and clinical improvements were seen in 69%. Adverse events known to be associated with bevacizumab occurred in 37%. Outcomes observed in this population resemble those reported for bevacizumab in older pediatric patients. The overall pattern and frequency of adverse events observed was similar to those seen in reports of older pediatric patients with a variety of conditions. The highest level of efficacy observed was seen among patients with vascular malformations or with low-grade CNS tumors. CONCLUSIONS Our results suggest that the use of bevacizumab is safe for the youngest children.
Collapse
Affiliation(s)
- N C Millan
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu num 2, Esplugues del Llobregat, 08950, Barcelona, Spain
| | - M J Poveda
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu num 2, Esplugues del Llobregat, 08950, Barcelona, Spain
| | - O Cruz
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu num 2, Esplugues del Llobregat, 08950, Barcelona, Spain
| | - J Mora
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu num 2, Esplugues del Llobregat, 08950, Barcelona, Spain.
| |
Collapse
|
47
|
Capitini CM, Otto M, DeSantes KB, Sondel PM. Immunotherapy in pediatric malignancies: current status and future perspectives. Future Oncol 2015; 10:1659-78. [PMID: 25145434 DOI: 10.2217/fon.14.62] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Novel immune-based therapies are becoming available as additions to, and in some cases as alternatives to, the traditional treatment modalities such as chemotherapy, surgery and radiation that have improved outcomes for childhood cancer for decades. In this article, we will discuss what immunotherapies are being tested in the clinic, barriers to widespread application, and the future of immuno-oncology for childhood cancer. While in many cases, these therapies have shown dramatic responses in the setting of refractory or relapsed cancer, much remains to be learned about how to integrate these therapies into existing upfront regimens. The progress and challenges of developing immunotherapies for childhood cancer in a timely and cost-effective fashion will be discussed.
Collapse
Affiliation(s)
- Christian M Capitini
- Department of Pediatrics & Carbone Cancer Center, University of Wisconsin School of Medicine & Public Health, 1111 Highland Avenue, WIMR 4137, Madison, WI 53705, USA
| | | | | | | |
Collapse
|
48
|
Saletta F, Wadham C, Ziegler DS, Marshall GM, Haber M, McCowage G, Norris MD, Byrne JA. Molecular profiling of childhood cancer: Biomarkers and novel therapies. BBA CLINICAL 2014; 1:59-77. [PMID: 26675306 PMCID: PMC4633945 DOI: 10.1016/j.bbacli.2014.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 06/16/2014] [Accepted: 06/24/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Technological advances including high-throughput sequencing have identified numerous tumor-specific genetic changes in pediatric and adolescent cancers that can be exploited as targets for novel therapies. SCOPE OF REVIEW This review provides a detailed overview of recent advances in the application of target-specific therapies for childhood cancers, either as single agents or in combination with other therapies. The review summarizes preclinical evidence on which clinical trials are based, early phase clinical trial results, and the incorporation of predictive biomarkers into clinical practice, according to cancer type. MAJOR CONCLUSIONS There is growing evidence that molecularly targeted therapies can valuably add to the arsenal available for treating childhood cancers, particularly when used in combination with other therapies. Nonetheless the introduction of molecularly targeted agents into practice remains challenging, due to the use of unselected populations in some clinical trials, inadequate methods to evaluate efficacy, and the need for improved preclinical models to both evaluate dosing and safety of combination therapies. GENERAL SIGNIFICANCE The increasing recognition of the heterogeneity of molecular causes of cancer favors the continued development of molecularly targeted agents, and their transfer to pediatric and adolescent populations.
Collapse
Key Words
- ALK, anaplastic lymphoma kinase
- ALL, acute lymphoblastic leukemia
- AML, acute myeloid leukemia
- ARMS, alveolar rhabdomyosarcoma
- AT/RT, atypical teratoid/rhabdoid tumor
- AURKA, aurora kinase A
- AURKB, aurora kinase B
- BET, bromodomain and extra terminal
- Biomarkers
- CAR, chimeric antigen receptor
- CML, chronic myeloid leukemia
- Childhood cancer
- DFMO, difluoromethylornithine
- DIPG, diffuse intrinsic pontine glioma
- EGFR, epidermal growth factor receptor
- ERMS, embryonal rhabdomyosarcoma
- HDAC, histone deacetylases
- Hsp90, heat shock protein 90
- IGF-1R, insulin-like growth factor type 1 receptor
- IGF/IGFR, insulin-like growth factor/receptor
- Molecular diagnostics
- NSCLC, non-small cell lung cancer
- ODC1, ornithine decarboxylase 1
- PARP, poly(ADP-ribose) polymerase
- PDGFRA/B, platelet derived growth factor alpha/beta
- PI3K, phosphatidylinositol 3′-kinase
- PLK1, polo-like kinase 1
- Ph +, Philadelphia chromosome-positive
- RMS, rhabdomyosarcoma
- SHH, sonic hedgehog
- SMO, smoothened
- SYK, spleen tyrosine kinase
- TOP1/TOP2, DNA topoisomerase 1/2
- TRAIL, TNF-related apoptosis-inducing ligand
- Targeted therapy
- VEGF/VEGFR, vascular endothelial growth factor/receptor
- mAb, monoclonal antibody
- mAbs, monoclonal antibodies
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Federica Saletta
- Children's Cancer Research Unit, Kids Research Institute, Westmead 2145, New South Wales, Australia
| | - Carol Wadham
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Randwick 2031, New South Wales, Australia
| | - David S. Ziegler
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Randwick 2031, New South Wales, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia
| | - Glenn M. Marshall
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Randwick 2031, New South Wales, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Randwick 2031, New South Wales, Australia
| | - Geoffrey McCowage
- The Children's Hospital at Westmead, Westmead 2145, New South Wales, Australia
| | - Murray D. Norris
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Randwick 2031, New South Wales, Australia
| | - Jennifer A. Byrne
- Children's Cancer Research Unit, Kids Research Institute, Westmead 2145, New South Wales, Australia
- The University of Sydney Discipline of Paediatrics and Child Health, The Children's Hospital at Westmead, Westmead 2145, New South Wales, Australia
| |
Collapse
|
49
|
Overcoming multiple drug resistance mechanisms in medulloblastoma. Acta Neuropathol Commun 2014; 2:57. [PMID: 24887326 PMCID: PMC4229867 DOI: 10.1186/2051-5960-2-57] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 05/17/2014] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Medulloblastoma (MB) is the most common malignant paediatric brain tumour. Recurrence and progression of disease occurs in 15-20% of standard risk and 30-40% of high risk patients. We analysed whether circumvention of chemoresistance pathways (drug export, DNA repair and apoptotic inhibition) can restore chemotherapeutic efficacy in a panel of MB cell lines. RESULTS We demonstrate, by immunohistochemistry in patient tissue microarrays, that ABCB1 is expressed in 43% of tumours and is significantly associated with high-risk. We show that ABCB1, O6-methylguanine-DNA-methyltransferase (MGMT) and BCL2 family members are differentially expressed (by quantitative reverse transcription polymerase chain reaction, Western blotting and flow cytometry) in MB cell lines. Based on these findings, each pathway was then inhibited or circumvented and cell survival assessed using clonogenic assays. Inhibition of ABCB1 using vardenafil or verapamil resulted in a significant increase in sensitivity to etoposide in ABCB1-expressing MB cell lines. Sensitivity to temozolomide (TMZ) was MGMT-dependent, but two novel imidazotetrazine derivatives (N-3 sulfoxide and N-3 propargyl TMZ analogues) demonstrated ≥7 fold and ≥3 fold more potent cytotoxicity respectively compared to TMZ in MGMT-expressing MB cell lines. Activity of the BAD mimetic ABT-737 was BCL2A1 and ABCB1 dependent, whereas the pan-BCL2 inhibitor obatoclax was effective as a single cytotoxic agent irrespective of MCL1, BCL2, BCL2A1, or ABCB1 expression. CONCLUSIONS ABCB1 is associated with high-risk MB; hence, inhibition of ABCB1 by vardenafil may represent a valid approach in these patients. Imidazotetrazine analogues of TMZ and the BH3 mimetic obatoclax are promising clinical candidates in drug resistant MB tumours expressing MGMT and BCL2 anti-apoptotic members respectively.
Collapse
|
50
|
Recent developments and current concepts in medulloblastoma. Cancer Treat Rev 2014; 40:356-65. [DOI: 10.1016/j.ctrv.2013.11.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 11/26/2013] [Accepted: 11/29/2013] [Indexed: 12/21/2022]
|