Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Oncologist ; 25(2): e198-e202, 2020 02.
Article in English | MEDLINE | ID: mdl-32043779

ABSTRACT

For pediatric patients with high-grade gliomas, standard-of-care treatment includes surgery, chemotherapy, and radiation therapy; however, most patients ultimately succumb to their disease. With advances in genomic characterization of pediatric high-grade gliomas, the use of targeted therapies in combination with current treatment modalities offer the potential to improve survival in this patient population. In this report, we present the case of a 3-year-old girl with glioblastoma who continues to experience an exceptional and durable response (>2 years) to the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib. Our patient presented with persistent and progressive seizure activity that upon workup was the result of a large heterogeneously enhancing, mixed cystic and solid mass in the left frontal-parietal-temporal region. Histopathologic analysis of resected tumor tissue confirmed the diagnosis of glioblastoma, and comprehensive genomic profiling demonstrated absence of any BRAF or H3F3A mutations. Genomic profiling, however, did reveal a probable germline heterozygous BRCA2 Lys3326Ter (K3226*) nonsense variant. After debulking surgery, the patient received standard-of-care treatment with radiation and temozolomide. Nine months later the PARP inhibitor olaparib was administered in combination with temozolomide for 16 cycles. This regimen was well tolerated by the patient and serial imaging showed reduction in tumor size. Since completion of the regimen, the patient remains neurologically intact with no evidence of tumor recurrence. To our knowledge, this represents the first case of a pediatric glioblastoma that maintains a durable response to a therapeutic strategy that included the PARP inhibitor olaparib and more generally highlights the potential clinical utility of incorporating these agents into the treatment of pediatric high-grade gliomas. KEY POINTS: Germline mutations detected in pediatric gliomas may represent a cancer predisposition syndrome. Integrating molecular testing into routine clinical care for pediatric patients with glioma is critical to identify therapeutic targets and patients with a cancer predisposition syndrome. Patients with glioma with defects in DNA repair pathway components (e.g., BRCA1/2) may show increased responsiveness to poly (ADP-ribose) polymerase (PARP) inhibitors. Combining PARP inhibitors with temozolomide (standard-of-care treatment) revealed no adverse events or toxicities over the course of 18 months.


Subject(s)
Antineoplastic Agents , Glioblastoma , Ovarian Neoplasms , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Neoplasm Recurrence, Local , Ovarian Neoplasms/drug therapy , Phthalazines/therapeutic use , Piperazines , Temozolomide/therapeutic use
2.
J Manag Care Spec Pharm ; 25(5): 601-611, 2019 May.
Article in English | MEDLINE | ID: mdl-30632889

ABSTRACT

BACKGROUND: Comprehensive genomic profiling (CGP) is a next-generation sequencing-based methodology that detects 4 classes of genomic alterations, as well as gene signature biomarkers such as microsatellite instability and tumor mutational burden. In the context of precision oncology, CGP can help to direct treatment to genomically matched therapies. OBJECTIVE: To describe the results of a 3-year observational analysis of patients undergoing testing with CGP assays (either FoundationOne or FoundationOne Heme) at a community oncology practice after a regional health plan implemented a medical policy that enabled coverage of CGP. METHODS: A retrospective analysis of medical records was completed at the oncology practice from November 2013 to January 2017; this date range was chosen to coincide with the regional health plan's medical policy implementation of CGP. The medical policy provided coverage of CGP for patients with advanced solid and hematologic cancers. A medical record review assessed all previous and current molecular test results, matched therapy or clinical trial enrollment, and clinical outcomes (clinical benefit or disease progression). The potential cost diversion, from payer to study sponsor, for patients who enrolled in clinical trials was explored. RESULTS: There were 96 patients in the community oncology practice who received CGP over the 3-year period, 86 of whom had clinically relevant genomic alterations. Of the 86, 15 patients were treated with genomically matched therapy, and 6 patients enrolled in clinical trials based on CGP results. In a subset of 32 patients who previously underwent conventional testing, most (84%) had clinically relevant genomic alterations detected by CGP that conventional testing did not identify, and a portion of these patients subsequently received treatment based on the CGP results. In the separate cost diversion analysis of 20 patients who enrolled in phase 1 clinical trials, an estimated $25,000 per-patient cost-benefit may have been accrued to the payer. CONCLUSIONS: This observational analysis characterized the use of CGP in a large community oncology practice among a group of patients insured by a regional health plan. Clinical trial enrollment was facilitated by CGP use in the community setting and may have contributed to cost diversion from the payer to study sponsors. DISCLOSURES: No separate study-related funding was provided by or to Priority Health, Foundation Medicine, and Cancer and Hematology Centers of West Michigan. Data analysis by Reitsma was conducted as part of an internship funded by Priority Health. Reitsma and Fox are employed by Priority Health. Anhorn, Vanden Borre, Cavanaugh, Chudnovsky, and Erlich are employed by Foundation Medicine.


Subject(s)
Biomarkers, Tumor/genetics , Community Health Services/organization & administration , Intersectoral Collaboration , Neoplasms/genetics , Public-Private Sector Partnerships/organization & administration , Adult , Aged , Aged, 80 and over , Female , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Insurance, Health/organization & administration , Male , Medical Oncology/organization & administration , Middle Aged , Mutation , Neoplasms/therapy , Precision Medicine/methods , Retrospective Studies , Stakeholder Participation
3.
Gynecol Oncol ; 147(3): 626-633, 2017 12.
Article in English | MEDLINE | ID: mdl-29102090

ABSTRACT

OBJECTIVE: Small cell carcinoma of the ovary, hypercalcemic-type (SCCOHT) is a rare, extremely aggressive neoplasm that usually occurs in young women and is characterized by deleterious germline or somatic SMARCA4 mutations. We performed comprehensive genomic profiling (CGP) to potentially identify additional clinically and pathophysiologically relevant genomic alterations in SCCOHT. METHODS: CGP assessment of all classes of coding alterations in up to 406 genes commonly altered in cancer and intronic regions for up to 31 genes commonly rearranged in cancer was performed on 18 SCCOHT cases (16 exhibiting classic morphology and 2 cases exhibiting exclusive a large cell variant morphology). In addition, a retrospective database search for clinically advanced ovarian tumors with genomic profiles similar to SCCOHT yielded 3 additional cases originally diagnosed as non-SCCOHT. RESULTS: CGP revealed inactivating SMARCA4 alterations and low tumor mutational burden (TMB) (<6mutations/Mb) in 94% (15/16) of SCCOHT with classic morphology. In contrast, both (2/2) cases exhibiting only large cell variant morphology were hypermutated (TMB scores of 90 and 360mut/Mb) and were wildtype for SMARCA4. In our retrospective search, an index ovarian cancer patient harboring inactivating SMARCA4 alterations, initially diagnosed as endometrioid carcinoma, was re-classified as SCCOHT and responded to an SCCOHT chemotherapy regimen. CONCLUSION: The vast majority of SCCOHT demonstrate genomic SMARCA4 loss with only rare co-occurring alterations. Our data support a role for CGP in the diagnosis and management of SCCOHT and of other lesions with overlapping histological and clinical features, since identifying the former by genomic profile suggests benefit from an appropriate regimen and treatment decisions, as illustrated by an index patient.


Subject(s)
Carcinoma, Small Cell/genetics , DNA Helicases/genetics , Hypercalcemia/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Transcription Factors/genetics , Adolescent , Adult , Carcinoma, Small Cell/blood , Carcinoma, Small Cell/enzymology , Carcinoma, Small Cell/pathology , Cohort Studies , DNA Helicases/metabolism , Female , Gene Silencing , Germ-Line Mutation , Humans , Hypercalcemia/enzymology , Hypercalcemia/pathology , Middle Aged , Nuclear Proteins/metabolism , Ovarian Neoplasms/blood , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Transcription Factors/metabolism , Transcriptome , Young Adult
4.
Oncologist ; 22(12): 1478-1490, 2017 12.
Article in English | MEDLINE | ID: mdl-28912153

ABSTRACT

BACKGROUND: Pediatric brain tumors are the leading cause of death for children with cancer in the U.S. Incorporating next-generation sequencing data for both pediatric low-grade (pLGGs) and high-grade gliomas (pHGGs) can inform diagnostic, prognostic, and therapeutic decision-making. MATERIALS AND METHODS: We performed comprehensive genomic profiling on 282 pediatric gliomas (157 pHGGs, 125 pLGGs), sequencing 315 cancer-related genes and calculating the tumor mutational burden (TMB; mutations per megabase [Mb]). RESULTS: In pLGGs, we detected genomic alterations (GA) in 95.2% (119/125) of tumors. BRAF was most frequently altered (48%; 60/125), and FGFR1 missense (17.6%; 22/125), NF1 loss of function (8.8%; 11/125), and TP53 (5.6%; 7/125) mutations were also detected. Rearrangements were identified in 35% of pLGGs, including KIAA1549-BRAF, QKI-RAF1, FGFR3-TACC3, CEP85L-ROS1, and GOPC-ROS1 fusions. Among pHGGs, GA were identified in 96.8% (152/157). The genes most frequently mutated were TP53 (49%; 77/157), H3F3A (37.6%; 59/157), ATRX (24.2%; 38/157), NF1 (22.2%; 35/157), and PDGFRA (21.7%; 34/157). Interestingly, most H3F3A mutations (81.4%; 35/43) were the variant K28M. Midline tumor analysis revealed H3F3A mutations (40%; 40/100) consisted solely of the K28M variant. Pediatric high-grade gliomas harbored oncogenic EML4-ALK, DGKB-ETV1, ATG7-RAF1, and EWSR1-PATZ1 fusions. Six percent (9/157) of pHGGs were hypermutated (TMB >20 mutations per Mb; range 43-581 mutations per Mb), harboring mutations deleterious for DNA repair in MSH6, MSH2, MLH1, PMS2, POLE, and POLD1 genes (78% of cases). CONCLUSION: Comprehensive genomic profiling of pediatric gliomas provides objective data that promote diagnostic accuracy and enhance clinical decision-making. Additionally, TMB could be a biomarker to identify pediatric glioblastoma (GBM) patients who may benefit from immunotherapy. IMPLICATIONS FOR PRACTICE: By providing objective data to support diagnostic, prognostic, and therapeutic decision-making, comprehensive genomic profiling is necessary for advancing care for pediatric neuro-oncology patients. This article presents the largest cohort of pediatric low- and high-grade gliomas profiled by next-generation sequencing. Reportable alterations were detected in 95% of patients, including diagnostically relevant lesions as well as novel oncogenic fusions and mutations. Additionally, tumor mutational burden (TMB) is reported, which identifies a subpopulation of hypermutated glioblastomas that harbor deleterious mutations in DNA repair genes. This provides support for TMB as a potential biomarker to identify patients who may preferentially benefit from immune checkpoint inhibitors.


Subject(s)
Genome, Human/genetics , Glioma/genetics , Neoplasm Proteins/genetics , Tumor Burden/genetics , Adolescent , Child , Child, Preschool , DNA Repair/genetics , Female , Glioma/pathology , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Mutation/genetics
7.
Nature ; 520(7547): 363-7, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25855294

ABSTRACT

Cancer cells adapt their metabolic processes to support rapid proliferation, but less is known about how cancer cells alter metabolism to promote cell survival in a poorly vascularized tumour microenvironment. Here we identify a key role for serine and glycine metabolism in the survival of brain cancer cells within the ischaemic zones of gliomas. In human glioblastoma multiforme, mitochondrial serine hydroxymethyltransferase (SHMT2) and glycine decarboxylase (GLDC) are highly expressed in the pseudopalisading cells that surround necrotic foci. We find that SHMT2 activity limits that of pyruvate kinase (PKM2) and reduces oxygen consumption, eliciting a metabolic state that confers a profound survival advantage to cells in poorly vascularized tumour regions. GLDC inhibition impairs cells with high SHMT2 levels as the excess glycine not metabolized by GLDC can be converted to the toxic molecules aminoacetone and methylglyoxal. Thus, SHMT2 is required for cancer cells to adapt to the tumour environment, but also renders these cells sensitive to glycine cleavage system inhibition.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Glycine Hydroxymethyltransferase/metabolism , Glycine/metabolism , Ischemia/metabolism , Acetone/analogs & derivatives , Acetone/metabolism , Acetone/toxicity , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/enzymology , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Female , Glioblastoma/blood supply , Glioblastoma/enzymology , Glycine Dehydrogenase (Decarboxylating)/antagonists & inhibitors , Glycine Dehydrogenase (Decarboxylating)/metabolism , Humans , Ischemia/enzymology , Ischemia/pathology , Mice , Necrosis , Oxygen Consumption , Pyruvaldehyde/metabolism , Pyruvaldehyde/toxicity , Pyruvate Kinase/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
9.
Cell Rep ; 6(2): 313-24, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24440720

ABSTRACT

Glioblastoma (GBM) harbors subpopulations of therapy-resistant tumor-initiating cells (TICs) that are self-renewing and multipotent. To understand the regulation of the TIC state, we performed an image-based screen for genes regulating GBM TIC maintenance and identified ZFHX4, a 397 kDa transcription factor. ZFHX4 is required to maintain TIC-associated and normal human neural precursor cell phenotypes in vitro, suggesting that ZFHX4 regulates differentiation, and its suppression increases glioma-free survival in intracranial xenografts. ZFHX4 interacts with CHD4, a core member of the nucleosome remodeling and deacetylase (NuRD) complex. ZFHX4 and CHD4 bind to overlapping sets of genomic loci and control similar gene expression programs. Using expression data derived from GBM patients, we found that ZFHX4 significantly affects CHD4-mediated gene expression perturbations, which defines ZFHX4 as a master regulator of CHD4. These observations define ZFHX4 as a regulatory factor that links the chromatin-remodeling NuRD complex and the GBM TIC state.


Subject(s)
Autoantigens/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Homeodomain Proteins/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Transcription Factors/metabolism , Animals , Autoantigens/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Glioblastoma/genetics , Homeodomain Proteins/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice, Inbred NOD , Protein Binding , Transcription Factors/genetics , Transcription, Genetic
11.
Nat Genet ; 37(7): 745-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15951821

ABSTRACT

Multiple genetic alterations occur in melanoma, a lethal skin malignancy of increasing incidence. These include mutations that activate Ras and two of its effector cascades, Raf and phosphoinositide 3-kinase (PI3K). Induction of Ras and Raf can be caused by active N-Ras and B-Raf mutants as well as by gene amplification. Activation of PI3K pathway components occurs by PTEN loss and by AKT3 amplification. Melanomas also commonly show impairment of the p16(INK4A)-CDK4-Rb and ARF-HDM2-p53 tumor suppressor pathways. CDKN2A mutations can produce p16(INK4A) and ARF protein loss. Rb bypass can also occur through activating CDK4 mutations as well as by CDK4 amplification. In addition to ARF deletion, p53 pathway disruption can result from dominant negative TP53 mutations. TERT amplification also occurs in melanoma. The extent to which these mutations can induce human melanocytic neoplasia is unknown. Here we characterize pathways sufficient to generate human melanocytic neoplasia and show that genetically altered human tissue facilitates functional analysis of mutations observed in human tumors.


Subject(s)
Melanoma/genetics , Mutation , Skin Neoplasms/genetics , Skin/pathology , Animals , Cells, Cultured , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/biosynthesis , Cyclin-Dependent Kinases/genetics , DNA-Binding Proteins , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Genes, ras , Humans , Melanocytes/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, SCID , Neoplasm Invasiveness , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/biosynthesis , Phosphoric Monoester Hydrolases/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Skin/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Telomerase/biosynthesis , Telomerase/genetics , Transplants , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
12.
J Clin Invest ; 115(4): 813-24, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15841168

ABSTRACT

Melanoma is a cancer of the neural crest-derived cells that provide pigmentation to skin and other tissues. Over the past 4 decades, the incidence of melanoma has increased more rapidly than that of any other malignancy in the United States. No current treatments substantially enhance patient survival once metastasis has occurred. This review focuses on recent insights into melanoma genetics and new therapeutic approaches being developed based on these advances.


Subject(s)
Melanoma/genetics , Melanoma/therapy , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Animals , Humans , Melanoma/pathology , Melanoma/physiopathology , Neoplasm Metastasis , Neoplasm Staging , Signal Transduction/genetics , Signal Transduction/physiology , Skin Neoplasms/pathology , Skin Neoplasms/physiopathology , ras Proteins/genetics , ras Proteins/metabolism
13.
J Immunol ; 171(11): 6244-50, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14634141

ABSTRACT

Pemphigus foliaceus (PF) is a life-threatening autoimmune blistering skin disease caused by pathogenic IgG autoantibodies against desmoglein 1 (dg1), a desmosomal cadherin-type adhesion glycoprotein. Using lectins and glycosidases, we have shown that dg1 displays an N-glycosylation pattern of the complex triantennary type. We have found that lectins and glycosidases interfere with N-bound sugar residues on the amino-terminal ectodomain of dg1 and completely abolish, in vitro, the antigenicity of dg1 in most of the patients' sera. Moreover, in an ex vivo model using punch biopsies from normal human skin, we demonstrate that preincubation of the epidermis in wheat germ agglutinin (WGA) prevents PF autoantibody binding, acantholysis, and subcorneal blistering. In addition, we show that topical treatment with WGA inhibits PF autoantibody binding to keratinocytes in both newborn BALB/c mice and in organotypic human epidermis grafted onto the back of SCID mice. The epidermis of these pretreated animals displays a regular morphology, whereas control animals develop the immunopathologic phenotype of PF. These findings suggest that WGA may interfere with autoantibody binding to dg1, preventing experimental PF without affecting the adhesive function of dg1. Our observations may provide a new approach to the therapy of PF.


Subject(s)
Autoantibodies/metabolism , Autoantigens/immunology , Binding Sites, Antibody , Cadherins/immunology , Cadherins/metabolism , Pemphigus/immunology , Pemphigus/prevention & control , Wheat Germ Agglutinins/pharmacology , Administration, Topical , Animals , Autoantibodies/administration & dosage , Autoantibodies/blood , Autoantigens/administration & dosage , Autoantigens/metabolism , Binding, Competitive/immunology , Cadherins/administration & dosage , Carbohydrates/immunology , Cells, Cultured , Desmoglein 1 , Disease Models, Animal , Glycosylation , Humans , Immune Sera/administration & dosage , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacology , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mice, SCID , Organ Culture Techniques , Pemphigus/pathology , Protein Binding/immunology , Transplantation, Heterologous , Wheat Germ Agglutinins/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...