272 results on '"Stewart Goldman"'
Search Results
2. Data from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Medulloblastoma is a highly malignant pediatric brain tumor associated with poor outcome. Developing treatments that target the cancer stem cell (CSC) population in medulloblastoma are important to prevent tumor relapse and induce long-lasting clinical responses. We utilized medulloblastoma neurospheres that display CSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. Of all class IA PI3Ks, only the PI3Kα isoform was required for sphere formation by medulloblastoma cells. Knockdown of p110α, but not p110β or p110δ, significantly disrupted cancer stem cell frequencies as determined by extreme limiting dilution analysis (ELDA), indicating an essential role for the PI3Kα catalytic isoform in medulloblastoma CSCs. Importantly, pharmacologic inhibition of the MAPK-interacting kinase (MNK) enhanced the antineoplastic effects of targeted PI3Kα inhibition in medulloblastoma. This indicates that MNK signaling promotes survival in medulloblastoma, suggesting dual PI3Kα and MNK inhibition may provide a novel approach to target and eliminate medulloblastoma CSCs. We also observed a significant reduction in tumor formation in subcutaneous and intracranial mouse xenograft models, which further suggests that this combinatorial approach may represent an efficient therapeutic strategy for medulloblastoma.Implications:These findings raise the possibility of a unique therapeutic approach for medulloblastoma, involving MNK targeting to sensitize medulloblastoma CSCs to PI3Kα inhibition.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Table S1 from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Pairwise tests for differences in stem cell frequencies.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure 1 from REST Is a Novel Prognostic Factor and Therapeutic Target for Medulloblastoma
- Author
-
Vidya Gopalakrishnan, Laurence Cooper, Alvaro Laureano, Lars Riedemann, Martin Hasselblatt, Tobey MacDonald, Irene B. Helenowski, Stewart Goldman, Veena Rajaram, Jason Fangusaro, and Pete Taylor
- Abstract
PDF file, 169KB, Measurement of Syn1 expression following SAHA treatment of murine NSC-M and NSC-MR cells.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figures S1 - S2 from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
These figures show the PN and MES specific drugs identified by the follow-up cell viability screen. Also shown are the enrichment of translation gene sets in MES GSCs. Relates to Figure 1.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table S1 from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
This table shows the initial cell viability screen in GBM cells. Relates to Figure 1C.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table S2 from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
This table shows the follow-up cell viability screen in MES and PN GBM cells. Relates to Figure 1D.
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
Mesenchymal (MES) and proneural (PN) are two distinct glioma stem cell (GSC) populations that drive therapeutic resistance in glioblastoma (GBM). We screened a panel of 650 small molecules against patient-derived GBM cells to discover compounds targeting specific GBM subtypes. Arsenic trioxide (ATO), an FDA-approved drug that crosses the blood–brain barrier, was identified as a potent PN-specific compound in the initial screen and follow-up validation studies. Furthermore, MES and PN GSCs exhibited differential sensitivity to ATO. As ATO has been shown to activate the MAPK-interacting kinase 1 (MNK1)-eukaryotic translation initiation factor 4E (eIF4E) pathway and subsequent mRNA translation in a negative regulatory feedback manner, the mechanistic role of ATO resistance in MES GBM was explored. In GBM cells, ATO-activated translation initiation cellular events via the MNK1–eIF4E signaling axis. Furthermore, resistance to ATO in intracranial PDX tumors correlated with high eIF4E phosphorylation. Polysomal fractionation and microarray analysis of GBM cells were performed to identify ATO's effect on mRNA translation and enrichment of anti-apoptotic mRNAs in the ATO-induced translatome was found. Additionally, it was determined that MNK inhibition sensitized MES GSCs to ATO in neurosphere and apoptosis assays. Finally, examination of the effect of ATO on patients from a phase I/II clinical trial of ATO revealed that PN GBM patients responded better to ATO than other subtypes as demonstrated by longer overall and progression-free survival.Implications: These findings raise the possibility of a unique therapeutic approach for GBM, involving MNK1 targeting to sensitize MES GSCs to drugs like arsenic trioxide. Mol Cancer Res; 16(1); 32–46. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Methods and Figure Legend from REST Is a Novel Prognostic Factor and Therapeutic Target for Medulloblastoma
- Author
-
Vidya Gopalakrishnan, Laurence Cooper, Alvaro Laureano, Lars Riedemann, Martin Hasselblatt, Tobey MacDonald, Irene B. Helenowski, Stewart Goldman, Veena Rajaram, Jason Fangusaro, and Pete Taylor
- Abstract
PDF file, 73KB.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table S3 from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
This table shows genes enriched in untreated and ATO-treated polysomes. Relates to Figure 4D.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure S1 from Potent Antineoplastic Effects of Combined PI3Kα–MNK Inhibition in Medulloblastoma
- Author
-
Leonidas C. Platanias, Rintaro Hashizume, Hidayatullah G. Munshi, Stewart Goldman, Craig Horbinski, David Z. Chen, Quanhong Ma, Ewa M. Kosciuczuk, Gavin T. Blyth, Jessica Clymer, Elspeth M. Beauchamp, Jonathan B. Bell, and Frank Eckerdt
- Abstract
Body weight of mice from flank tumor xenograft experiment
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Methods from Differential Response of Glioma Stem Cells to Arsenic Trioxide Therapy Is Regulated by MNK1 and mRNA Translation
- Author
-
Leonidas C. Platanias, Michael E. Berens, Jeffrey Raizer, Priya Kumthekar, Kristiina Vuori, Andrew P. Mazar, Craig Horbinski, Ichiro Nakano, C. David James, Shi-Yuan Cheng, Stewart Goldman, Jessica Clymer, Kristen Alley, Elspeth M. Beauchamp, Barbara Kroczynska, Seungchan Kim, Sen Peng, Darren Finlay, Harshil D. Dhruv, Frank Eckerdt, and Jonathan B. Bell
- Abstract
Relates to materials & methods.
- Published
- 2023
- Full Text
- View/download PDF
13. Figure S3 from Radiosensitization by Histone H3 Demethylase Inhibition in Diffuse Intrinsic Pontine Glioma
- Author
-
Rintaro Hashizume, Ali Shilatifard, Lihua Zou, Oren J. Becher, Sriram Venneti, Stewart Goldman, Rishi Lulla, Nitin R. Wadhwani, Akihide Kondo, Kathryn L. Laurie, Vera Gorbunova, Andrei Seluanov, Xiao Tian, Angel M. Carcaboso, Jonathan B. Lamano, Yosuke Shimazu, Andrea Piunti, Quanhong Ma, Ali Zhang, Xingyao He, Takahiro Sasaki, Dusten Unruh, Nundia Louis, and Hiroaki Katagi
- Abstract
Supplementary Figure S3
- Published
- 2023
- Full Text
- View/download PDF
14. Data from Phase I Study of Vismodegib in Children with Recurrent or Refractory Medulloblastoma: A Pediatric Brain Tumor Consortium Study
- Author
-
Tom Curran, Richard J. Gilbertson, James M. Boyett, Naoko Takebe, Dana Wallace, Murali Chintagumpala, Stewart Goldman, Roger J. Packer, Larry E. Kun, Sue Kaste, David W. Ellison, Clinton F. Stewart, and Amar Gajjar
- Abstract
Purpose: To investigate the safety, dose-limiting toxicities, and pharmacokinetics of the smoothened inhibitor vismodegib in children with refractory or relapsed medulloblastoma.Experimental design: Initially, vismodegib was administered daily at 85 mg/m2 and escalated to 170 mg/m2. The study was then revised to investigate a flat-dosing schedule of 150 mg for patients with small body surface area (BSA, 0.67–1.32 m2) or 300 mg for those who were larger (BSA, 1.33–2.20 m2). Pharmacokinetics were performed during the first course of therapy, and the right knees of all patients were imaged to monitor bone toxicity. Immunohistochemical analysis was done to identify patients with Sonic Hedgehog (SHH)-subtype medulloblastoma.Results: Thirteen eligible patients were enrolled in the initial study: 6 received 85 mg/m2 vismodegib, and 7 received 170 mg/m2. Twenty eligible patients were enrolled in the flat-dosing part of the study: 10 at each dosage level. Three dose-limiting toxicities were observed, but no drug-related bone toxicity was documented. The median (range) vismodegib penetration in the cerebrospinal fluid (CSF) was 0.53 (0.26–0.78), when expressed as a ratio of the concentration of vismodegib in the CSF to that of the unbound drug in plasma. Antitumor activity was seen in 1 of 3 patients with SHH-subtype disease whose tumors were evaluable, and in none of the patients in the other subgroups.Conclusions: Vismodegib was well tolerated in children with recurrent or refractory medulloblastoma; only two dose-limiting toxicities were observed with flat dosing. The recommended phase II study dose is 150 or 300 mg, depending on the patient's BSA. Clin Cancer Res; 19(22); 6305–12. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
15. Table S2 from Radiosensitization by Histone H3 Demethylase Inhibition in Diffuse Intrinsic Pontine Glioma
- Author
-
Rintaro Hashizume, Ali Shilatifard, Lihua Zou, Oren J. Becher, Sriram Venneti, Stewart Goldman, Rishi Lulla, Nitin R. Wadhwani, Akihide Kondo, Kathryn L. Laurie, Vera Gorbunova, Andrei Seluanov, Xiao Tian, Angel M. Carcaboso, Jonathan B. Lamano, Yosuke Shimazu, Andrea Piunti, Quanhong Ma, Ali Zhang, Xingyao He, Takahiro Sasaki, Dusten Unruh, Nundia Louis, and Hiroaki Katagi
- Abstract
Supplementary Table S2
- Published
- 2023
- Full Text
- View/download PDF
16. Figure S1 from Radiosensitization by Histone H3 Demethylase Inhibition in Diffuse Intrinsic Pontine Glioma
- Author
-
Rintaro Hashizume, Ali Shilatifard, Lihua Zou, Oren J. Becher, Sriram Venneti, Stewart Goldman, Rishi Lulla, Nitin R. Wadhwani, Akihide Kondo, Kathryn L. Laurie, Vera Gorbunova, Andrei Seluanov, Xiao Tian, Angel M. Carcaboso, Jonathan B. Lamano, Yosuke Shimazu, Andrea Piunti, Quanhong Ma, Ali Zhang, Xingyao He, Takahiro Sasaki, Dusten Unruh, Nundia Louis, and Hiroaki Katagi
- Abstract
Supplementary Figure S1
- Published
- 2023
- Full Text
- View/download PDF
17. Data from Radiosensitization by Histone H3 Demethylase Inhibition in Diffuse Intrinsic Pontine Glioma
- Author
-
Rintaro Hashizume, Ali Shilatifard, Lihua Zou, Oren J. Becher, Sriram Venneti, Stewart Goldman, Rishi Lulla, Nitin R. Wadhwani, Akihide Kondo, Kathryn L. Laurie, Vera Gorbunova, Andrei Seluanov, Xiao Tian, Angel M. Carcaboso, Jonathan B. Lamano, Yosuke Shimazu, Andrea Piunti, Quanhong Ma, Ali Zhang, Xingyao He, Takahiro Sasaki, Dusten Unruh, Nundia Louis, and Hiroaki Katagi
- Abstract
Purpose:Radiotherapy (RT) has long been and remains the only treatment option for diffuse intrinsic pontine glioma (DIPG). However, all patients show evidence of disease progression within months of completing RT. No further clinical benefit has been achieved using alternative radiation strategies. Here, we tested the hypothesis that histone demethylase inhibition by GSK-J4 enhances radiation-induced DNA damage, making it a potential radiosensitizer in the treatment of DIPG.Experimental Design: We evaluated the effects of GSK-J4 on genes associated with DNA double-strand break (DSB) repair in DIPG cells by RNA sequence, ATAC sequence, and quantitative real-time PCR. Radiation-induced DNA DSB repair was analyzed by immunocytochemistry of DSB markers γH2AX and 53BP1, DNA-repair assay, and cell-cycle distribution. Clonogenic survival assay was used to determine the effect of GSK-J4 on radiation response of DIPG cells. In vivo response to radiation monotherapy and combination therapy of RT and GSK-J4 was evaluated in patient-derived DIPG xenografts.Results:GSK-J4 significantly reduced the expression of DNA DSB repair genes and DNA accessibility in DIPG cells. GSK-J4 sustained high levels of γH2AX and 53BP1 in irradiated DIPG cells, thereby inhibiting DNA DSB repair through homologous recombination pathway. GSK-J4 reduced clonogenic survival and enhanced radiation effect in DIPG cells. In vivo studies revealed increased survival of animals treated with combination therapy of RT and GSK-J4 compared with either monotherapy.Conclusions:Together, these results highlight GSK-J4 as a potential radiosensitizer and provide a rationale for developing combination therapy with radiation in the treatment of DIPG.
- Published
- 2023
- Full Text
- View/download PDF
18. Figure S2 from Radiosensitization by Histone H3 Demethylase Inhibition in Diffuse Intrinsic Pontine Glioma
- Author
-
Rintaro Hashizume, Ali Shilatifard, Lihua Zou, Oren J. Becher, Sriram Venneti, Stewart Goldman, Rishi Lulla, Nitin R. Wadhwani, Akihide Kondo, Kathryn L. Laurie, Vera Gorbunova, Andrei Seluanov, Xiao Tian, Angel M. Carcaboso, Jonathan B. Lamano, Yosuke Shimazu, Andrea Piunti, Quanhong Ma, Ali Zhang, Xingyao He, Takahiro Sasaki, Dusten Unruh, Nundia Louis, and Hiroaki Katagi
- Abstract
Supplementary Figure S2
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Tables 1 - 3 from Phase I Study of Vismodegib in Children with Recurrent or Refractory Medulloblastoma: A Pediatric Brain Tumor Consortium Study
- Author
-
Tom Curran, Richard J. Gilbertson, James M. Boyett, Naoko Takebe, Dana Wallace, Murali Chintagumpala, Stewart Goldman, Roger J. Packer, Larry E. Kun, Sue Kaste, David W. Ellison, Clinton F. Stewart, and Amar Gajjar
- Abstract
PDF file - 104K, Supplementary Table S1. Vismodegib Dosing Strategy Supplementary Table S2. Commonly Reported Adverse Events According to Grade Supplementary Table S3. Pharmacokinetics of Unbound Vismodegib in Pediatric Patients with Medulloblastoma
- Published
- 2023
- Full Text
- View/download PDF
20. Multi-institutional study of the frequency, genomic landscape, and outcome of IDH-mutant glioma in pediatrics
- Author
-
Kee Kiat Yeo, Sanda Alexandrescu, Jennifer A Cotter, Jayne Vogelzang, Varun Bhave, Marilyn M Li, Jianling Ji, Jamal K Benhamida, Marc K Rosenblum, Tejus A Bale, Nancy Bouvier, Kristiyana Kaneva, Tom Rosenberg, Mary Jane Lim-Fat, Hia Ghosh, Migdalia Martinez, Dolly Aguilera, Amy Smith, Stewart Goldman, Eli L Diamond, Igor Gavrilovic, Tobey J MacDonald, Matthew D Wood, Kellie J Nazemi, AiLien Truong, Andrew Cluster, Keith L Ligon, Kristina Cole, Wenya Linda Bi, Ashley S Margol, Matthias A Karajannis, and Karen D Wright
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) ,Pediatric Neuro-Oncology - Abstract
Background The incidence and biology of IDH1/2 mutations in pediatric gliomas are unclear. Notably, current treatment approaches by pediatric and adult providers vary significantly. We describe the frequency and clinical outcomes of IDH1/2-mutant gliomas in pediatrics. Methods We performed a multi-institutional analysis of the frequency of pediatric IDH1/2-mutant gliomas, identified by next-generation sequencing (NGS). In parallel, we retrospectively reviewed pediatric IDH1/2-mutant gliomas, analyzing clinico-genomic features, treatment approaches, and outcomes. Results Incidence: Among 851 patients with pediatric glioma who underwent NGS, we identified 78 with IDH1/2 mutations. Among patients 0–9 and 10–21 years old, 2/378 (0.5%) and 76/473 (16.1%) had IDH1/2-mutant tumors, respectively. Frequency of IDH mutations was similar between low-grade glioma (52/570, 9.1%) and high-grade glioma (25/277, 9.0%). Four tumors were graded as intermediate histologically, with one IDH1 mutation. Outcome: Seventy-six patients with IDH1/2-mutant glioma had outcome data available. Eighty-four percent of patients with low-grade glioma (LGG) were managed observantly without additional therapy. For low-grade astrocytoma, 5-year progression-free survival (PFS) was 42.9% (95%CI:20.3–63.8) and, despite excellent short-term overall survival (OS), numerous disease-related deaths after year 10 were reported. Patients with high-grade astrocytoma had a 5-year PFS/OS of 36.8% (95%CI:8.8–66.4) and 84% (95%CI:50.1–95.6), respectively. Patients with oligodendroglioma had excellent OS. Conclusions A subset of pediatric gliomas is driven by IDH1/2 mutations, with a higher rate among adolescents. The majority of patients underwent upfront observant management without adjuvant therapy. Findings suggest that the natural history of pediatric IDH1/2-mutant glioma may be similar to that of adults, though additional studies are needed.
- Published
- 2022
- Full Text
- View/download PDF
21. Characteristics of children ≤36 months of age with DIPG: A report from the international DIPG registry
- Author
-
Allison L Bartlett, Adam Lane, Brooklyn Chaney, Nancy Yanez Escorza, Katie Black, Anne Cochrane, Jane Minturn, Ute Bartels, Kathy Warren, Jordan Hansford, David Ziegler, Blanca Diez, Stewart Goldman, Roger Packer, Mark Kieran, Mariko DeWire-Schottmiller, Craig Erker, Michelle Monje-Deisseroth, Lars Wagner, Carl Koschmann, Kathleen Dorris, Chie-Schin Shih, Tim Hassall, Yvan Samson, Paul Fisher, Stacie S Wang, Karen Tsui, Gustavo Sevlever, Xiaoting Zhu, Phillip Dexheimer, Anthony Asher, Christine Fuller, Rachid Drissi, Blaise Jones, James Leach, and Maryam Fouladi
- Subjects
Cancer Research ,Oncology ,Child, Preschool ,Humans ,Brain Stem Neoplasms ,Glioma ,Registries ,Neurology (clinical) ,Astrocytoma ,Child ,Pediatric Neuro-Oncology - Abstract
Background Children ≤36 months with diffuse intrinsic pontine glioma (DIPG) have increased long-term survival (LTS, overall survival (OS) ≥24 months). Understanding distinguishing characteristics in this population is critical to improving outcomes. Methods Patients ≤36 months at diagnosis enrolled on the International DIPG Registry (IDIPGR) with central imaging confirmation were included. Presentation, clinical course, imaging, pathology and molecular findings were analyzed. Results Among 1183 patients in IDIPGR, 40 were eligible (median age: 29 months). Median OS was 15 months. Twelve patients (30%) were LTS, 3 (7.5%) very long-term survivors ≥5 years. Among 8 untreated patients, median OS was 2 months. Patients enrolled in the registry but excluded from our study by central radiology review or tissue diagnosis had median OS of 7 months. All but 1 LTS received radiation. Among 32 treated patients, 1-, 2-, 3-, and 5-year OS rates were 68.8%, 31.2%, 15.6% and 12.5%, respectively. LTS had longer duration of presenting symptoms (P = .018). No imaging features were predictive of outcome. Tissue and genomic data were available in 18 (45%) and 10 patients, respectively. Among 9 with known H3K27M status, 6 had a mutation. Conclusions Children ≤36 months demonstrated significantly more LTS, with an improved median OS of 15 months; 92% of LTS received radiation. Median OS in untreated children was 2 months, compared to 17 months for treated children. LTS had longer duration of symptoms. Excluded patients demonstrated a lower OS, contradicting the hypothesis that children ≤36 months with DIPG show improved outcomes due to misdiagnosis.
- Published
- 2022
- Full Text
- View/download PDF
22. Accuracy of central neuro-imaging review of DIPG compared with histopathology in the International DIPG Registry
- Author
-
Jane E. Minturn, Ayman El-Sheikh, Gustavo Sevlever, Michelle Monje-Deisseroth, Hetal Dholaria, Karen Tsui, Maryam Fouladi, Pratiti Bandopadhayay, Cynthia Hawkins, Scott L Coven, Lindsay Kilburn, Christopher L. Tinkle, David S. Ziegler, Eric Sandler, Yvan Samson, Jordan R. Hansford, Eric Bouffet, Sylvia Cheng, Sridharan Gururangan, Kathleen Dorris, Tim Hassall, Mohamed S. Zaghloul, Carl Koschmann, Sarah Leary, Mercedes Garcia Lombardi, Blaise V. Jones, Paul G. Fisher, Anthony Asher, Rachid Drissi, Blanca Diez, Kenneth J. Cohen, Jie Ma, Adriana Fonseca, Katie Black, Nicholas G. Gottardo, Stewart Goldman, Christine E. Fuller, Tabitha Cooney, Moatasem El-Ayadi, Adam Lane, Brooklyn Chaney, Mariko DeWire, Robert J. Greiner, Ute Bartels, Margot A Lazow, James L. Leach, Lars M. Wagner, and Roger J. Packer
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Cancer ,Autopsy ,Glioma ,Astrocytoma ,medicine.disease ,Tissue acquisition ,Glutamates ,Oncology ,Neuroimaging ,Biopsy ,medicine ,Medical imaging ,Brain Stem Neoplasms ,Humans ,Histopathology ,Registries ,Neurology (clinical) ,Radiology ,Medical diagnosis ,business ,Pediatric Neuro-Oncology - Abstract
Background Diffuse intrinsic pontine glioma (DIPG) remains a clinico-radiologic diagnosis without routine tissue acquisition. Reliable imaging distinction between DIPG and other pontine tumors with potentially more favorable prognoses and treatment considerations is essential. Methods Cases submitted to the International DIPG registry (IDIPGR) with histopathologic and/or radiologic data were analyzed. Central imaging review was performed on diagnostic brain MRIs (if available) by two neuro-radiologists. Imaging features suggestive of alternative diagnoses included nonpontine origin, Results Among 286 patients with pathology from biopsy and/or autopsy, 23 (8%) had histologic diagnoses inconsistent with DIPG, most commonly nondiffuse low-grade gliomas and embryonal tumors. Among 569 patients with centrally-reviewed diagnostic MRIs, 40 (7%) were classified as non-DIPG, alternative diagnosis suspected. The combined analysis included 151 patients with both histopathology and centrally-reviewed MRI. Of 77 patients with imaging classified as characteristic of DIPG, 76 (99%) had histopathologic diagnoses consistent with DIPG (infiltrating grade II-IV gliomas). Of 57 patients classified as likely DIPG with some unusual imaging features, 55 (96%) had histopathologic diagnoses consistent with DIPG. Of 17 patients with imaging features suggestive of an alternative diagnosis, eight (47%) had histopathologic diagnoses inconsistent with DIPG (remaining patients were excluded due to nonpontine tumor origin). Association between central neuro-imaging review impression and histopathology was significant (p < 0.001), and central neuro-imaging impression was prognostic of overall survival. Conclusions The accuracy and important role of central neuro-imaging review in confirming the diagnosis of DIPG is demonstrated.
- Published
- 2021
- Full Text
- View/download PDF
23. Characteristics of patients ≥10 years of age with diffuse intrinsic pontine glioma: a report from the International DIPG/DMG Registry
- Author
-
Craig Erker, Adam Lane, Brooklyn Chaney, Sarah Leary, Jane E Minturn, Ute Bartels, Roger J Packer, Kathleen Dorris, Nicholas G Gottardo, Katherine E Warren, Alberto Broniscer, Mark W Kieran, Xiaoting Zhu, Peter White, Phillip J Dexheimer, Katie Black, Anthony Asher, Mariko DeWire, Jordan R Hansford, Sridharan Gururangan, Javad Nazarian, David S Ziegler, Eric Sandler, Allison Bartlett, Stewart Goldman, Chie-Schin Shih, Tim Hassall, Hetal Dholaria, Pratiti Bandopadhayay, Yvan Samson, Michelle Monje, Paul G Fisher, Andrew Dodgshun, Sarah Parkin, Murali Chintagumpala, Karen Tsui, David Gass, Valerie Larouche, Emmett Broxson, Mercedes Garcia Lombardi, Stacie Shiqi Wang, Jie Ma, Cynthia Hawkins, Dima Hamideh, Lars Wagner, Carl Koschmann, Christine Fuller, Rachid Drissi, Blaise V Jones, James Leach, and Maryam Fouladi
- Subjects
Adult ,Cancer Research ,Pediatrics ,medicine.medical_specialty ,IDH1 ,Adolescent ,Population ,Clinical Investigations ,Autopsy ,Astrocytoma ,Young Adult ,Biopsy ,medicine ,Brain Stem Neoplasms ,Humans ,In patient ,Registries ,Young adult ,Child ,education ,ATRX ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Diffuse Intrinsic Pontine Glioma ,Glioma ,Oncology ,Neurology (clinical) ,business ,Median survival - Abstract
BackgroundDiffuse intrinsic pontine gliomas (DIPG) generally occur in young school-age children, although can occur in adolescents and young adults. The purpose of this study was to describe clinical, radiological, pathologic, and molecular characteristics in patients ≥10 years of age with DIPG enrolled in the International DIPG Registry (IDIPGR).MethodsPatients ≥10 years of age at diagnosis enrolled in the IDIPGR with imaging confirmed DIPG diagnosis were included. The primary outcome was overall survival (OS) categorized as long-term survivors (LTS) (≥24 months) or short-term survivors (STS) (ResultsAmong 1010 patients, 208 (21%) were ≥10 years of age at diagnosis; 152 were eligible with a median age of 12 years (range 10-26.8). Median OS was 13 (2-82) months. The 1-, 3-, and 5-year OS was 59.2%, 5.3%, and 3.3%, respectively. The 18/152 (11.8%) LTS were more likely to be older (P < .01) and present with longer symptom duration (P < .01). Biopsy and/or autopsy were performed in 50 (33%) patients; 77%, 61%, 33%, and 6% of patients tested had H3K27M (H3F3A or HIST1H3B), TP53, ATRX, and ACVR1 mutations/genome alterations, respectively. Two of 18 patients with IDH1 testing were IDH1-mutant and 1 was a LTS. The presence or absence of H3 alterations did not affect survival.ConclusionPatients ≥10 years old with DIPG have a median survival of 13 months. LTS present with longer symptom duration and are likely to be older at presentation compared to STS. ATRX mutation rates were higher in this population than the general DIPG population.
- Published
- 2021
- Full Text
- View/download PDF
24. Electronic Dashboard to Improve Outcomes in Pediatric Patients With Type 1 Diabetes Mellitus
- Author
-
Lily Sandblom, Chirag Kapadia, Vinay Vaidya, Melissa Chambers, Rob Gonsalves, Lea Ann Holzmeister, Fran Hoekstra, and Stewart Goldman
- Subjects
Endocrinology, Diabetes and Metabolism ,Biomedical Engineering ,Internal Medicine ,Bioengineering - Abstract
Background and Objectives: Incidence of type 1 diabetes mellitus (T1DM) is increasing, and these patients often have poor glycemic control. Electronic dashboards summating patient data have been shown to improve patient outcomes in other conditions. In addition, educating patients on T1DM has shown to improve glycated hemoglobin (A1C) levels. We hypothesized that using data from the electronic dashboard to monitor defined diabetes management activities to implement population-based interventions would improve patient outcomes. Methods: Inclusion criteria included patients aged 0 to 18 years at Phoenix Children’s Hospital with T1DM. Patient data were collected via the electronic dashboard, and both diabetes management activities (A1C, patient admissions, and visits to the emergency department) and patient outcomes (patient education, appointment compliance, follow-up after hospital admission) were analyzed. Results: This study revealed that following implementation of the electronic dashboard, the percentage of patients receiving appropriate education increased from 48% to 80% (Z-score = 23.55, P < .0001), the percentage of patients attending the appropriate number of appointments increased from 50% to 68.2%, and the percentage of patients receiving follow-up care within 40 days after a hospital admission increased from 43% to 70%. The median A1C level decreased from 9.1% to 8.2% (Z-score = −6.74, P < .0001), and patient admissions and visits to the emergency department decreased by 20%. Conclusions: This study shows, with the implementation of an electronic dashboard, we were able to improve outcomes for our pediatric patients with T1DM. This tool can be used at other institutions to improve care and outcomes for pediatric patients with T1DM and other chronic conditions.
- Published
- 2023
- Full Text
- View/download PDF
25. Therapeutic targeting of transcriptional elongation in diffuse intrinsic pontine glioma
- Author
-
Amanda Saratsis, Edwin R. Smith, Yongzhan Zhang, Gavin T. Blyth, Hiroaki Katagi, Nozomu Takata, Akihide Kondo, Stewart Goldman, Lihua Zou, Yuki Aoi, Rintaro Hashizume, Yusuke Tomita, Frank Eckerdt, Ali Shilatifard, Oren J. Becher, Takahiro Sasaki, and Emily J. Rendleman
- Subjects
Cancer Research ,biology ,Cell growth ,Chemistry ,RNA polymerase II ,Cell cycle ,Chromatin ,Oncology ,Apoptosis ,Transcription (biology) ,Gene expression ,Cancer research ,biology.protein ,Neurology (clinical) ,Viability assay - Abstract
BackgroundDiffuse intrinsic pontine glioma (DIPG) is associated with transcriptional dysregulation driven by H3K27 mutation. The super elongation complex (SEC) is required for transcriptional elongation through release of RNA polymerase II (Pol II). Inhibition of transcription elongation by SEC disruption can be an effective therapeutic strategy of H3K27M-mutant DIPG. Here, we tested the effect of pharmacological disruption of the SEC in H3K27M-mutant DIPG to advance understanding of the molecular mechanism and as a new therapeutic strategy for DIPG.MethodsShort hairpin RNAs (shRNAs) were used to suppress the expression of AF4/FMR2 4 (AFF4), a central SEC component, in H3K27M-mutant DIPG cells. A peptidomimetic lead compound KL-1 was used to disrupt a functional component of SEC. Cell viability assay, colony formation assay, and apoptosis assay were utilized to analyze the effects of KL-1 treatment. RNA- and ChIP-sequencing were used to determine the effects of KL-1 on gene expression and chromatin occupancy. We treated mice bearing H3K27M-mutant DIPG patient-derived xenografts (PDXs) with KL-1. Intracranial tumor growth was monitored by bioluminescence image and therapeutic response was evaluated by animal survival.ResultsDepletion of AFF4 significantly reduced the cell growth of H3K27M-mutant DIPG. KL-1 increased genome-wide Pol II occupancy and suppressed transcription involving multiple cellular processes that promote cell proliferation and differentiation of DIPG. KL-1 treatment suppressed DIPG cell growth, increased apoptosis, and prolonged animal survival with H3K27M-mutant DIPG PDXs.ConclusionsSEC disruption by KL-1 increased therapeutic benefit in vitro and in vivo, supporting a potential therapeutic activity of KL-1 in H3K27M-mutant DIPG.
- Published
- 2021
- Full Text
- View/download PDF
26. Safety of Tumor Treating Fields (TTFields) therapy in pediatric patients with malignant brain tumors: Post-marketing surveillance data
- Author
-
Stewart Goldman, Ashley Margol, Eugene I. Hwang, Kazuhiro Tanaka, Bogdana Suchorska, John R. Crawford, and Santosh Kesari
- Subjects
Cancer Research ,Oncology - Abstract
There is an unmet need to develop effective and tolerable treatments for pediatric patients with malignant central nervous system tumors. This is especially essential for pediatric patients with aggressive brain tumors such as high-grade gliomas, which have a typical survival rate of under 2 years. Tumor Treating Fields (TTFields) are locoregional, noninvasive electric fields that produce an antimitotic effect on cancerous cells when applied to the skin via arrays. TTFields therapy (200 kHz) is currently approved in adult patients with newly diagnosed glioblastoma (GBM), with temozolomide, and recurrent GBM as monotherapy. Positive preclinical and clinical data have encouraged off-label use of TTFields therapy in pediatric patients with brain tumors, and this study aims to explore the safety of TTFields therapy in pediatric patients (0–18 years of age) based on data from an unsolicited post-marketing surveillance safety database. The real-world data reported here demonstrate that TTFields therapy has a favorable safety profile for pediatric patients with brain tumors, with no new safety signals observed. Findings from this study warrant further research into the efficacy of TTFields therapy, as well as its potential impact on the quality of life in pediatric patients.
- Published
- 2022
27. Convection-Enhanced Delivery of Enhancer of Zeste Homolog-2 (EZH2) Inhibitor for the Treatment of Diffuse Intrinsic Pontine Glioma
- Author
-
Stewart Goldman, Takahiro Sasaki, Oren J. Becher, Rintaro Hashizume, and Hiroaki Katagi
- Subjects
Drug ,media_common.quotation_subject ,Brain Stem Neoplasm ,Convection ,Infusion Site ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Brain Stem Neoplasms ,Humans ,Medicine ,Distribution (pharmacology) ,Enhancer of Zeste Homolog 2 Protein ,media_common ,business.industry ,Diffuse Intrinsic Pontine Glioma ,EZH2 ,Research—Animal ,Xenograft Model Antitumor Assays ,030220 oncology & carcinogenesis ,Cancer research ,Systemic administration ,Surgery ,Neurology (clinical) ,Brainstem ,business ,Convection-Enhanced Delivery ,030217 neurology & neurosurgery - Abstract
Background Diffuse intrinsic pontine glioma (DIPG) is a fatal childhood brain tumor and the majority of patients die within 2 yr after initial diagnosis. Factors that contribute to the dismal prognosis of these patients include the infiltrative nature and anatomic location in an eloquent area of the brain, which precludes total surgical resection, and the presence of the blood-brain barrier (BBB), which reduces the distribution of systemically administered agents. Convection-enhanced delivery (CED) is a direct infusion technique to deliver therapeutic agents into a target site in the brain and able to deliver a high concentration drug to the infusion site without systemic toxicities. Objective To assess the efficacy of enhancer of zeste homolog-2 (EZH2) inhibitor by CED against human DIPG xenograft models. Methods The concentration of EZH2 inhibitor (EPZ-6438) in the brainstem tumor was evaluated by liquid chromatography-mass spectrometry (LC/MS). We treated mice-bearing human DIPG xenografts with EPZ-6438 using systemic (intraperitoneal) or CED administration. Intracranial tumor growth was monitored by bioluminescence image, and the therapeutic response was evaluated by animal survival. Results LC/MS analysis showed that the concentration of EPZ-6438 in the brainstem tumor was 3.74% of serum concentration after systemic administration. CED of EPZ-6438 suppressed tumor growth and significantly extended animal survival when compared to systemic administration of EPZ-6438 (P = .0475). Conclusion Our results indicate that CED of an EZH2 inhibitor is a promising strategy to bypass the BBB and to increase the efficacy of an EZH2 inhibitor for the treatment of DIPG.
- Published
- 2020
- Full Text
- View/download PDF
28. MR imaging features of diffuse intrinsic pontine glioma and relationship to overall survival: report from the International DIPG Registry
- Author
-
Jane E. Minturn, Ute Bartels, Nicholas G. Gottardo, Jordan R. Hansford, Christine Fuller, Cynthia Hawkins, Renee Doughman, Mariko DeWire-Schottmiller, Sarah Leary, Brooklyn Chaney, Blaise V. Jones, Michelle Monje-Deisseroth, James Roebker, Katherine E. Warren, Hetal Dholaria, Austin Schafer, Rachid Drissi, Adam Lane, Joshua J Baugh, Roger J. Packer, James L. Leach, Paul G. Fisher, Stewart Goldman, Stacie S. Wang, Maryam Fouladi, and David S. Ziegler
- Subjects
Imaging Feature ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Diffuse Intrinsic Pontine Glioma ,Magnetic resonance imaging ,Magnetic Resonance Imaging ,Mr imaging ,Text mining ,Oncology ,Basic and Translational Investigations ,medicine ,Overall survival ,Medical imaging ,Brain Stem Neoplasms ,Humans ,Prospective Studies ,Registries ,Neurology (clinical) ,Radiology ,Medical diagnosis ,Prospective cohort study ,business - Abstract
Abtract Background This study describes imaging features of diffuse intrinsic pontine glioma (DIPG) and correlates with overall survival (OS) and histone mutation status in the International DIPG Registry (IDIPGR). Methods Four hundred cases submitted to the IDIPGR with a local diagnosis of DIPG and baseline MRI were evaluated by consensus review of 2 neuroradiologists; 43 cases were excluded (inadequate imaging or alternative diagnoses). Agreement between reviewers, association with histone status, and univariable and multivariable analyses relative to OS were assessed. Results On univariable analysis imaging features significantly associated with worse OS included: extrapontine extension, larger size, enhancement, necrosis, diffusion restriction, and distant disease. On central review, 9.5% of patients were considered not to have DIPG. There was moderate mean agreement of MRI features between reviewers. On multivariable analysis, chemotherapy, age, and distant disease were predictors of OS. There was no difference in OS between wild-type and H3 mutated cases. The only imaging feature associated with histone status was the presence of ill-defined signal infiltrating pontine fibers. Conclusions Baseline imaging features are assessed in the IDIPGR. There was a 9.5% discordance in DIPG diagnosis between local and central review, demonstrating need for central imaging confirmation for prospective trials. Although several imaging features were significantly associated with OS (univariable), only age and distant disease were significant on multivariable analyses. There was limited association of imaging features with histone mutation status, although numbers are small and evaluation exploratory.
- Published
- 2020
- Full Text
- View/download PDF
29. Phase II study of peginterferon alpha-2b for patients with unresectable or recurrent craniopharyngiomas: a Pediatric Brain Tumor Consortium report
- Author
-
Nathan Robison, Giles W. Robinson, Tina Young Poussaint, Lindsay Kilburn, Stewart Goldman, Maryam Fouladi, Adekunle M. Adesina, Sonia Partap, Donald W Parsons, Ian F. Pollack, Ira J. Dunkel, Arzu Onar-Thomas, Catherine A. Billups, Ashok Panigrahy, Regina I. Jakacki, and Alberto Broniscer
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Pediatric Brain Tumor Consortium ,Adolescent ,medicine.medical_treatment ,Phases of clinical research ,Interferon alpha-2 ,Polyethylene Glycols ,Craniopharyngioma ,Pegylated interferon ,medicine ,Humans ,Pituitary Neoplasms ,Progression-free survival ,Young adult ,Child ,Brain Neoplasms ,business.industry ,Infant ,Interferon-alpha ,medicine.disease ,Recombinant Proteins ,Surgery ,Radiation therapy ,Treatment Outcome ,Oncology ,Child, Preschool ,Peginterferon alfa-2b ,Female ,Neurology (clinical) ,business ,Pediatric Neuro-Oncology ,medicine.drug - Abstract
BackgroundCraniopharyngiomas account for approximately 1.2–4% of all CNS tumors. They are typically treated with a combination of surgical resection and focal radiotherapy. Unfortunately, treatment can lead to permanent deleterious effects on behavior, learning, and endocrine function.MethodsThe Pediatric Brain Tumor Consortium performed a multicenter phase 2 study in children and young adults with unresectable or recurrent craniopharyngioma (PBTC-039). Between December 2013 and November 2017, nineteen patients (median age at enrollment, 13.1 y; range, 2–25 y) were enrolled in one of 2 strata: patients previously treated with surgery alone (stratum 1) or who received radiation (stratum 2).ResultsEighteen eligible patients (8 male, 10 female) were treated with weekly subcutaneous pegylated interferon alpha-2b for up to 18 courses (108 wk). Therapy was well tolerated with no grade 4 or 5 toxicities. 2 of the 7 eligible patients (28.6%) in stratum 1 had a partial response, but only one response was sustained for more than 3 months. None of the 11 stratum 2 patients had an objective radiographic response, although median progression-free survival was 19.5 months.ConclusionsPegylated interferon alpha-2b treatment, in lieu of or following radiotherapy, was well tolerated in children and young adults with recurrent craniopharyngiomas. Although objective responses were limited, progression-free survival results are encouraging, warranting further studies.
- Published
- 2020
- Full Text
- View/download PDF
30. A phase II study of continuous oral mTOR inhibitor everolimus for recurrent, radiographic-progressive neurofibromatosis type 1–associated pediatric low-grade glioma: a Neurofibromatosis Clinical Trials Consortium study
- Author
-
Peter E. Manley, Nathan Robison, Stewart Goldman, Michael Fisher, John P. Perentesis, Alan B. Cantor, Coretta Thomas, Bruce R. Korf, Alyssa Reddy, Mark W. Kieran, Susan N. Chi, Sanjay P. Prabhu, Nicole J. Ullrich, Tomoyuki Mizuno, Jeffrey C. Allen, Alexander A. Vinks, David Viskochil, Gary Cutter, Roger J. Packer, and David H. Gutmann
- Subjects
Oncology ,Cancer Research ,Phases of clinical research ,Clinical endpoint ,Child ,Cancer ,Pediatric ,education.field_of_study ,low-grade glioma ,TOR Serine-Threonine Kinases ,Glioma ,6.1 Pharmaceuticals ,Biotechnology ,medicine.drug ,Pediatric Research Initiative ,medicine.medical_specialty ,Neurofibromatosis 1 ,Neurofibromatoses ,Combination therapy ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Population ,Antineoplastic Agents ,Malignant peripheral nerve sheath tumor ,Neurofibromatosis ,Rare Diseases ,Clinical Research ,Internal medicine ,medicine ,Humans ,Everolimus ,Oncology & Carcinogenesis ,education ,RAD001 ,PI3K/AKT/mTOR pathway ,Sirolimus ,business.industry ,Editorials ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Brain Disorders ,Brain Cancer ,Orphan Drug ,NF1 ,Neurology (clinical) ,PIK3K/mTOR pathway ,business ,Pediatric Neuro-Oncology - Abstract
Background Activation of the mammalian target of rapamycin (mTOR) pathway is observed in neurofibromatosis type 1 (NF1) associated low-grade gliomas (LGGs), but agents that inhibit this pathway, including mTOR inhibitors, have not been studied in this population. We evaluate the efficacy of the orally administered mTOR inhibitor everolimus for radiographically progressive NF1-associated pediatric LGGs. Methods Children with radiologic-progressive, NF1-associated LGG and prior treatment with a carboplatin-containing chemotherapy were prospectively enrolled on this phase II clinical trial to receive daily everolimus. Whole blood was analyzed for everolimus and markers of phosphatidylinositol-3 kinase (PI3K)/mTOR pathway inhibition. Serial MRIs were obtained during treatment. The primary endpoint was progression-free survival at 48 weeks. Results Twenty-three participants (median age, 9.4 y; range, 3.2–21.6 y) were enrolled. All participants were initially evaluable for response; 1 patient was removed from study after development of a malignant peripheral nerve sheath tumor. Fifteen of 22 participants (68%) demonstrated a response, defined as either shrinkage (1 complete response, 2 partial response) or arrest of tumor growth (12 stable disease). Of these, 10/15 remained free of progression (median follow-up, 33 mo). All remaining 22 participants were alive at completion of therapy. Treatment was well tolerated; no patient discontinued therapy due to toxicity. Pharmacokinetic parameters and pre-dose concentrations showed substantial between-subject variability. PI3K/mTOR pathway inhibition markers demonstrating blood mononuclear cell mTOR pathway inactivation was achieved in most participants. Conclusion Individuals with recurrent/progressive NF1-associated LGG demonstrate significant disease stability/shrinkage during treatment with oral everolimus with a well-tolerated toxicity profile. Everolimus is well suited for future consideration as upfront or combination therapy in this patient population.
- Published
- 2020
- Full Text
- View/download PDF
31. Tolerability of Tumor Treating Fields (TTFields) in pediatric high-grade glioma patients illustrated by post-market surveillance data
- Author
-
Stewart Goldman
- Published
- 2022
- Full Text
- View/download PDF
32. Phase I study of ribociclib and everolimus in children with newly diagnosed DIPG and high-grade glioma: A CONNECT pediatric neuro-oncology consortium report
- Author
-
Mariko DeWire, Margot Lazow, Olivia Campagne, James Leach, Christine Fuller, Shiva Senthil Kumar, Joseph Stanek, Peter de Blank, Trent R Hummel, Natasha Pillay-Smiley, Ralph Salloum, Charles B Stevenson, Patricia Baxter, David Gass, Stewart Goldman, Sarah E S Leary, Adam Carle, Leonie Mikael, Dorothy Crabtree, Brooklyn Chaney, Adam Lane, Rachid Drissi, Clinton F Stewart, and Maryam Fouladi
- Subjects
General Medicine - Abstract
Background Genomic aberrations in the cell cycle and PI3K/Akt/mTOR pathways have been reported in diffuse intrinsic pontine glioma (DIPG) and high-grade glioma (HGG). Dual inhibition of CDK4/6 and mTOR has biologic rationale and minimal overlapping toxicities. This study determined the recommended phase 2 dose (RP2D) of ribociclib and everolimus following radiotherapy in children with DIPG and HGG. Methods Patients were enrolled according to a Rolling-6 design and received ribociclib and everolimus once daily for 21 and 28 days, respectively. All patients with HGG and biopsied DIPG were screened for retinoblastoma protein presence by immunohistochemistry. Pharmacokinetics were analyzed. Results Nineteen patients enrolled (median age: 8 years [range: 2-18]). Three patients enrolled at each dose level 1 and 2 without dose-limiting toxicities (DLT). Thirteen patients were enrolled at dose level 3, with one patient experiencing a DLT (grade 3 infection). One patient came off therapy before cycle 9 due to cardiac toxicity. The most common grade 3/4 toxicities were neutropenia (33%), leucopenia (17%), and lymphopenia (11%). Steady-state everolimus exposures in combination were 1.9 ± 0.9-fold higher than single-agent administration. Median overall survival for 15 patients with DIPG was 13.9 months; median event-free survival for four patients with HGG was 10.5 months. Two longer survivors had tumor molecular profiling identifying CDKN2A/B deletion and CDK4 overexpression. Conclusion The combination of ribociclib and everolimus following radiotherapy in children with newly diagnosed DIPG and HGG was well tolerated, with a RP2D of ribociclib 170 mg/m2 and everolimus 1.5 mg/m2. Results will inform a molecularly guided phase II study underway to evaluate efficacy.
- Published
- 2022
- Full Text
- View/download PDF
33. EANO, SNO and Euracan consensus review on the current management and future development of intracranial germ cell tumors in adolescents and young adults
- Author
-
Thomas Czech, Mark M. Souweidane, D Haas-Kogen, Alexandre Vasiljevic, P Wen, C Faure Conter, Eric Bouffet, Jonathan L. Finlay, D. Frappaz, Matthew J. Murray, RD Kortmann, Ute Bartels, Dennis W. W. Shaw, Ching C. Lau, David Schiff, S Schöenberger, Jeffrey C. Allen, Girish Dhall, James Nicholson, P Robertson, Gabriele Calaminus, Claire Alapetite, Giovanni Morana, A Albanese, Stewart Goldman, Murray, Matthew J [0000-0002-4480-1147], Bartels, Ute [0000-0003-2112-5251], Albanese, Assunta [0000-0003-4051-6661], Czech, Thomas [0000-0001-8112-2795], Bouffet, Eric [0000-0002-6832-6539], and Apollo - University of Cambridge Repository
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Consensus ,Adolescent ,medicine.medical_treatment ,Medizin ,germinoma ,Disease ,Craniospinal Irradiation ,Young Adult ,Testicular Neoplasms ,Medicine ,Humans ,Young adult ,Radical surgery ,Retrospective Studies ,non-germinomatous germ cell tumor ,Chemotherapy ,Germinoma ,adolescents and young adults ,brain tumors ,germ cell tumor ,business.industry ,Brain Neoplasms ,Neoplasms, Germ Cell and Embryonal ,medicine.disease ,Radiation therapy ,Oncology ,Neurology (clinical) ,Germ cell tumors ,Radiology ,business - Abstract
The incidence of intracranial germ cell tumors (iGCT) is much lower in European and North American (E&NA) than in Asian population. However, E&NA cooperative groups have simultaneously developed with success treatment strategies with specific attention paid to long-term sequelae. Neurological sequelae may be reduced by establishing a diagnosis with an endoscopic biopsy and/or cerebrospinal fluid (CSF) and/or serum analysis, deferring the need to perform a radical surgery. Depending on markers and/or histological characteristics, patients are treated as either germinoma or non-germinomatous germ cell tumors (NGGCT). Metastatic disease is defined by a positive CSF cytology and/or distant drops in craniospinal MRI. The combination of surgery and/or chemotherapy and radiation therapy is tailored according to grouping and staging. With more than 90% 5-year event-free survival (EFS), localized germinomas can be managed without aggressive surgery, and benefit from chemotherapy followed by whole ventricular irradiation with local boost. Bifocal germinomas are treated as non-metastatic entities. Metastatic germinomas may be cured with craniospinal irradiation. With a 5-year EFS over 70%, NGGCT benefit from chemotherapy followed by delayed surgery in case of residual disease, and some form of radiotherapy. Future strategies will aim at decreasing long-term side effects while preserving high cure rates.
- Published
- 2022
34. Mass cytometry detects H3.3K27M-specific vaccine responses in diffuse midline glioma
- Author
-
Erin R Bonner, Lisa H. Butterfield, Annette M. Molinaro, Nicholas S Whipple, Michael D. Prados, Jared Taitt, Andres M. Salazar, Ryan Gilbert, Sabine Mueller, Javad Nazarian, Neil D. Almeida, Anu Banerjee, Hideho Okada, Susan N. Chi, Karen Gauvain, Javier Villanueva-Meyer, Kellie J. Nazemi, John Robertson Crawford, Rishi Lulla, Payal Watchmaker, Takahide Nejo, Kaori Okada, Stewart Goldman, University of Zurich, and Okada, Hideho
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer immunotherapy ,2700 General Medicine ,CD8-Positive T-Lymphocytes ,Brain cancer ,Medical and Health Sciences ,Histones ,0302 clinical medicine ,Brain Stem Neoplasms ,Child ,Stroke ,Cancer ,Pediatric ,Vaccines ,Immunity, Cellular ,Brain Neoplasms ,General Medicine ,Glioma ,Flow Cytometry ,Neoplasm Proteins ,medicine.anatomical_structure ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Child, Preschool ,Female ,Corrigendum ,medicine.drug ,Adult ,medicine.medical_specialty ,Adolescent ,T cell ,Immunology ,Clinical Trials and Supportive Activities ,Mutation, Missense ,610 Medicine & health ,Peripheral blood mononuclear cell ,Cancer Vaccines ,Vaccine Related ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,Injection site reaction ,medicine ,Humans ,Preschool ,Adverse effect ,Dexamethasone ,business.industry ,Immunity ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Brain Disorders ,Good Health and Well Being ,030104 developmental biology ,Amino Acid Substitution ,10036 Medical Clinic ,Mutation ,Immunization ,Cellular ,Missense ,Clinical Medicine ,business ,CD8 - Abstract
BACKGROUND: Patients with diffuse midline gliomas (DMGs), including diffuse intrinsic pontine glioma (DIPG), have dismal outcomes. We previously described the H3.3K27M mutation as a shared neoantigen in HLA-A*02.01(+), H3.3K27M(+) DMGs. Within the Pacific Pediatric Neuro-Oncology Consortium, we assessed the safety and efficacy of an H3.3K27M-targeted peptide vaccine. METHODS: Newly diagnosed patients, aged 3–21 years, with HLA-A*02.01(+) and H3.3K27M(+) status were enrolled in stratum A (DIPG) or stratum B (nonpontine DMG). Vaccine was administered in combination with polyinosinic-polycytidylic acid-poly-I-lysine carboxymethylcellulose (poly-ICLC) every 3 weeks for 8 cycles, followed by once every 6 weeks. Immunomonitoring and imaging were performed every 3 months. Imaging was centrally reviewed. Immunological responses were assessed in PBMCs using mass cytometry. RESULTS: A total of 19 patients were enrolled in stratum A (median age,11 years) and 10 in stratum B (median age, 13 years). There were no grade-4 treatment-related adverse events (TRAEs). Injection site reaction was the most commonly reported TRAE. Overall survival (OS) at 12 months was 40% (95% CI, 22%–73%) for patients in stratum A and 39% (95% CI, 16%–93%) for patients in stratum B. The median OS was 16.1 months for patients who had an expansion of H3.3K27M-reactive CD8(+) T cells compared with 9.8 months for their counterparts (P = 0.05). Patients with DIPG with below-median baseline levels of myeloid-derived suppressor cells had prolonged OS compared with their counterparts (P < 0.01). Immediate pretreatment dexamethasone administration was inversely associated with H3.3K27M-reactive CD8(+) T cell responses. CONCLUSION: Administration of the H3.3K27M-specific vaccine was well tolerated. Patients with H3.3K27M-specific CD8(+) immunological responses demonstrated prolonged OS compared with nonresponders. TRIAL REGISTRATION: ClinicalTrials.gov NCT02960230. FUNDING: The V Foundation, the Pacific Pediatric Neuro-Oncology Consortium Foundation, the Pediatric Brain Tumor Foundation, the Mithil Prasad Foundation, the MCJ Amelior Foundation, the Anne and Jason Farber Foundation, Will Power Research Fund Inc., the Isabella Kerr Molina Foundation, the Parker Institute for Cancer Immunotherapy, and the National Institute of Neurological Disorders and Stroke (NINDS), NIH (R35NS105068).
- Published
- 2022
- Full Text
- View/download PDF
35. DIPG-25. Patterns of cerebrospinal fluid diversion and survival in children with diffuse intrinsic pontine glioma: a report from the International Diffuse Intrinsic Pontine Glioma Registry
- Author
-
Tabitha Cooney, Mariko DeWire-Schottmiller, Adam Lane, Raya Saab, Pratiti Bandopadhayay, Kathleen Dorris, Roger Packer, Lindsay Kilburn, Jane Minturn, Andrew Dodgshun, Sara Parkin, Stewart Goldman, Eric Sandler, Robert Greiner, Nicholas Gottardo, Hetal Dholaria, Scott Coven, Tim Hassall, Jordan Hansford, Yvan Samson, Sarah Leary, Ute Bartels, Adriana Fonseca, Eric Bouffet, Christopher Tinkle, Michelle Monje, Paul Fisher, David Ziegler, Murali Chintagumpala, Lars Wagner, Carl Koschmann, James Leach, Blaise Jones, Elisa Carrasquedo Benito, Hailey Bond, Brooklyn Chaney, Katie Black, Anthony Asher, Maryam Fouladi, Lindsey Hoffman, and Katherine Warren
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: There are no standard practice guidelines for cerebrospinal (CSF) diversion for diffuse intrinsic pontine glioma (DIPG), nor clear understanding of potential for palliation and life-prolongation. We evaluated CSF diversion characteristics in children with DIPG to determine incidence, indications, symptom effects, and survival. METHODS: Data were extracted from subjects registered in the International DIPG registry (IDIPGR). Univariable analyses was performed using the Fisher’s exact test or Wilcoxon rank sum test. Survival was estimated using the Kaplan-Meier method. RESULTS: Evaluable patients (n=542) met criteria for DIPG diagnosis by central radiologic review; of those, 126 (23%) had permanent CSF diversion. Median time from diagnosis to diversion was 0.5 months (IQR 0.1-4.5 months). Those with permanent diversion were significantly younger (median 5.4 years vs 7.0 years, p
- Published
- 2022
- Full Text
- View/download PDF
36. EPCT-06. Phase I study of ribociclib and everolimus post-radiotherapy in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG) and high-grade glioma (HGG): Updated report from the COllaborative Network for NEuro-Oncology Clinical Trials (CONNECT)
- Author
-
Margot A Lazow, Mariko DeWire, Olivia Campagne, James L Leach, Christine Fuller, Shiva Senthil Kumar, Joseph Stanek, Peter de Blank, Trent R Hummel, Natasha Pillay-Smiley, Ralph Salloum, Charles B Stevenson, Patricia Baxter, David Gass, Stewart Goldman, Sarah E S Leary, Adam Carle, Adam Lane, Rachid Drissi, Clinton Stewart, and Maryam Fouladi
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: Dual inhibition of CDK4/6 and mTOR in DIPG and pediatric HGG has strong biologic rationale, given prevalent genetic alterations resulting in upregulated cell cycle and PI3K/mTOR pathways in these diseases, as well as non-overlapping agent toxicities. This study sought to evaluate safety/tolerability and determine the recommended phase 2 dose (RP2D) of ribociclib and everolimus among children with newly diagnosed DIPG and HGG post-radiotherapy. METHODS: Patients were enrolled according to a Rolling-6 design and received oral ribociclib and everolimus once daily for 21 and 28 days, respectively, starting 2-4 weeks post-completion of radiotherapy. All HGG and biopsied DIPG patients were screened for RB protein presence by immunohistochemistry. Pharmacokinetics and survival data were analyzed. RESULTS: Nineteen patients enrolled (median age: 8 years [range: 2-18]). Three patients enrolled at each of dose levels 1 and 2 without dose-limiting toxicities (DLTs). Thirteen patients enrolled at dose level 3, with one patient experiencing a DLT (grade 3 infection). One patient came off therapy prior to cycle 9 due to cardiac toxicity. The most common grade 3/4 toxicities were neutropenia (33%), leucopenia (17%), and lymphopenia (11%). Steady-state everolimus exposures in combination were 1.9±0.9-fold higher than single-agent administration. Median overall survival (OS) for 15 patients with DIPG was 13.9 months, with 12-, 24-, and 36-month OS of 53.3%, 38.9%, and 38.9%. Median event-free survival for four patients with HGG was 10.5 months. Among patients with tumor molecular profiling, two longer survivors (OS: 20, >37 months) had evidence of cell cycle upregulation with CDKN2A/B deletion and CDK4 overexpression identified. CONCLUSIONS: The combination of ribociclib and everolimus was well-tolerated post-radiotherapy in children with newly diagnosed DIPG and HGG, with a RP2D of ribociclib 170 mg/m2 days 1-21 and everolimus 1.5 mg/m2 days 1-28. Results will inform a molecularly-guided phase II study currently underway to evaluate efficacy.
- Published
- 2022
- Full Text
- View/download PDF
37. Adaptive Convergence of Methylomes Reveals Epigenetic Drivers and Boosters of Repeated Relapses in Patient-matched Childhood Ependymomas and Identifies Targets for Anti-Recurrence Therapies
- Author
-
Yuanda Hua, Deqiang Sun, Stewart Goldman, Lin Qi, Frank K. Braun, Sibo Zhao, Murali Chintagumpala, Jack Su, Adekunle M. Adesina, Paola Genevini, Xiao-Nan Li, Shirong Ding, Holly Lindsay, Sophie Xiao, Yun-Fei Xia, Yulun Huang, Jia Li, Jianfan Li, Yun Huang, Donald W. Parsons, Laszlo Perlaky, Miklos Miklos, Kogiso Mari, Huiyuan Zhang, Clifford Stephan, Peter F. Davies, Jianhua Yang, Anne-Clémence Veillard, Yuchen Du, Sol Schvartzman, Yongcheng Song, Tsz Kwong Man, Patricia Baxter, and Wan-Yee Teo
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,In patient ,Childhood ependymoma ,Convergence (relationship) ,Epigenetics ,business - Abstract
Ependymoma (EPN) is the third most common brain tumor in children and frequently recurs. Here, we report an integrated longitudinal analysis of epigenetic, genetic and tumorigenic changes in 30 patient-matched repeated relapses obtained from 10 pediatric patients to understand the mechanism of recurrences. Genome-wide DNA methylation analysis revealed stable molecular subtypes and convergent epigenetic reprogramming during serial relapses of the 5 RELA and 5 PFA EPNs that paralleled with elevated patient-derived orthotopic xenograft (PDOX) (13/27) formation in the late relapses. Differentially methylated CpGs (DMCs) preexisted in the primary tumors and persisted in the relapses (driver DMCs) were detected, ranging from 51 hypo-methylated in RELA to 148 hyper-methylated DMCs in PFA tumors; while newly acquired DMCs sustained in all the relapses but was absent in the primary tumors (booster DMCs) ranged from 38- 323 hyper-methylated DMCs in RELA and PFA EPNs, respectively. Integrated analysis of these DMC associated DNA methylation regions (DMRs) and RNAseq in both patient and PDOX tumors identified a small fraction of the differentially expressed genes (4.6±4.4% in RELA and 4.5±1.1% in PFA) as regulated by driver DMRs (e.g., up-regulated CACNA1H, SLC12A7, RARA in RELA and HSPB8, GMPR, ITGB4 in PFA) and booster DMRs (including the sole upregulated PLEKHG1 in RELA and NOTCH, EPHA2, SUFU, FOXJ1 in PFA tumors). Most these genes were novel to EPN relapses. Seven DMCs in RELA and 22 in PFA tumors were also identified as potential relapse predictors. Finally, integrating DNA methylation with histone modification identified LSD1 as a relapse driver gene. Combined treatment of a novel inhibitor SYC-836 with radiation significantly prolonged survival times in two PDOX models of recurrent PFA. This high-resolution epigenetic and genetic roadmap of EPN relapse and our 13 new PDOX models should significantly facilitate biological and preclinical studies of pediatric EPN recurrences.
- Published
- 2021
- Full Text
- View/download PDF
38. QOL-16. SYMON-SAYS (SYMPTOM MONITORING & SYSTEMATIC ASSESSMENT AND REPORTING SYSTEM IN YOUNG SURVIVORS) PROGRAM HAS THE POTENTIAL TO DECREASE SYMPTOM BURDEN ON CHILDREN WITH CANCER
- Author
-
Jin-Shei Lai, Devin Peipert, Sally Jensen, Megan Urban, Stewart Goldman, and Alicia Lenzen
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
AIMS The unrelieved symptoms and side effects of often-aggressive cancer treatments can lead to poor outcomes. SyMon-SAYS was developed to minimize symptom management barriers and symptom burdens by routinely collecting and interpreting patient-reported outcomes in pediatric oncology ambulatory settings. This paper reports the preliminary results of the 16-week SyMon-SAYS trial. METHODS Children (ages 8-17) with cancer (on-therapy or within 6-M post-treatment) were randomly assigned to either intervention (IG; weeks 1-16 intervention) or waitlist (WG; weeks 1-8 waitlist, weeks 9-16 intervention) group. Children in the intervention phase reported on 9 symptoms (fatigue, sadness, itch, pain, worry, appetite, nausea, sleep, headache) weekly via an electronic medical record patient portal. Scores exceeding a pre-defined threshold triggered an alert to the treatment team. Parents completed a symptom management barriers questionnaire (SMBQ) at baseline, weeks 8 (primary time-point and analyzed in this paper) and 16. Mixed-effects models were used to evaluate symptom burden over time. RESULTS Data from 75 children (37 IG, 38 WG) were analyzed (mean age=13.3 years, 58.8% male, 74.7% white). Of them, 43.9% had leukemia, and 17.5% brain tumor. On average, the IG completed 11 (possible min=0 max=16) and the WG completed 5 (possible min=0 max=8) symptom checklists; of them, 60% triggered symptom alerts. Results of the mixed-effects models showed significantly (p< 0.05) improvement in fatigue, sadness, worry, appetite and headache. No significant changes were found on others. For SMBQ, IG parents reported significantly in favor of “enough time with my child's doctors/nurses to talk about symptoms” than WG parents from baseline to week 8. No significant differences between IG and WG over time on other SMBQ items. CONCLUSIONS Our preliminary findings showed SyMon-SAYS alleviated emotional related symptoms over time. Physical symptoms might be related more to disease severity and treatment intensity, which we plan to investigate when more data is available.
- Published
- 2022
- Full Text
- View/download PDF
39. Review of the genomic landscape of common pediatric CNS tumors and how data sharing will continue to shape this landscape in the future
- Author
-
Kristiyana Kaneva and Stewart Goldman
- Subjects
Somatic cell ,medicine.medical_treatment ,Central nervous system ,Brain tumor ,Bioinformatics ,Molecular oncology ,Targeted therapy ,Genetics ,medicine ,Humans ,Cerebellar Neoplasms ,Child ,Molecular Biology ,Gene ,business.industry ,Point mutation ,General Medicine ,Genomics ,Glioma ,medicine.disease ,Data sharing ,medicine.anatomical_structure ,Ependymoma ,Mutation ,business ,Medulloblastoma - Abstract
Over the past decade we have witnessed a rapid increase in our understanding of the molecular characteristics of pediatric central nervous system (CNS) tumors. Studies that utilize genomic sequencing have revealed a heterogeneous group of genetic drivers in pediatric CNS tumors including point mutations, gene fusions, and copy number alterations. This manuscript provides an overview of somatic genomic alterations in the most common pediatric CNS tumors including low grade gliomas, high grade gliomas, medulloblastomas, and ependymomas. Additionally, we will discuss the need and opportunity for genomic and clinical data sharing through the children's brain tumor network and other international initiatives.
- Published
- 2021
40. Phase I study of gene-mediated cytotoxic immunotherapy with AdV-tk as adjuvant to surgery and radiation for pediatric malignant glioma and recurrent ependymoma
- Author
-
Liliana Goumnerova, Stewart Goldman, Rishi Lulla, Estuardo Aguilar-Cordova, Susan N. Chi, Peter E. Manley, Andrea G. Manzanera, Mark W. Kieran, Tadanori Tomita, Karen J. Marcus, Laura K. Aguilar, and Arthur J DiPatri
- Subjects
0301 basic medicine ,Ependymoma ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,Monitoring, Immunologic ,Glioma ,medicine ,Humans ,Child ,Chemotherapy ,Temozolomide ,Brain Neoplasms ,business.industry ,Common Terminology Criteria for Adverse Events ,Genetic Therapy ,medicine.disease ,Chemotherapy regimen ,Surgery ,Radiation therapy ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Immunotherapy ,Neurology (clinical) ,business ,Pediatric Neuro-Oncology ,medicine.drug ,Anaplastic astrocytoma - Abstract
Background Gene-mediated cytotoxic immunotherapy (GMCI) is a tumor-specific immune stimulatory strategy implemented through local delivery of aglatimagene besadenovec (AdV-tk) followed by anti-herpetic prodrug. GMCI induces T-cell dependent tumor immunity and synergizes with radiotherapy. Clinical trials in adult malignant gliomas demonstrated safety and potential efficacy. This is the first trial of GMCI in pediatric brain tumors. Methods This phase I dose escalation study was conducted to evaluate GMCI in patients 3 years of age or older with malignant glioma or recurrent ependymoma. AdV-tk at doses of 1 × 1011 and 3 × 1011 vector particles (vp) was injected into the tumor bed at the time of surgery followed by 14 days of valacyclovir. Radiation started within 8 days of surgery, and if indicated, chemotherapy began after completion of valacyclovir. Results Eight patients (6 glioblastoma, 1 anaplastic astrocytoma, 1 recurrent ependymoma) were enrolled and completed therapy: 3 on dose level 1 and 5 on dose level 2. Median age was 12.5 years (range 7-17) and Lansky/Karnofsky performance scores were 60-100. Five patients had multifocal/extensive tumors that could not be resected completely and 3 had gross total resection. There were no dose-limiting toxicities. The most common possibly GMCI-related adverse events included Common Terminology Criteria for Adverse Events grade 1-2 fever, fatigue, and nausea/vomiting. Three patients, in dose level 2, lived more than 24 months, with 2 alive without progression 37.3 and 47.7 months after AdV-tk injection. Conclusions GMCI can be safely combined with radiation therapy with or without temozolomide in pediatric patients with brain tumors and the present results strongly support further investigation. Clinical trial registry ClinicalTrials.gov NCT00634231.
- Published
- 2019
- Full Text
- View/download PDF
41. QOL-09. SyMon-SAYS: A symptom monitoring and reporting program for children with cancer
- Author
-
Jin-Shei Lai, Sally Jensen, Megan Urban, Stewart Goldman, and Alicia Lenzen
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Unrelieved symptom burden due to cancer treatments can lead to poor psychosocial functioning and decreased health-related quality of life (HRQOL) for patients and their families. Barriers at the patient, healthcare provider and system levels can contribute to poor symptom management. Funded by the US National Cancer Institute, we have developed the Symptom Monitoring & Systematic Assessment and Reporting System in Young Survivors (SyMon-SAYS) program. SyMon-SAYS is a technology-based program with the potential to minimize symptom management barriers by routinely collecting and interpreting patient-reported outcomes in pediatric oncology ambulatory settings in a manner that is efficient, actionable by clinicians, supports engagement of patients and families with their health and care, and improves clinical processes and outcomes. This is a single institution modified waitlist control 16-week randomized trial of 200 children (ages 8-17) with cancer and their parents/guardians. Participants in the intervention phase will complete a symptom checklist weekly via the electronic health record patient portal. Scores exceeding a pre-defined threshold will trigger an alert to the treatment team, which will review the report and take appropriate actions. Participants will complete a separate battery of questionnaires assessing HRQOL at baseline and weeks 8 and 16. The recruitment is in progress. As of today, we have recruited 57 patients/parents. 29 completed 16-week study (15 intervention & 14 wait-list). Preliminary results showed SyMon-SAYS system was easy (92%) and convenient (85%) to use. Parents were satisfied (74.1%) with the SyMon-SAYS program. Comparing to the waitlist control, intervention group parents reported significantly less concerns on not having enough time to discuss their child’s symptoms with treating clinicians (p=0.0022), and disagreed that it is not necessary to treat their child’s symptoms as they will go away (p=0.04). We anticipate completing the recruitment by the end of 2023.
- Published
- 2022
- Full Text
- View/download PDF
42. LGG-06. Selumetinib in pediatric patients with non-neurofibromatosis type 1-associated, non-optic pathway (OPG) and non-pilocytic recurrent/progressive low-grade glioma harboring BRAFV600E mutation or BRAF-KIAA1549 fusion: a multicenter prospective Pediatric Brain Tumor Consortium (PBTC) Phase 2 trial
- Author
-
Jason Fangusaro, Arzu Onar-Thomas, Tina Young Poussaint, Shelly Lensing, Shengjie Wu, Azra H Ligon, Neal Lindeman, Clinton F Stewart, David T W Jones, Stefan M Pfister, Natasha Pillay Smiley, James Leach, Roger Packer, Gilbert Vezina, Alicia Lenzen, Alok Jaju, Stewart Goldman, Laurence Austin Doyle, Malcolm Smith, Maryam Fouladi, and Ira Dunkel
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: A greater understanding of the Ras-MAP kinase pathway in pediatric low-grade glioma (LGG) paired with the availability of selective inhibitors has enhanced the ability to target this pathway with therapeutic intent. METHODS: The PBTC conducted a multi-institutional phase II study (NCT01089101) evaluating selumetinib (AZD6244, ARRY-142886), a MEK I/II inhibitor, in children with recurrent/progressive LGG assigned to 6 strata and treated at a dose of 25 mg/m2/dose PO BID for up to two years. Here we present stratum 5 which enrolled children without NF1, non-OPG and non-pilocytic LGG harboring either a BRAFV600E mutation or BRAF-KIAA1549 fusion. RESULTS: Twenty-four of 25 children enrolled were eligible; 23 were evaluable for the primary radiologic response endpoint. Enrollment stopped early due to slow accrual and initiation of COG ACNS1931. The most common histologies were ganglioglioma (42%) and astrocytoma NOS (33%). Thirteen tumors (54%) had BRAF-KIAA1549 fusion; 11 (46%) had the BRAFV600E mutation. Five of 23 (22%) evaluable patients achieved a centrally confirmed partial response (PR), 12 (52%) had stable disease and 6 (26%) had progression with a 2-year progression-free survival of 75 + 9%. Four of 11 (36%) patients with a BRAFV600E mutation and 1/12 (8%) with a BRAF-KIAA1549 fusion achieved a PR. The 2-year PFS did not significantly differ between tumors with BRAFV600E mutation (82 + 12%) versus BRAF-KIAA1549 fusion (68 + 13%) (n=24, p=0.548). No patient remains on therapy. The most common attributable toxicities were grade 1/2 ALT/AST elevation, dry skin and leukopenia. Rare grade 3/4 toxicities included elevated CPK, rash, paronychia, fever, weight gain and sinus tachycardia. CONCLUSIONS: Despite lower than planned accrual, selumetinib met the design threshold for success in treating children with recurrent/progressive non-pilocytic, non-OPG LGG without NF1 that harbored the common BRAF aberrations. Ongoing phase 3 prospective studies will better determine the role of this agent in this population.
- Published
- 2022
- Full Text
- View/download PDF
43. GCT-04. Pattern of Treatment Failures in Central Nervous System Non-Germinomatous Germ Cell Tumors (CNS-NGGCT): A Pooled Analysis of Clinical Trials
- Author
-
Adriana Fonseca, Cecile Faure-Conter, Matthew Murray, Jason Fangusaro, Shivani Bailey, Stewart Goldman, Soumen Khatua, Didier Frapaz, Gabriele Calaminus, Girish Dhall, James Nicholson, Eric Bouffet, and Ute Bartels
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: Central Nervous System- Non-Germinomatous Germ Cell Tumors (CNS-NGGCT) are rare but curable tumors. Due to their rarity, treatment failures remain a poorly characterized disease with unfavorable outcomes. In this study, we sought to characterize the treatment failures in a large cohort of prospectively treated patients. METHODS: European and North American clinical trials for patients with CNS-NGGCT (SIOP-GCT96, SFOP-TGM TC 90/92, COG-ACNS0122 and COG-ACNS1123) were pooled for analysis. Additionally, patients included and treated in the UK and France national registries under strict protocol-guidelines were included as an independent, non-overlapping cohort. RESULTS: A total of 118 patients experienced a treatment failure. Twenty-four patients had progressive disease during therapy and additional eleven patients were diagnosed with growing teratoma syndrome (GTS). Patients with GTS are significantly younger and present with local failures and negative tumor markers. Eighty-three individuals experienced disease relapses after treatment ended. Patients’ metastatic relapses presented significantly earlier than local relapses and were associated with tumor marker elevation (OR: 4.39; p=0.026). In our analysis, focal or whole ventricular (WVI) radiation therapy was not associated with an increased risk of metastatic relapses. CONCLUSIONS: Herein, we present the largest pooled dataset of prospectively treated patients with relapsed CNS-NGGCT. Our study identified younger age and negative tumor markers to be characteristic of GTS. Additionally, we elucidated that metastatic relapses occur earlier than local relapses, are associated with elevated tumor markers, and are not associated with the field of radiation therapy. These findings are of utmost importance for the planning of future clinical trials and the implementation of surveillance strategies in these patients.
- Published
- 2022
- Full Text
- View/download PDF
44. RARE-13. Clinical management and functional and survival outcomes in pediatric craniopharyngioma, a patient and family perspective
- Author
-
Emily Marshall, Sylvia Cheng, Julia Crowley, Shana McCormack, Brian Rood, Todd Hankinson, Michael DeCuypere, Sandy Lam, Stewart Goldman, Svenja Boekhoff, Hermann L Muller, Ryan Velasco, Phillip B Storm, Adam Resnick, Michael Prados, Sabine Mueller, Cassie Kline, and Fatema Malbari
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Craniopharyngiomas are rare, histologically benign, sellar/parasellar tumors with significant tumor and therapy related morbidity and impairment in quality of life (QOL). We report survey results from patients/families affected by childhood-onset craniopharyngioma to identify opportunities for improvement in management. An anonymous REDCap survey was distributed via social media and clinic visits to patients/families of craniopharyngioma survivors. Survey questions investigated perspectives on clinical management and functional and survival outcomes at initial diagnosis and recurrence. A total of 159 patients/families completed the survey, 40% (n=64) reported craniopharyngioma recurrence. For primary craniopharyngioma, maximal safe resection was the most frequent treatment reported (n=84), followed by partial resection (n=40), radiation (n=8), biopsy (n=5), and chemotherapy (n=3). Most patients (n=120) decided on a treatment plan within one week, 63 (40%) decided in one day. For recurrent craniopharyngioma, maximal safe resection and radiation were the most frequent interventions (n=33 each), followed by partial resection (n=13), chemotherapy (n=4) and biopsy (n=2). Multiple treatment options and/or participation in a clinical trial were offered to similar numbers of patients across primary and recurrent diagnoses (~21% for each). Most recurrent craniopharyngioma patients decided on management within one week (n=43). Long term effects related to tumor and treatment were identified as the primary concern in all respondents. The most common deficits for all patients were neuro-endocrine followed by vision and neurocognition problems. Neuro-endocrine complications were self-reported as the biggest impact on QOL. Families reported that they would prefer treatment options with the potential for improved QOL, even if these options also carried an increased risk of recurrence. Craniopharyngioma continues to be predominantly treated with surgery and radiation initially and with recurrence. Survivors have multiple comorbidities, with an interest in targeted therapies that preserve QOL. Novel therapies to prevent co-morbidities and provide long term benefits are necessary and upcoming.
- Published
- 2022
- Full Text
- View/download PDF
45. OTHR-08. Pediatric Neurologic Assessment in Neuro-oncology (pNANO) Scale: A tool to assess neurologic function for Response Assessment in Neuro-oncology (RAPNO)
- Author
-
Fatema Malbari, Craig Erker, Pablo Hernáiz Driever, Natasha Pillay-Smiley, Robert A Avery, Robert Craig Castellino, Antoinette A Y N Schouten-van Meeteren, Alicia C Lenzen, Rick Brandsma, Kim Kramer, Patricia A Baxter, Girish Dhall, Stewart Goldman, Patrick Y Wen, Michael Prados, Roger J Packer, Katherine E Warren, and Lakshmi Nayak
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Background: The Neurologic Assessment in Neuro-Oncology (NANO) scale, a standardized metric to objectively measure neurologic function in adult brain tumor patients, complements radiographic assessment in evaluating outcomes of neuro-oncology patients in clinical trials and clinical practice. Currently, there is no standardized measure for neurologic function in pediatric neuro-oncology patients despite their distinct clinical presentations and tumor locations. Therefore, we developed a dedicated pediatric NANO (pNANO) scale. Methods: An international group of pediatric neurologists and adult and pediatric neuro-oncologists convened bi-weekly over 5 months to draft the pNANO scale as an objective and quantifiable measure of neurologic function in children that can be administered during routine examination by pediatric-trained providers of any sub-specialty and be utilized together with other indicators to assess response in clinical trials. Results: Ten relevant domains of neurologic function were identified based on common pediatric brain tumor locations: gait, strength, cerebellar function, visual fields, visual acuity, facial strength, level of consciousness, extraocular movements, dysarthria and dysphagia. For each domain, developmental age appropriate levels of function were defined and categorized. Each domain, based on direct observation and testing during any routine neurological exam, can be used for longitudinal monitoring. Conclusions: The pNANO scale has been developed and aims to provide an objective metric of neurologic function for pediatric brain tumor patients. This scale will be tested for reliability, feasibility and inter-observer variability. Consistent evaluation of neurologic function using pNANO along with radiographic assessment will enable more comprehensive and standardized response assessment in pediatric neuro-oncology patients enrolled in clinical trials.
- Published
- 2022
- Full Text
- View/download PDF
46. A phase I trial of the CDK 4/6 inhibitor palbociclib in pediatric patients with progressive brain tumors: A Pediatric Brain Tumor Consortium study (PBTC-042)
- Author
-
Stewart Goldman, Tong Lin, Oren J. Becher, Sridharan Gururangan, David Van Mater, Patricia Baxter, Joanna J. Phillips, Ira J. Dunkel, Tina Young Poussaint, Giles W. Robinson, Girish Dhall, Clinton F. Stewart, Mariko DeWire-Schottmiller, Sarah Leary, Jie Huang, Olivia Campagne, Maryam Fouladi, Arzu Onar-Thomas, and Eugene Hwang
- Subjects
Oncology ,Male ,Pediatric Brain Tumor Consortium ,Pyridines ,PBTC-042 ,Piperazines ,0302 clinical medicine ,Child ,Cancer ,Pediatric ,Leukopenia ,Brain Neoplasms ,Hematology ,030220 oncology & carcinogenesis ,Child, Preschool ,6.1 Pharmaceuticals ,Toxicity ,Disease Progression ,Female ,medicine.symptom ,pharmacokinetics ,brain tumor ,PBTC‐ ,Adult ,medicine.medical_specialty ,Pediatric Research Initiative ,Adolescent ,palbociclib ,Pediatric Cancer ,Clinical Trials and Supportive Activities ,Clinical Sciences ,Oncology and Carcinogenesis ,Brain tumor ,Antineoplastic Agents ,Palbociclib ,Neutropenia ,Article ,Paediatrics and Reproductive Medicine ,03 medical and health sciences ,Young Adult ,Rare Diseases ,Pharmacokinetics ,Clinical Research ,Internal medicine ,medicine ,pharmacodynamics ,Humans ,Oncology & Carcinogenesis ,Preschool ,Protein Kinase Inhibitors ,business.industry ,Neurosciences ,Cyclin-Dependent Kinase 4 ,Evaluation of treatments and therapeutic interventions ,Cyclin-Dependent Kinase 6 ,medicine.disease ,Orphan Drug ,Pharmacodynamics ,Pediatrics, Perinatology and Child Health ,business ,030215 immunology - Abstract
BackgroundDisruption of cell-cycle regulators is a potential therapeutic target for brain tumors in children and adolescents. The aim of this study was to determine the maximum tolerated dose (MTD) and describe toxicities related to palbociclib, a selective cyclin-dependent kinase 4/6 (CDK4/6) inhibitor in pediatric patients with progressive/refractory brain tumors with intact retinoblastoma protein.MethodsPalbociclib was administered orally starting at 50mg/m2 daily for the first 21 days of a 28-day course. Dose escalation was according to the Rolling-6 statistical design in less heavily (stratum I) and heavily pretreated (stratum II) patients, and MTD was determined separately for each group. Pharmacokinetic studies were performed during the first course, and pharmacodynamic studies were conducted to evaluate relationships between drug levels and toxicities.ResultsA total of 21 patients were enrolled on stratum I and 14 patients on stratum II. The MTD for both strata was 75mg/m2 . Palbociclib absorption (mean Tmax between 4.9 and 6.6 h) and elimination (mean half-life between 11.3 and 19.5 h) were assessed. The most common toxicity was myelosuppression. Higher palbociclib exposure was associated with grade 3/4 neutropenia and leukopenia. Dose limiting toxicities included grade 4 neutropenia and grade 3 thrombocytopenia and dehydration. No patients had an objective response to palbociclib therapy.ConclusionsPalbociclib was safely administered to children and adolescents at a dosage of 75mg/m2 for 21 consecutive days followed by seven days of rest in both strata. Future studies will establish its optimal utilization in pediatric patients with brain tumors.
- Published
- 2021
47. A phase II trial of selumetinib in children with recurrent optic pathway and hypothalamic low-grade glioma without NF1: a Pediatric Brain Tumor Consortium study
- Author
-
Ibrahim Qaddoumi, Laurence Austin Doyle, Ira J. Dunkel, Jeremy Jones, Azra H. Ligon, Shengjie Wu, Sridharan Gururangan, Maryam Fouladi, Patricia Baxter, Zoltan Patay, Stewart Goldman, Lindsay Kilburn, Malcolm A. Smith, Olivia Campagne, Neal I. Lindeman, Corey Bregman, Clinton F. Stewart, Anu Banerjee, Jason Fangusaro, Arzu Onar-Thomas, Tina Young Poussaint, Gilbert Vezina, and Michael Fisher
- Subjects
Optic Nerve Glioma ,Cancer Research ,medicine.medical_specialty ,Pediatric Brain Tumor Consortium ,Visual acuity ,Neurofibromatosis 1 ,Nausea ,Phases of clinical research ,Gastroenterology ,Glioma ,Internal medicine ,medicine ,Humans ,Child ,business.industry ,Brain Neoplasms ,Editorials ,medicine.disease ,Rash ,Oncology ,Selumetinib ,Benzimidazoles ,Neurology (clinical) ,medicine.symptom ,business ,Pediatric Neuro-Oncology ,Progressive disease - Abstract
Background Pediatric low-grade gliomas (pLGGs) are the most common childhood brain tumor. Progression-free survival (PFS) is much lower than overall survival, emphasizing the need for alternative treatments. Sporadic (without neurofibromatosis type 1) optic pathway and hypothalamic gliomas (OPHGs) are often multiply recurrent and cause significant visual deficits. Recently, there has been a prioritization of functional outcomes. Methods We present results from children with recurrent/progressive OPHGs treated on a PBTC (Pediatric Brain Tumor Consortium) phase II trial evaluating efficacy of selumetinib (AZD6244, ARRY-142886) a MEK-1/2 inhibitor. Stratum 4 of PBTC-029 included patients with sporadic recurrent/progressive OPHGs treated with selumetinib at the recommended phase II dose (25mg/m2/dose BID) for a maximum of 26 courses. Results Twenty-five eligible and evaluable patients were enrolled with a median of 4 (1-11) previous therapies. Six of 25 (24%) had partial response, 14/25 (56%) had stable disease, and 5 (20%) had progressive disease while on treatment. The median treatment courses were 26 (2-26); 14/25 patients completed all 26 courses. Two-year PFS was 78 ± 8.5%. Nineteen of 25 patients were evaluable for visual acuity which improved in 4/19 patients (21%), was stable in 13/19 (68%), and worsened in 2/19 (11%). Five of 19 patients (26%) had improved visual fields and 14/19 (74%) were stable. The most common toxicities were grade 1/2 CPK elevation, anemia, diarrhea, headache, nausea/emesis, fatigue, AST and ALT increase, hypoalbuminemia, and rash. Conclusions Selumetinib was tolerable and led to responses and prolonged disease stability in children with recurrent/progressive OPHGs based upon radiographic response, PFS, and visual outcomes.
- Published
- 2021
48. NF106: A Neurofibromatosis Clinical Trials Consortium Phase II Trial of the MEK Inhibitor Mirdametinib (PD-0325901) in Adolescents and Adults With NF1-Related Plexiform Neurofibromas
- Author
-
Chie Emoto, Nathan Robison, Brigitte C. Widemann, Brian Weiss, Stewart Goldman, Jeffrey C. Allen, James H. Tonsgard, Alexander A. Vinks, Nancy Ratner, Michael Fisher, Jaishri O. Blakeley, Bruce R. Korf, Pamela L. Wolters, Coretta Thomas Robinson, Lloyd J. Edwards, Eva Dombi, Elizabeth K. Schorry, Scott R. Plotkin, Tsuyoshi Fukuda, Gary Cutter, and Roger J. Packer
- Subjects
MAPK/ERK pathway ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Neurofibromatosis 1 ,Time Factors ,Adolescent ,Extracellular signal-regulated kinases ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Plexiform neurofibroma ,Internal medicine ,medicine ,Humans ,Neurofibromatosis ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase Kinases ,Neurofibroma, Plexiform ,business.industry ,MEK inhibitor ,Diphenylamine ,medicine.disease ,Magnetic Resonance Imaging ,United States ,Clinical trial ,Treatment Outcome ,030220 oncology & carcinogenesis ,Benzamides ,Female ,business ,030217 neurology & neurosurgery - Abstract
PURPOSE Patients with neurofibromatosis type 1 (NF1) frequently develop plexiform neurofibromas (PNs), which can cause significant morbidity. We performed a phase II trial of the MAPK/ERK kinase inhibitor, mirdametinib (PD-0325901), in patients with NF1 and inoperable PNs. The primary objective was response rate based on volumetric magnetic resonance imaging analysis. METHODS Inclusion criteria included age ≥ 16 years and a PN that was either progressive or causing significant morbidity. First-dose pharmacokinetics were performed. Patients completed patient-reported outcome measures. Patients received mirdametinib by mouth twice a day at 2 mg/m2/dose (maximum dose = 4 mg twice a day) in a 3-week on/1-week off sequence. Each course was 4 weeks in duration. Evaluations were performed after four courses for the first year and then after every six courses. Patients could receive a maximum of 24 total courses. RESULTS Nineteen patients were enrolled, and all 19 received mirdametinib. The median age was 24 years (range, 16-39 years); the median baseline tumor volume was 363.8 mL (range, 3.9-5,161 mL). Eight of the 19 patients (42%) achieved a partial response of the target PN by course 12, and 10 (53%) had stable disease. One patient (5%) developed progressive disease at course 8. Significant and durable decreases were observed in pain ratings. CONCLUSION To our knowledge, this analysis represents the first characterization of the activity and pharmacokinetics of mirdametinib in patients with NF1 and PNs and is the first published response study for MAPK/ERK kinase inhibitors in adults with NF1 and PNs. Mirdametinib given at 2 mg/m2/dose (maximum dose, 4 mg) twice daily in a 3-week on/1-week off sequence resulted in a 42% partial response rate with preliminary evidence of reduction in pain.
- Published
- 2021
49. Visual outcomes following everolimus targeted therapy for neurofibromatosis type 1‐associated optic pathway gliomas in children
- Author
-
Nicole J. Ullrich, Stewart Goldman, Jeffrey C. Allen, Mark W. Kieran, Nathan Robison, Sanjay P. Prabhu, David H. Gutmann, Michael Fisher, Bruce R. Korf, David Viskochil, Roger J. Packer, and John P. Perentesis
- Subjects
Adult ,Male ,Optic Nerve Glioma ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Neurofibromatosis 1 ,Visual acuity ,Adolescent ,genetic structures ,Optic glioma ,medicine.medical_treatment ,Visual Acuity ,Antineoplastic Agents ,Targeted therapy ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Ophthalmology ,medicine ,Humans ,Everolimus ,Neurofibromatosis ,Child ,Prospective cohort study ,Retrospective Studies ,business.industry ,Infant ,Hematology ,medicine.disease ,eye diseases ,nervous system diseases ,Treatment Outcome ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Female ,medicine.symptom ,business ,After treatment ,030215 immunology ,medicine.drug - Abstract
Data for visual acuity (VA) after treatment of neurofibromatosis type 1-associated optic pathway gliomas (NF1-OPGs) are limited. We retrospectively collected VA, converted to logMAR, before and after targeted therapy with everolimus for NF1-OPG, and compared to radiologic outcomes (14/18 with NF1-OPG, 25 eyes [three without quantifiable vision]). Upon completion of treatment, VA was stable in 19 eyes, improved in four eyes, and worsened in two eyes; visual and radiologic outcomes were discordant. In summary, the majority of children with NF1-OPG exhibited stabilization of their VA after everolimus treatment. A larger, prospective study will help delineate visual outcomes after targeted therapy.
- Published
- 2020
- Full Text
- View/download PDF
50. EPID-11. A MULTI-INSTITUTIONAL COMPARATIVE ANALYSIS OF THE CLINICAL, GENOMIC, AND SURVIVAL CHARACTERISTICS OF PEDIATRIC, YOUNG ADULT AND OLDER ADULT PATIENTS WITH IDH-MUTANT GLIOMA
- Author
-
Sanda Alexandrescu, Katherine E. Warren, Wenya Linda Bi, Stewart Goldman, Patrick Y. Wen, Mehdi Touat, Keith L. Ligon, Emily Burton, Mary Jane Lim-Fat, Jennifer A. Cotter, Jayne Vogelzang, Ashley Margol, Kristiyana Kaneva, Karen Wright, Clement Ma, Kristina A. Cole, Amy Smith, Kee Kiat Yeo, Kellie Nazemi, Emily Owens, Marilyn M. Li, Hasan Al-Sayegh, Alana McGovern, David A. Reardon, and Eleanor Woodward
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Adult patients ,business.industry ,Mutant ,O-6-methylguanine-DNA methyltransferase ,medicine.disease ,Copy Number Polymorphism ,Internal medicine ,Glioma ,medicine ,Neurology (clinical) ,Progression-free survival ,Epidemiology & Biostatistics ,Young adult ,business - Abstract
BACKGROUND Prognostic significance of IDH-mutation in glioma is incompletely understood in children and adolescents/young adults (YAs). We compared the clinico-genomic features, outcomes and prognostic factors observed in IDH-mutant gliomas across age groups. METHODS Clinical, histologic and molecular data of patients with IDH-mutant gliomas from 8 pediatric institutions (spanning twenty years) and adult patients from two institutions (from 2013–2019) were identified. Patients were grouped as pediatric (< 19y), YA (19y to < 40y) or older adult (≥ 40y). Genomic alterations, including somatic mutations and copy number variants, were captured with institutional next generation sequencing. Factors were compared across age categories using Fisher’s exact test or analysis-of-variance. Cox proportional-hazards regression tested factors for association with overall (OS) and progression-free survival (PFS). RESULTS Of 379 patients, 48(13%) were pediatric, 204(54%) YA and 127(33%) older adult. Histological subtype differed significantly by age group (p< 0.0001). YAs had higher rates of malignant transformation (p=0.01) and shorter time-from-diagnosis-to-malignant transformation (p=0.01) compared to other age groups. Analysis of genomic alterations revealed an age-related difference in distribution in ATRX mutations only (p=0.0018). Median PFS and OS for the entire cohort were 4.62 and 17.19 years. In univariate models, PFS differed by age group (p=0.0012), with YAs having the worst outcomes. Lack of MGMT methylation (p=0.024) predicted poorer OS. Upfront observant management was predictive of poorer PFS. Gene mutations were not associated with PFS. In multi-variable models, YAs had shorter PFS compared to pediatric (hazard ratio [HR]=2.03, p=0.01) and older adults (HR=1.59, p=0.003) after adjusting for histology, extent of resection, and initial therapy. Age at diagnosis was not associated with OS in multi-variable analysis. CONCLUSIONS Within our cohort, YA with IDH-mutant tumors progressed more quickly compared to their pediatric counterparts. Further study of YA patients with IDH-mutant glioma is critical to better define best practices for this group.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.