350 results on '"Tracy T. Batchelor"'
Search Results
2. Diagnostic, therapeutic, and prognostic implications of the 2021 World Health Organization classification of tumors of the central nervous system
- Author
-
Tracy T. Batchelor, L. Nicolas Gonzalez Castro, and Simon Gritsch
- Subjects
Adult ,Central Nervous System ,Cancer Research ,Central nervous system ,Disease ,World Health Organization ,Bioinformatics ,World health ,Central Nervous System Neoplasms ,Glioma ,Humans ,Medicine ,CNS TUMORS ,Child ,Grading (tumors) ,Brain Neoplasms ,business.industry ,Prognosis ,medicine.disease ,Isocitrate Dehydrogenase ,Clinical trial ,medicine.anatomical_structure ,Oncology ,Mutation ,business ,Who classification - Abstract
The 2016 revised fourth edition of the World Health Organization (WHO) classification of central nervous system (CNS) tumors incorporated molecular features with histologic grading, revolutionizing how oncologists conceptualize primary brain and spinal cord tumors as well as providing new insights into their management and prognosis. The 2021 revised fifth edition of the WHO classification further integrates molecular alterations for CNS tumor categorization, updating current understanding of the pathophysiology of many of these disease entities. Here, the authors review changes in the new classification for the most common primary adult tumors-gliomas (including astrocytomas, oligodendrogliomas, and ependymomas) and meningiomas-highlighting the key genomic alterations for each group classification to help clinicians interpret them as they consider therapeutic options-including clinical trials and targeted therapies-and discuss the prognosis of these tumors with their patients. The revised, updated 2021 WHO classification also further integrates molecular alterations in the classification of pediatric CNS tumors, but those are not covered in the current review.
- Published
- 2021
3. Body CT and PET/CT detection of extracranial lymphoma in patients with newly diagnosed central nervous system lymphoma
- Author
-
Sung Tae Kim, Minjung Seong, Tracy T. Batchelor, Ji Eun Park, Choong Gon Choi, Sung Soo Ahn, Lakshmi Nayak, Seung Koo Lee, Chong Hyun Suh, Kichang Han, Sang Min Lee, Seung Chai Jung, Jeffrey P. Guenette, Raymond Y. Huang, Seung Hong Choi, Ho Sung Kim, Sang Joon Kim, and Jeong Hoon Kim
- Subjects
Cancer Research ,medicine.medical_specialty ,PET-CT ,medicine.diagnostic_test ,business.industry ,Brain biopsy ,Central nervous system ,Disease ,medicine.disease ,Lymphoma ,medicine.anatomical_structure ,Oncology ,medicine ,In patient ,Neurology (clinical) ,Radiology ,business ,Pathological ,Cohort study - Abstract
Background We aimed to investigate the detection rate of body CT or PET/CT for sites of extracranial disease in patients with a new pathological diagnosis of CNS DLBCL and to identify factors associated with sites of extracranial disease. Methods An international multicenter cohort study of consecutive immunocompetent patients with a new diagnosis of CNS DLBCL confirmed by brain biopsy who underwent CT and/or PET/CT to evaluate for sites of extracranial disease between 1998 and 2019. The primary outcome was the detection rate of extracranial lymphoma by CT or PET/CT. Subgroup analyses according to age and EBV status were also performed. Logistic regression analyses were performed to determine factors related to sites of extracranial disease. Detection rates of CT and PET/CT were compared. Results One thousand and forty-three patients were included. The overall detection rate of CT or PET/CT was 2.6% (27/1043). The treatment approach was adjusted in 74% of these patients. Multivariable analysis demonstrated that age >61 years (OR, 3.10; P = .016) and EBV positivity (OR, 3.78; P = .045) were associated with greater odds of extracranial lymphoma. There was no statistically significant difference in detection rate between CT and PET/CT (P = .802). In patients ≤61 years old, the false-referral rates were significantly higher than the detection rates (P < .001). Conclusion Our results showed increased odds of extracranial lymphoma in patients with older age or EBV-positive lymphoma. Treatment was adjusted in a majority of patients diagnosed with extracranial lymphoma, thereby supporting the current guidelines for the use of contrast-enhanced body CT or PET/CT in patients with newly diagnosed CNS DLBCL.
- Published
- 2021
4. Palbociclib demonstrates intracranial activity in progressive brain metastases harboring cyclin-dependent kinase pathway alterations
- Author
-
Anita Giobbie-Hurder, Tracy T. Batchelor, Maura Mahar, Michael White, Daniel P. Cahill, Rebecca S. Heist, Justine V. Cohen, Justin F. Gainor, Helen A. Shih, Priscilla K. Brastianos, Scott L. Carter, Ryan J. Sullivan, Deborah Forst, Donald P. Lawrence, Nancy Wang, David P. Ryan, Kevin S. Oh, Sandro Santagata, A. John Iafrate, Albert E. Kim, Jennifer A. Ligibel, Ugonma Chukwueke, Elizabeth R. Gerstner, and Eudocia Q. Lee
- Subjects
Cancer Research ,biology ,Kinase ,business.industry ,medicine.medical_treatment ,Palbociclib ,Interim analysis ,Targeted therapy ,Oncology ,Cyclin-dependent kinase ,Cancer research ,medicine ,Clinical endpoint ,biology.protein ,Metastatic brain cancer ,business ,CDK inhibitor - Abstract
Recent studies suggest that the cyclin-dependent kinase (CDK) pathway may be a therapeutic target for brain metastases (BM). Here, we present interim analysis of a basket trial evaluating the intracranial efficacy of the CDK inhibitor palbociclib in patients with progressive BM and CDK alterations. Our study met its primary endpoint and provides evidence for performing molecular testing of archival BM tissue, if available, to inform the choice of CNS-penetrant targeted therapy. Brastianos and colleagues report interim trial results on the intracranial clinical benefit of palbociclib for patients with progressive metastatic brain cancer carrying cyclin-dependent kinase pathway alterations.
- Published
- 2021
5. Consensus recommendations for MRI and PET imaging of primary central nervous system lymphoma: guideline statement from the International Primary CNS Lymphoma Collaborative Group (IPCG)
- Author
-
Maciej M. Mrugala, Edward A. Neuwelt, Heiko Schöder, Benjamin M. Ellingson, C. Chad Quarles, Gerald Illerhaus, Tracy T. Batchelor, Timothy J. Kaufmann, Andrés J.M. Ferreri, James L. Rubenstein, Christopher P. Fox, Nicoletta Anzalone, Christian Grommes, Leland S. Hu, Letterio S. Politi, Ovidiu C. Andronesi, Dorothee P. Auer, Jerrold L. Boxerman, Motoo Nagane, Ramon F. Barajas, Prakash Ambady, Barajas, R. F., Politi, L. S., Anzalone, N., Schoder, H., Fox, C. P., Boxerman, J. L., Kaufmann, T. J., Quarles, C. C., Ellingson, B. M., Auer, D., Andronesi, O. C., Ferreri, A. J. M., Mrugala, M. M., Grommes, C., Neuwelt, E. A., Ambady, P., Rubenstein, J. L., Illerhaus, G., Nagane, M., Batchelor, T. T., and Hu, L. S.
- Subjects
Central Nervous System ,Cancer Research ,Lymphoma ,Central Nervous System Neoplasms ,0302 clinical medicine ,hemic and lymphatic diseases ,Cancer ,medicine.diagnostic_test ,Primary central nervous system lymphoma ,imaging ,Hematology ,Guideline Statement ,Magnetic Resonance Imaging ,Oncology ,Positron emission tomography ,030220 oncology & carcinogenesis ,Biomedical Imaging ,Patient Safety ,4.2 Evaluation of markers and technologies ,MRI ,medicine.medical_specialty ,Consensus ,Oncology and Carcinogenesis ,Brain tumor ,Context (language use) ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,medicine ,Medical imaging ,AcademicSubjects/MED00300 ,Humans ,Medical physics ,Oncology & Carcinogenesis ,PCNSL ,primary central nervous system lymphoma ,business.industry ,Neurosciences ,Reproducibility of Results ,Magnetic resonance imaging ,Guideline ,medicine.disease ,Clinical trial ,PET ,Positron-Emission Tomography ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Advanced molecular and pathophysiologic characterization of primary central nervous system lymphoma (PCNSL) has revealed insights into promising targeted therapeutic approaches. Medical imaging plays a fundamental role in PCNSL diagnosis, staging, and response assessment. Institutional imaging variation and inconsistent clinical trial reporting diminishes the reliability and reproducibility of clinical response assessment. In this context, we aimed to: (1) critically review the use of advanced positron emission tomography (PET) and magnetic resonance imaging (MRI) in the setting of PCNSL; (2) provide results from an international survey of clinical sites describing the current practices for routine and advanced imaging, and (3) provide biologically based recommendations from the International PCNSL Collaborative Group (IPCG) on adaptation of standardized imaging practices. The IPCG provides PET and MRI consensus recommendations built upon previous recommendations for standardized brain tumor imaging protocols (BTIP) in primary and metastatic disease. A biologically integrated approach is provided to addresses the unique challenges associated with the imaging assessment of PCNSL. Detailed imaging parameters facilitate the adoption of these recommendations by researchers and clinicians. To enhance clinical feasibility, we have developed both “ideal” and “minimum standard” protocols at 3T and 1.5T MR systems that will facilitate widespread adoption.
- Published
- 2021
6. A Hyperactive RelA/p65-Hexokinase 2 Signaling Axis Drives Primary Central Nervous System Lymphoma
- Author
-
Daniel P. Cahill, Kensuke Tateishi, Hiroyuki Mano, Shoji Yamanaka, Alexandria Fink, Koichi Ichimura, Takashi Yamamoto, Andrew S. Chi, Makoto Ohtake, Shilpa S. Tummala, Motoo Nagane, Tracy T. Batchelor, Masahito Kawazu, Akio Miyake, Ryohei Miyazaki, Akihide Ryo, Naoko Udaka, Nobuyoshi Sasaki, Manabu Natsumeda, Hidetoshi Murata, Jun Suenaga, Kentaro Ohki, Toshihide Ueno, Yukie Yoshii, Hiroaki Wakimoto, Ichio Aoki, Mayuko Nishi, Jun Watanabe, Taishi Nakamura, Yukihiko Fujii, Yohei Miyake, Jo Sasame, Norio Shiba, Julie J. Miller, Naoki Ikegaya, and Yuko Matsushita
- Subjects
0301 basic medicine ,Cancer Research ,Lymphoma ,Central nervous system ,Mice, SCID ,medicine.disease_cause ,Central Nervous System Neoplasms ,Viral Matrix Proteins ,03 medical and health sciences ,0302 clinical medicine ,Hexokinase ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Mutation ,business.industry ,NF-kappa B ,Transcription Factor RelA ,Primary central nervous system lymphoma ,CD79B ,medicine.disease ,Xenograft Model Antitumor Assays ,Phenotype ,NIMA-Interacting Peptidylprolyl Isomerase ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Myeloid Differentiation Factor 88 ,Cancer research ,Female ,Signal transduction ,business ,Glycolysis ,CD79 Antigens ,Signal Transduction - Abstract
Primary central nervous system lymphoma (PCNSL) is an isolated type of lymphoma of the central nervous system and has a dismal prognosis despite intensive chemotherapy. Recent genomic analyses have identified highly recurrent mutations of MYD88 and CD79B in immunocompetent PCNSL, whereas LMP1 activation is commonly observed in Epstein–Barr virus (EBV)-positive PCNSL. However, a lack of clinically representative preclinical models has hampered our understanding of the pathogenic mechanisms by which genetic aberrations drive PCNSL disease phenotypes. Here, we establish a panel of 12 orthotopic, patient-derived xenograft (PDX) models from both immunocompetent and EBV-positive PCNSL and secondary CNSL biopsy specimens. PDXs faithfully retained their phenotypic, metabolic, and genetic features, with 100% concordance of MYD88 and CD79B mutations present in PCNSL in immunocompetent patients. These models revealed a convergent functional dependency upon a deregulated RelA/p65-hexokinase 2 signaling axis, codriven by either mutated MYD88/CD79B or LMP1 with Pin1 overactivation in immunocompetent PCNSL and EBV-positive PCNSL, respectively. Notably, distinct molecular alterations used by immunocompetent and EBV-positive PCNSL converged to deregulate RelA/p65 expression and to drive glycolysis, which is critical for intracerebral tumor progression and FDG-PET imaging characteristics. Genetic and pharmacologic inhibition of this key signaling axis potently suppressed PCNSL growth in vitro and in vivo. These patient-derived models offer a platform for predicting clinical chemotherapeutics efficacy and provide critical insights into PCNSL pathogenic mechanisms, accelerating therapeutic discovery for this aggressive disease. Significance: A set of clinically relevant CNSL xenografts identifies a hyperactive RelA/p65-hexokinase 2 signaling axis as a driver of progression and potential therapeutic target for treatment and provides a foundational preclinical platform.
- Published
- 2020
7. Clinical utility of targeted next-generation sequencing assay in IDH-wildtype glioblastoma for therapy decision-making
- Author
-
Mary Jane Lim-Fat, Gilbert C Youssef, Mehdi Touat, J Bryan Iorgulescu, Sydney Whorral, Marie Allen, Rifaquat Rahman, Ugonma Chukwueke, J Ricardo McFaline-Figueroa, Lakshmi Nayak, Eudocia Q Lee, Tracy T Batchelor, Omar Arnaout, Pier Paolo Peruzzi, E Antonio Chiocca, David A Reardon, David Meredith, Sandro Santagata, Rameen Beroukhim, Wenya Linda Bi, Keith L Ligon, and Patrick Y Wen
- Subjects
Adult ,Cancer Research ,Clinical Trials as Topic ,Oncology ,Mutation ,Clinical Investigations ,High-Throughput Nucleotide Sequencing ,Humans ,Neurology (clinical) ,Pathology, Molecular ,Glioblastoma ,Retrospective Studies - Abstract
Background Targeted gene NGS testing is available through many academic institutions and commercial entities and is increasingly incorporated in practice guidelines for glioblastoma (GBM). This single-center retrospective study aimed to evaluate the clinical utility of incorporating NGS results in the management of GBM patients at a clinical trials-focused academic center. Methods We identified 1011 consecutive adult patients with pathologically confirmed GBM (IDHwt or IDHmut) who had somatic tumor sequencing (Oncopanel, ~500 cancer gene panel) at DFCI from 2013–2019. Clinical records of all IDHwt GBM patients were reviewed to capture clinical trial enrollment and off-label targeted therapy use based on NGS results. Results Of the 557 IDHwt GBM patients with sequencing, 182 entered clinical trials at diagnosis (32.7%) and 213 (38.2%) entered after recurrence. Sequencing results for 130 patients (23.3%) were utilized for clinical trial enrollment for either targeted therapy indications (6.9 % upfront and 27.7% at recurrent clinical trials and 3.1% for off-label targeted therapy) or exploratory studies (55.4% upfront and 6.9% recurrent clinical trials). Median overall survival was 20.1 months with no survival difference seen between patients enrolled in clinical trials compared to those who were not, in a posthoc analysis. Conclusions While NGS testing has become essential for improved molecular diagnostics, our study illustrates that targeted gene panels remain underutilized for selecting therapy in GBM-IDHwt. Targeted therapy and clinical trial design remain to be improved to help leverage the potential of NGS in clinical care.
- Published
- 2021
8. TAMI-29. MR SPECTROSCOPY MEASURES OF LAC/NAA AND NAA/CHO DIFFERENTIATE SURVIVORSHIP IN PATIENTS WITH RECURRENT GLIOBLASTOMA TREATED WITH ANTI-ANGIOGENIC THERAPY
- Author
-
Elizabeth R. Gerstner, Bruce R. Rosen, Eva-Maria Ratai, Jorg Dietrich, Pratik Talati, Otto Rapalino, Deborah Forst, Ovidiu C. Andronesi, Sharif N Natheir, Jayashree Kalpathy-Cramer, Mark Vangel, Yi-Fen Yen, Daniel Kim, Ramon Gonzalez, Tracy T. Batchelor, Melanie Fu, Michael Wenke, Mohamed El-Abtah, Isabel Arrillaga-Romany, Julian He, and Anna Vaynrub
- Subjects
In vivo magnetic resonance spectroscopy ,Cancer Research ,business.industry ,Recurrent glioblastoma ,Anti angiogenic ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,nervous system diseases ,nervous system ,Oncology ,Survivorship curve ,mental disorders ,Cancer research ,Medicine ,In patient ,Neurology (clinical) ,business - Abstract
Patients with recurrent glioblastoma (rGBM) are often started on anti-angiogenic therapy such as bevacizumab. However, determining treatment failure using conventional MRI methods remains challenging. We prospectively collected longitudinal MR spectroscopy data in 33 patients with rGBM and quantified various metabolites including N-acetylaspartate (NAA), Choline (Cho), and Lactate (Lac). After stratifying patients by 9 month survival, we found that longer-term survivors had decreased Lac/NAA and increased NAA/Cho compared to shorter-term survivors. ROC analyses illustrated that intratumoral changes in NAA/Cho were predictive of survival at 1 day (AUC 0.92), 2 weeks (AUC 0.75), 8 weeks (AUC 0.71) and 16 weeks (AUC 0.85) but not 4 weeks (AUC 0.60). Intratumoral changes in Lac/NAA were predictive of survival at all time points tested (AUCs > 0.76 for all time points). At 8 weeks, 90% of patients with increased Lac/NAA from baseline and 88% of patients with decreased NAA/Cho did not survive 9 months. Changes in NAA/Cho and Lac/NAA may serve as early biomarkers of anti-angiogenic treatment failure.
- Published
- 2021
9. BIOM-09. MYO-INOSITOL LEVELS ON MR SPECTROSCOPY CAN PREDICT FAILURE OF ANTI-ANGIOGENIC TREATMENT IN RECURRENT GLIOBLASTOMA
- Author
-
Elizabeth R. Gerstner, Pratik Talati, Ramon Gonzalez, Otto Rapalino, Ovidiu C. Andronesi, Mark Vangel, Akila Weerasekera, Eva-Maria Ratai, Julian He, Daniel Kim, Yi-Fen Yen, Anna Vaynrub, Tracy T. Batchelor, Michael Wenke, Melanie Fu, Isabel Arrillaga-Romany, Jorg Dietrich, Bruce R. Rosen, Deborah Forst, and Mohamed El-Abtah
- Subjects
In vivo magnetic resonance spectroscopy ,Cancer Research ,Tumor microenvironment ,Bevacizumab ,business.industry ,Angiogenesis ,Recurrent glioblastoma ,Anti angiogenic ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,medicine.disease ,chemistry.chemical_compound ,Oncology ,chemistry ,Cancer research ,Medicine ,Inositol ,Neurology (clinical) ,business ,medicine.drug ,Glioblastoma - Abstract
BACKGROUND Recurrent glioblastoma (rGBM) patients are often treated with anti-angiogenic agents such as bevacizumab (BEV). Despite therapeutic promise, conventional MR methods fail to determine which patients may not benefit. PURPOSE: The purpose of this study was to utilize magnetic resonance spectroscopic imaging (MRSI) with intermediate and short echo time to generate corrected Myo-inositol normalized by contralateral creatine (mI/c-Cr) in patients with rGBM treated with BEV and investigate whether it can predict survivorship prior to BEV initiation (baseline) and at 1-day, 4-weeks, and 8-weeks thereafter. METHODS We conducted a prospective, longitudinal study and evaluated spectroscopic data of myo-inositol (mI), a glial marker and osmoregulator within the brain, normalized to contralateral-creatine (mI/c-Cr) in the intratumoral, contralateral normal appearing white matter, and peritumoral volumes of rGBM patients. Area under the ROC curve (AUC) was calculated for all volumes at baseline, 1-day, 4-weeks, and 8-weeks after treatment to determine mI/c-Cr’s ability to predict survivorship. RESULTS 21 participants (62 ± 12 years, 15 men) were evaluated. Lower mI/c-Cr in the tumor prior to and during BEV treatment predicted poor survivorship, with ROC analyses illustrating an AUC of 0.75 at baseline, 0.87 at 1-day, and 1 at 8 weeks. Lower levels of mI/c-Cr were also observed in the contralateral and the peritumoral volumes for shorter-term survivors. In the contralateral volume, lower mI/Cr was predictive of shorter-term survival at baseline and all other timepoints. Within the peritumoral volume, lower mI/c-Cr was predictive of shorter-term survival at baseline (AUC=0.80), 1-day (AUC=0.93), and 4-weeks (AUC=0.68). CONCLUSIONS Lower levels of mI/c-Cr within intratumoral, contralateral, and peritumoral volumes were predictive of poor survivorship and anti-angiogenic treatment failure as early as one month before BEV treatment. Acquiring MRSI alongside conventional MR imaging modalities can convey critical information regarding tumor microenvironment that informs management of patients with rGBM.
- Published
- 2021
10. SARS‐COV‐2 INFECTION IN 50 PATIENTS WITH PRIMARY CNS LYMPHOMA: PRESENTATION, EFFECTS ON TUMOR TREATMENT AND OUTCOME IN A SERIES OF THE INTERNATIONAL PCNSL COLLABORATIVE GROUP
- Author
-
K. Hoang Xuan, Anna Ferreri, Sara Steffanoni, Kate Cwynarski, J. Boot, Gilad Itchaki, Mehdi Touat, Christian Grommes, A. Alencar, Marianna Sassone, N. Crosbie, S. Chaganti, Alice Laurenge, Carole Soussain, Christopher P. Fox, J. Dietrich, Tracy T. Batchelor, and T. Calimeri
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Tumor therapy ,Hematology ,General Medicine ,Collaborative group ,Primary CNS Lymphoma ,Internal medicine ,medicine ,Presentation (obstetrics) ,business - Published
- 2021
11. Biological activity of weekly ONC201 in adult recurrent glioblastoma patients
- Author
-
Patrick Y. Wen, Isabel Arrillaga-Romany, Krystal Merdinger, Varun V. Prabhu, Minesh P. Mehta, Rohinton Tarapore, Martin Stogniew, Joshua E. Allen, Yazmin Odia, Wolfgang Oster, and Tracy T. Batchelor
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pyridines ,Clinical Investigations ,Antineoplastic Agents ,Histones ,Young Adult ,Dopamine receptor D2 ,Internal medicine ,Clinical endpoint ,Humans ,Medicine ,Receptors, Dopamine D5 ,Dosing ,Aged ,Brain Neoplasms ,business.industry ,Imidazoles ,Antagonist ,Middle Aged ,Progression-Free Survival ,Pyrimidines ,Dopamine receptor ,Pharmacodynamics ,Mutation ,Cohort ,Toxicity ,Female ,Neurology (clinical) ,Neoplasm Recurrence, Local ,Glioblastoma ,business - Abstract
Background ONC201 is a dopamine receptor D2 (DRD2) antagonist that penetrates the blood–brain barrier. ONC201 efficacy has been shown in glioblastoma animal models and is inversely correlated with dopamine receptor DRD5 expression. ONC201 is well tolerated in adult recurrent glioblastoma patients with dosing every 3 weeks and has achieved an objective radiographic response in a patient harboring the H3 K27M mutation. Methods In a window-of-opportunity arm, 6 adult subjects initiated ONC201 prior to re-resection of recurrent glioblastoma with intratumoral concentrations as the primary endpoint. An additional 20 adults with recurrent glioblastoma received single agent weekly oral ONC201 at 625 mg, with progression-free survival at 6 months (PFS6) by Response Assessment in Neuro-Oncology (RANO) criteria as the primary endpoint. Results The window-of-opportunity arm achieved its primary endpoint with intratumoral ONC201 concentrations at ~24 hours following the second weekly dose ranging from 600 nM to 9.3 µM. Intratumoral pharmacodynamics assessed by activating transcriptional factor 4, death receptor 5, and apoptosis induction relative to archival samples were observed with the strongest intensity and uniformity among patients with low DRD5 tumor expression. The primary endpoint of PFS6 by RANO was not achieved at 5% in this molecularly unselected cohort; however, 1 of 3 patients enrolled with the H3 K27M mutation had a complete regression of enhancing multifocal lesions that remained durable for >1.5 years. No treatment modifications or discontinuations due to toxicity were observed, including in those who underwent re-resection. Conclusions Weekly ONC201 is well tolerated, and meaningful intratumoral concentrations were achieved. ONC201 may be biologically active in a subset of adult patients with recurrent glioblastoma.
- Published
- 2019
12. Pediatric and adult H3 K27M-mutant diffuse midline glioma treated with the selective DRD2 antagonist ONC201
- Author
-
Isabel Arillaga-Romany, Yoshie Umemura, Ziad Khatib, Krystal Merdinger, Rohinton Tarapore, Andrew S. Chi, Sabine Mueller, Martin Stogniew, Matthias A. Karajannis, Angela J. Waanders, David N. Korones, Cassie Kline, Yazmin Odia, Joshua E. Allen, Matthew Hall, Wafik Zaky, Irene Cherrick, Jane E. Minturn, Minesh P. Mehta, Shiao Pei Weathers, Toba N. Niazi, Sharon Gardner, Wolfgang Oster, Soumen Khatua, Doured Daghistani, Lee Schalop, Nicole Shonka, Xiao-Tang Kong, Ashley Sumrall, Tracy T. Batchelor, and Patrick Y. Wen
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Neurology ,Adolescent ,Pyridines ,Radiography ,Antineoplastic Agents ,Disease ,Malignancy ,Heterocyclic Compounds, 4 or More Rings ,Article ,Histones ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Median follow-up ,Glioma ,Internal medicine ,Humans ,Medicine ,Young adult ,Child ,Brain Neoplasms ,Receptors, Dopamine D2 ,business.industry ,Imidazoles ,Prognosis ,medicine.disease ,Survival Rate ,Pyrimidines ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Expanded access ,Mutation ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Follow-Up Studies - Abstract
BACKGROUND: H3 K27M-mutant diffuse midline glioma is a fatal malignancy with no proven medical therapies. The entity predominantly occurs in children and young adults. ONC201 is a small molecule selective antagonist of dopamine receptor D2/3 (DRD2/3) with an exceptional safety profile. Following up on a durable response in the first H3 K27M-mutant diffuse midline glioma patient who received ONC201 (NCT02525692), an expanded access program was initiated. METHODS: Patients with H3 K27M-mutant gliomas who received at least prior radiation were eligible. Patients with leptomeningeal spread were excluded. All patients received open-label ONC201 orally once every week. Safety, radiographic assessments, and overall survival were regularly assessed at least every 8 weeks by investigators. As of August 2018, a total of 18 patients with H3 K27M-mutant diffuse midline glioma or DIPG were enrolled to single patient expanded access ONC201 protocols. Among the 18 patients: seven adult (>20 years old) and seven pediatric (
- Published
- 2019
13. Long-term outcomes and late adverse effects of a prospective study on proton radiotherapy for patients with low-grade glioma
- Author
-
Barbara C. Fullerton, Beow Y. Yeap, Helen A. Shih, Tracy T. Batchelor, Lisa B. Nachtigall, William T. Curry, Jay S. Loeffler, Trevor J. Royce, Janet C. Sherman, Shervin Tabrizi, Michael Dworkin, J Daartz, Mary K. Colvin, and Kevin S. Oh
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Neurocognitive Disorders ,Disease ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,Quality of life ,Internal medicine ,Proton Therapy ,Humans ,Medicine ,Radiology, Nuclear Medicine and imaging ,Prospective Studies ,Radiation Injuries ,Adverse effect ,Prospective cohort study ,Proton therapy ,Brain Neoplasms ,business.industry ,Glioma ,Hematology ,Middle Aged ,Neurosecretory Systems ,Progression-Free Survival ,Radiation therapy ,030220 oncology & carcinogenesis ,Toxicity ,Disease Progression ,Quality of Life ,Female ,Neoplasm Grading ,business ,Neurocognitive - Abstract
Background Patients with low-grade gliomas (LGG) can survive years with their illness. Proton radiotherapy (PRT) can reduce off-target dose and decrease the risk of treatment-related morbidity. We examined long-term morbidity following proton therapy in this updated prospective cohort of patients with LGG. Methods Twenty patients with LGG were enrolled prospectively and received PRT to 54 Gy(RBE) in 30 fractions. Comprehensive baseline and longitudinal assessments of toxicity, neurocognitive and neuroendocrine function, quality of life, and survival outcomes were performed up to 5 years following treatment. Results Six patients died (all of disease) and six had progression of disease. Median follow-up was 6.8 years for the 14 patients alive at time of reporting. Median progression-free survival (PFS) was 4.5 years. Of tumors tested for molecular markers, 71% carried the IDH1-R132H mutation and 29% had 1p/19q co-deletion. There was no overall decline in neurocognitive function; however, a subset of five patients with reported cognitive symptoms after radiation therapy had progressively worse function by neurocognitive testing. Six patients developed neuroendocrine deficiencies, five of which received Dmax ≥20 Gy(RBE) to the hypothalamus–pituitary axis (HPA). Most long-term toxicities developed within 2 years after radiation therapy. Conclusions The majority of patients with LGG who received proton therapy retained stable cognitive and neuroendocrine function. The IDH1-R132H mutation was present in the majority, while 1p/19q loss was present in a minority. A subset of patients developed neuroendocrine deficiencies and was more common in those with higher dose to the HPA.
- Published
- 2019
14. PI3K/AKT/mTOR Pathway Alterations Promote Malignant Progression and Xenograft Formation in Oligodendroglial Tumors
- Author
-
Yohei Miyake, A. John Iafrate, Daniel P. Cahill, Jo Sasame, Yuko Matsushita, Tracy T. Batchelor, William T. Curry, Julie J. Miller, Erik A. Williams, Mara V.A. Koerner, Shigeo Mukaihara, Ryogo Minamimoto, Kenji Fujimoto, Akihide Ryo, Shigeta Miyake, Hiroki Taguchi, Alexandria Fink, Tareq A. Juratli, Takashi Shuto, Andrew S. Chi, Nina Lelic, Shigeo Matsunaga, Shilpa S. Tummala, Naoko Udaka, Takahiro Tanaka, Dora Dias-Santagata, Koichi Ichimura, Takashi Yamamoto, Mayuko Nishi, Hidetoshi Murata, Hiroaki Wakimoto, Taishi Nakamura, Kensuke Tateishi, and Shoji Yamanaka
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,medicine.disease ,In vitro ,nervous system diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,In vivo ,Tumor progression ,Cell culture ,Cancer research ,Medicine ,Oligodendroglial Tumor ,Oligodendroglioma ,business ,neoplasms ,Protein kinase B ,030217 neurology & neurosurgery ,PI3K/AKT/mTOR pathway - Abstract
Purpose: Oligodendroglioma has a relatively favorable prognosis, however, often undergoes malignant progression. We hypothesized that preclinical models of oligodendroglioma could facilitate identification of therapeutic targets in progressive oligodendroglioma. We established multiple oligodendroglioma xenografts to determine if the PI3K/AKT/mTOR signaling pathway drives tumor progression. Experimental Design: Two anatomically distinct tumor samples from a patient who developed progressive anaplastic oligodendroglioma (AOD) were collected for orthotopic transplantation in mice. We additionally implanted 13 tumors to investigate the relationship between PI3K/AKT/mTOR pathway alterations and oligodendroglioma xenograft formation. Pharmacologic vulnerabilities were tested in newly developed AOD models in vitro and in vivo. Results: A specimen from the tumor site that subsequently manifested rapid clinical progression contained a PIK3CA mutation E542K, and yielded propagating xenografts that retained the OD/AOD-defining genomic alterations (IDH1R132H and 1p/19q codeletion) and PIK3CAE542K, and displayed characteristic sensitivity to alkylating chemotherapeutic agents. In contrast, a xenograft did not engraft from the region that was clinically stable and had wild-type PIK3CA. In our panel of OD/AOD xenografts, the presence of activating mutations in the PI3K/AKT/mTOR pathway was consistently associated with xenograft establishment (6/6, 100%). OD/AOD that failed to generate xenografts did not have activating PI3K/AKT/mTOR alterations (0/9, P < 0.0001). Importantly, mutant PIK3CA oligodendroglioma xenografts were vulnerable to PI3K/AKT/mTOR pathway inhibitors in vitro and in vivo—evidence that mutant PIK3CA is a tumorigenic driver in oligodendroglioma. Conclusions: Activation of the PI3K/AKT/mTOR pathway is an oncogenic driver and is associated with xenograft formation in oligodendrogliomas. These findings have implications for therapeutic targeting of PI3K/AKT/mTOR pathway activation in progressive oligodendrogliomas.
- Published
- 2019
15. Automatic assessment of glioma burden: a deep learning algorithm for fully automated volumetric and bidimensional measurement
- Author
-
Otto Rapalino, Andrew Beers, Bruce R. Rosen, Bo Xiao, Marco C. Pinho, Paul J. Zhang, Harrison X. Bai, Jerrold L. Boxerman, Yinyan Wang, Alessandro Boaro, Jayashree Kalpathy-Cramer, Ken Chang, Tracy T. Batchelor, James M. Brown, Patrick Y. Wen, Xuejun Li, Joeky T. Senders, Li Yang, Raymond Y. Huang, Weihua Liao, Hao Zhou, Elizabeth R. Gerstner, Wenya Linda Bi, Vasileios K. Kavouridis, Omar Arnaout, Chang Su, Qin Shen, and K. Ina Ly
- Subjects
G740 Computer Vision ,RANO ,Cancer Research ,Treatment response ,A300 Clinical Medicine ,Intraclass correlation ,Fluid-attenuated inversion recovery ,Preoperative care ,longitudinal response assessment ,Automation ,03 medical and health sciences ,Deep Learning ,0302 clinical medicine ,Artificial Intelligence ,G730 Neural Computing ,Glioma ,Image Processing, Computer-Assisted ,medicine ,Humans ,Longitudinal Studies ,Postoperative Care ,medicine.diagnostic_test ,Brain Neoplasms ,business.industry ,segmentation ,Editorials ,Magnetic resonance imaging ,Prognosis ,medicine.disease ,Magnetic Resonance Imaging ,Hyperintensity ,Tumor Burden ,Oncology ,Fully automated ,030220 oncology & carcinogenesis ,Basic and Translational Investigations ,Neurology (clinical) ,G760 Machine Learning ,B140 Neuroscience ,business ,Algorithm ,Algorithms ,030217 neurology & neurosurgery - Abstract
Background Longitudinal measurement of glioma burden with MRI is the basis for treatment response assessment. In this study, we developed a deep learning algorithm that automatically segments abnormal fluid attenuated inversion recovery (FLAIR) hyperintensity and contrast-enhancing tumor, quantitating tumor volumes as well as the product of maximum bidimensional diameters according to the Response Assessment in Neuro-Oncology (RANO) criteria (AutoRANO). Methods Two cohorts of patients were used for this study. One consisted of 843 preoperative MRIs from 843 patients with low- or high-grade gliomas from 4 institutions and the second consisted of 713 longitudinal postoperative MRI visits from 54 patients with newly diagnosed glioblastomas (each with 2 pretreatment “baseline” MRIs) from 1 institution. Results The automatically generated FLAIR hyperintensity volume, contrast-enhancing tumor volume, and AutoRANO were highly repeatable for the double-baseline visits, with an intraclass correlation coefficient (ICC) of 0.986, 0.991, and 0.977, respectively, on the cohort of postoperative GBM patients. Furthermore, there was high agreement between manually and automatically measured tumor volumes, with ICC values of 0.915, 0.924, and 0.965 for preoperative FLAIR hyperintensity, postoperative FLAIR hyperintensity, and postoperative contrast-enhancing tumor volumes, respectively. Lastly, the ICCs for comparing manually and automatically derived longitudinal changes in tumor burden were 0.917, 0.966, and 0.850 for FLAIR hyperintensity volume, contrast-enhancing tumor volume, and RANO measures, respectively. Conclusions Our automated algorithm demonstrates potential utility for evaluating tumor burden in complex posttreatment settings, although further validation in multicenter clinical trials will be needed prior to widespread implementation.
- Published
- 2019
16. PIK3CA activating mutations are associated with more disseminated disease at presentation and earlier recurrence in glioblastoma
- Author
-
Daniel P. Cahill, Andrew S. Chi, Shota Tanaka, Daniel Yang, Tracy T. Batchelor, Dora Dias-Santagata, David N. Louis, Darrell R. Borger, A. John Iafrate, and Leif W. Ellisen
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Methyltransferase ,Neurology ,Multivariate analysis ,Class I Phosphatidylinositol 3-Kinases ,Dissemination ,Gliomatosis ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,0302 clinical medicine ,Internal medicine ,Gene duplication ,medicine ,Humans ,Disseminated disease ,Clinical significance ,Genotyping ,neoplasms ,lcsh:Neurology. Diseases of the nervous system ,Aged ,Retrospective Studies ,Aged, 80 and over ,medicine.diagnostic_test ,business.industry ,Brain Neoplasms ,Research ,Brain ,PIK3CA ,DNA Methylation ,Middle Aged ,medicine.disease ,Survival Analysis ,030104 developmental biology ,Multicentric ,Mutation ,Female ,Neurology (clinical) ,Neoplasm Recurrence, Local ,business ,Glioblastoma ,030217 neurology & neurosurgery ,Fluorescence in situ hybridization - Abstract
Phosphatidylinositol 3-kinase signaling promotes cell growth and survival and is frequently activated in infiltrative gliomas. Activating mutations in PIK3CA gene are observed in 6–15% of glioblastomas, although their clinical significance is largely undescribed. The objective of this study was to examine whether PIK3CA mutations are associated with a specific clinical phenotype in glioblastoma. We retrospectively reviewed 157 consecutive newly diagnosed glioblastoma patients from December 2009 to June 2012 who underwent molecular profiling consisting of targeted hotspot genotyping, fluorescence in situ hybridization for gene amplification, and methylation-specific PCR for O6-methylguanine-DNA methyltransferase promoter methylation. Molecular alterations were correlated with clinical features, imaging and outcome. The Cancer Genome Atlas data was analyzed as a validation set. There were 91 males; median age was 58 years (range, 23–85). With a median follow-up of 20.9 months, median progression-free survival (PFS) and estimated overall survival (OS) were 11.9 and 24.0 months, respectively. Thirteen patients (8.3%) harbored PIK3CA mutation, which was associated with younger age (mean 49.4 vs. 58.1 years, p = 0.02). PIK3CA mutation correlated with shorter PFS (median 6.9 vs. 12.4 months, p = 0.01) and OS (median 21.2 vs. 24.2 months, p = 0.049) in multivariate analysis. A significant association between PIK3CA mutation and more disseminated disease at diagnosis, as defined by gliomatosis, multicentric lesions, or distant leptomeningeal lesions, was observed (46.2% vs. 11.1%, p = 0.004). In conclusion, despite the association with younger age, PIK3CA activating mutations are associated with earlier recurrence and shorter survival in adult glioblastoma. The aggressive course of these tumors may be related to their propensity for disseminated presentation. Electronic supplementary material The online version of this article (10.1186/s40478-019-0720-8) contains supplementary material, which is available to authorized users.
- Published
- 2019
17. Treatment-induced brain tissue necrosis: a clinical challenge in neuro-oncology
- Author
-
Tracy T. Batchelor, Jorg Dietrich, Franziska Loebel, Maria Martinez-Lage, Peter Vajkoczy, Sebastian F Winter, and Jay S. Loeffler
- Subjects
Cancer Research ,medicine.medical_specialty ,Necrosis ,Neuro oncology ,Reviews ,Brain tissue ,Diagnostic tools ,Diagnosis, Differential ,Temozolomide ,medicine ,Humans ,Radiation Injuries ,Intensive care medicine ,Adverse effect ,Antineoplastic Agents, Alkylating ,Protein Kinase Inhibitors ,Radiotherapy ,Neurologic Oncology ,Brain Neoplasms ,business.industry ,Brain ,Cancer ,Chemoradiotherapy ,medicine.disease ,Patient management ,Oncology ,Disease Progression ,Neurotoxicity Syndromes ,Neurology (clinical) ,Neoplasm Recurrence, Local ,medicine.symptom ,Glioblastoma ,business - Abstract
Cancer therapy-induced adverse effects on the brain are a major challenge in neuro-oncology. Brain tissue necrosis (treatment necrosis [TN]) as a consequence of brain directed cancer therapy remains an insufficiently characterized condition with diagnostic and therapeutic difficulties and is frequently associated with significant patient morbidity. A better understanding of the underlying mechanisms, improvement of diagnostic tools, development of preventive strategies, and implementation of evidence-based therapeutic practices are pivotal to improve patient management. In this comprehensive review, we address existing challenges associated with current TN-related clinical and research practices and highlight unanswered questions and areas in need of further research with the ultimate goal to improve management of patients affected by this important neuro-oncological condition.
- Published
- 2019
18. Upfront Surgical Resection of Melanoma Brain Metastases Provides a Bridge Toward Immunotherapy-Mediated Systemic Control
- Author
-
Jackson Stocking, Alexander Kaplan, Anita Giobbie-Hurder, Mia Bertalan, Christopher Alvarez-Breckenridge, Priscilla K. Brastianos, Daniel P. Cahill, Donald P. Lawrence, Corey M. Gill, Tracy T. Batchelor, Kevin S. Oh, Helen A. Shih, Naema Nayyar, Keith T. Flaherty, and Ryan J. Sullivan
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Skin Neoplasms ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Kaplan-Meier Estimate ,Radiosurgery ,Young Adult ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,CTLA-4 Antigen ,Neuro‐Oncology ,Prospective Studies ,Melanoma ,Aged ,Retrospective Studies ,Aged, 80 and over ,Brain Neoplasms ,business.industry ,Proportional hazards model ,Hazard ratio ,Age Factors ,Brain ,Retrospective cohort study ,Immunotherapy ,Middle Aged ,Prognosis ,medicine.disease ,Neoadjuvant Therapy ,Immune checkpoint ,Blockade ,Treatment Outcome ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Female ,business ,030217 neurology & neurosurgery ,Follow-Up Studies ,Brain metastasis - Abstract
Background Immune checkpoint blockade has systemic efficacy in patients with metastatic melanoma, including those with brain metastases (MBMs). However, immunotherapy-induced intracranial tumoral inflammation can lead to neurologic compromise, requiring steroids, which abrogate the systemic efficacy of this approach. We investigated whether upfront neurosurgical resection of MBM is associated with a therapeutic advantage when performed prior to initiation of immunotherapy. Material and methods An institutional review board-approved, retrospective study identified 142 patients with MBM treated with immune checkpoint blockade between 2010 and 2016 at Massachusetts General Hospital, of whom 79 received surgery. Patients were classified based on the temporal relationship between immunotherapy, surgery, and development of central nervous system metastases. Overall survival (OS) was calculated from the date of diagnosis of MBM until death from any cause. Multivariate model building included a prognostic Cox model of OS, the effect of immunotherapy and surgical sequencing on OS, and the effect of immunotherapy and radiation sequencing on OS. Results The 2-year overall survival for patients treated with cytotoxic T-lymphocyte antigen 4, programmed death 1, or combinatorial blockade was 19%, 54%, and 57%, respectively. Among immunotherapy-naive melanoma brain metastases, surgery followed by immunotherapy had a median survival of 22.7 months (95% confidence interval [CI], 12.6-39.2) compared with 10.8 months for patients treated with immunotherapy alone (95% CI, 7.8-16.3) and 9.4 months for patients treated with immunotherapy followed by surgery (95% CI, 4.1 to ∞; p = .12). On multivariate analysis, immunotherapy-naive brain metastases treated with immunotherapy alone were associated with increased risk of death (hazard ratio, 1.72; 95% CI, 1.00-2.99) compared with immunotherapy-naive brain metastases treated with surgery followed by immunotherapy. Conclusion In treatment-naive patients, early surgical resection for local control should be considered prior to commencing immunotherapy. A prospective, randomized trial comparing the sequence of surgery and immunotherapy for treatment-naive melanoma brain metastases is warranted. Implications for practice In this retrospective study of 142 patients with melanoma brain metastases treated with immune checkpoint blockade, the development of melanoma brain metastases following immunotherapy was associated with decreased survival compared with diagnosis of immunotherapy-naive brain metastases. The benefit of surgical intervention was seen in immunotherapy-naive brain metastases in contrast to brain metastases that developed on immunotherapy. These results suggest that upfront local control with surgery for immunotherapy-naive melanoma brain metastasis may provide a bridge toward immunotherapy-mediated systemic control.
- Published
- 2019
19. CTNI-40. EVALUATING FEASIBILITY AND EFFICIENCY OF PHASE II ADAPTIVE PLATFORM TRIAL DESIGNS BASED ON THE INDIVIDUALIZED SCREENING TRIAL OF INNOVATIVE GLIOBLASTOMA THERAPY (INSIGhT) EXPERIENCE
- Author
-
Jennifer Bruno, L. Burt Nabors, Shyam K. Tanguturi, Howard Colman, Mary Welch, Brittany Fisher-Longden, William Pisano, Eudocia Q. Lee, Mehdi Touat, Tracy T. Batchelor, Geffrey Fell, Jan Drappatz, Emily Lapinskas, Rifaquat Rahman, David A. Reardon, Manmeet Ahluwalia, Wenya Linda Bi, Isabel Arrillaga-Romany, Ugonma Chukwueke, Thomas Kaley, Jaroslaw T. Hepel, David Schiff, Christine McCluskey, Heinrich Elinzano, Lakshmi Nayak, E. Antonio Chiocca, Evanthia Galanis, Christine Lu-Emerson, Daphne A. Haas-Kogan, Brian M. Alexander, David Meredith, J Ricardo McFaline-Figueroa, Lorenzo Trippa, Mikael L. Rinne, Daniel N. Cagney, Rameen Beroukhim, Maria Lavallee, Ayal A. Aizer, Keith L. Ligon, Omar Arnaout, Lisa Doherty, Sarah Gaffey, Andrew B. Lassman, Shanna Dowling, and Patrick Y. Wen
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,O-6-methylguanine-DNA methyltransferase ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,Gene expression profiling ,Radiation therapy ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Troponin I ,Neratinib ,medicine ,Neurology (clinical) ,Progression-free survival ,business ,Abemaciclib ,medicine.drug - Abstract
BACKGROUND The Individualized Screening Trial of Innovative Glioblastoma Therapy (INSIGhT) is a phase II platform trial with Bayesian adaptive randomization and deep genomic profiling to more efficiently test experimental agents in newly diagnosed glioblastoma and to prioritize therapies for late-stage testing. METHODS In the ongoing INSIGhT trial, patients with newly diagnosed MGMT-unmethylated glioblastoma are randomized to the control arm or one of three experimental therapy arms (CC-115, abemaciclib, and neratinib). The control arm therapy is radiotherapy with concomitant and adjuvant temozolomide, and primary endpoint is overall survival. Randomization has been adapted based on Bayesian estimation of biomarker-specific probability of treatment impact on progression-free survival (PFS). All tumors undergo detailed molecular sequencing, and this is facilitated with the companion ALLELE protocol. To evaluate feasibility of this approach, we assessed the status of this ongoing trial. RESULTS Since INSIGhT was activated 4.3 years ago, it has expanded to include 12 sites across the United States. A total of 247 patients have been enrolled. Randomization probabilities have been repeatedly adjusted over time based upon early PFS results to alter the randomization ratio from standard 1:1:1:1 randomization. All three arms have completed accrual and efficacy estimates are available based upon comparison to the common control arm in context of relevant biomarkers. There are 87 patients alive and in follow-up, and there are ongoing plans to add additional arms to evaluate further treatments in the future. CONCLUSION The INSIGhT trial demonstrates that a multi-center Bayesian adaptive platform trial is a feasible and effective approach to help prioritize therapies and biomarkers for newly diagnosed GBM. The trial has maintained robust accrual, and the simultaneous testing of multiple agents, sharing a common control arm and adaptive randomization serve as features to increase trial efficiency relative to traditional clinical trial designs.
- Published
- 2021
20. MR spectroscopic imaging predicts early response to anti-angiogenic therapy in recurrent glioblastoma
- Author
-
Anna Vaynrub, Mark Vangel, Deborah Forst, Ovidiu C. Andronesi, Jayashree Kalpathy-Cramer, R. Gilberto Gonzalez, Eva-Maria Ratai, Mohamed El-Abtah, Julian He, Daniel Kim, Otto Rapalino, Tracy T. Batchelor, Michael Wenke, Bruce R. Rosen, Pratik Talati, Sharif N Natheir, Jorg Dietrich, Yi-Fen Yen, Elizabeth R. Gerstner, Melanie Fu, and Isabel Arrillaga-Romany
- Subjects
0301 basic medicine ,In vivo magnetic resonance spectroscopy ,Oncology ,medicine.medical_specialty ,Bevacizumab ,Angiogenesis ,Clinical Investigations ,Brain tumor ,bevacizumab ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,mental disorders ,AcademicSubjects/MED00300 ,Medicine ,Choline ,lactate ,medicine.diagnostic_test ,business.industry ,Recurrent glioblastoma ,MR spectroscopy ,Magnetic resonance imaging ,medicine.disease ,nervous system diseases ,030104 developmental biology ,nervous system ,chemistry ,030220 oncology & carcinogenesis ,biomarker ,Biomarker (medicine) ,AcademicSubjects/MED00310 ,business ,brain tumor ,medicine.drug - Abstract
Background Determining failure to anti-angiogenic therapy in recurrent glioblastoma (GBM) (rGBM) remains a challenge. The purpose of the study was to assess treatment response to bevacizumab-based therapy in patients with rGBM using MR spectroscopy (MRS). Methods We performed longitudinal MRI/MRS in 33 patients with rGBM to investigate whether changes in N-acetylaspartate (NAA)/Choline (Cho) and Lactate (Lac)/NAA from baseline to subsequent time points after treatment can predict early failures to bevacizumab-based therapies. Results After stratifying based on 9-month survival, longer-term survivors had increased NAA/Cho and decreased Lac/NAA levels compared to shorter-term survivors. ROC analyses for intratumoral NAA/Cho correlated with survival at 1 day, 2 weeks, 8 weeks, and 16 weeks. Intratumoral Lac/NAA ROC analyses were predictive of survival at all time points tested. At the 8-week time point, 88% of patients with decreased NAA/Cho did not survive 9 months; furthermore, 90% of individuals with an increased Lac/NAA from baseline did not survive at 9 months. No other metabolic ratios tested significantly predicted survival. Conclusions Changes in metabolic levels of tumoral NAA/Cho and Lac/NAA can serve as early biomarkers for predicting treatment failure to anti-angiogenic therapy as soon as 1 day after bevacizumab-based therapy. The addition of MRS to conventional MR methods can provide better insight into how anti-angiogenic therapy affects tumor microenvironment and predict patient outcomes.
- Published
- 2021
21. Intracranial Foreign Body Granuloma Mimicking Brain Tumor Recurrence: A Case Series
- Author
-
Tracy T. Batchelor, Deborah Forst, Jorg Dietrich, Derek H. Oakley, and Sebastian F Winter
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Brain tumor ,Gossypiboma ,Asymptomatic ,03 medical and health sciences ,0302 clinical medicine ,Glioma ,medicine ,Enhancing Lesion ,Humans ,Retrospective Studies ,business.industry ,Brain Neoplasms ,Granuloma, Foreign-Body ,Brain ,medicine.disease ,Magnetic Resonance Imaging ,Oncology ,030220 oncology & carcinogenesis ,Granuloma ,Histopathology ,Female ,Radiology ,medicine.symptom ,Neoplasm Recurrence, Local ,business ,Brief Communications ,030217 neurology & neurosurgery ,Foreign body granuloma - Abstract
Background Intracranial foreign body granuloma (FBG) is a rare inflammatory reaction to retained foreign material, manifesting acutely or months to years following neurosurgical procedures. Radiographically, FBG can mimic tumor progression, and tissue biopsy may be required to guide management. Materials and Methods In this retrospective case series, we present unique clinico-radiographic and histopathological features of six neuro-oncological patients diagnosed with FBG between 2007 and 2019. Results All six patients (4 women and 2 men, aged 29–54 [median, 30.5] years) had undergone surgical resection of a low- (n = 4) or high-grade (n = 2) glioma. FBG manifestation postsurgery ranged from 1 day to 4 years and was predominantly asymptomatic (n = 5/6). Magnetic resonance imaging universally demonstrated one or multiple peripherally enhancing lesion(s) adjacent to the resection cavity. Histopathology in all (n = 4/4) resected specimens demonstrated an inflammatory reaction to foreign material, confirming FBG. Conclusion Intracranial FBG constitutes a rare but challenging treatment-related condition effectively managed by surgery, with important therapeutic implications in neuro-oncology.
- Published
- 2020
22. PATH-03. CLINICAL UTILITY OF NEXT GENERATION SEQUENCING IN IDH-WILDTYPE GLIOBLASTOMA: THE DANA-FARBER CANCER INSTITUTE EXPERIENCE
- Author
-
Sydney Whorral, Wenya Linda Bi, Lakshmi Nayak, David Meredith, Marie Allen, Mehdi Touat, Bryan Iorgulescu, Tracy T. Batchelor, Rifaquat Rahman, Jayne Vogelzang, William Pisano, Eudocia Q. Lee, E. Antonio Chiocca, Gilbert Youssef, Mary Jane Lim-Fat, David A. Reardon, Eleanor Woodward, Ugonma Chukwueke, Patrick Y. Wen, Jose Mcfaline-Figueroa, Keith L. Ligon, Sandro Santagata, and Rameen Beroukhim
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Dana-Farber Cancer Institute ,Cancer ,Molecular Pathology & Classification ,medicine.disease ,DNA sequencing ,PIK3CA gene ,Internal medicine ,Medicine ,Neurology (clinical) ,Msh2 gene ,business ,Exome ,Platelet-Derived Growth Factor alpha Receptor ,Glioblastoma - Abstract
BACKGROUND Comprehensive next generation sequencing (NGS) is available through many academic institutions and commercial entities, and is incorporated in practice guidelines for glioblastoma (GBM). We retrospective evaluated the practice patterns and utility of incorporating NGS data into routine care of GBM patients at a clinical trials-focused academic center. METHODS We identified 1,011 consecutive adult patients with histologically confirmed GBM with OncoPanel testing, a targeted exome NGS platform of 447 cancer-associated genes at Dana Farber Cancer Institute (DFCI), from 2013-2019. We selected and retrospectively reviewed clinical records of all IDH-wildtype GBM patients treated at DFCI. RESULTS We identified 557 GBM IDH-wildtype patients, of which 227 were male (40.7%). OncoPanel testing revealed 833 single nucleotide variants and indels in 44 therapeutically relevant genes (Tier 1 or 2 mutations) including PIK3CA (n=51), BRAF (n=9), FGFR1 (n=8), MSH2 (n=4), MSH6 (n=2) and MLH1 (n=1). Copy number analysis revealed 509 alterations in 18 therapeutically relevant genes including EGFR amplification (n= 186), PDGFRA amplification (N=39) and CDKN2A/2B homozygous loss (N=223). Median overall survival was 17.5 months for the whole cohort. Seventy-four therapeutic clinical trials accrued 144 patients in the upfront setting (25.9%) and 203 patients (36.4%) at recurrence. Altogether, NGS data for 107 patients (19.2%) were utilized for clinical trial enrollment or targeted therapy indications. High mutational burden (>17mutations/Mb) was identified in 11/464 samples (2.4%); of whom 3/11 received immune checkpoint blockade. Four patients received compassionate use therapy targeting EGFRvIII (rindopepimut, n=2), CKD4/6 (abemaciclib, n=1) and BRAFV600E (dabrafenib/trametinib, n=1). CONCLUSION While NGS has greatly improved diagnosis and molecular classification, we highlight that NGS remains underutilized in selecting therapy in GBM, even in a setting where clinical trials and off-label therapies are relatively accessible. Continued efforts to develop better targeted therapies and efficient clinical trial design are required to maximize the potential benefits of genomically-stratified data.
- Published
- 2020
23. CTNI-12. PRELIMINARY RESULTS OF THE ABEMACICLIB ARM IN THE INDIVIDUALIZED SCREENING TRIAL OF INNOVATIVE GLIOBLASTOMA THERAPY (INSIGHT): A PHASE II PLATFORM TRIAL USING BAYESIAN ADAPTIVE RANDOMIZATION
- Author
-
Lakshmi Nayak, Shanna Dowling, Brian M. Alexander, David Meredith, Sandro Santagata, E. Antonio Chiocca, Omar Arnaout, Brittany Fisher-Longden, Daphne H. Haas-Kogan, Geffrey Fell, Jack Geduldig, Rifaquat Rahman, Daniel Cagney, Ayal Aizer, Howard Colman, Christine Lu-Emerson, Tracy T. Batchelor, Thomas Kaley, Jan Drappatz, Patrick Y. Wen, Andrew B. Lassman, Maria Lavallee, Mehdi Touat, Ingo K. Mellinghoff, David Schiff, Manmeet Ahluwalia, Rameen Beroukhim, Shyam Tanguturi, Lisa Doherty, Lorenzo Trippa, Jaroslaw T. Hepel, Wenya Linda Bi, David A. Reardon, Jennifer Bruno, Sarah Gaffey, Keith L. Ligon, Ugonma Chukwueke, Christine McCluskey, Heinrich Elinzano, Evanthia Galanis, Mary Welch, Isabel Arrillaga-Romany, Fiona Watkinson, Pierpaolo Peruzzi, J Ricardo McFaline-Figueroa, Eudocia Q. Lee, and L. Burt Nabors
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,Screening Trial ,Bayesian probability ,Clinical Trials: Non-Immunologic ,Adaptive randomization ,medicine.disease ,chemistry.chemical_compound ,Pharmaceutical Adjuvants ,chemistry ,Internal medicine ,medicine ,Neurology (clinical) ,business ,Abemaciclib ,neoplasms ,medicine.drug ,Glioblastoma - Abstract
BACKGROUND The cyclin D-CDK4/6-Rb pathway is activated in most glioblastomas. Abemaciclib is a potent CDK4/6 inhibitor with good brain penetration approved for ER/PR/HER2- breast cancer. In order to efficiently evaluate the potential impact of abemaciclib on overall survival (OS) in newly diagnosed glioblastoma and to simultaneously develop information regarding potential genomic biomarker associations, abemaciclib was included as an arm on the Individualized Screening Trial of Innovative Glioblastoma Therapy (INSIGhT) trial. INSIGhT is a phase II platform trial using response adaptive randomization and deep genomic profiling to more efficiently test experimental agents in MGMT unmethylated glioblastoma and potentially accelerate identification of novel therapies for phase III testing. Initial randomization was equal between abemaciclib, control, and two other experimental arms but subsequent randomization was adapted based on efficacy as determined by progression-free survival (PFS). Ineffective arms were discontinued and new arms added by protocol amendment. We report preliminary results for the abemaciclib arm which has completed accrual. METHODS Patients with newly diagnosed MGMT-unmethylated glioblastoma were randomized to receive either radiotherapy with concomitant and adjuvant temozolomide at standard doses or standard radiochemotherapy followed by adjuvant abemaciclib (150–200 mg orally BID) without temozolomide. Treatment continued until progression or development of unacceptable toxicities. The primary endpoint was OS. Association between abemaciclib efficacy and cyclin D-CDK4/6-Rb pathway genomic alterations was also investigated. RESULTS There were 123 patients (50 control; 73 treated with abemaciclib). Abemaciclib was generally well-tolerated with no new toxicity signals identified. PFS was significantly longer (p=0.03, logrank test) with abemaciclib (median 6.31 months 95% CI [5.29, 8.18]) compared to the control arm (5.16 months 95% CI [4.37, 6.28]). 28/50 control and 36/73 abemaciclib patients remain alive. CONCLUSION Preliminary analysis suggests that abemaciclib increases PFS compared to control. Updated toxicity, PFS and survival data and potential genomic biomarker associations will be presented.
- Published
- 2020
24. CTNI-11. CC-115 IN NEWLY DIAGNOSED MGMT UNMETHYLATED GLIOBLASTOMA IN THE INDIVIDUALIZED SCREENING TRIAL OF INNOVATIVE GLIOBLASTOMA THERAPY (INSIGHT): A PHASE II RANDOMIZED BAYESIAN ADAPTIVE PLATFORM TRIAL
- Author
-
Lorenzo Trippa, David Meredith, E. Antonio Chiocca, Mehdi Touat, Lakshmi Nayak, Mary Welch, David Schiff, Jack Geduldig, Omar Arnaout, Louis B. Nabors, Christine McCluskey, Rameen Beroukhim, Thomas Kaley, Mikael L. Rinne, Sandro Santagata, Howard Colman, Shyam K. Tanguturi, Manmeet Ahluwalia, Brian M. Alexander, Rifaquat Rahman, Daniel N. Cagney, Andrew B. Lassman, David A. Reardon, Patrick Y. Wen, Maria Lavallee, Geoffrey Fell, Ugonma Chukwueke, Heinrich Elinzano, Ayal A. Aizer, Keith L. Ligon, Jose Mcfaline-Figueroa, Fiona Watkinson, Isabel Arrillaga-Romany, Jaroslaw T. Hepel, Daphne A. Haas-Kogan, Lisa Doherty, Christine Lu-Emerson, Tracy T. Batchelor, Shanna Dowling, Wenya Linda Bi, Jennifer Brunno, Evanthia Galanis, Brittany Fisher-Longden, Jan Drappatz, Sarah C. Gaffey, and Eudocia Q. Lee
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,Screening Trial ,medicine.medical_treatment ,Clinical Trials: Non-Immunologic ,O-6-methylguanine-DNA methyltransferase ,medicine.disease ,Radiation therapy ,Internal medicine ,medicine ,MGMT-Unmethylated Glioblastoma ,Neurology (clinical) ,Liver function ,Progression-free survival ,business ,medicine.drug ,Glioblastoma - Abstract
BACKGROUND CC-115 is an oral, CNS-penetrant, selective inhibitor of mammalian target of rapamycin kinase (mTOR) and deoxyribonucleic acid-dependent protein kinase (DNA-PK). Both targets are important in glioblastoma; PI3K/Akt/mTOR signaling is hyperactive in most glioblastomas, and DNA-PK is integral to repair of radiotherapy-mediated DNA damage. To investigate CC-115 in newly diagnosed glioblastoma and explore potential genomic biomarker associations, CC-115 was evaluated in the Individualized Screening Trial of Innovative Glioblastoma Therapy (INSIGhT) trial, an adaptive platform trial designed to efficiently test experimental agents. METHODS Adults with newly diagnosed MGMT-unmethylated glioblastoma, with genomic data available, are eligible for this ongoing trial. Patients are adaptively randomized to one of several experimental arms or the control arm: standard radiotherapy with concurrent and adjuvant temozolomide. The primary endpoint is overall survival (OS). Patients randomized to CC-115 (10mg po BID) received it concurrently with radiotherapy and as adjuvant monotherapy. As the first in-human use of CC-115 with radiation, a safety lead-in 3 + 3 design was used. RESULTS Twelve patients were randomized to CC-115; seven patients had possible treatment-related CTCAE grade > 3 toxicity, including four pre-specified dose-limiting toxicities: liver function abnormality (n=1), hyperlipidemia (n=1), lipase elevation (n=1) and cerebral edema (n=1). There was no significant difference in progression-free survival (PFS, median 4.2 months [CC-115] vs. 5.2 months, p=0.9) or OS (median 10.1 months [CC-115] vs. 14.5 months, p=0.9) compared to the 50 patients randomized to the control arm. Based on early PFS results, randomization probability to CC-115 decreased from 25% to < 10% at time of the trial arm closure. CONCLUSION Concurrent and adjuvant CC-115 was associated with toxicity and failed to improve PFS or OS. The INSIGhT trial design allowed for more efficient testing of CC-115, decreasing patients and resources allocated to a therapy that was discontinued due to concerns about toxicity and unfavorable risk-to-benefit ratio.
- Published
- 2020
25. Exploring Predictors of Response to Dacomitinib in EGFR-Amplified Recurrent Glioblastoma
- Author
-
Tom Mikkelsen, Jorg Dietrich, Lindsay K. Klofas, Hiroaki Wakimoto, Johan Skog, David A. Reardon, Sudipto K. Chakrabortty, Michael D. Valentino, Patrick Y. Wen, Eric T. Wong, Elizabeth R. Gerstner, Andrew S. Chi, William T. Curry, Leonora Balaj, Shota Tanaka, Nina Lelic, Robert R. Kitchen, Lakshmi Nayak, Tracy T. Batchelor, Isabel Arrillaga-Romany, David M. Peereboom, Eudocia Q. Lee, Mara V.A. Koerner, Xandra O. Breakefield, Andrew D. Norden, Mia Bertalan, A. John Iafrate, Scott R. Plotkin, Rebecca A. Betensky, Darrel R. Borger, and Daniel P. Cahill
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Recurrent glioblastoma ,Disease ,medicine.disease ,Dacomitinib ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Text mining ,chemistry ,030220 oncology & carcinogenesis ,Internal medicine ,Original Reports ,Medicine ,business ,Glioblastoma - Abstract
PURPOSE Despite the high frequency of EGFR genetic alterations in glioblastoma (GBM), EGFR-targeted therapies have not had success in this disease. To improve the likelihood of efficacy, we targeted adult patients with recurrent GBM enriched for EGFR gene amplification, which occurs in approximately half of GBM, with dacomitinib, a second-generation, irreversible epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor that penetrates the blood-brain barrier, in a multicenter phase II trial. PATIENTS AND METHODS We retrospectively explored whether previously described EGFR extracellular domain (ECD)–sensitizing mutations in the context of EGFR gene amplification could predict response to dacomitinib, and in a predefined subset of patients, we measured post-treatment intratumoral dacomitinib levels to verify tumor penetration. RESULTS We found that dacomitinib effectively penetrates contrast-enhancing GBM tumors. Among all 56 treated patients, 8 (14.3%) had a clinical benefit as defined by a duration of treatment of at least 6 months, of whom 5 (8.9%) remained progression free for at least 1 year. Presence of EGFRvIII or EGFR ECD missense mutation was not associated with clinical benefit. We evaluated the pretreatment transcriptome in circulating extracellular vesicles (EVs) by RNA sequencing in a subset of patients and identified a signature that distinguished patients who had durable benefit versus those with rapid progression. CONCLUSION While dacomitinib was not effective in most patients with EGFR-amplified GBM, a subset experienced a durable, clinically meaningful benefit. Moreover, EGFRvIII and EGFR ECD mutation status in archival tumors did not predict clinical benefit. RNA signatures in circulating EVs may warrant investigation as biomarkers of dacomitinib efficacy in GBM.
- Published
- 2020
26. Pemetrexed in Recurrent or Progressive Central Nervous System Lymphoma: A Phase I Multicenter Clinical Trial
- Author
-
Andrew D. Norden, April F. Eichler, Tracy T. Batchelor, SooAe Jones, Jorg Dietrich, Jan Drappatz, Laura Versmée, Eric T. Wong, Bruce A. Chabner, Lakshmi Nayak, Michelle Pisapia, Fred H. Hochberg, and Amanda Gordon
- Subjects
Central Nervous System ,Cancer Research ,medicine.medical_specialty ,Lymphoma ,Maximum Tolerated Dose ,Pemetrexed ,Neutropenia ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Adverse effect ,business.industry ,Lymphoma, Non-Hodgkin ,Clinical Trial Results ,Middle Aged ,medicine.disease ,Clinical trial ,Oncology ,Tolerability ,030220 oncology & carcinogenesis ,Cohort ,Methotrexate ,Neoplasm Recurrence, Local ,business ,030217 neurology & neurosurgery ,Progressive disease ,medicine.drug - Abstract
Lessons Learned The findings from this study using monotherapy with pemetrexed in a pretreated patient population are, overall, encouraging. Unlike high-dose methotrexate, which requires several days of inpatient hospitalization, pemetrexed is relatively easy to administer in the outpatient setting and remains a viable treatment option in this patient population. The maximum tolerated dose of pemetrexed administered (900 mg/m2 every 2 weeks) was generally well tolerated and showed activity in patients with relapsed or refractory CNSL. Background There is currently no standard salvage treatment for patients with relapsed/refractory central nervous system (CNS) lymphoma (CNSL). We report the results of a phase I study of pemetrexed, an antifolate drug with broader activity than methotrexate (MTX). We provide the safety, tolerability, and maximum tolerated dose (MTD) of pemetrexed in patients with recurrent CNSL. Methods Through October 2015, 17 patients with relapsed/refractory CNSL received pemetrexed every 2 weeks with the first cohort receiving 600 mg/m2 and dose escalation in increments of 300 mg/m2 to a maximum of 1,200 mg/m2. Three patients were to enroll at each dose level with expansion to six patients in the event of dose-limiting toxicity. Patients with both primary CNS lymphoma (PCNSL) and secondary CNS lymphoma (SCNSL) could be enrolled. Results Seventeen patients were evaluable with a median age of 63.7 years. Main adverse events included fatigue (82.4%), anemia (82.4%), and neutropenia (70.6%). The MTD was established at 900 mg/m2. Dose-limiting toxicities were recorded in one patient in the 600 mg/m2 cohort and in two patients in the 1,200 mg/m2 cohort. Fourteen patients were evaluable for response assessment; 21.4% achieved a complete response, 35.7% had a partial response, 14.3% had stable disease, and 28.6% had progressive disease. The median progression-free survival was 4.2 months. The median overall survival was 44.5 months. In the original study protocol, the plan was to add an expansion cohort of six patients at MTD level. However, the first phase of the study was characterized by slow recruitment. Therefore, after achieving the primary objective of the study and establishing the MTD, the investigators decided to amend the protocol and to close the study. Conclusion Pemetrexed administered at 900 mg/m2 every 2 weeks exhibits single-agent activity in patients with recurrent CNSL; it is well tolerated, and side effects are manageable.
- Published
- 2020
27. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions
- Author
-
Patrick Roth, Jill S. Barnholtz-Sloan, Marc Sanson, Jonathan Tsang, Tracy T. Batchelor, Ingo K. Mellinghoff, Minesh P. Mehta, Kenneth Aldape, Martin J B Taphoorn, Floris P. Barthel, Ranjit S. Bindra, Emilie Le Rhun, David Nathanson, Matthias Preusser, David A. Reardon, Michael Platten, Susan M. Chang, Brian M. Alexander, Andreas von Deimling, Martin J. van den Bent, Andrew B. Lassman, Craig Horbinski, Gelareh Zadeh, Roel G.W. Verhaak, E. Antonio Chiocca, Evanthia Galanis, Susan C Short, Eudocia Q. Lee, Patrick Y. Wen, John DeGroot, Raymond Y. Huang, Michael Weller, Mark R. Gilbert, Joerg C. Tonn, Wolfgang Wick, Monika E. Hegi, David Schiff, Michael Lim, Giuseppe Minniti, Timothy F. Cloughesy, INSERM, Université de Lille, Harvard Medical School [Boston] [HMS], University hospital of Zurich [Zurich], Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM) - U 1192 [PRISM], Dana-Farber Cancer Institute [Boston], Brigham & Women’s Hospital [Boston] (BWH), Harvard Medical School [Boston] (HMS), Universität Zürich [Zürich] = University of Zurich (UZH), Case Western Reserve University [Cleveland], The Jackson Laboratory [Bar Harbor] (JAX), Yale University School of Medicine, University of California [San Francisco] (UCSF), University of California, University of California [Los Angeles] (UCLA), MD Anderson Cancer Center [Houston], The University of Texas Health Science Center at Houston (UTHealth), Mayo Clinic [Rochester], National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH), Université de Lausanne (UNIL), Centre Hospitalier Universitaire Vaudois [Lausanne] (CHUV), Northwestern University Feinberg School of Medicine, New York Presbyterian Hospital, Columbia University Irving Medical Center (CUIMC), Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM) - U 1192 (PRISM), Université de Lille-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Centre Régional de Lutte contre le Cancer Oscar Lambret [Lille] (UNICANCER/Lille), Université Lille Nord de France (COMUE)-UNICANCER, Johns Hopkins University School of Medicine [Baltimore], Miami Cancer Institute [Florida], Memorial Sloane Kettering Cancer Center [New York], Università degli Studi di Siena = University of Siena (UNISI), Heidelberg University, Medizinische Universität Wien = Medical University of Vienna, Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), University of Virginia [Charlottesville], University of Leeds, Leiden University Medical Center (LUMC), Ludwig Maximilian University [Munich] (LMU), German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), Heidelberg University Hospital [Heidelberg], Princess Margaret Cancer Centre [Toronto, Canada], Erasmus University Medical Center [Rotterdam] (Erasmus MC), SALZET, Michel, Yale School of Medicine [New Haven, Connecticut] (YSM), University of California [San Francisco] (UC San Francisco), University of California (UC), Université de Lausanne = University of Lausanne (UNIL), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Université de Lille-UNICANCER, Institut du Cerveau = Paris Brain Institute (ICM), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), University of Virginia, Universiteit Leiden, and Neurology
- Subjects
Oncology ,clinical trials ,diagnosis ,glioblastoma ,therapy ,Cancer Research ,medicine.medical_specialty ,Consensus ,Neuro oncology ,[SDV]Life Sciences [q-bio] ,Oncology and Carcinogenesis ,Brain tumor ,Review ,03 medical and health sciences ,0302 clinical medicine ,Rare Diseases ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Randomized Controlled Trials as Topic ,Cancer ,Brain Neoplasms ,Molecular pathogenesis ,Neurosciences ,Chemoradiotherapy ,medicine.disease ,Isocitrate Dehydrogenase ,3. Good health ,Brain Disorders ,[SDV] Life Sciences [q-bio] ,Clinical trial ,Phase III as Topic ,Brain Cancer ,Good Health and Well Being ,Current management ,030220 oncology & carcinogenesis ,Neurology (clinical) ,Glioblastoma ,Societies ,030217 neurology & neurosurgery - Abstract
Glioblastomas are the most common form of malignant primary brain tumor and an important cause of morbidity and mortality. In recent years there have been important advances in understanding the molecular pathogenesis and biology of these tumors, but this has not translated into significantly improved outcomes for patients. In this consensus review from the Society for Neuro-Oncology (SNO) and the European Association of Neuro-Oncology (EANO), the current management of isocitrate dehydrogenase wildtype (IDHwt) glioblastomas will be discussed. In addition, novel therapies such as targeted molecular therapies, agents targeting DNA damage response and metabolism, immunotherapies, and viral therapies will be reviewed, as well as the current challenges and future directions for research.
- Published
- 2020
28. Financially effective test algorithm to identify an aggressive, EGFR-amplified variant of IDH-wildtype, lower-grade diffuse glioma
- Author
-
Otto Rapalino, Daniel P. Cahill, Nisha Ramamurthy, John C. DeWitt, Tejus A Bale, Jochen K. Lennerz, David N. Louis, Nicholas A. Jessop, Valentina Nardi, Maria Martinez-Lage Alvarez, Justin T. Jordan, Matthew P. Frosch, A. John Iafrate, and Tracy T. Batchelor
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lower grade ,medicine.diagnostic_test ,biology ,business.industry ,Wild type ,medicine.disease ,03 medical and health sciences ,Diffuse Glioma ,0302 clinical medicine ,Isocitrate dehydrogenase ,Test algorithm ,030220 oncology & carcinogenesis ,Internal medicine ,Glioma ,medicine ,biology.protein ,Neurology (clinical) ,Epidermal growth factor receptor ,business ,030217 neurology & neurosurgery ,Fluorescence in situ hybridization - Abstract
Background Update 3 of the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT-NOW) recognizes amplification of epidermal growth factor receptor (EGFR) as one important aberration in diffuse gliomas (World Health Organization [WHO] grade II/III). While these recommendations endorse testing, a cost-effective, clinically relevant testing paradigm is currently lacking. Here, we use real-world clinical data to propose a financially effective diagnostic test algorithm in the context of new guidelines. Methods To determine the prevalence, distribution, neuroradiographic features (Visually Accessible REMBRANDT Images [VASARI]), and prognostic relevance of EGFR amplification in lower-grade gliomas, we assembled a consecutive series of diffuse gliomas. For validation we included publicly available data from The Cancer Genome Atlas. For a cost-utility analysis we compared combined EGFR and isocitrate dehydrogenase (IDH) testing, EGFR testing based on IDH results, and no EGFR testing. Results In n = 71 WHO grade II/III gliomas, we identified EGFR amplification in 28.2%. With one exception, all EGFR amplifications occurred in IDH-wildtype gliomas. Comparison of overall survival showed that EGFR amplification denotes a significantly more aggressive subset of tumors (P 5% tumor contrast enhancement (75%, P = 0.016), and mild (not avid) enhancement (P = 0.016). The proposed testing algorithm reserves EGFR fluorescence in situ hybridization (FISH) testing for IDH-wildtype cases. Implementation would result in ~37.9% cost reduction at our institution, or about $1.3-4 million nationally. Conclusion EGFR-amplified diffuse gliomas are "glioblastoma-like" in their behavior and may represent undersampled glioblastomas, or subsets of IDH-wildtype diffuse gliomas with inherently aggressive biology. EGFR FISH after IDH testing is a financially effective and clinically relevant test algorithm for routine clinical practice.
- Published
- 2018
29. Phase I and Biomarker Study of Plerixafor and Bevacizumab in Recurrent High-Grade Glioma
- Author
-
David A. Reardon, Jennifer Stefanik, Christine McCluskey, Dan G. Duda, Alona Muzikansky, Andrew D. Norden, Tracy T. Batchelor, John G. Kuhn, Rakesh K. Jain, Patrick Y. Wen, Eudocia Q. Lee, Debra LaFrankie, Sarah C. Gaffey, Elizabeth R. Gerstner, Lisa Doherty, Katrina H. Smith, Lakshmi Nayak, and Trupti Vardam
- Subjects
Adult ,Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Oncology ,Benzylamines ,Receptors, CXCR4 ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,CD34 ,Cyclams ,CXCR4 ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Heterocyclic Compounds ,Internal medicine ,Glioma ,Biomarkers, Tumor ,medicine ,Humans ,Progression-free survival ,Aged ,Dose-Response Relationship, Drug ,Hepatocyte Growth Factor ,business.industry ,Plerixafor ,Middle Aged ,Proto-Oncogene Proteins c-met ,Neoplastic Cells, Circulating ,medicine.disease ,Progression-Free Survival ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,Neoplasm Recurrence, Local ,business ,Signal Transduction ,medicine.drug - Abstract
Purpose: Although antiangiogenic therapy for high-grade glioma (HGG) is promising, responses are not durable. Correlative clinical studies suggest that the SDF-1α/CXCR4 axis may mediate resistance to VEGFR inhibition. Preclinical data have demonstrated that plerixafor (a reversible CXCR4 inhibitor) could inhibit glioma progression after anti-VEGF pathway inhibition. We conducted a phase I study to determine the safety of plerixafor and bevacizumab in recurrent HGG. Patients and Methods: Part 1 enrolled 23 patients with a 3 × 3 dose escalation design to a maximum planned dose of plerixafor 320 μg/kg subcutaneously on days 1 to 21 and bevacizumab 10 mg/kg intravenously on days 1 and 15 of each 28-day cycle. Cerebrospinal fluid (CSF) and plasma samples were obtained for pharmacokinetic analyses. Plasma and cellular biomarkers were evaluated before and after treatment. Part 2 enrolled 3 patients and was a surgical study to determine plerixafor's penetration in tumor tissue. Results: In Part 1, no dose-limiting toxicities were seen at the maximum planned dose of plerixafor + bevacizumab. Treatment was well tolerated. After plerixafor 320 μg/kg treatment, the average CSF drug concentration was 26.8 ± 19.6 ng/mL. Plerixafor concentration in resected tumor tissue from patients pretreated with plerixafor was 10 to 12 μg/g. Circulating biomarker data indicated that plerixafor + bevacizumab induces rapid and persistent increases in plasma SDF-1α and placental growth factor. Progression-free survival correlated with pretreatment plasma soluble mesenchymal–epithelial transition receptor and sVEGFR1, and overall survival with the change during treatment in CD34+ progenitor/stem cells and CD8 T cells. Conclusions: Plerixafor + bevacizumab was well tolerated in HGG patients. Plerixafor distributed to both the CSF and brain tumor tissue, and treatment was associated with biomarker changes consistent with VEGF and CXCR4 inhibition. Clin Cancer Res; 24(19); 4643–9. ©2018 AACR.
- Published
- 2018
30. Wide Range of Clinical Outcomes in Patients with Gliomatosis Cerebri Growth Pattern: A Clinical, Radiographic, and Histopathologic Study
- Author
-
Fred H. Hochberg, Tracy T. Batchelor, Rebecca A. Betensky, Alexander B Pine, Sy Han Chiou, Daniel P. Cahill, Matthew P. Frosch, Derek H. Oakley, K. Ina Ly, Stuart R. Pomerantz, and Jorg Dietrich
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Radiography ,Gliomatosis cerebri ,World health ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Neuro‐Oncology ,In patient ,Child ,Aged ,Retrospective Studies ,Aged, 80 and over ,medicine.diagnostic_test ,business.industry ,Hazard ratio ,Histopathologic Study ,Magnetic resonance imaging ,Middle Aged ,medicine.disease ,Neoplasms, Neuroepithelial ,Confidence interval ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,Radiology ,business ,030217 neurology & neurosurgery - Abstract
Background The 2016 World Health Organization Classification of Central Nervous System Tumors categorizes gliomatosis cerebri growth pattern (GC) as a subgroup of diffuse infiltrating gliomas, defined by extent of brain involvement on magnetic resonance imaging (MRI). Clinical and radiographic features in GC patients are highly heterogeneous; however, prognosis has historically been considered poor. Subjects, materials, and methods We performed a retrospective search for patients at our institution meeting radiographic criteria of primary, type I GC (defined as diffuse tumor infiltration without associated tumor mass and contrast enhancement on MRI) and analyzed their clinical, imaging, and histopathologic features. Results A total of 34 patients met radiographic criteria of primary, type I GC, and 33 had a confirmed histologic diagnosis of an infiltrating glial neoplasm. Age >47 years at diagnosis was associated with worse overall survival (OS) compared with age ≤47 years (hazard ratio [HR] 1.04, 95% confidence interval [CI] 1.01-1.07, p = .003). Patients with grade 2 tumors demonstrated a trend for improved OS compared with those with grade 3 tumors (HR 2.65, 95% CI 0.99-7.08, p = .051). Except for brainstem involvement, extent or location of radiographic involvement did not detectably affect clinical outcome. IDH mutation status identified a subgroup of GC patients with particularly long survival up to 25 years and was associated with longer time to progression (HR 4.81, 95% CI 0.99-23.47, p = .052). Conclusion Patients with primary, type I GC do not uniformly carry a poor prognosis, even in the presence of widespread radiographic involvement. Consistent with other reports, IDH mutation status may identify patients with improved clinical outcome. Molecular characterization, rather than MRI features, may be most valuable for prognostication and management of GC patients. Implications for practice Patients with gliomatosis cerebri growth pattern (GC) constitute a challenge to clinicians, given their wide range of clinical, histologic, and radiographic presentation, heterogeneous outcome patterns, and the lack of consensus on a standardized treatment approach. This study highlights that radiographic extent of disease-albeit category-defining-does not detectably influence survival and that IDH mutations may impact clinical outcome. Practicing oncologists should be aware that select GC patients may demonstrate exceptionally favorable survival times and prognosticate patients based on molecular markers, rather than imaging features alone.
- Published
- 2018
31. Primary Central Nervous System Lymphoma
- Author
-
Sarah Löw and Tracy T. Batchelor
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Antineoplastic Agents ,Transplantation, Autologous ,Central Nervous System Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Autologous stem-cell transplantation ,hemic and lymphatic diseases ,Internal medicine ,Humans ,Medicine ,Chemotherapy ,Radiotherapy ,business.industry ,Lymphoma, Non-Hodgkin ,Primary central nervous system lymphoma ,Immunotherapy ,medicine.disease ,Lymphoma ,Radiation therapy ,Transplantation ,Neurology ,030220 oncology & carcinogenesis ,Rituximab ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Stem Cell Transplantation ,medicine.drug - Abstract
Primary central nervous system lymphoma (PCNSL) is an extranodal non-Hodgkin lymphoma limited to the brain, spinal cord, leptomeninges, and eyes. The majority of patients are immunocompetent, with a median age of 65 years at diagnosis. Historically, whole-brain radiation therapy (WBRT) was the first and sole treatment for PCNSL. Today, due to the recognized neurotoxicity of WBRT, this modality is usually avoided in the treatment. Most chemotherapy regimens are based on high-dose methotrexate plus the anti-CD20 monoclonal antibody rituximab, leading to high response rates, but 5-year survival is still poor at approximately 30% compared with other extranodal lymphomas. New treatment strategies including high-dose chemotherapy/autologous stem cell transplantation, targeted therapies focusing on, for example, genetic alterations in B cells or mammalian target of rapamycin signaling, and immunotherapy with inhibitors of the programmed cell death 1 receptor are only a few options to improve the armamentarium against PCNSL.
- Published
- 2018
32. INNV-40. REAL WORLD INTEGRATION OF THE NEUROLOGIC ASSESSMENT IN NEURO-ONCOLOGY (NANO) SCALE IN CLINICAL PRACTICE IN PATIENTS WITH IDH-WT GBM
- Author
-
Mary Jane Lim-Fat, Oluwatosin Akintola, Tamar Berger, Gilbert Youssef, Lakshmi Nayak, Marie Allen, Elisa Aquilanti, David A. Reardon, Annie Hsieh, Ugonma Chukwueke, Timothy R. Smith, L. Nicolas Gonzalez Castro, J Ricardo McFaline-Figueroa, Joshua Budhu, Tracy T. Batchelor, Lisa Doherty, Rameen Beroukhim, Christina Taubert, Patrick Y. Wen, Jennifer Stefanik, Brian M Andersen, Eudocia Q. Lee, and Alexandra Torres
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Neuro oncology ,Innovations in Patient Care ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,Clinical Practice ,Internal medicine ,medicine ,AcademicSubjects/MED00300 ,In patient ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business - Abstract
BACKGROUND The neurologic assessment in neuro-oncology (NANO) scale was developed as a standardized metric to objectively measure neurologic function in patients with brain tumors and complement radiographic assessment in defining overall outcome. The scale has been incorporated in clinical trials. Early data is suggestive of feasibility and that NANO contributes to overall outcome assessment. However, real-world use of the NANO scale to drive clinical-decision making and the predictive value of the NANO scale to determine overall survival remains unclear in IDH-wt GBM. METHODS We report on an ongoing study using the NANO scale to evaluate neurologic function in patients with IDH-wt GBM, seen at Dana-Farber Cancer Institute (DFCI). Patient demographics, tumor histology and molecular status, treatment history and progression dates are being captured. NANO score, as collected by a built-in scale in our institutional electronic medical record (EMR), functional status (Karnofsky performance status) and corticosteroid dose are collected at prespecified time points (prior to start of therapy, and during each subsequent MRI visit). Changes in the NANO score will be correlated to overall survival. Statistical analyses including descriptive data analysis and generalized linear models will be performed using R (version 3.4.3). RESULTS Since June 2020, 50 patients have been enrolled in this study, including 42 (84%) with ≥2 follow up visits. Study accrual was initially impacted by the COVID-19 pandemic, but adaptation to a virtual platform for NANO allowed for improved recruitment and follow up of patients. Study results will be available for discussion at the 2021 SNO conference. CONCLUSIONS Evaluation of neurologic function by NANO is feasible in a virtual framework in a prospective study in patients with GBM, aided by integration of the scale in our institutional EMR. NANO is able to objectively track neurologic function throughout disease course in IDH-wt GBM.
- Published
- 2021
33. Diffusion MRI Phenotypes Predict Overall Survival Benefit from Anti-VEGF Monotherapy in Recurrent Glioblastoma: Converging Evidence from Phase II Trials
- Author
-
Benjamin M. Ellingson, Dana T. Aftab, Tracy T. Batchelor, Robert J. Harris, Raymond Y. Huang, Rivka R. Colen, Elizabeth R. Gerstner, Lauren E. Abrey, Colin Hessel, Kevin Leu, Timothy F. Cloughesy, John de Groot, Gisela Schwab, Davis C. Woodworth, Martin J. van den Bent, Marion Smits, Ararat Chakhoyan, Renske Gahrmann, Whitney B. Pope, Catalina Raymond, Patrick Y. Wen, Radiology & Nuclear Medicine, Internal Medicine, and Neurology
- Subjects
Vascular Endothelial Growth Factor A ,Male ,Oncology ,Cancer Research ,Imaging biomarker ,Pyridines ,Phases of clinical research ,Angiogenesis Inhibitors ,chemistry.chemical_compound ,0302 clinical medicine ,Lomustine ,Receptors ,Medicine ,Anilides ,Cancer ,Aflibercept ,Tumor ,Vascular Endothelial Growth Factor ,Middle Aged ,Bevacizumab ,Local ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Biomedical Imaging ,Female ,medicine.drug ,medicine.medical_specialty ,Cabozantinib ,Recombinant Fusion Proteins ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Article ,Disease-Free Survival ,Cediranib ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,Biomarkers, Tumor ,Humans ,Effective diffusion coefficient ,Oncology & Carcinogenesis ,business.industry ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,Brain Disorders ,Brain Cancer ,Receptors, Vascular Endothelial Growth Factor ,Neoplasm Recurrence ,Diffusion Magnetic Resonance Imaging ,chemistry ,Quinazolines ,Neoplasm Recurrence, Local ,Glioblastoma ,business ,Nuclear medicine ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Purpose: Anti-VEGF therapies remain controversial in the treatment of recurrent glioblastoma (GBM). In the current study, we demonstrate that recurrent GBM patients with a specific diffusion MR imaging signature have an overall survival (OS) advantage when treated with cediranib, bevacizumab, cabozantinib, or aflibercept monotherapy at first or second recurrence. These findings were validated using a separate trial comparing bevacizumab with lomustine. Experimental Design: Patients with recurrent GBM and diffusion MRI from the monotherapy arms of 5 separate phase II clinical trials were included: (i) cediranib (NCT00035656); (ii) bevacizumab (BRAIN Trial, AVF3708g; NCT00345163); (iii) cabozantinib (XL184-201; NCT00704288); (iv) aflibercept (VEGF Trap; NCT00369590); and (v) bevacizumab or lomustine (BELOB; NTR1929). Apparent diffusion coefficient (ADC) histogram analysis was performed prior to therapy to estimate “ADCL,” the mean of the lower ADC distribution. Pretreatment ADCL, enhancing volume, and clinical variables were tested as independent prognostic factors for OS. Results: The coefficient of variance (COV) in double baseline ADCL measurements was 2.5% and did not significantly differ (P = 0.4537). An ADCL threshold of 1.24 μm2/ms produced the largest OS differences between patients (HR ∼ 0.5), and patients with an ADCL > 1.24 μm2/ms had close to double the OS in all anti-VEGF therapeutic scenarios tested. Training and validation data confirmed that baseline ADCL was an independent predictive biomarker for OS in anti-VEGF therapies, but not in lomustine, after accounting for age and baseline enhancing tumor volume. Conclusions: Pretreatment diffusion MRI is a predictive imaging biomarker for OS in patients with recurrent GBM treated with anti-VEGF monotherapy at first or second relapse. Clin Cancer Res; 23(19); 5745–56. ©2017 AACR.
- Published
- 2017
34. Diagnosis and management of primary central nervous system lymphoma
- Author
-
Catherine H. Han and Tracy T. Batchelor
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Performance status ,business.industry ,medicine.medical_treatment ,Primary central nervous system lymphoma ,Cancer ,Induction chemotherapy ,medicine.disease ,Surgery ,Lymphoma ,Radiation therapy ,03 medical and health sciences ,0302 clinical medicine ,Autologous stem-cell transplantation ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Rituximab ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Primary central nervous system lymphoma (PCNSL) is a rare and aggressive extranodal non-Hodgkin lymphoma (NHL) that is confined to the brain, eyes, spinal cord, or leptomeninges without systemic involvement. The overall prognosis, diagnosis, and management of PCNSL differ from those for other types of NHL. Prompt diagnosis and initiation of treatment are vital for improving clinical outcomes. PCNSL is responsive to radiation therapy; however, whole-brain radiotherapy (WBRT) inadequately controls the disease when it is used alone, and its delayed neurotoxicity causes neurocognitive impairment, especially in elderly patients. High-dose methotrexate (HD-MTX)-based induction chemotherapy with or without autologous stem cell transplantation (ASCT) or reduced-dose WBRT leads to durable disease control and less neurotoxicity. The optimal treatment has yet to be defined; however, HD-MTX-based induction chemotherapy is considered standard for newly diagnosed PCNSL. Ongoing randomized trials are addressing the roles of rituximab and consolidative treatment with ASCT or reduced-dose WBRT. Despite high tumor response rates with the initial treatment, many patients relapse with a very poor prognosis. The optimal treatment for refractory or relapsed PCNSL is poorly defined. The choice of salvage treatment depends on a patient's age, previous treatment and response, performance status, and comorbidities at the time of relapse. This review provides an overview of the clinical features, diagnosis, pathology, and management of PCNSL in immunocompetent patients, and it focuses on recent advances in treatment. Cancer 2017;123:4314-24. © 2017 American Cancer Society.
- Published
- 2017
35. Resolving the phylogenetic origin of glioblastoma via multifocal genomic analysis of pre-treatment and treatment-resistant autopsy specimens
- Author
-
Naema Nayyar, Elizabeth R. Gerstner, Andrew S. Chi, Corey M. Gill, Mario L. Suvà, Daniel Rosebrock, Ugonma Chukwueke, Gad Getz, Daniel P. Cahill, Elisa Aquilanti, Andrew Kaneb, Dimitri Livitz, Ignaty Leshchiner, Mathew P. Frosch, Mia Bertalan, Scott R. Plotkin, Kaitlin Hoang, Priscilla K. Brastianos, Megan R. D'Andrea, and Tracy T. Batchelor
- Subjects
0301 basic medicine ,Chromosome 7 (human) ,Genetics ,Cancer Research ,education.field_of_study ,IDH1 ,Phylogenetic tree ,Population ,Cancer ,Biology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Somatic evolution in cancer ,lcsh:RC254-282 ,Article ,Human genetics ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,education ,Exome sequencing - Abstract
Glioblastomas are malignant neoplasms composed of diverse cell populations. This intratumoral diversity has an underlying architecture, with a hierarchical relationship through clonal evolution from a common ancestor. Therapies are limited by emergence of resistant subclones from this phylogenetic reservoir. To characterize this clonal ancestral origin of recurrent tumors, we determined phylogenetic relationships using whole exome sequencing of pre-treatment IDH1/2 wild-type glioblastoma specimens, matched to post-treatment autopsy samples (n = 9) and metastatic extracranial post-treatment autopsy samples (n = 3). We identified “truncal” genetic events common to the evolutionary ancestry of the initial specimen and later recurrences, thereby inferring the identity of the precursor cell population. Mutations were identified in a subset of cases in known glioblastoma genes such as NF1(n = 3), TP53(n = 4) and EGFR(n = 5). However, by phylogenetic analysis, there were no protein-coding mutations as recurrent truncal events across the majority of cases. In contrast, whole copy-loss of chromosome 10 (12 of 12 cases), copy-loss of chromosome 9p21 (11 of 12 cases) and copy-gain in chromosome 7 (10 of 12 cases) were identified as shared events in the majority of cases. Strikingly, mutations in the TERT promoter were also identified as shared events in all evaluated pairs (9 of 9). Thus, we define four truncal non-coding genomic alterations that represent early genomic events in gliomagenesis, that identify the persistent cellular reservoir from which glioblastoma recurrences emerge. Therapies to target these key early genomic events are needed. These findings offer an evolutionary explanation for why precision therapies that target protein-coding mutations lack efficacy in GBM., Brain cancer: Non-coding genomic changes fuel glioblastoma growth Non-coding and structural alterations may be early drivers of brain cancer development. A team led by Priscilla Brastianos and Tracy Batchelor from Massachusetts General Hospital, Boston, USA, analyzed the genetic landscape of glioblastoma by comparing pre-treatment and autopsy tumor specimens from 12 patients who died of the aggressive brain cancer. They identified a common set of four genetic events that occurred early in the evolution of nearly every patient’s cancer: three losses or gains of chromosome regions or entire chromosomes, and mutations in the gene-activating promoter of TERT, which encodes an enzyme implicated in the cancer’s growth. The findings help explain why therapies that target protein-coding mutations don’t work in brain cancer when they do in other tumor types. They also point to new drug targets.
- Published
- 2017
36. The Alkylating Chemotherapeutic Temozolomide Induces Metabolic Stress in IDH1-Mutant Cancers and Potentiates NAD+ Depletion–Mediated Cytotoxicity
- Author
-
Kensuke Tateishi, Tracy T. Batchelor, Ganesh M. Shankar, Shota Tanaka, A. John Iafrate, Julie J. Miller, Mara V.A. Koerner, Fumi Higuchi, Andrew S. Chi, Nina Lelic, Daniel P. Cahill, Hiroaki Wakimoto, and David E. Fisher
- Subjects
0301 basic medicine ,Cancer Research ,Temozolomide ,IDH1 ,DNA damage ,Poly ADP ribose polymerase ,Nicotinamide phosphoribosyltransferase ,Biology ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Isocitrate dehydrogenase ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,medicine ,NAD+ kinase ,medicine.drug - Abstract
IDH1-mutant gliomas are dependent upon the canonical coenzyme NAD+ for survival. It is known that PARP activation consumes NAD+ during base excision repair (BER) of chemotherapy-induced DNA damage. We therefore hypothesized that a strategy combining NAD+ biosynthesis inhibitors with the alkylating chemotherapeutic agent temozolomide could potentiate NAD+ depletion–mediated cytotoxicity in mutant IDH1 cancer cells. To investigate the impact of temozolomide on NAD+ metabolism, patient-derived xenografts and engineered mutant IDH1-expressing cell lines were exposed to temozolomide, in vitro and in vivo, both alone and in combination with nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, which block NAD+ biosynthesis. The acute time period (
- Published
- 2017
37. A phase 2 study of the first imipridone ONC201, a selective DRD2 antagonist for oncology, administered every three weeks in recurrent glioblastoma
- Author
-
Patrick Y. Wen, Wolfgang Oster, Tracy T. Batchelor, Isabel Arrillaga-Romany, Joshua E. Allen, and Andrew S. Chi
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Phases of clinical research ,Imipridone ,Lesion ,03 medical and health sciences ,GPCR ,Glioma ,Internal medicine ,glioma ,medicine ,Surrogate endpoint ,business.industry ,Antagonist ,glioblastoma ,Cancer ,ONC201 ,medicine.disease ,3. Good health ,Discontinuation ,030104 developmental biology ,Every Three Weeks ,medicine.symptom ,Clinical Research Paper ,business - Abstract
// Isabel Arrillaga-Romany 1 , Andrew S. Chi 3 , Joshua E. Allen 4 , Wolfgang Oster 4 , Patrick Y. Wen 2 and Tracy T. Batchelor 1 1 Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, MA, USA 2 Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA, USA 3 Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, USA 4 Oncoceutics, Philadelphia, PA, USA Correspondence to: Isabel Arrillaga-Romany, email: // Keywords : ONC201, Imipridone, GPCR, glioblastoma, glioma Received : April 25, 2017 Accepted : April 28, 2017 Published : May 12, 2017 Abstract ONC201 is an oral, small molecule selective antagonist of the G protein-coupled receptor DRD2 that causes p53-independent apoptosis in tumor cells via integrated stress response activation and Akt/ERK inactivation. We performed a Phase II study that enrolled 17 patients with recurrent, bevacizumab-naive, IDH1/2 WT glioblastoma who received 625mg ONC201 every three weeks. Median OS was 41.6 weeks with OS6 of 71% and OS9 of 53%. Seven of 17 patients are alive. PFS6 was 11.8% with two patients remaining on study who continue to receive ONC201 for >12 months. One of these patients had a durable objective response with a secondary glioblastoma possessing a H3.3 K27M mutation, exhibiting regression by 85% in one lesion and 76% in the second lesion. The second patient who continues to receive ONC201 for >12 months remains disease-free after enrolling on this trial following a re-resection. No drug-related SAEs or treatment discontinuation due to toxicity occurred. Plasma PK at 2 hours post-dose was 2.6 ug/mL, serum prolactin induction was observed as a surrogate marker of target engagement, and DRD2 was expressed in all evaluated archival tumor specimens. In summary, ONC201 is well tolerated and may have single agent activity in recurrent glioblastoma patients.
- Published
- 2017
38. Phase I trial of aflibercept (VEGF trap) with radiation therapy and concomitant and adjuvant temozolomide in patients with high-grade gliomas
- Author
-
Patrick Y. Wen, Susan M. Chang, Alfred Yung, Tracy T. Batchelor, Stuart A. Grossman, Lakshmi Nayak, Alice P. Chen, Timothy F. Cloughesy, Frank S. Lieberman, Antonio Omuro, Xiaobu Ye, Joy D. Fisher, Mark R. Gilbert, Lisa M. DeAngelis, Jeffrey S. Wefel, Jan Drappatz, Michael D. Prados, John de Groot, and Kenneth Aldape
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_treatment ,Neuropsychological Tests ,0302 clinical medicine ,Receptors ,Adjuvant ,Cancer ,Aflibercept ,Brain Neoplasms ,Vascular Endothelial Growth Factor ,Hematology ,Glioma ,Middle Aged ,Alkylating ,Combined Modality Therapy ,Dacarbazine ,Treatment Outcome ,Neurology ,Chemotherapy, Adjuvant ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Combination ,Drug Therapy, Combination ,Female ,VEGF trap ,medicine.drug ,Adult ,medicine.medical_specialty ,Recombinant Fusion Proteins ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Antineoplastic Agents ,Neutropenia ,Article ,Vaccine Related ,03 medical and health sciences ,Rare Diseases ,Drug Therapy ,Clinical Research ,Internal medicine ,Temozolomide ,medicine ,Chemotherapy ,Humans ,Oncology & Carcinogenesis ,Antineoplastic Agents, Alkylating ,Aged ,business.industry ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,Newly diagnosed glioblastoma ,medicine.disease ,Brain Disorders ,Surgery ,Brain Cancer ,Radiation therapy ,Regimen ,Orphan Drug ,Receptors, Vascular Endothelial Growth Factor ,030104 developmental biology ,Concomitant ,Dose-dense ,Neurology (clinical) ,business - Abstract
Anti-vascular endothelial growth factor (VEGF) therapy has shown promise in the treatment of high-grade gliomas (HGG). Aflibercept is a recombinant human fusion protein that acts as a soluble decoy receptor for VEGF-A, VEGF-B and placental growth factor, depleting circulating levels of these growth factors. The Adult Brain Tumor Consortium conducted a phase I trial of aflibercept and temozolomide (TMZ) in patients with newly diagnosed HGG with 2 dose levels and a 3+3 design. Three arms using aflibercept were examined; with radiation and concomitant temozolomide; with adjuvant temozolomide using the 5/28 regimen; and with adjuvant temozolomide using the 21/28day regimen. Fifty-nine patients were enrolled, 21 in arm 1, 20 in arm 2 and 18 in arm 3. Median age was 56 years (24-69); median KPS 90 (60-100). The maximum tolerated dose (MTD) of aflibercept for all 3 arms was 4mg/kg every 2 weeks. Dose limiting toxicities at the MTD were: Arm 1: 0/21 patients; Arm 2: 2/20 patients (G3 deep vein thrombosis, G4 neutropenia; Arm 3: 3/18 patients) (G4 biopsy-confirmed thrombotic microangiopathy, G3 rash, G4 thrombocytopenia). The median number of cycles of aflibercept was 5 (range, 1-16). All patients stopped treatment; 28 (47%) for disease progression, 21 (36%) for toxicities, 8 (14%) for other reasons, and 2 (3%) patients completed the full treatment course. This study met its primary endpoint and the MTD of aflibercept with radiation and concomitant and adjuvant temozolomide is 4mg/kg every 2 weeks.
- Published
- 2017
39. Radiation Therapy for Glioblastoma: American Society of Clinical Oncology Clinical Practice Guideline Endorsement of the American Society for Radiation Oncology Guideline
- Author
-
Matthew Mumber, Vinai Gondi, Mark R. Gilbert, Terri S. Armstrong, Nofisat Ismaila, Mary Lovely, Andrew E. Sloan, Minesh P. Mehta, Timothy F. Cloughesy, Susan M. Chang, Evanthia Galanis, Tracy T. Batchelor, Erik P. Sulman, and Christina Tsien
- Subjects
Cancer Research ,medicine.medical_specialty ,Consensus ,Biopsy ,medicine.medical_treatment ,MEDLINE ,03 medical and health sciences ,0302 clinical medicine ,Quality of life ,Temozolomide ,Humans ,Medicine ,Medical physics ,Antineoplastic Agents, Alkylating ,Evidence-Based Medicine ,Performance status ,Brain Neoplasms ,business.industry ,Patient Selection ,Chemoradiotherapy ,Guideline ,Evidence-based medicine ,Dacarbazine ,Radiation therapy ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Dose Fractionation, Radiation ,Cranial Irradiation ,Glioblastoma ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Purpose The American Society for Radiation Oncology (ASTRO) produced an evidence-based guideline on radiation therapy for glioblastoma. Because of its relevance to the ASCO membership, ASCO reviewed the guideline and applied a set of procedures and policies used to critically examine guidelines developed by other organizations. Methods The ASTRO guideline on radiation therapy for glioblastoma was reviewed for developmental rigor by methodologists. An ASCO endorsement panel updated the literature search and reviewed the content and recommendations. Results The ASCO endorsement panel determined that the recommendations from the ASTRO guideline, published in 2016, are clear, thorough, and based on current scientific evidence. ASCO endorsed the ASTRO guideline on radiation therapy for glioblastoma and added qualifying statements. Recommendations Partial-brain fractionated radiotherapy with concurrent and adjuvant temozolomide is the standard of care after biopsy or resection of newly diagnosed glioblastoma in patients up to 70 years of age. Hypofractionated radiotherapy for elderly patients with fair to good performance status is appropriate. The addition of concurrent and adjuvant temozolomide to hypofractionated radiotherapy seems to be safe and efficacious without impairing quality of life for elderly patients with good performance status. Reasonable options for patients with poor performance status include hypofractionated radiotherapy alone, temozolomide alone, or best supportive care. Focal reirradiation represents an option for select patients with recurrent glioblastoma, although this is not supported by prospective randomized evidence. Additional information is available at www.asco.org/glioblastoma-radiotherapy-endorsement and www.asco.org/guidelineswiki .
- Published
- 2017
40. New Directions in Anti-Angiogenic Therapy for Glioblastoma
- Author
-
Nancy Wang, Tracy T. Batchelor, and Rakesh K. Jain
- Subjects
Vascular Endothelial Growth Factor A ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Bevacizumab ,Angiogenesis ,Angiogenesis Inhibitors ,Review ,Pharmacology ,Metastasis ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Randomized controlled trial ,law ,Glioma ,Internal medicine ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Progression-free survival ,Clinical Trials as Topic ,Neovascularization, Pathologic ,Brain Neoplasms ,business.industry ,medicine.disease ,Vascular endothelial growth factor ,Clinical trial ,Treatment Outcome ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Neurology (clinical) ,Glioblastoma ,business ,medicine.drug - Abstract
Anti-angiogenic therapy has become an important component in the treatment of many solid tumors given the importance of adequate blood supply for tumor growth and metastasis. Despite promising preclinical data and early clinical trials, anti-angiogenic agents have failed to show a survival benefit in randomized controlled trials of patients with glioblastoma. In particular, agents targeting vascular endothelial growth factor (VEGF) appear to prolong progression free survival, possibly improve quality of life, and decrease steroid usage, yet the trials to date have demonstrated no extension of overall survival. In order to improve duration of response and convey a survival benefit, additional research is still needed to explore alternative pro-angiogenic pathways, mechanisms of resistance, combination strategies, and biomarkers to predict therapeutic response.
- Published
- 2017
41. CTNI-17. CLINICAL EFFICACY AND PREDICTIVE BIOMARKERS OF ONC201 IN H3 K27M-MUTANT DIFFUSE MIDLINE GLIOMA
- Author
-
Pankaj Vats, Alyssa Paul, Yoshie Umemura, Timothy F. Cloughesy, Chase Thomas, Nicole Shonka, Daniel Martinez, Isabel Arrillaga-Romany, Ian Wolfe, Benjamin M. Ellingson, Arul M. Chinnaiyan, Evan Cantor, Zachary Miklja, John de Groot, Abed Rahman Kawakibi, Andrew S. Chi, Chandan Kumar-Sinha, Rebecca Harrison, Adam C. Resnick, Nicholas Butowski, Guangrong Lu, Minesh P. Mehta, Joshua E. Allen, Ashley Sumrall, Matthew Hall, Rajen Mody, Carl Koschmann, Mariella G. Filbin, Ruby Siada, Sylvia Kurz, Rohinton Tarapore, Sebastian M Waszak, Doured Daghistani, Hugh J. L. Garton, Viveka Nand Yadav, Javad Nazarian, Amy K. Bruzek, Patrick Y. Wen, Sabine Mueller, Tracy T. Batchelor, Sharon Gardner, Jessica R. Cummings, Sriram Venneti, Jonathan Schwartz, Brendan Mullan, Patricia L. Robertson, Li Jiang, Andrea Franson, Yazmin Odia, and Rodrigo Cartaxo
- Subjects
Drd2 gene ,Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,Mutant ,Clinical Trials: Non-Immunologic ,medicine.disease ,Oncology ,Circulating tumor DNA ,Glioma ,Troponin I ,medicine ,Neurology (clinical) ,Clinical efficacy ,business ,Predictive biomarker - Abstract
Patients with diffuse midline glioma (DMG) harboring H3 K27M mutation rarely survive longer than two years and have no proven therapies following first-line radiation. ONC201, a bitopic DRD2 antagonist and allosteric ClpP agonist, has shown encouraging efficacy in early phase studies in H3 K27M-mutant DMG. In order to define response rates in H3 K27M DMG patients and to clarify the genomic, anatomic and molecular predictors of response, we performed an integrated pre-clinical and clinical analysis of ONC201 treatment. ONC201 was effective in intra-uterine electroporation (IUE)-generated H3 K27M-mutant murine glioma models with excellent CNS penetration and survival benefit. Patients with H3 K27M-mutant DMG treated with ONC201 on active clinical trials (n=50, 27 thalamic, 23 brainstem) showed an overall survival (OS) of 28.1 (range: 5.9–105) months from diagnosis (enrollment by 4/29/19, data cut-off 12/28/19), compared to historical median OS of 12 months. Median OS for non-recurrent patients has not been reached (n=16, median follow-up: 16.8 from diagnosis). For non-recurrent thalamic patients (n=8), median PFS is 20.1 (range: 9.3–27.6) months from diagnosis (median time on drug: 14.5 months). Best response for thalamic patients by RANO: 1 CR, 5 PR, 7 SD, 8 PD, 6 not reported. Decreased H3 K27M cell-free tumor DNA in plasma and CSF at 6 months correlated with long-term response. Baseline tumor gene expression profiling in patients treated with ONC201 (n=14) identified EGFR and the cortical developmental transcription factor FOXG1 as the strongest biomarkers of radiographic response to ONC201. Analysis of 541 ONC201-treated human cancer cell lines from DepMap, provided evidence for an EGFR-dependent ONC201 resistance mechanism. Analysis of 38 glioma cell lines further supports FOXG1 as a glioma-specific predictive biomarker of ONC201 response. The unprecedented survival results and radiographic responses to ONC201 in H3K27M DMG make a compelling case for later phase and combinatorial studies.
- Published
- 2020
42. CTNI-37. EFFICACY OF ONC201 IN PATIENTS WITH ONC201 FOR RECURRENT H3 K27M-MUTANT DIFFUSE MIDLINE GLIOMA
- Author
-
Rebecca Harrison, Tracy T. Batchelor, Guangrong Lu, Andrew B. Lassman, Patrick Y. Wen, Rohinton Tarapore, Minesh P. Mehta, Lindsay Kilburn, John de Groot, Yoshie Umemura, Timothy F. Cloughesy, Karan Dixit, Nicole Shonka, Andrew S. Chi, Jerome J. Graber, Joshua E. Allen, Isabel Arrillaga-Romany, Nicholas Butowski, Lynne Taylor, Phioanh Nghiemphu, Yazmin Odia, Ashley Sumrall, and Sylvia Kurz
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,Thalamus ,Clinical Trials: Non-Immunologic ,Fluid-attenuated inversion recovery ,Spinal cord ,medicine.disease ,medicine.anatomical_structure ,Cerebrospinal fluid ,Oncology ,Glioma ,Troponin I ,medicine ,Neurology (clinical) ,Progression-free survival ,business ,Dexamethasone ,medicine.drug - Abstract
H3 K27M-mutant diffuse midline gliomas (DMG) have a dismal prognosis. We report an integrated analysis for ONC201, a DRD2 antagonist and ClpP agonist, in patients with recurrent H3 K27M DMG administered as monotherapy in 3 clinical studies (NCT03295396, n=20; NCT02525692, n=6; or expanded access, n=4) by independent, central radiology review among 30 patients. At baseline, patients had measurable contrast-enhancing disease by RANO criteria, KPS>60, and were >90 days from prior radiation. Patients with primary lesions that involved the pons or spinal cord or had evidence of leptomeningeal or cerebrospinal fluid dissemination were excluded. ONC201 was orally administered at 625 mg (scaled by body weight for patients < 18 years old) weekly in 29 patients and every 3 weeks in 1 patient. Median age was 31 years (range 8–70; two patients < 18). There were 16 women and 14 men. Tumors were predominantly thalamic (73%), with other locations including the cerebellum (5%), brainstem (non-pontine) (3%), basal ganglia (3%), and midbrain (3%). Median time from prior radiation was 7.5 months. The most frequent drug-related adverse events were low grade nausea (10%) and fatigue (10%). Nine patients (30%) had >50% regression of T1 contrast enhancement and 11 (36.7%) patients had regression of T2/FLAIR. Objective response rate and progression-free survival by RANO criteria, as well overall survival will be reported. Among 8 patients with sustained radiographic regressions, 6 were tapered off dexamethasone and 4 had improvement in KPS. Five patients (16.7%) remain on treatment with a median of 9.5 months (range 7.3–12). In conclusion, single agent ONC201 is well tolerated and clinically active in recurrent H3 K27M DMG patients.
- Published
- 2020
43. Diagnosis and treatment of epidural metastases
- Author
-
Julie J. Miller, Tracy T. Batchelor, and M. Loreto Yáñez
- Subjects
Cancer Research ,medicine.medical_specialty ,Weakness ,Cord ,business.industry ,Cancer ,Cauda equina ,medicine.disease ,Spinal cord ,Surgery ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Spinal cord compression ,030220 oncology & carcinogenesis ,Epidemiology ,medicine ,Paralysis ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Epidural metastases occur in 5% to 10% of cancer patients and represent a neurological emergency. Patients most commonly present with an acute onset of motor weakness, and restoration of neurological function is critically dependent on prompt diagnosis and treatment. This review discusses the clinical, epidemiological, and radiological features associated with epidural metastases and resulting spinal cord compression. Moreover, current treatment paradigms are reviewed. The timely initiation of radiation as well as surgery in select cases is critical for preserving neurological function and achieving local tumor control and pain control. Future studies investigating surgical and radiation treatment for metastatic epidural cord compression are urgently needed. Cancer 2017;123:1106-1114. © 2016 American Cancer Society.
- Published
- 2016
44. Standard chemoradiation in combination with VEGF targeted therapy for glioblastoma results in progressive gray and white matter volume loss
- Author
-
Pavlina Polaskova, Morgan L Prust, Tracy T. Batchelor, Elizabeth R. Gerstner, Jayashree Kalpathy-Cramer, Kourosh Jafari-Khouzani, and Jorg Dietrich
- Subjects
Adult ,Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,VEGF receptors ,Gray (unit) ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Neuroimaging ,White matter volume loss ,Internal medicine ,medicine ,Humans ,Gray Matter ,Letter to the Editor ,Aged ,Aged, 80 and over ,biology ,business.industry ,Neurotoxicity ,Chemoradiotherapy ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,White Matter ,030104 developmental biology ,biology.protein ,Female ,Neurology (clinical) ,Glioblastoma ,business ,030217 neurology & neurosurgery - Published
- 2018
45. ACTR-61. A RANDOMIZED PHASE 2 TRIAL OF CEDIRANIB IN COMBINATION WITH OLAPARIB VERSUS BEVACIZUMAB IN PATIENTS WITH RECURRENT GLIOBLASTOMA
- Author
-
Andrew B. Nixon, Douglas E. Ney, Jan Drappatz, Alona Muzikansky, S. Percy Ivy, Mary Welch, Biswajit Das, Isabel Arrillaga-Romany, P. Mickey Williams, John L. Villano, Eudocia Q. Lee, Elizabeth M. Swisher, Robert Aiken, Pierre Giglio, Chris Karlovich, Elizabeth R. Gerstner, Tracy T. Batchelor, David Picconi, Benjamin M. Ellingson, Jian Campian, Kevin P. Becker, and Solmaz Sahebjam
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,Bevacizumab ,business.industry ,Surrogate endpoint ,Phases of clinical research ,Olaparib ,Cediranib ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Adult Clinical Trials - Non-Immunologic ,Medicine ,In patient ,Neurology (clinical) ,Progression-free survival ,business ,medicine.drug - Abstract
BACKGROUND Like most proliferating tumors, GBM relies heavily on accurate DNA repair for maintenance of genome stability. Dysfunction in repair of both single and double strand DNA breaks by PARP inhibition and impairment of homologous recombination, respectively, would be synthetically lethal. In this study we combined the PARP inhibitor olaparib with cediranib, a pan VEGF receptor inhibitor. Cediranib may mediate disruption in the homologous recombination pathway through its antiangiogenic properties. METHODS Through the Experimental Therapeutics Clinical Trials Network, we performed an open-label randomized phase II study of bevacizumab (BEV)- naive adult patients with first or second recurrence of glioblastoma after radiation and temozolomide. Patients were randomized 1:1 to receive either olaparib 200 mg by mouth twice daily with cediranib 30 mg by mouth daily or BEV 10 mg/kg IV every 2 weeks. The primary endpoint was progression-free survival at 6 months (PFS6). Secondary endpoints included safety and overall survival. Exploratory objectives included blood, tissue and imaging-based biomarkers of response to treatment. RESULTS Seventy patients were enrolled. Median age was 60.5 years (range: 19–79), 39% females, median KPS was 90 (range: 60–100). Baseline characteristics were well balanced. With a data cut-off of 5/2/2019, PFS6 was 14% [95% CI 4–30%] in the cediranib/olaparib arm vs 30.9% [95% CI 12.7–51.2%] in the BEV arm. Median OS was 247 days in the cediranib/olaparib arm vs 201 days in the BEV arm, HR 0.816, 95% CI (0.431, 1.546). Related grade 3, 4 or 5 toxicity was experienced in 29% vs 12% of patients for the cediranib/olaparib vs BEV arm. CONCLUSION Treatment with cediranib/olaparib failed to increase PFS and OS in patients with recurrent GBM. Blood, tissue and imaging correlates will be presented to help understand why this treatment combination was unsuccessful.
- Published
- 2019
46. GENE-63. GENOMIC CHARACTERIZATION OF HUMAN BRAIN METASTASES IDENTIFIES NOVEL DRIVERS OF LUNG ADENOCARCINOMA PROGRESSION
- Author
-
Juan Carlos Martinez-Gutierrez, Michael White, Elizabeth R. Gerstner, Corey M. Gill, Darrell R. Borger, Benjamin Kaufman, Kaitlin Hoang, Scott L. Carter, Ibiayi Dagogo-Jack, Naema Nayyar, Sandro Santagata, Daniel P. Cahill, Tracy T. Batchelor, Matthias Preusser, Megan R. D'Andrea, Ivanna Bihun, Franziska M. Ippen, Priscilla K. Brastianos, A. John Iafrate, Maria Martinez-Lage, Emily Batchelor, Devin McCabe, Ryan P. Frazier, Anna S. Berghoff, Matthew Lastrapes, Parker H. Merrill, Matthew R. Strickland, Christopher Alvarez-Breckenridge, Sung Hye Park, Deepika Nagabhushan, Mia Bertalan, Sun Ha Paek, Nicholas D. Camarda, Elisa Aquilanti, David Shih, Andrew Kaneb, Benjamin M. Kuter, Bruce E. Johnson, Alexander Kaplan, Ugonma Chukwueke, Brandyn A. Castro, Matthew P. Frosch, and Magali De Sauvage
- Subjects
Genetics and Epigenetics ,Cancer Research ,Lung ,Tumor cells ,Human brain ,Biology ,medicine.disease ,Genome ,Intracardiac injection ,medicine.anatomical_structure ,Oncology ,Cancer research ,medicine ,Adenocarcinoma ,Neurology (clinical) ,Gene ,Exome sequencing - Abstract
Although lung adenocarcinomas frequently metastasize to the brain, treatment options for lung adenocarcinoma brain metastases are limited. We discovered novel candidate drivers of progression by using case-control analyses to compare whole-exome sequencing data from a cohort of 73 lung adenocarcinoma brain metastases to a control cohort of 503 primary lung adenocarcinomas. We identified 3 genomic regions with significantly more frequent amplifications in brain metastases compared to the control cohort: MYC (12% vs 6%), YAP1 (7% vs 0.8%) and MMP13 (10% vs 0.6%). We also identified CDKN2A/B as a region deleted at a significantly greater frequency in brain metastases compared to primary lung adenocarcinomas (27% vs 13%, respectively). We confirmed frequent amplifications of MYC and YAP1/MMP13 in an independent validation cohort of 105 lung adenocarcinoma brain metastasis samples using fluorescence in situ hybridization. We further validated that MYC, YAP1 and MMP13 can drive brain metastases in a patient-derived xenograft mouse model. We found a higher incidence of metastases to the brain in mice receiving intracardiac injections of tumor cells expressing the candidate drivers compared to tumor cells expressing LacZ as a control. These results indicate that somatic alterations can drive lung adenocarcinomas to metastasize to the brain. The candidate brain metastasis drivers that we identified may serve as therapeutic targets in patients with lung adenocarcinomas who develop this devastating complication.
- Published
- 2019
47. Primary dural lymphomas: Clinical presentation, management, and outcome
- Author
-
Justin T. Jordan, Joerg-Christian Tonn, Philipp Karschnia, Joachim M. Baehring, Frank J Barbiero, Robert K. Fulbright, Leon D. Kaulen, Jay S. Loeffler, Anita Huttner, Jorg Dietrich, Sebastian F Winter, Niklas Thon, Brian A. Shaw, and Tracy T. Batchelor
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Lymphoma ,Dura mater ,medicine.medical_treatment ,Follicular lymphoma ,Neuroimaging ,Kaplan-Meier Estimate ,Radiosurgery ,Meningioma ,Central Nervous System Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,030212 general & internal medicine ,Aged ,Chemotherapy ,business.industry ,Cerebrospinal Fluid Proteins ,Middle Aged ,medicine.disease ,Prognosis ,Magnetic Resonance Imaging ,Radiation therapy ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Female ,Radiology ,Dura Mater ,business - Abstract
Background Clinical experience is limited for primary central nervous system (CNS) lymphoma that arises from the dura mater, which is denoted with the term primary dural lymphoma (PDL). This study was aimed at determining the relative incidence, presentation, and outcomes of PDL. Methods The institutional databases of the Divisions of Neuro-Oncology at the Massachusetts General Hospital and the Yale School of Medicine were retrospectively searched for patients with primary CNS lymphoma. Patients with pathologically confirmed dural lymphoma and no evidence of primary cerebral or systemic involvement were identified. Clinical data, diagnostic findings, treatments, and outcomes were recorded. Results A total of 20 patients with PDL were identified, and they represented 6.3% of the individuals with primary CNS lymphomas (20 of 316). Histopathological examination of PDL revealed the following underlying subtypes: diffuse large B-cell lymphoma (10 of 20 patients), marginal zone lymphoma (6 of 20), follicular lymphoma (2 of 20), undefined B-cell non-Hodgkin lymphoma (1 of 20), and T-cell non-Hodgkin lymphoma (1 of 20). On imaging, all tumors appeared as extra-axial masses with avid contrast enhancement and mostly mimicked meningioma. The median apparent diffusion coefficient value was 667 ± 26 mm2 /s. Cerebrospinal fluid analyses and symptoms were nonspecific, and the diagnosis rested on tissue analysis. Therapeutic approaches included surgery, radiotherapy, and chemotherapy. The median overall survival was not reached after 5 years. Three patients were deceased at database closure because of tumor progression. The extent of tumor resection correlated positively with overall survival (P = .044). Conclusions PDL is a rare variant of primary CNS lymphoma that can be radiographically mistaken for meningioma. The outcome is excellent with multimodality treatment, and aggressive surgery may convey a survival advantage in select cases.
- Published
- 2019
48. OS12.4 In vivo dynamics and targeting of vessel co-option in glioma
- Author
-
Tracy T. Batchelor, Shanmugarajan Krishnan, Rakesh K. Jain, Manish K. Aghi, David H. Rowitch, Emmanuelle Huillard, Samuel J. Shelton, Amelie Griveau, Jonas Kloepper, Anat Stemmer-Rachamimov, Giorgio Seano, and Nancy Wang
- Subjects
Cancer Research ,Oncology ,In vivo ,Chemistry ,Glioma ,Dynamics (mechanics) ,Cancer research ,medicine ,Oral Presentations ,Neurology (clinical) ,medicine.disease - Abstract
BACKGROUND Gliomas comprise heterogeneous malignant glial and stromal cells. While blood vessel co-option is a potential mechanism to escape anti-angiogenic therapy, the relevance of glial phenotype in this process is unclear. MATERIAL AND METHODS Here, we intravitally study preclinical syngenetic models of glioma as well as patient-derived cells transplanted orthotopically. Moreover, we profoundly confirm our preclinical results with histological studies on patient specimens. RESULTS We show that Olig2+ oligodendrocyte precursor-like glioma cells invade by single-cell vessel co-option and preserve the blood-brain barrier (BBB). Conversely, Olig2-negative glioma cells form dense perivascular collections and promote angiogenesis and BBB breakdown, leading to innate immune cell activation. Experimentally, Olig2 promotes Wnt7b expression, a finding that correlates in human glioma profiling. Targeted Wnt7a/7b deletion or pharmacologic Wnt inhibition blocks Olig2+ glioma single-cell vessel co-option and enhances responses to temozolomide. Finally, Olig2 and Wnt7 become upregulated after anti-VEGF treatment in preclinical models and patients. CONCLUSION Here, we show that glioma is able to employ vessel co-option, i.e. the movement of tumor cells towards and along the pre-existing vasculature. Glioma oligodendrocyte-like (OPCL) cells express Wnt7 that is necessary for vessel co-option and Wnt inhibitors significantly improve survival with temozolomide. Moreover, we demonstrated that anti-VEGF-treatment of glioma selects for Olig2/Wnt7+ cells
- Published
- 2019
49. Primary central nervous system lymphoma: A curable disease
- Author
-
Tracy T. Batchelor
- Subjects
Cancer Research ,Lymphoma ,medicine.medical_treatment ,Disease ,Multimodal Imaging ,Central Nervous System Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Autologous stem-cell transplantation ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Humans ,Chemotherapy ,business.industry ,Incidence ,Neurotoxicity ,Primary central nervous system lymphoma ,Induction chemotherapy ,Disease Management ,Hematology ,General Medicine ,medicine.disease ,Prognosis ,Combined Modality Therapy ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Practice Guidelines as Topic ,Cancer research ,Methotrexate ,business ,030215 immunology ,medicine.drug - Abstract
Primary central nervous system lymphoma is a rare subtype of non-Hodgkin lymphoma that is confined to the brain, leptomeninges, or the eye and is associated with a relatively poor prognosis compared to other extranodal diffuse large B-cell lymphomas. However, methotrexate-based induction chemotherapy followed by consolidative chemotherapy or high-dose therapy and autologous stem cell transplantation is associated with improved survival and reduced neurotoxicity. Aberrant activation of B-cell receptor signaling and activation of nuclear factor kappa beta is a frequent genetic alteration and offers opportunities for targeted therapies in this lymphoma subtype.
- Published
- 2019
50. Publisher Correction: Probing tumor microenvironment in patients with newly diagnosed glioblastoma during chemoradiation and adjuvant temozolomide with functional MRI
- Author
-
Kyrre E. Emblem, Gregory A. Sorensen, Dan G. Duda, Yangming Ou, Jayashree Kalpathy-Cramer, Elizabeth R. Gerstner, Andrew S. Chi, Rebecca A. Betensky, Bella Vakulenko-Lagun, Bruce R. Rosen, Rakesh K. Jain, Xiao Da, Scott R. Plotkin, K. Ina Ly, and Tracy T. Batchelor
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,lcsh:Medicine ,Contrast Media ,Newly diagnosed ,Text mining ,Internal medicine ,medicine ,Temozolomide ,Tumor Microenvironment ,Cerebral Blood Volume ,Humans ,In patient ,Prospective Studies ,lcsh:Science ,Antineoplastic Agents, Alkylating ,Aged ,Tumor microenvironment ,Multidisciplinary ,business.industry ,lcsh:R ,Chemoradiotherapy ,Middle Aged ,medicine.disease ,Prognosis ,Publisher Correction ,Survival Rate ,Diffusion Magnetic Resonance Imaging ,Chemotherapy, Adjuvant ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,lcsh:Q ,Female ,business ,Glioblastoma ,Adjuvant ,Biomarkers ,medicine.drug ,Follow-Up Studies - Abstract
Functional MRI may identify critical windows of opportunity for drug delivery and distinguish between early treatment responders and non-responders. Using diffusion-weighted, dynamic contrast-enhanced, and dynamic susceptibility contrast MRI, as well as pro-angiogenic and pro-inflammatory blood markers, we prospectively studied the physiologic tumor-related changes in fourteen newly diagnosed glioblastoma patients during standard therapy. 153 MRI scans and blood collection were performed before chemoradiation (baseline), weekly during chemoradiation (week 1-6), monthly before each cycle of adjuvant temozolomide (pre-C1-C6), and after cycle 6. The apparent diffusion coefficient, volume transfer coefficient (K
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.