Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Blood ; 133(5): 436-445, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30567753

ABSTRACT

Ibrutinib is a first-in-class inhibitor of Bruton tyrosine kinase (BTK) and has shown single-agent activity in recurrent/refractory central nervous system (CNS) lymphoma. Clinical responses are often transient or incomplete, suggesting a need for a combination therapy approach. We conducted a phase 1b clinical trial to explore the sequential combination of ibrutinib (560 or 840 mg daily dosing) with high-dose methotrexate (HD-MTX) and rituximab in patients with CNS lymphoma (CNSL). HD-MTX was given at 3.5 g/m2 every 2 weeks for a total of 8 doses (4 cycles; 1 cycle = 28 days). Ibrutinib was held on days of HD-MTX infusion and resumed 5 days after HD-MTX infusion or after HD-MTX clearance. Single-agent daily ibrutinib was administered continuously after completion of induction therapy until disease progression, intolerable toxicity, or death. We also explored next-generation sequencing of circulating tumor DNA (ctDNA) in cerebrospinal fluid (CSF) before and during treatment. The combination of ibrutinib, HD-MTX, and rituximab was tolerated with an acceptable safety profile (no grade 5 events, 3 grade 4 events). No dose-limiting toxicity was observed. Eleven of 15 patients proceeded to maintenance ibrutinib after completing 4 cycles of the ibrutinib/HD-MTX/rituximab combination. Clinical responses occurred in 12 of 15 patients (80%). Sustained tumor responses were associated with clearance of ctDNA from the CSF. This trial was registered at www.clinicaltrials.gov as #NCT02315326.


Subject(s)
Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/drug therapy , Lymphoma/drug therapy , Methotrexate/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Rituximab/therapeutic use , Adenine/analogs & derivatives , Adult , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Circulating Tumor DNA/genetics , Female , Humans , Lymphoma/genetics , Lymphoma/pathology , Methotrexate/adverse effects , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Piperidines , Protein Kinase Inhibitors/adverse effects , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Rituximab/adverse effects , Treatment Outcome , Young Adult
2.
Clin Cancer Res ; 25(18): 5537-5547, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31263031

ABSTRACT

PURPOSE: The genomic landscape of gliomas has been characterized and now contributes to disease classification, yet the relationship between molecular profile and disease progression and treatment response remain poorly understood.Experimental Design: We integrated prospective clinical sequencing of 1,004 primary and recurrent tumors from 923 glioma patients with clinical and treatment phenotypes. RESULTS: Thirteen percent of glioma patients harbored a pathogenic germline variant, including a subset associated with heritable genetic syndromes and variants mediating DNA repair dysfunctions (29% of the total) that were associated with somatic biallelic inactivation and mechanism-specific somatic phenotypes. In astrocytomas, genomic alterations in effectors of cell-cycle progression correlated with aggressive disease independent of IDH mutation status, arose preferentially in enhancing tumors (44% vs. 8%, P < 0.001), were associated with rapid disease progression following tumor recurrence (HR = 2.6, P = 0.02), and likely preceded the acquisition of alkylating therapy-associated somatic hypermutation. Thirty-two percent of patients harbored a potentially therapeutically actionable lesion, of whom 11% received targeted therapies. In BRAF-mutant gliomas, response to agents targeting the RAF/MEK/ERK signaling axis was influenced by the type of mutation, its clonality, and its cellular and genomic context. CONCLUSIONS: These data reveal genomic correlates of disease progression and treatment response in diverse types of glioma and highlight the potential utility of incorporating genomic information into the clinical decision-making for patients with glioma.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Genetic Variation , Genomics , Glioma/genetics , Glioma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Child , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Disease Progression , Female , Genomics/methods , Germ-Line Mutation , Glioma/diagnostic imaging , Glioma/therapy , High-Throughput Nucleotide Sequencing , Humans , Image Enhancement , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Middle Aged , Models, Biological , Mutation , Precision Medicine/methods , Prognosis , Promoter Regions, Genetic , Treatment Outcome , Tumor Suppressor Proteins/genetics , Young Adult
3.
Semin Oncol Nurs ; 34(5): 569-572, 2018 12.
Article in English | MEDLINE | ID: mdl-30409552

ABSTRACT

OBJECTIVE: To present and describe three emerging areas of glioma research: volumetric imaging analysis, molecular profiling, and the use of liquid biopsies. DATA SOURCES: Published manuscripts and textbook chapters. CONCLUSION: As new imaging and molecular technologies for the patient with glioma become main-stream, precise care can be individualized and personalized. IMPLICATIONS FOR NURSING PRACTICE: Oncology nurses caring for patients with gliomas can gain knowledge of emerging research and help increase accessibility to this technology by referring patients to clinical trials that apply this technology.


Subject(s)
Glioma/nursing , Oncology Nursing/methods , Oncology Nursing/standards , Patient-Centered Care/standards , Practice Guidelines as Topic , Precision Medicine/methods , Precision Medicine/standards , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged
4.
J Clin Oncol ; 36(17): 1702-1709, 2018 06 10.
Article in English | MEDLINE | ID: mdl-29683790

ABSTRACT

Purpose Carboxyamidotriazole orotate (CTO) is a novel oral inhibitor of non-voltage-dependent calcium channels with modulatory effects in multiple cell-signaling pathways and synergistic effects with temozolomide (TMZ) in glioblastoma (GBM) models. We conducted a phase IB study combining CTO with two standard TMZ schedules in GBM. Methods In cohort 1, patients with recurrent anaplastic gliomas or GBM received escalating doses of CTO (219 to 812.5 mg/m2 once daily or 600 mg fixed once-daily dose) combined with TMZ (150 mg/m2 5 days during each 28-day cycle). In cohort 2, patients with newly diagnosed GBM received escalating doses of CTO (219 to 481 mg/m2/d once daily) with radiotherapy and TMZ 75 mg/m2/d, followed by TMZ 150 mg to 200 mg/m2 5 days during each 28-day cycle. Results Forty-seven patients were enrolled. Treatment was well tolerated; toxicities included fatigue, constipation, nausea, and hypophosphatemia. Pharmacokinetics showed that CTO did not alter TMZ levels; therapeutic concentrations were achieved in tumor and brain. No dose-limiting toxicities were observed; the recommended phase II dose was 600 mg/d flat dose. Signals of activity in cohort 1 (n = 27) included partial (n = 6) and complete (n = 1) response, including in O6-methylguanine-DNA methyltransferase unmethylated and bevacizumab-refractory tumors. In cohort 2 (n = 15), median progression-free survival was 15 months and median overall survival was not reached (median follow-up, 28 months; 2-year overall survival, 62%). Gene sequencing disclosed a high rate of responses among EGFR-amplified tumors ( P = .005), with mechanisms of acquired resistance possibly involving mutations in mismatch-repair genes and/or downstream components TSC2, NF1, NF2, PTEN, and PIK3CA. Conclusion CTO can be combined safely with TMZ or chemoradiation in GBM and anaplastic gliomas, displaying favorable brain penetration and promising signals of activity in this difficult-to-treat population.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioma/drug therapy , Triazoles/administration & dosage , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/adverse effects , Chemoradiotherapy , Cohort Studies , Dose-Response Relationship, Drug , Female , Glioblastoma/pathology , Glioblastoma/radiotherapy , Glioma/pathology , Glioma/radiotherapy , Humans , Male , Middle Aged , Triazoles/adverse effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL