1
|
Jiang J, Li WB, Xiao SW. Prognostic factors analysis of diffuse midline glioma. J Neurooncol 2024; 167:285-292. [PMID: 38381257 PMCID: PMC11023999 DOI: 10.1007/s11060-024-04605-6] [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: 01/05/2024] [Accepted: 02/12/2024] [Indexed: 02/22/2024]
Abstract
PURPOSE This study retrospectively analyzes cases of diffuse midline glioma treated with radiotherapy, with the aim of investigating the prognosis of the tumor and its influencing factors. METHODS From January 2018 to November 2022, we treated 64 patients who were pathologically diagnosed with diffuse midline glioma. Among them, 41 underwent surgical resection, and 23 underwent biopsy procedures. All patients received postoperative radiotherapy. We followed up with the patients to determine the overall survival rate and conducted univariate and multivariate analyses on relevant indicators. RESULTS The median survival time for the entire patient group was 33.3 months, with overall survival rates of 92.9%, 75.4%, and 45.0% at 1 year, 2 years, and 3 years, respectively. Univariate and multivariate analyses indicated that older patients had a better prognosis. CONCLUSION Patient age is an independent prognostic factor for patients with diffuse midline glioma undergoing radiation therapy.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100071, China
- Department of Radiation Oncology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Wen-Bin Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100071, China.
| | - Shao-Wen Xiao
- Department of Radiation Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
2
|
Malik JR, Podany AT, Khan P, Shaffer CL, Siddiqui JA, Baranowska‐Kortylewicz J, Le J, Fletcher CV, Ether SA, Avedissian SN. Chemotherapy in pediatric brain tumor and the challenge of the blood-brain barrier. Cancer Med 2023; 12:21075-21096. [PMID: 37997517 PMCID: PMC10726873 DOI: 10.1002/cam4.6647] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/18/2023] [Accepted: 10/12/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Pediatric brain tumors (PBT) stand as the leading cause of cancer-related deaths in children. Chemoradiation protocols have improved survival rates, even for non-resectable tumors. Nonetheless, radiation therapy carries the risk of numerous adverse effects that can have long-lasting, detrimental effects on the quality of life for survivors. The pursuit of chemotherapeutics that could obviate the need for radiotherapy remains ongoing. Several anti-tumor agents, including sunitinib, valproic acid, carboplatin, and panobinostat, have shown effectiveness in various malignancies but have not proven effective in treating PBT. The presence of the blood-brain barrier (BBB) plays a pivotal role in maintaining suboptimal concentrations of anti-cancer drugs in the central nervous system (CNS). Ongoing research aims to modulate the integrity of the BBB to attain clinically effective drug concentrations in the CNS. However, current findings on the interaction of exogenous chemical agents with the BBB remain limited and do not provide a comprehensive explanation for the ineffectiveness of established anti-cancer drugs in PBT. METHODS We conducted our search for chemotherapeutic agents associated with the blood-brain barrier (BBB) using the following keywords: Chemotherapy in Cancer, Chemotherapy in Brain Cancer, Chemotherapy in PBT, BBB Inhibition of Drugs into CNS, Suboptimal Concentration of CNS Drugs, PBT Drugs and BBB, and Potential PBT Drugs. We reviewed each relevant article before compiling the information in our manuscript. For the generation of figures, we utilized BioRender software. FOCUS We focused our article search on chemical agents for PBT and subsequently investigated the role of the BBB in this context. Our search criteria included clinical trials, both randomized and non-randomized studies, preclinical research, review articles, and research papers. FINDING Our research suggests that, despite the availability of potent chemotherapeutic agents for several types of cancer, the effectiveness of these chemical agents in treating PBT has not been comprehensively explored. Additionally, there is a scarcity of studies examining the role of the BBB in the suboptimal outcomes of PBT treatment, despite the effectiveness of these drugs for other types of tumors.
Collapse
Affiliation(s)
- Johid Reza Malik
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Anthony T. Podany
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
- Pediatric Clinical Pharmacology ProgramChild Health Research Institute, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Parvez Khan
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Christopher L. Shaffer
- Pediatric Clinical Pharmacology ProgramChild Health Research Institute, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Jawed A. Siddiqui
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | | | - Jennifer Le
- University of California San Diego Skaggs School of Pharmacy and Pharmaceutical SciencesSan DiegoCaliforniaUSA
| | - Courtney V. Fletcher
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Sadia Afruz Ether
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Sean N. Avedissian
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
- Pediatric Clinical Pharmacology ProgramChild Health Research Institute, University of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
3
|
Wang LM, Englander ZK, Miller ML, Bruce JN. Malignant Glioma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1405:1-30. [PMID: 37452933 DOI: 10.1007/978-3-031-23705-8_1] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
This chapter provides a comprehensive overview of malignant gliomas, the most common primary brain tumor in adults. These tumors are varied in their cellular origin, genetic profile, and morphology under the microscope, but together they share some of the most dismal prognoses of all neoplasms in the body. Although there is currently no cure for malignant glioma, persistent efforts to improve outcomes in patients with these tumors have led to modest increases in survival, and researchers worldwide continue to strive toward a deeper understanding of the factors that influence glioma development and response to treatment. In addition to well-established epidemiology, clinical manifestations, and common histopathologic and radiologic features of malignant gliomas, this section considers recent advances in molecular biology that have led to a more nuanced understanding of the genetic changes that characterize the different types of malignant glioma, as well as their implications for treatment. Beyond the traditional classification of malignant gliomas based on histopathological features, this chapter incorporates the World Health Organization's 2016 criteria for the classification of brain tumors, with special focus on disease-defining genetic alterations and newly established subcategories of malignant glioma that were previously unidentifiable based on microscopic examination alone. Traditional therapeutic modalities that form the cornerstone of treatment for malignant glioma, such as aggressive surgical resection followed by adjuvant chemotherapy and radiation therapy, and the studies that support their efficacy are reviewed in detail. This provides a foundation for additional discussion of novel therapeutic methods such as immunotherapy and convection-enhanced delivery, as well as new techniques for enhancing extent of resection such as fluorescence-guided surgery.
Collapse
Affiliation(s)
- Linda M Wang
- Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Michael L Miller
- Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jeffrey N Bruce
- Department of Neurosurgery, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
4
|
Immunogenic Cell Death Enhances Immunotherapy of Diffuse Intrinsic Pontine Glioma: From Preclinical to Clinical Studies. Pharmaceutics 2022; 14:pharmaceutics14091762. [PMID: 36145510 PMCID: PMC9502387 DOI: 10.3390/pharmaceutics14091762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/02/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is the most lethal tumor involving the pediatric central nervous system. The median survival of children that are diagnosed with DIPG is only 9 to 11 months. More than 200 clinical trials have failed to increase the survival outcomes using conventional cytotoxic or myeloablative chemotherapy. Immunotherapy presents exciting therapeutic opportunities against DIPG that is characterized by unique and heterogeneous features. However, the non-inflammatory DIPG microenvironment greatly limits the role of immunotherapy in DIPG. Encouragingly, the induction of immunogenic cell death, accompanied by the release of damage-associated molecular patterns (DAMPs) shows satisfactory efficacy of immune stimulation and antitumor strategies. This review dwells on the dilemma and advances in immunotherapy for DIPG, and the potential efficacy of immunogenic cell death (ICD) in the immunotherapy of DIPG.
Collapse
|
5
|
Carcamo B, Francia G. Cyclic Metronomic Chemotherapy for Pediatric Tumors: Six Case Reports and a Review of the Literature. J Clin Med 2022; 11:jcm11102849. [PMID: 35628975 PMCID: PMC9144744 DOI: 10.3390/jcm11102849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/06/2022] [Accepted: 05/13/2022] [Indexed: 12/03/2022] Open
Abstract
We report a retrospective case series of six Hispanic children with tumors treated with metronomic chemotherapy. The six cases comprised one rhabdoid tumor of the kidney, one ependymoma, two medulloblastomas, one neuroblastoma, and a type II neurocytoma of the spine. Treatment included oral cyclophosphamide daily for 21 days alternating with oral etoposide daily for 21 days in a backbone of daily valproic acid and celecoxib. In one case, celecoxib was substituted with sulindac. Of the six patients, three showed complete responses, and all patients showed some response to metronomic therapy with only minor hematologic toxicity. One patient had hemorrhagic gastritis likely associated with NSAIDs while off prophylactic antacids. These data add to a growing body of evidence suggesting that continuous doses of valproic acid and celecoxib coupled with alternating metronomic chemotherapy of agents such as etoposide and cyclophosphamide can produce responses in pediatric tumors relapsing to conventional dose chemotherapy.
Collapse
Affiliation(s)
- Benjamin Carcamo
- Department of Pediatric Hematology Oncology, El Paso Children’s Hospital, El Paso, TX 79905, USA
- Department of Pediatrics, Texas Tech University Health Science Center, El Paso, TX 79430, USA
- Correspondence: (B.C.); (G.F.); Tel.: +1-915-479-8970 (B.C.); +1-915-747-8025 (G.F.); Fax: +1-915-242-8437 (B.C.); +1-915-747-5808 (G.F.)
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso (UTEP), El Paso, TX 79968, USA
- Correspondence: (B.C.); (G.F.); Tel.: +1-915-479-8970 (B.C.); +1-915-747-8025 (G.F.); Fax: +1-915-242-8437 (B.C.); +1-915-747-5808 (G.F.)
| |
Collapse
|
6
|
Cazzaniga ME, Cordani N, Capici S, Cogliati V, Riva F, Cerrito MG. Metronomic Chemotherapy. Cancers (Basel) 2021; 13:cancers13092236. [PMID: 34066606 PMCID: PMC8125766 DOI: 10.3390/cancers13092236] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The present article reviews the state of the art of metronomic chemotherapy use to treat the principal types of cancers, namely breast, non-small cell lung cancer and colorectal ones, and of the most recent progresses in understanding the underlying mechanisms of action. Areas of novelty, in terms of new regimens, new types of cancer suitable for Metronomic chemotherapy (mCHT) and the overview of current ongoing trials, along with a critical review of them, are also provided. Abstract Metronomic chemotherapy treatment (mCHT) refers to the chronic administration of low doses chemotherapy that can sustain prolonged, and active plasma levels of drugs, producing favorable tolerability and it is a new promising therapeutic approach in solid and in hematologic tumors. mCHT has not only a direct effect on tumor cells, but also an action on cell microenvironment, by inhibiting tumor angiogenesis, or promoting immune response and for these reasons can be considered a multi-target therapy itself. Here we review the state of the art of mCHT use in some classical tumour types, such as breast and no small cell lung cancer (NSCLC), see what is new regarding most recent data in different cancer types, such as glioblastoma (GBL) and acute myeloid leukemia (AML), and new drugs with potential metronomic administration. Finally, a look at the strategic use of mCHT in the context of health emergencies, or in low –and middle-income countries (LMICs), where access to adequate healthcare is often not easy, is mandatory, as we always need to bear in in mind that equity in care must be a compulsory part of our medical work and research.
Collapse
Affiliation(s)
- Marina Elena Cazzaniga
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy;
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.)
- Correspondence: (M.E.C.); (M.G.C.); Tel.: +39-0392-339-037 (M.E.C.)
| | - Nicoletta Cordani
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy;
| | - Serena Capici
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.)
| | - Viola Cogliati
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.)
| | - Francesca Riva
- Unit of Clinic Oncology, ASST-Monza (MB), 20900 Monza, Italy;
| | - Maria Grazia Cerrito
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy;
- Correspondence: (M.E.C.); (M.G.C.); Tel.: +39-0392-339-037 (M.E.C.)
| |
Collapse
|
7
|
Yoon HI, Wee CW, Kim YZ, Seo Y, Im JH, Dho YS, Kim KH, Hong JB, Park JS, Choi SH, Kim MS, Moon J, Hwang K, Park JE, Cho JM, Yoon WS, Kim SH, Kim YI, Kim HS, Sung KS, Song JH, Lee MH, Han MH, Lee SH, Chang JH, Lim DH, Park CK, Lee YS, Gwak HS. The Korean Society for Neuro-Oncology (KSNO) Guideline for Adult Diffuse Midline Glioma: Version 2021.1. Brain Tumor Res Treat 2021; 9:1-8. [PMID: 33913265 PMCID: PMC8082289 DOI: 10.14791/btrt.2021.9.e8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 02/15/2021] [Accepted: 03/03/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND There have been no guidelines for the management of adult patients with diffuse midline glioma (DMG), H3K27M-mutant in Korea since the 2016 revised WHO classification newly defined this disease entity. Thus, the Korean Society for Neuro-Oncology (KSNO), a multidisciplinary academic society, had begun preparing guidelines for DMG since 2019. METHODS The Working Group was composed of 27 multidisciplinary medical experts in Korea. References were identified through searches of PubMed, MEDLINE, EMBASE, and Cochrane CENTRAL using specific and sensitive keywords as well as combinations of keywords. As 'diffuse midline glioma' was recently defined, and there was no international guideline, trials and guidelines of 'diffuse intrinsic pontine glioma' or 'brain stem glioma' were thoroughly reviewed first. RESULTS The core contents are as follows. The DMG can be diagnosed when all of the following three criteria are satisfied: the presence of the H3K27M mutation, midline location, and infiltrating feature. Without identification of H3K27M mutation by diagnostic biopsy, DMG cannot be diagnosed. For the primary treatment, maximal safe resection should be considered for tumors when feasible. Radiotherapy is the primary option for tumors in case the total resection is not possible. A total dose of 54 Gy to 60 Gy with conventional fractionation prescribed at 1-2 cm plus gross tumor volume is recommended. Although no chemotherapy has proven to be effective in DMG, concurrent chemoradiotherapy (± maintenance chemotherapy) with temozolomide following WHO grade IV glioblastoma's protocol is recommended. CONCLUSION The detection of H3K27M mutation is the most important diagnostic criteria for DMG. Combination of surgery (if amenable to surgery), radiotherapy, and chemotherapy based on comprehensive multidisciplinary discussion can be considered as the treatment options for DMG.
Collapse
Affiliation(s)
- Hong In Yoon
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Chan Woo Wee
- Department of Radiation Oncology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Young Zoon Kim
- Division of Neurooncology and Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Youngbeom Seo
- Department of Neurosurgery, Yeungnam University Hospital, Yeungnam University College of Medicine, Daegu, Korea
| | - Jung Ho Im
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - Yun Sik Dho
- Department of Neurosurgery, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Kyung Hwan Kim
- Department of Neurosurgery, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
| | - Je Beom Hong
- Department of Neurosurgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Sung Park
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seo Hee Choi
- Department of Radiation Oncology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Min Sung Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jangsup Moon
- Department of Genomic Medicine, Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Kihwan Hwang
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin Mo Cho
- Department of Neurosurgery, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Korea
| | - Wan Soo Yoon
- Department of Neurosurgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Il Kim
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyoung Su Sung
- Department of Neurosurgery, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Korea
| | - Jin Ho Song
- Department of Radiation Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Ho Lee
- Department of Neurosurgery, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Uijeongbu, Korea
| | - Myung Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Se Hoon Lee
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chul Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| | - Youn Soo Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Ho Shin Gwak
- Department of Cancer Control, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.
| |
Collapse
|
8
|
Williams JR, Young CC, Vitanza NA, McGrath M, Feroze AH, Browd SR, Hauptman JS. Progress in diffuse intrinsic pontine glioma: advocating for stereotactic biopsy in the standard of care. Neurosurg Focus 2021; 48:E4. [PMID: 31896081 DOI: 10.3171/2019.9.focus19745] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 09/20/2019] [Indexed: 11/06/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a universally fatal pediatric brainstem tumor affecting approximately 300 children in the US annually. Median survival is less than 1 year, and radiation therapy has been the mainstay of treatment for decades. Recent advances in the biological understanding of the disease have identified the H3K27M mutation in nearly 80% of DIPGs, leading to the 2016 WHO classification of diffuse midline glioma H3K27M-mutant, a grade IV brainstem tumor. Developments in epigenetic targeting of transcriptional tendencies have yielded potential molecular targets for clinical trials. Chimeric antigen receptor T cell therapy has also shown preclinical promise. Recent clinical studies, including prospective trials, have demonstrated the safety and feasibility of pediatric brainstem biopsy in the setting of DIPG and other brainstem tumors. Given developments in the ability to analyze DIPG tumor tissue to deepen biological understanding of this disease and develop new therapies for treatment, together with the increased safety of stereotactic brainstem biopsy, the authors present a case for offering biopsy to all children with suspected DIPG. They also present their standard operative techniques for image-guided, frameless stereotactic biopsy.
Collapse
Affiliation(s)
- John R Williams
- 1Department of Neurological Surgery, University of Washington
| | | | - Nicholas A Vitanza
- 2Division of Hematology/Oncology, Department of Pediatrics, Seattle Children's Hospital; and
| | | | | | - Samuel R Browd
- 3Division of Neurosurgery, Seattle Children's Hospital, Seattle, Washington
| | - Jason S Hauptman
- 3Division of Neurosurgery, Seattle Children's Hospital, Seattle, Washington
| |
Collapse
|
9
|
Fonseca A, Afzal S, Bowes L, Crooks B, Larouche V, Jabado N, Perreault S, Johnston DL, Zelcer S, Fleming A, Scheinemann K, Silva M, Vanan MI, Mpofu C, Wilson B, Eisenstat DD, Lafay-Cousin L, Hukin J, Hawkins C, Bartels U, Bouffet E. Pontine gliomas a 10-year population-based study: a report from The Canadian Paediatric Brain Tumour Consortium (CPBTC). J Neurooncol 2020; 149:45-54. [PMID: 32632896 DOI: 10.1007/s11060-020-03568-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/22/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Diffuse intrinsic pontine gliomas (DIPG) are midline gliomas that arise from the pons and the majority are lethal within a few months after diagnosis. Due to the lack of histological diagnosis the epidemiology of DIPG is not completely understood. The aim of this report is to provide population-based data to characterize the descriptive epidemiology of this condition in Canadian children. PATIENTS AND METHODS A national retrospective study of children and adolescents diagnosed with DIPG between 2000 and 2010 was undertaken. All cases underwent central review to determine clinical and radiological diagnostic characteristics. Crude incidence figures were calculated using age-adjusted (0-17 year) population data from Statistics Canada. Survival analyses were performed using the Kaplan-Meier method. RESULTS A total of 163 patients with pontine lesions were identified. Central review determined one-hundred and forty-three patients who met clinical, radiological and/or histological criteria for diagnosis. We estimate an incidence rate of 1.9 DIPG/1,000,000 children/year in the Canadian population over a 10 years period. Median age at diagnosis was 6.8 years and 50.3% of patients were female. Most patients presented with cranial nerve palsies (76%) and ataxia (66%). Despite typical clinical and radiological characteristics, histological confirmation reported three lesions to be low-grade gliomas and three were diagnosed as CNS embryonal tumor not otherwise specified (NOS). CONCLUSIONS Our study highlights the challenges associated with epidemiology studies on DIPG and the importance of central review for incidence rate estimations. It emphasizes that tissue biopsies are required for accurate histological and molecular diagnosis in patients presenting with pontine lesions and reinforces the limitations of radiological and clinical diagnosis in DIPG. Likewise, it underscores the urgent need to increase the availability and accessibility to clinical trials.
Collapse
Affiliation(s)
- Adriana Fonseca
- Division of Haematology Oncology, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, M5G 1X8, Canada.
| | - Samina Afzal
- IWK Health Center, Dalhousie University, Halifax, B3K 6R8, Canada.,Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates
| | - Lynette Bowes
- Janeway Child Health Center, Memorial University, St. John's, A1B 3V6, Canada
| | - Bruce Crooks
- IWK Health Center, Dalhousie University, Halifax, B3K 6R8, Canada
| | - Valerie Larouche
- CHU de Québec- Université Laval, Laval University, Québec, G1V 4G2, Canada
| | - Nada Jabado
- Montreal Children's Hospital, McGill University, Montreal, H4A 3J1, Canada
| | - Sebastien Perreault
- Centre Hospitalier Universitaire Sainte, Justine, Université de Montreal, Montreal, H3T 1C5, Canada
| | - Donna L Johnston
- Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, K1H 8L1, Canada
| | - Shayna Zelcer
- London Health Sciences Children's Hospital, Western University, London, N6A 5A5, Canada
| | - Adam Fleming
- McMaster Children's Hospital, McMaster University, Hamilton, L8P 1H1, Canada
| | - Katrin Scheinemann
- McMaster Children's Hospital, McMaster University, Hamilton, L8P 1H1, Canada.,Department of Pediatrics, Kantonsspital Aarau, Aarau, Switzerland.,University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Mariana Silva
- Kingston General Hospital, Queens University, Kingston, K7L 2V7, Canada
| | - Magimairajan Issai Vanan
- Cancer Care Manitoba, Pediatrics and Child Health, University of Manitoba, Winnipeg, R3E 0V9, Canada
| | - Chris Mpofu
- Jim Pattison Children's Hospital, University of Saskatchewan, Saskatoon, S7N 0W8, Canada
| | - Beverly Wilson
- Stollery Children's Hospital, University of Alberta, Edmonton, T6G 2B7, Canada
| | - David D Eisenstat
- Stollery Children's Hospital, University of Alberta, Edmonton, T6G 2B7, Canada
| | - Lucie Lafay-Cousin
- Alberta Children's Hospital, University of Calgary, Calgary, T3B 6A8, Canada
| | - Juliette Hukin
- BC Children's Hospital, University of British Columbia, Vancouver, V6H 3N1, Canada
| | - Cynthia Hawkins
- Division of Haematology Oncology, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, M5G 1X8, Canada
| | - Ute Bartels
- Division of Haematology Oncology, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, M5G 1X8, Canada
| | - Eric Bouffet
- Division of Haematology Oncology, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, M5G 1X8, Canada
| |
Collapse
|
10
|
Pramanik R, Bakhshi S. Metronomic therapy in pediatric oncology: A snapshot. Pediatr Blood Cancer 2019; 66:e27811. [PMID: 31207063 DOI: 10.1002/pbc.27811] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/16/2022]
Abstract
Metronomic chemotherapy transitioned from the bench to bedside in the early 2000s and since then has carved a niche for itself in pediatric oncology. It has been used solely or in combination with other modalities such as radiotherapy, maximum tolerated dose chemotherapy, and targeted agents in adjuvant, palliative, as well as maintenance settings. No wonder, the resulting medical literature is extremely heterogeneous. In this review, the authors review and synthesize the published literature in pediatric metronomics giving a glimpse of its history, varied applications, and evolution of this genre of chemotherapy in pediatric cancers. Limitations, future prospects, and grey areas are also highlighted.
Collapse
Affiliation(s)
- Raja Pramanik
- Department of Medical Oncology, Dr. B. R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B. R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
11
|
Diffuse Intrinsic Pontine Glioma: From Diagnosis to Next-Generation Clinical Trials. Curr Treat Options Neurol 2019; 21:37. [PMID: 31290035 DOI: 10.1007/s11940-019-0577-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE OF REVIEW This review of diffuse intrinsic pontine glioma (DIPG) provides clinical background, a systematic approach to diagnosis and initial care, and synthesizes historical, modern, and future directions for treatment. We present evidence supporting neurosurgical biopsy, early palliative care involvement, limitation of glucocorticoid use, and the leveraging of preclinical DIPG models as a pipeline to next-generation clinical trials. RECENT FINDINGS New molecular understanding of pediatric high-grade gliomas has led to the reclassification of DIPG as one member of a family of diffuse gliomas occurring in the midline of the central nervous system that exhibit pathognomonic mutations in genes encoding histone 3 (H3 K27M). DIPG remains a clinically relevant term, though diagnostically the 80% of DIPG cases that exhibit the H3 K27M mutation have been reclassified as diffuse midline glioma, H3 K27M-mutant. Re-irradiation has been shown to be well-tolerated and of potential benefit. Epigenetic targeting of transcriptional dependencies in preclinical models is fueling molecularly targeted clinical trials. Chimeric antigen receptor T cell immunotherapy has also demonstrated efficacy in preclinical models and provides a promising new clinical strategy. DIPG is a universally fatal, epigenetically driven tumor of the pons that is considered part of a broader class of diffuse midline gliomas sharing H3 K27M mutations. Radiation remains the standard of care, single-agent temozolomide is not recommended, and glucocorticoids should be used only sparingly. A rapid evolution of understanding in the chromatin, signaling, and immunological biology of DIPG may soon result in clinical breakthroughs.
Collapse
|
12
|
Pediatric diffuse intrinsic anaplastic astrocytoma of the medulla oblongata. INTERDISCIPLINARY NEUROSURGERY 2018. [DOI: 10.1016/j.inat.2018.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
13
|
Pre-irradiation intensive induction and marrow-ablative consolidation chemotherapy in young children with newly diagnosed high-grade brainstem gliomas: report of the "head-start" I and II clinical trials. J Neurooncol 2018; 140:717-725. [PMID: 30392092 DOI: 10.1007/s11060-018-03003-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/22/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND The dismal outcome in children with high-grade brainstem gliomas (BSG) accentuates the need for effective therapeutic strategies. We investigated the role of intensive, including marrow-ablative, chemotherapy regimens in the treatment of young children with newly-diagnosed high-grade BSG. METHODS Between 1991-and-2002, 15 eligible children less than 10 years of age with a diagnosis of high-grade BSG were treated on "Head-Start" I and II protocols (HSI and HSII). Treatment included Induction with 4-5 cycles of one of three intensive chemotherapy regimens followed by Consolidation with one cycle of marrow-ablative chemotherapy (thiotepa, carboplatin and etoposide) with autologous hematopoietic cell rescue (AHCR). Irradiation was required for children over 6 years of age or for those with residual tumor at the end of Consolidation. RESULTS We had two long-term survivors who were found retrospectively to harbor low-grade glial tumors and thus were not included in the survival analysis. Of the remaining 13 patients, the 1-year event-free (EFS) and overall (OS) survival for these children were 31% (95% CI 9-55%) and 38% (95% CI 14-63%), respectively. Median EFS and OS were 6.6 (95% CI 2.7, 12.7) and 8.7 months (95% CI 6.9, 20.9), respectively. Eight patients developed progressive disease during study treatment (seven during Induction and one at the end of Consolidation). Ten children received focal irradiation, five for residual tumor (three following Induction and two following Consolidation) and five due to disease progression. CONCLUSIONS Children with high-grade BSG did not benefit from this intensive chemotherapy strategy administered prior to irradiation.
Collapse
|
14
|
Clymer J, Kieran MW. The Integration of Biology Into the Treatment of Diffuse Intrinsic Pontine Glioma: A Review of the North American Clinical Trial Perspective. Front Oncol 2018; 8:169. [PMID: 29868485 PMCID: PMC5968382 DOI: 10.3389/fonc.2018.00169] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/01/2018] [Indexed: 11/25/2022] Open
Abstract
Dramatic advances in the molecular analysis of diffuse intrinsic pontine glioma have occurred over the last decade and resulted in the identification of potential therapeutic targets. In spite of these advances, no significant improvement in the outcome has been achieved and median survival remains approximately 10 months. An understanding of the approaches that have been taken to date, why they failed, and how that information can lead the field forward is critical if we are to change the status quo. In this review, we will discuss the clinical trial landscape in North America with an overview of historical approaches that failed and what might account for this failure. We will then provide a discussion of how our understanding of the genotype of this disease has led to the development of a number of trials targeting the mutations and epigenome of diffuse intrinsic pontine gliomas and the issues related to these trials. Similarly, the introduction of methodologies to address penetration across the blood–brain barrier will be considered in the context of both targeted approaches, epigenetic modification, and immune surveillance of these tumors. The comprehensive analysis of these data, generated through cooperative groups, collaborative clinical trials, and pilot studies in North America will be the focus of the IVth Memorial Alicia Pueyo international symposium in Barcelona on March 12th, 2018 and will be compared and contrasted with a similar comprehensive analysis of the European data with the goal of bringing all of these data together to develop a uniform platform on which new rational trials can be based.
Collapse
Affiliation(s)
- Jessica Clymer
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, United States
| | - Mark W Kieran
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Kossatz S, Carney B, Schweitzer M, Carlucci G, Miloushev VZ, Maachani UB, Rajappa P, Keshari KR, Pisapia D, Weber WA, Souweidane MM, Reiner T. Biomarker-Based PET Imaging of Diffuse Intrinsic Pontine Glioma in Mouse Models. Cancer Res 2017; 77:2112-2123. [PMID: 28108511 DOI: 10.1158/0008-5472.can-16-2850] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/09/2017] [Accepted: 01/17/2017] [Indexed: 12/20/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a childhood brainstem tumor with a universally poor prognosis. Here, we characterize a positron emission tomography (PET) probe for imaging DIPG in vivo In human histological tissues, the probes target, PARP1, was highly expressed in DIPG compared to normal brain. PET imaging allowed for the sensitive detection of DIPG in a genetically engineered mouse model, and probe uptake correlated to histologically determined tumor infiltration. Imaging with the sister fluorescence agent revealed that uptake was confined to proliferating, PARP1-expressing cells. Comparison with other imaging technologies revealed remarkable accuracy of our biomarker approach. We subsequently demonstrated that serial imaging of DIPG in mouse models enables monitoring of tumor growth, as shown in modeling of tumor progression. Overall, this validated method for quantifying DIPG burden would serve useful in monitoring treatment response in early phase clinical trials. Cancer Res; 77(8); 2112-23. ©2017 AACR.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brandon Carney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Chemistry, Hunter College and PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, New York
| | - Melanie Schweitzer
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Giuseppe Carlucci
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vesselin Z Miloushev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Uday B Maachani
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Prajwal Rajappa
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Kayvan R Keshari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David Pisapia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Mark M Souweidane
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York.,Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Radiology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
16
|
Abstract
Brainstem gliomas are not nearly as common in adults as they are in children. They are likely the final common consequence not of a single disease process but of several. They can be difficult to diagnose, and are challenging to treat. Clinical studies of this diagnosis are few and generally small. Because of these factors, our understanding of the biology of adult brainstem glioma is incomplete. However, the knowledge base is growing and progress is being made. In this article, we review the current state of knowledge for brainstem glioma in adults and identify key areas for which additional information is required.
Collapse
Affiliation(s)
- Jethro Hu
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Santosh Kesari
- Department of Translational Neuro-Oncology and Neurotherapeutics, John Wayne Cancer Institute, Pacific Neuroscience Institute, Providence Saint John's Health Center , Santa Monica, CA , USA
| |
Collapse
|
17
|
Lee MJ. Overview of CNS Gliomas in Childhood. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2016. [DOI: 10.15264/cpho.2016.23.1.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Mee Jeong Lee
- Department of Pediatrics, Dankook University College of Medicine, Cheonan, Korea
| |
Collapse
|
18
|
Luan X, Guan YY, Lovell JF, Zhao M, Lu Q, Liu YR, Liu HJ, Gao YG, Dong X, Yang SC, Zheng L, Sun P, Fang C, Chen HZ. Tumor priming using metronomic chemotherapy with neovasculature-targeted, nanoparticulate paclitaxel. Biomaterials 2016; 95:60-73. [PMID: 27130953 DOI: 10.1016/j.biomaterials.2016.04.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 02/06/2023]
Abstract
Normalization of the tumor microenvironment is a promising approach to render conventional chemotherapy more effective. Although passively targeted drug nanocarriers have been investigated to this end, actively targeted tumor priming remains to be explored. In this work, we demonstrate an effective tumor priming strategy using metronomic application of nanoparticles actively targeted to tumor neovasculature. F56 peptide-conjugated paclitaxel-loaded nanoparticles (F56-PTX-NP) were formulated from PEGylated polylactide using an oil in water emulsion approach. Metronomic F56-PTX-NP specifically targeted tumor vascular endothelial cells (ECs), pruned vessels with strong antiangiogenic activity and induced thrombospondin-1 (TSP-1) secretion from ECs. The treatment induced tumor vasculature normalization as evidenced by significantly increased coverage of basement membrane and pericytes. The tumor microenvironment was altered with enhanced pO2, lower interstitial fluid pressure, and enhanced vascular perfusion and doxorubicin delivery. A "normalization window" of at least 9 days was induced, which was longer than other approaches using antiangiogenic agents. Together, these results show that metronomic, actively-targeted nanomedicine can induce tumor vascular normalization and modulate the tumor microenvironment, opening a window of opportunity for effective combination chemotherapies.
Collapse
Affiliation(s)
- Xin Luan
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Ying-Yun Guan
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China; Department of Pharmacy, Ruijin Hospital, SJTU-SM, 197 Rui Jin Er Road, Shanghai 200025, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Mei Zhao
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Road, Shanghai 201318, China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Ya-Rong Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Hai-Jun Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Yun-Ge Gao
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Xiao Dong
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Si-Cong Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Lin Zheng
- Pathology Center, Shanghai First People's Hospital, SJTU-SM, 280 South Chongqing Road, Shanghai 200025, China
| | - Peng Sun
- Department of General Surgery, Shanghai Tongren Hospital, SJTU-SM, 1111 Xianxia Road, Shanghai 200336, China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China.
| | - Hong-Zhuan Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
19
|
Yoshida K, Sulaiman NS, Miyawaki D, Ejima Y, Nishimura H, Ishihara T, Matsuo Y, Nishikawa R, Sasayama T, Hayakawa A, Kohmura E, Sasaki R. Radiotherapy for brainstem gliomas in children and adults: A single-institution experience and literature review. Asia Pac J Clin Oncol 2016; 13:e153-e160. [DOI: 10.1111/ajco.12451] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/10/2015] [Accepted: 11/25/2015] [Indexed: 01/30/2023]
Affiliation(s)
- Kenji Yoshida
- Division of Radiation Oncology; Kobe University Graduate School of Medicine; Kobe Japan
| | - Nor Shazrina Sulaiman
- Division of Radiation Oncology; Kobe University Graduate School of Medicine; Kobe Japan
| | - Daisuke Miyawaki
- Division of Radiation Oncology; Kobe University Graduate School of Medicine; Kobe Japan
| | - Yasuo Ejima
- Division of Radiation Oncology; Kobe University Graduate School of Medicine; Kobe Japan
| | - Hideki Nishimura
- Department of Radiation Oncology; Kobe Minimally Invasive Cancer Center; Kobe Japan
| | - Takeaki Ishihara
- Division of Radiation Oncology; Kobe University Graduate School of Medicine; Kobe Japan
| | - Yoshiro Matsuo
- Division of Radiation Oncology; Kobe University Graduate School of Medicine; Kobe Japan
| | - Ryo Nishikawa
- Division of Radiation Oncology; Kobe University Graduate School of Medicine; Kobe Japan
| | - Takashi Sasayama
- Department of Neurosurgery; Kobe University Graduate School of Medicine; Kobe Japan
| | - Akira Hayakawa
- Department of Pediatrics; Kobe University Graduate School of Medicine; Kobe Japan
| | - Eiji Kohmura
- Department of Neurosurgery; Kobe University Graduate School of Medicine; Kobe Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology; Kobe University Graduate School of Medicine; Kobe Japan
| |
Collapse
|
20
|
Vanan MI, Eisenstat DD. DIPG in Children - What Can We Learn from the Past? Front Oncol 2015; 5:237. [PMID: 26557503 PMCID: PMC4617108 DOI: 10.3389/fonc.2015.00237] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 10/08/2015] [Indexed: 02/02/2023] Open
Abstract
Brainstem tumors represent 10–15% of pediatric central nervous system tumors and diffuse intrinsic pontine glioma (DIPG) is the most common brainstem tumor of childhood. DIPG is almost uniformly fatal and is the leading cause of brain tumor-related death in children. To date, radiation therapy (RT) is the only form of treatment that offers a transient benefit in DIPG. Chemotherapeutic strategies including multi-agent neoadjuvant chemotherapy, concurrent chemotherapy with RT, and adjuvant chemotherapy have not provided any survival advantage. To overcome the restrictive ability of the intact blood–brain barrier (BBB) in DIPG, several alternative drug delivery strategies have been proposed but have met with minimal success. Targeted therapies either alone or in combination with RT have also not improved survival. Five decades of unsuccessful therapies coupled with recent advances in the genetics and biology of DIPG have taught us several important lessons (1). DIPG is a heterogeneous group of tumors that are biologically distinct from other pediatric and adult high grade gliomas (HGG). Adapting chemotherapy and targeted therapies that are used in pediatric or adult HGG for the treatment of DIPG should be abandoned (2). Biopsy of DIPG is relatively safe and informative and should be considered in the context of multicenter clinical trials (3). DIPG probably represents a whole brain disease so regular neuraxis imaging is important at diagnosis and during therapy (4). BBB permeability is of major concern in DIPG and overcoming this barrier may ensure that drugs reach the tumor (5). Recent development of DIPG tumor models should help us accurately identify and validate therapeutic targets and small molecule inhibitors in the treatment of this deadly tumor.
Collapse
Affiliation(s)
- Magimairajan Issai Vanan
- Department of Pediatrics and Child Health, University of Manitoba , Winnipeg, MB , Canada ; Department of Biochemistry and Medical Genetics, University of Manitoba , Winnipeg, MB , Canada
| | - David D Eisenstat
- Department of Pediatrics, University of Alberta , Edmonton, AB , Canada ; Department of Medical Genetics, University of Alberta , Edmonton, AB , Canada ; Department of Oncology, University of Alberta , Edmonton, AB , Canada
| |
Collapse
|
21
|
Pediatric brainstem gliomas: new understanding leads to potential new treatments for two very different tumors. Curr Oncol Rep 2015; 17:436. [PMID: 25702179 DOI: 10.1007/s11912-014-0436-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Pediatric brainstem gliomas include low-grade focal brainstem gliomas (FBSG) and high-grade diffuse intrinsic pontine gliomas (DIPG). These tumors share a crucial and eloquent area of the brain as their location, which carries common challenges for treatment. Otherwise, though, these two diseases are very different in terms of presentation, biology, treatment, and prognosis. FBSG usually present with greater than 3 months of symptoms, while DIPG are usually diagnosed within 3 months of symptom onset. Surgery remains the preferred initial treatment for FBSG, with chemotherapy used for persistent, recurrent, or inoperable disease; conversely, radiation is the only known effective treatment for DIPG. Recent developments in biological understanding of both tumors have led to new treatment possibilities. In FBSG, two genetic changes related to BRAF characterize the majority of tumors, and key differences in their biological effects are informing strategies for targeted chemotherapy use. In DIPG, widespread histone H3 and ACVR1 mutations have led to new hope for effective targeted treatments. FBSG has an excellent prognosis, while the long-term survival rate of DIPG tragically remains near zero. In this review, we cover the epidemiology, biology, presentation, imaging characteristics, multimodality treatment, and prognosis of FBSG and DIPG, with a focus on recent biological discoveries.
Collapse
|
22
|
André N, Carré M, Pasquier E. Metronomics: towards personalized chemotherapy? Nat Rev Clin Oncol 2014; 11:413-31. [PMID: 24913374 DOI: 10.1038/nrclinonc.2014.89] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Since its inception in 2000, metronomic chemotherapy has undergone major advances as an antiangiogenic therapy. The discovery of the pro-immune properties of chemotherapy and its direct effects on cancer cells has established the intrinsic multitargeted nature of this therapeutic approach. The past 10 years have seen a marked rise in clinical trials of metronomic chemotherapy, and it is increasingly combined in the clinic with conventional treatments, such as maximum-tolerated dose chemotherapy and radiotherapy, as well as with novel therapeutic strategies, such as drug repositioning, targeted agents and immunotherapy. We review the latest advances in understanding the complex mechanisms of action of metronomic chemotherapy, and the recently identified factors associated with disease resistance. We comprehensively discuss the latest clinical data obtained from studies performed in both adult and paediatric populations, and highlight ongoing clinical trials. In this Review, we foresee the future developments of metronomic chemotherapy and specifically its potential role in the era of personalized medicine.
Collapse
Affiliation(s)
- Nicolas André
- Service d'Hématologie & Oncologie Pédiatrique, AP-HM, 264 rue Saint Pierre, 13385 Marseille, France
| | - Manon Carré
- INSERM UMR 911, Centre de Recherche en Oncologie Biologique et Oncopharmacologie, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005 Marseille, France
| | - Eddy Pasquier
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, PO Box 81, Randwick NSW 2031, Australia
| |
Collapse
|
23
|
Palmieri D, Duchnowska R, Woditschka S, Hua E, Qian Y, Biernat W, Sosińska-Mielcarek K, Gril B, Stark AM, Hewitt SM, Liewehr DJ, Steinberg SM, Jassem J, Steeg PS. Profound prevention of experimental brain metastases of breast cancer by temozolomide in an MGMT-dependent manner. Clin Cancer Res 2014; 20:2727-39. [PMID: 24634373 DOI: 10.1158/1078-0432.ccr-13-2588] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
PURPOSE Brain metastases of breast cancer cause neurocognitive damage and are incurable. We evaluated a role for temozolomide in the prevention of brain metastases of breast cancer in experimental brain metastasis models. EXPERIMENTAL DESIGN Temozolomide was administered in mice following earlier injection of brain-tropic HER2-positive JIMT-1-BR3 and triple-negative 231-BR-EGFP sublines, the latter with and without expression of O(6)-methylguanine-DNA methyltransferase (MGMT). In addition, the percentage of MGMT-positive tumor cells in 62 patient-matched sets of breast cancer primary tumors and resected brain metastases was determined immunohistochemically. RESULTS Temozolomide, when dosed at 50, 25, 10, or 5 mg/kg, 5 days per week, beginning 3 days after inoculation, completely prevented the formation of experimental brain metastases from MGMT-negative 231-BR-EGFP cells. At a 1 mg/kg dose, temozolomide prevented 68% of large brain metastases, and was ineffective at a dose of 0.5 mg/kg. When the 50 mg/kg dose was administered beginning on days 18 or 24, temozolomide efficacy was reduced or absent. Temozolomide was ineffective at preventing brain metastases in MGMT-transduced 231-BR-EGFP and MGMT-expressing JIMT-1-BR3 sublines. In 62 patient-matched sets of primary breast tumors and resected brain metastases, 43.5% of the specimens had concordant low MGMT expression, whereas in another 14.5% of sets high MGMT staining in the primary tumor corresponded with low staining in the brain metastasis. CONCLUSIONS Temozolomide profoundly prevented the outgrowth of experimental brain metastases of breast cancer in an MGMT-dependent manner. These data provide compelling rationale for investigating the preventive efficacy of temozolomide in a clinical setting.
Collapse
Affiliation(s)
- Diane Palmieri
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Renata Duchnowska
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Stephan Woditschka
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Emily Hua
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Yongzhen Qian
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Wojciech Biernat
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Katarzyna Sosińska-Mielcarek
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Brunilde Gril
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Andreas M Stark
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Stephen M Hewitt
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - David J Liewehr
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Seth M Steinberg
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Jacek Jassem
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, GermanyAuthors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| | - Patricia S Steeg
- Authors' Affiliations: Women's Malignancies Branch; Laboratory of Pathology, Center for Cancer Research; Biostatistics and Data Management Section, NCI, NIH, Bethesda; Laboratory Animal Sciences Program, SAIC-Frederick, NCI, NIH, Frederick, Maryland; Department of Oncology, Military Institute of Medicine, Warsaw; Departments of Pathomorphology, and Oncology and Radiotherapy, Medical University; Regional Cancer Center, Gdańsk, Poland; and Klinik fur Neurochirurgie UKSH Campus Kiel, Kiel, Germany
| |
Collapse
|
24
|
Zaghloul MS, Eldebawy E, Ahmed S, Mousa AG, Amin A, Refaat A, Zaky I, Elkhateeb N, Sabry M. Hypofractionated conformal radiotherapy for pediatric diffuse intrinsic pontine glioma (DIPG): a randomized controlled trial. Radiother Oncol 2014; 111:35-40. [PMID: 24560760 DOI: 10.1016/j.radonc.2014.01.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 01/13/2014] [Accepted: 01/18/2014] [Indexed: 01/10/2023]
Abstract
BACKGROUND The pediatric diffuse intrinsic pontine glioma (DIPG) outcome remains dismal despite multiple therapeutic attempts. PURPOSE To compare the results of treatment of pediatric diffuse intrinsic pontine glioma (DIPG) using hypofractionated versus conventional radiotherapy. PATIENTS AND METHODS Seventy-one newly diagnosed DIPG children were randomized into hypofractionated (HF) (39Gy/13 fractions in 2.6weeks) and conventional (CF) arm (54Gy/30 fractions in 6weeks). RESULTS The median and one-year overall survival (OS) was 7.8months and 36.4±8.2% for the hypofractionated arm, and 9.5 and 26.2±7.4% for the conventional arm respectively. The 18-month OS difference was 2.2%. The OS hazard ratio (HR) was 1.14 (95% CI: 0.70-1.89) (p=0.59). The hypofractionated arm had a median and one-year progression-free survival (PFS) of 6.6months and 22.5±7.1%, compared to 7.3 and 17.9±7.1% for the conventional arm. The PFS HR was 1.10 (95% CI: 0.67-1.90) (p=0.71). The 18-month PFS difference was 1.1%. These differences exceed the non-inferiority margin. The immediate and delayed side effects were not different in the 2 arms. CONCLUSIONS Hypofractionated radiotherapy offers lesser burden on the patients, their families and the treating departments, with nearly comparable results to conventional fractionation, though not fulfilling the non-inferiority assumption.
Collapse
Affiliation(s)
- Mohamed S Zaghloul
- Radiation Oncology Department, Children's Cancer Hospital, Egypt (CCHE) and National Cancer Institute, Cairo University, Egypt.
| | - Eman Eldebawy
- Radiation Oncology Department, Children's Cancer Hospital, Egypt (CCHE) and National Cancer Institute, Cairo University, Egypt
| | - Soha Ahmed
- Radiation Oncology Department, Children's Cancer Hospital, Egypt (CCHE) and National Cancer Institute, Cairo University, Egypt
| | - Amr G Mousa
- Radiation Oncology Department, Children's Cancer Hospital, Egypt (CCHE) and National Cancer Institute, Cairo University, Egypt
| | - Amr Amin
- Radiation Oncology Department, Children's Cancer Hospital, Egypt (CCHE) and National Cancer Institute, Cairo University, Egypt
| | - Amal Refaat
- Radiology Department, Children's Cancer Hospital, Egypt (CCHE) and National Cancer Institute, Cairo University, Egypt
| | - Iman Zaky
- Radiology Department, Children's Cancer Hospital, Egypt (CCHE) and National Cancer Institute, Cairo University, Egypt
| | | | - Mohamed Sabry
- Research Department, Children's Cancer Hospital, Egypt
| |
Collapse
|
25
|
Bailey S, Howman A, Wheatley K, Wherton D, Boota N, Pizer B, Fisher D, Kearns P, Picton S, Saran F, Gibson M, Glaser A, Connolly D, Hargrave D. Diffuse intrinsic pontine glioma treated with prolonged temozolomide and radiotherapy--results of a United Kingdom phase II trial (CNS 2007 04). Eur J Cancer 2013; 49:3856-62. [PMID: 24011536 PMCID: PMC3853623 DOI: 10.1016/j.ejca.2013.08.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 06/30/2013] [Accepted: 08/08/2013] [Indexed: 12/03/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) has a dismal prognosis with no chemotherapy regimen so far resulting in any significant improvement over standard radiotherapy. In this trial, a prolonged regimen (21/28d) of temozolomide was studied with the aim of overcoming O(6)-methylguanine methyltransferase (MGMT) mediated resistance. Forty-three patients with a defined clinico-radiological diagnosis of DIPG received radiotherapy and concomitant temozolomide (75 mg/m(2)) after which up to 12 courses of 21d of adjuvant temozolomide (75-100mg/m(2)) were given 4 weekly. The trial used a 2-stage design and passed interim analysis. At diagnosis median age was 8 years (2-20 years), 81% had cranial nerve abnormalities, 76% ataxia and 57% long tract signs. Median Karnofsky/Lansky score was 80 (10-100). Patients received a median of three courses of adjuvant temozolomide, five received all 12 courses and seven did not start adjuvant treatment. Three patients were withdrawn from study treatment due to haematological toxicity and 10 had a dose reduction. No other significant toxicity related to temozolomide was noted. Overall survival (OS) (95% confidence interval (CI)) was 56% (40%, 69%) at 9 months, 35% (21%, 49%) at 1 year and 17% (7%, 30%) at 2 years. Median survival was 9.5 months (range 7.5-11.4 months). There were five 2-year survivors with a median age of 13.6 years at diagnosis. This trial demonstrated no survival benefit of the addition of dose dense temozolomide, to standard radiotherapy in children with classical DIPG. However, a subgroup of adolescent DIPG patients did have a prolonged survival, which needs further exploration.
Collapse
Affiliation(s)
- S. Bailey
- Great North Childrens Hospital, Newcastle upon Tyne, United Kingdom
| | - A. Howman
- CRCTU, University of Birmingham, Birmingham, United Kingdom
| | - K. Wheatley
- CRCTU, University of Birmingham, Birmingham, United Kingdom
| | - D. Wherton
- CRCTU, University of Birmingham, Birmingham, United Kingdom
| | - N. Boota
- Nottingham Clinical Trials Unit, Nottingham, United Kingdom
| | - B. Pizer
- Alder Hey Childrens Hospital, Liverpool, United Kingdom
| | - D. Fisher
- Addenbroookes Hopsital, Cambridge, United Kingdom
| | - P. Kearns
- CRCTU, University of Birmingham, Birmingham, United Kingdom
| | - S. Picton
- Leeds General Infirmary, Leeds, United Kingdom
| | - F. Saran
- Royal Marsden Hospital, Surrey, London, United Kingdom
| | - M. Gibson
- CRCTU, University of Birmingham, Birmingham, United Kingdom
| | - A. Glaser
- Leeds General Infirmary, Leeds, United Kingdom
| | | | - D. Hargrave
- Great Ormond Street Hospital For Sick Children, London, United Kingdom
| |
Collapse
|
26
|
Abstract
Brainstem gliomas (BGs) are a heterogenous group of gliomas that occur predominately in children. They can be separated into groups on the basis of anatomy and clinical behavior: diffuse intrinsic pontine glioma (DIPG), exophytic medullary glioma, and tectal glioma. DIPG is the commonest BG. Median age at onset is 6.5 years and median survival is less than 1 year. Adults with DIPG survive longer, suggesting a less aggressive and biologically different tumor from that in children. Patients present with cranial nerve dysfunction, long tract signs, or ataxia, either in isolation or in combination. Magnetic resonance imaging shows an infiltrative lesion occupying most of the pons and contrast enhancement is usually not prominent. Standard treatment is fractionated radiotherapy. Platelet-derived growth factor receptor alpha and epidermal growth factor receptor mutations have been identified. Inhibitors of these growth factor receptors are being evaluated in clinical trials. Exophytic medullary and tectal gliomas are relatively indolent tumors that can often be followed closely without treatment.
Collapse
Affiliation(s)
- Sean A Grimm
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | |
Collapse
|
27
|
Wang Y, Tian Y, Wan H, Li D, Wu W, Yin L, Jiang J, Wan W, Zhang L. Differences between brainstem gliomas in juvenile and adult rats. Oncol Lett 2013; 6:246-250. [PMID: 23946812 PMCID: PMC3742815 DOI: 10.3892/ol.2013.1319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 04/16/2013] [Indexed: 11/25/2022] Open
Abstract
Clinical studies have shown that gliomas of the brainstem behave differently in children and adults. The aim of the present study was to compare and analyze the differences between these gliomas in juvenile and adult rats with regard to tumor growth, survival, pathology and magnetic resonance imaging (MRI). A total of 25 juvenile and 25 adult Wistar rats were divided into groups A (15 juvenile rats), B (10 juvenile rats), C (15 adult rats) and D (10 adult rats). The rats of groups A and C (experimental) were injected with glioma cells, while groups B and D (control) were injected with a physiological saline solution. Rat neurological signs, survival time, tumor size, hematoxylin and eosin (HE) staining and immunohistochemical staining for MMP-2, MMP-9 and β-catenin were compared. The survival time of group A was 19.47±2.232 days, whereas that of group C was 21.47±2.232 days (P<0.05). The tumor sizes were 4.55 and 4.62 mm (P>0.05) in groups A and C, respectively. HE and immunohistochemical staining revealed no differences between the groups. The results suggest that the growth patterns and invasiveness of brainstem gliomas may vary in children compared with adults due to the varied biological behaviors of the tumor cells.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Warren KE. Diffuse intrinsic pontine glioma: poised for progress. Front Oncol 2012; 2:205. [PMID: 23293772 PMCID: PMC3531714 DOI: 10.3389/fonc.2012.00205] [Citation(s) in RCA: 207] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/11/2012] [Indexed: 12/21/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are amongst the most challenging tumors to treat. Surgery is not an option, the effects of radiation therapy are temporary, and no chemotherapeutic agent has demonstrated significant efficacy. Numerous clinical trials of new agents and novel therapeutic approaches have been performed over the course of several decades in efforts to improve the outcome of children with DIPG, yet without success. The diagnosis of DIPG is based on radiographic findings in the setting of a typical clinical presentation, and tissue is not routinely obtained as the standard of care. The paradigm for treating children with these tumors has been based on that for supratentorial high-grade gliomas in adults as the biology of these lesions were presumed to be similar. However, recent pivotal studies demonstrate that DIPGs appear to be their own entity. Simply identifying this fact releases a number of constraints and opens opportunities for biologic investigation of these lesions, setting the stage to move forward in identifying DIPG-specific treatments. This review will summarize the current state of knowledge of DIPG, discuss obstacles to therapy, and summarize results of recent biologic studies.
Collapse
Affiliation(s)
- Katherine E Warren
- Pediatric Neuro-Oncology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
29
|
Hashizume R, Smirnov I, Liu S, Phillips JJ, Hyer J, McKnight TR, Wendland M, Prados M, Banerjee A, Nicolaides T, Mueller S, James CD, Gupta N. Characterization of a diffuse intrinsic pontine glioma cell line: implications for future investigations and treatment. J Neurooncol 2012; 110:305-13. [PMID: 22983601 DOI: 10.1007/s11060-012-0973-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 09/11/2012] [Indexed: 10/27/2022]
Abstract
Diffuse intrinsic pontine gliomas arise almost exclusively in children, and despite advances in treatment, the majority of patients die within 2 years after initial diagnosis. Because of their infiltrative nature and anatomic location in an eloquent area of the brain, most pontine gliomas are treated without a surgical biopsy. The corresponding lack of tissue samples has resulted in a limited understanding of the underlying genetic and molecular biologic abnormalities associated with pontine gliomas, and is a substantial obstacle for the preclinical testing of targeted therapeutic agents for these tumors. We have established a human glioma cell line that originated from surgical biopsy performed on a patient with a pontine glioma. To insure sustainable in vitro propagation, tumor cells were modified with hTERT (human telomerase ribonucleoprotein reverse transcriptase), and with a luciferase reporter to enable non-invasive bioluminescence imaging. The hTERT modified cells are tumorigenic in athymic rodents, and produce brainstem tumors that recapitulate the infiltrative growth of brainstem gliomas in patients.
Collapse
Affiliation(s)
- Rintaro Hashizume
- Department of Neurological Surgery, Brain Tumor Research Center, University of California San Francisco, San Francisco, CA 94143-0520, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Waterhouse DN, Yapp D, Verreault M, Anantha M, Sutherland B, Bally MB. Lipid-based nanoformulation of irinotecan: dual mechanism of action allows for combination chemo/angiogenic therapy. Nanomedicine (Lond) 2012; 6:1645-54. [PMID: 22077466 DOI: 10.2217/nnm.11.140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A number of studies have outlined the antiangiogenic effects of cytotoxic agents when administered frequently at low doses. These studies suggest that the effect of the cytotoxic agent is on the vasculature within the tumor and it is assumed that there is little or negligible cytotoxicity. Liposomal drug delivery systems have the ability to provide a dual mechanism of activity where tumor accumulation can deliver high local concentrations of the drug at the site of action with concomitant slow release of the drug from carriers in the blood compartment that results in antivascular effects, similar to that achieved when dosing frequently at low levels. Although this dual mechanism of activity may be linked to other lipid nanoparticle formulations of anticancer drugs, this article summarizes the evidence supporting direct (cytotoxic) and indirect (antivascular) actions of a liposomal formulation of irinotecan.
Collapse
Affiliation(s)
- Dawn N Waterhouse
- BC Cancer Agency, Department of Experimental Therapeutics, 675 West 10th Avenue, Vancouver BC Canada, V5Z 1L3.
| | | | | | | | | | | |
Collapse
|
31
|
Jansen M, van Vuurden D, Vandertop W, Kaspers G. Diffuse intrinsic pontine gliomas: A systematic update on clinical trials and biology. Cancer Treat Rev 2012; 38:27-35. [DOI: 10.1016/j.ctrv.2011.06.007] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 05/17/2011] [Accepted: 06/25/2011] [Indexed: 11/28/2022]
|
32
|
Proceedings of the diffuse intrinsic pontine glioma (DIPG) Toronto Think Tank: advancing basic and translational research and cooperation in DIPG. J Neurooncol 2011; 105:119-25. [DOI: 10.1007/s11060-011-0704-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 08/16/2011] [Indexed: 10/17/2022]
|
33
|
Moving forward with metronomic chemotherapy: meeting report of the 2nd International Workshop on Metronomic and Anti-Angiogenic Chemotherapy in Paediatric Oncology. Transl Oncol 2011; 4:203-11. [PMID: 21804915 DOI: 10.1593/tlo.11124] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 05/15/2011] [Accepted: 05/16/2011] [Indexed: 12/21/2022] Open
Abstract
Metronomic chemotherapy, which is defined by the frequent, repetitive administration of chemotherapeutic drugs at relatively low doses, and without prolonged drug-free break, is an emerging strategy to fight cancer. Initially thought to act by targeting tumor angiogenesis, additional mechanisms have been recently unveiled, and metronomic chemotherapy is now considered to represent a form of multitargeted therapy. Despite representing a genuine alternative for advanced and/or high-risk cancer therapy, the development of metronomic approaches in pediatric oncology is still in the early stage. The few numbers of large-scale state-of-the-art clinical trials, issues regarding terminology and the limited understanding of the complex and intertwined mechanisms of action of metronomic treatments have limited progress in this important field of research. On March 18 and 19, 2010, the 2nd International Workshop on Metronomic and Anti-Angiogenic Chemotherapy in Paediatric Oncology was held in Marseille, France, and brought together clinicians, basic scientists, physician-scientists, trainees, and students from all around the world. The main aim of this international meeting was to provide a unique forum to 1) reflect on the major advances that have been made in this field of research since its creation, 2) communicate results from the most recent clinical trials and preclinical studies, 3) discuss the current and future challenges of the field, and 4) set forth a solid framework for future collaborative biologic and clinical studies. The present report documents the main preclinical and clinical data that were presented in the keynote and best abstract sessions and delivers the key messages from the meeting.
Collapse
|
34
|
Bartels U, Baruchel S, Carret A, Crooks B, Hukin J, Johnston D, Silva M, Strother D, Wilson B, Zelcer S, Eisenstat D, Sung L, Bouffet E. The use and effectiveness of temozolomide in children with central nervous system tumours: a survey from the Canadian Paediatric Brain Tumour Consortium. Curr Oncol 2011; 18:e19-24. [PMID: 21331268 PMCID: PMC3031361 DOI: 10.3747/co.v18i1.675] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To describe the use of temozolomide (tmz) in Canadian children treated for brain tumours and to evaluate survival and predictors of survival for children treated with this agent. METHODS A survey was conducted within the Canadian Paediatric Brain Tumour Consortium (cpbtc), a group of tertiary care centres in pediatric neuro-oncology (n = 16) in Canada that are involved in the treatment of children with central nervous system tumours. RESULTS In 10 of the 16 participating pediatric oncology centres of the cpbtc, 137 children with brain tumours were treated with tmz between January 2000 and March 2006. Although 33% of the children were enrolled into a clinical trial, 67% were treated outside open studies. Most patients (72%) received tmz treatment on recurrence of their brain tumour (first or subsequent). The most commonly administered regimen was single-agent tmz 150-200 mg/m(2) administered on 5 consecutive days every 28 days. The median duration of tmz treatment was 141 days (range: 4-1102 days). Response data were provided for 127 of the 137 patients, of whom 6 showed a complete response. Sixteen patients experienced a minor or partial response, 53 had stable disease, and 52 had progressive disease. Of 32 patients alive at last follow-up, 19 had a diagnosis of low-grade glioma. CONCLUSIONS Temozolomide is used in a variety of pediatric brain tumours, often at the time of recurrence. The lack of insight into clear indications for this agent in pediatric brain tumours-used either alone or in combination therapy-may be a result of suboptimal design of phase i and ii studies and a lack of phase iii trials in the pediatric brain tumour population.
Collapse
Affiliation(s)
| | | | | | | | - J. Hukin
- British Columbia’s Children’s Hospital, Vancouver, BC
| | - D. Johnston
- Children’s Hospital of Easter n Ontario, Ottawa, ON
| | - M. Silva
- Kingston General Hospital, Kingston, ON
| | - D. Strother
- University of Calgary, Alberta Hospital, Calgary, AB
| | - B. Wilson
- Stollery Children’s Hospital, Edmonton, AB
| | - S. Zelcer
- Children’s Hospital of Western Ontario, London, ON
| | | | - L. Sung
- Hospital for Sick Children, Toronto, ON
| | | |
Collapse
|
35
|
Massimino M, Bode U, Biassoni V, Fleischhack G. Nimotuzumab for pediatric diffuse intrinsic pontine gliomas. Expert Opin Biol Ther 2010; 11:247-56. [PMID: 21171927 DOI: 10.1517/14712598.2011.546341] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Diffuse intrinsic pontine gliomas (DIPG) have a poor prognosis: the median survival rate is less than one year. Radiotherapy is the only effective treatment affording an overall survival of 6 - 9 months. So far, no improvement has been achieved with the addition of single/poly-chemotherapy regimens. An urgent need is to advance in this field, from both the biological and the clinical points of view. AREAS COVERED Among the few studies providing biological information on DIPG, Gilbertson's group demonstrated a significant increase in EGFR expression. The activity of nimotuzumab, a humanized anti-EGFR monoclonal antibody, was therefore studied within a Phase II trial in 47 relapsing pediatric patients with DIPG and high-grade gliomas, showing an interesting, persistent response, especially in the first group treated. A multicenter exploratory study combining nimotuzumab and radiotherapy showed disease control and an overall patient survival similar to previous experiences along with an improvement in the quality of patient survival and no severe side effects. EXPERT OPINION We recommend considering this combination in the armamentarium against DIPG. It might be improved by adding other target drugs/low-toxicity chemotherapy regimens with a synergistic effect with the anti-EGFR component.
Collapse
Affiliation(s)
- Maura Massimino
- Pediatric Unit, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian, 1 20133 Milano, Italy.
| | | | | | | |
Collapse
|