251
|
Bian C, Zhao X, Liu Y, Chen M, Zheng S, Tian X, Xu KF. Case report of neurofibromatosis type 1 combined with primary ciliary dyskinesia. Front Med 2021; 15:933-937. [PMID: 34432223 DOI: 10.1007/s11684-021-0860-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/08/2021] [Indexed: 10/20/2022]
Abstract
Neurofibromatosis (NF) is a genetic disease in which the lungs are rarely involved. However, in NF cases with lung involvement, chest computed tomography may show bilateral basal reticulations, apical bullae, and cysts without bronchiectasis. Herein, we report a patient diagnosed with NF on the basis of the results of genetic testing who presented with early-onset wet cough and bronchiectasis. Considering the differential diagnosis of bronchiectasis combined with his early-onset wet cough, sinusitis, and sperm quality decline, we considered the possibility of primary ciliary dyskinesia (PCD). Further electron microscopy analysis of cilia and identification of homozygous mutations in the RSPH4A gene confirmed the diagnosis of PCD. Therefore, for patients with NF, when an image change exists in the lungs that does not correspond to NF, the possibility of other diagnoses, including PCD, must be considered.
Collapse
Affiliation(s)
- Chun Bian
- Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Xinyue Zhao
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Yaping Liu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Minjiang Chen
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Shuying Zheng
- Department of Electron Microscope Laboratory, Peking University People's Hospital, Beijing, 100034, China
| | - Xinlun Tian
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Kai-Feng Xu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
252
|
Riva M, Martorana D, Uliana V, Caleffi E, Boschi E, Garavelli L, Ponti G, Sangiorgi L, Graziano C, Bigoni S, Rocchetti LM, Madeo S, Soli F, Grosso E, Carli D, Goldoni M, Pisani F, Percesepe A. Recurrent NF1 gene variants and their genotype/phenotype correlations in patients with Neurofibromatosis type I. Genes Chromosomes Cancer 2021; 61:10-21. [PMID: 34427956 PMCID: PMC9291954 DOI: 10.1002/gcc.22997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/08/2022] Open
Abstract
Neurofibromatosis type I, a genetic condition due to pathogenic variants in the NF1 gene, is burdened by a high rate of complications, including neoplasms, which increase morbidity and mortality for the disease. We retrospectively re-evaluated the NF1 gene variants found in the period 2000-2019 and we studied for genotype/phenotype correlations of disease complications and neoplasms 34 variants, which were shared by at least two unrelated families (range 2-11) for a total 141 of probands and 21 relatives affected by Neurofibromatosis type I. Recurrent variants could be ascribed to the most common mutational mechanisms (C to T transition, microsatellite slippage, non-homologous recombination). In genotype/phenotype correlations, the variants p.Arg440*, p.Tyr489Cys, and p.Arg1947*, together with the gross gene deletions, displayed the highest rates of complications. When considering neoplasms, carriers of variants falling in the extradomain region at the 5' end of NF1 had a lower age-related cancer frequency than the rest of the gene sequence, showing a borderline significance (p = 0.045), which was not conserved after correction with covariates. We conclude that (1) hotspots in NF1 occur via different mutational mechanisms, (2) several variants are associated with high rates of complications and cancers, and (3) there is an initial evidence toward a lower cancer risk for carriers of variants in the 5' end of the NF1 gene although not significant at the multivariate analysis.
Collapse
Affiliation(s)
- Matteo Riva
- Medical Genetics, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | | | - Elena Boschi
- Plastic Surgery, University Hospital of Parma, Parma, Italy
| | - Livia Garavelli
- Medical Genetics Unit, Azienda USL-IRCCS, Reggio Emilia, Italy
| | - Giovanni Ponti
- Division of Clinical Pathology, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Sangiorgi
- Medical Genetics and Skeletal Rare Diseases, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Claudio Graziano
- Medical Genetics, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Stefania Bigoni
- Medical Genetics, Ferrara University Hospital, Ferrara, Italy
| | | | - Simona Madeo
- Pediatrics, University Hospital of Modena, Italy
| | - Fiorenza Soli
- Medical Genetics, Santa Chiara Hospital, Trento, Italy
| | - Enrico Grosso
- Medical Genetics, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Diana Carli
- Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
| | - Matteo Goldoni
- Statistics, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Francesco Pisani
- Children's Neuropsycological Services, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Antonio Percesepe
- Medical Genetics, Department of Medicine and Surgery, University of Parma, Parma, Italy.,University Hospital of Parma, Parma, Italy
| |
Collapse
|
253
|
Ha C, Kim JW, Jang JH. Performance Evaluation of SpliceAI for the Prediction of Splicing of NF1 Variants. Genes (Basel) 2021; 12:1308. [PMID: 34573290 PMCID: PMC8472818 DOI: 10.3390/genes12091308] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/16/2022] Open
Abstract
Neurofibromatosis type 1, characterized by neurofibromas and café-au-lait macules, is one of the most common genetic disorders caused by pathogenic NF1 variants. Because of the high proportion of splicing mutations in NF1, identifying variants that alter splicing may be an essential issue for laboratories. Here, we investigated the sensitivity and specificity of SpliceAI, a recently introduced in silico splicing prediction algorithm in conjunction with other in silico tools. We evaluated 285 NF1 variants identified from 653 patients. The effect on variants on splicing alteration was confirmed by complementary DNA sequencing followed by genomic DNA sequencing. For in silico prediction of splicing effects, we used SpliceAI, MaxEntScan (MES), and Splice Site Finder-like (SSF). The sensitivity and specificity of SpliceAI were 94.5% and 94.3%, respectively, with a cut-off value of Δ Score > 0.22. The area under the curve of SpliceAI was 0.975 (p < 0.0001). Combined analysis of MES/SSF showed a sensitivity of 83.6% and specificity of 82.5%. The concordance rate between SpliceAI and MES/SSF was 84.2%. SpliceAI showed better performance for the prediction of splicing alteration for NF1 variants compared with MES/SSF. As a convenient web-based tool, SpliceAI may be helpful in clinical laboratories conducting DNA-based NF1 sequencing.
Collapse
Affiliation(s)
| | | | - Ja-Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (C.H.); (J.-W.K.)
| |
Collapse
|
254
|
Wolters PL, Vranceanu AM, Thompson HL, Martin S, Merker VL, Baldwin A, Barnett C, Koetsier KS, Hingtgen CM, Funes CJ, Tonsgard JH, Schorry EK, Allen T, Smith T, Franklin B, Reeve S. Current Recommendations for Patient-Reported Outcome Measures Assessing Domains of Quality of Life in Neurofibromatosis Clinical Trials. Neurology 2021; 97:S50-S63. [PMID: 34230198 PMCID: PMC8594008 DOI: 10.1212/wnl.0000000000012421] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/13/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To review and recommend patient-reported outcome (PRO) measures assessing multidimensional domains of quality of life (QoL) to use as clinical endpoints in medical and psychosocial trials for children and adults with neurofibromatosis (NF) type 1, NF2, and schwannomatosis. METHODS The PRO working group of the Response Evaluation in Neurofibromatosis and Schwannomatosis (REiNS) International Collaboration used systematic methods to review, rate, and recommend existing self-report and parent-report PRO measures of generic and disease-specific QoL for NF clinical trials. Recommendations were based on 4 main criteria: patient characteristics, item content, psychometric properties, and feasibility. RESULTS The highest-rated generic measures were (1) the Pediatric Quality of Life Inventory (PedsQL) Generic Core Scales for NF clinical trials for children or for children through adults, (2) the Functional Assessment of Cancer Therapy-General for adult medical trials, and (3) the World Health Organization Quality of Life-BREF for adult psychosocial trials. The highest-rated disease-specific measures were (1) the PedsQL NF1 Module for NF1 trials, (2) the NF2 Impact on Quality of Life Scale for NF2 trials, and (3) the Penn Acoustic Neuroma Quality of Life Scale for NF2 trials targeting vestibular schwannomas. To date, there are no disease-specific tools assessing multidimensional domains of QoL for schwannomatosis. CONCLUSIONS The REiNS Collaboration currently recommends these generic and disease-specific PRO measures to assess multidimensional domains of QoL for NF clinical trials. Additional research is needed to further evaluate the use of these measures in both medical and psychosocial trials.
Collapse
Affiliation(s)
- Pamela L Wolters
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.).
| | - Ana-Maria Vranceanu
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Heather L Thompson
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Staci Martin
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Vanessa L Merker
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Andrea Baldwin
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Carolina Barnett
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Kimberley S Koetsier
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Cynthia M Hingtgen
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Christopher J Funes
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - James H Tonsgard
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Elizabeth K Schorry
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Taryn Allen
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Taylor Smith
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Barbara Franklin
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| | - Stephanie Reeve
- From the Pediatric Oncology Branch (P.L.W., S.M.), National Cancer Institute, NIH, Bethesda, MD; Integrated Brain Health Clinical and Research Program (A.-M.V., C.J.F.), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; Center for Healthcare Organization and Implementation Research (V.L.M.), Edith Nourse Rogers Memorial Veterans Hospital, Bedford; Clinical Monitoring Research Program Directorate (A.B., T.A.), Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD; Division of Neurology (C.B.), Department of Medicine, University Health Network and University of Toronto, Ontario, Canada; Department of Otolaryngology/Head and Neck Surgery (K.S.K.), Leiden University Medical Center, the Netherlands; Department of Clinical Neurosciences (C.M.H.), Spectrum Health Medical Group and College of Human Medicine, Michigan State University, East Lansing; University of Chicago Pritzker School of Medicine (J.H.T.), IL; Division of Human Genetics (E.K.S.), Cincinnati Children's Hospital, OH; Department of Psychology and Child Development (T.S.), California Polytechnic State University, San Luis Obispo; and REiNS International Collaboration Patient Representative (B.F., S.R.)
| |
Collapse
|
255
|
Geoffray MM, Robinson L, Ramamurthy K, Manderson L, O'Flaherty J, Lehtonen A, Tordjman S, Green J, Vassallo G, Garg S. Predictors of cognitive, behavioural and academic difficulties in NF1. J Psychiatr Res 2021; 140:545-550. [PMID: 34182240 DOI: 10.1016/j.jpsychires.2021.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/17/2021] [Accepted: 06/04/2021] [Indexed: 11/16/2022]
Abstract
The impact of the Neurofibromatosis type 1 (NF1) on cognition have been subject to much clinical investigation, but environmental modifiers of disease expression have not yet been systematically investigated. The aim of this paper is to determine the role of demographic and environmental factors such as age, sex, socioeconomic status, parental NF1 status and neurological complications on the cognitive, behavioural and academic outcomes in NF1. Participants included 206 children aged 4-18 years seen within the Manchester clinical research NF1 service. Multiple linear regression models were used to study the effect of the hypothesized predictor variables on cognitive, behavioural and academic outcomes. Relative to population norms, 80% of the NF1 sample demonstrated significantly lower scores in at least one cognitive, behavioural or academic domains. Family history of NF1 and lower SES were independently associated with poorer cognitive, behavioural and academic outcomes. Neurological problems such as epilepsy and hydrocephalus were associated with lower IQ and academic skills. Cognitive and behavioural phenotypes emerge commonly via a complex interplay between genes and environmental factors, and this is true also of a monogenic condition such as NF1. Early interventions and remedial education may be targeted to risk groups such those with familial NF1, families with lower SES and those with associated neurological comorbidities.
Collapse
Affiliation(s)
- Marie-Maude Geoffray
- Centre Hospitalier le Vinatier, 95 Bd Pinel, 69500 Bron, France; Division of Neuroscience and Experimental Psychology, Faculty of Biological Medical & Health Sciences, University of Manchester, Oxford Road, M13 9PL, UK
| | - Louise Robinson
- Dept of Child and Adolescent Mental Health, Manchester University NHS Foundation Trust, Manchester, UK
| | - Kavitha Ramamurthy
- Dept of Child and Adolescent Mental Health, Manchester University NHS Foundation Trust, Manchester, UK
| | - Lauren Manderson
- Dept of Child and Adolescent Mental Health, Manchester University NHS Foundation Trust, Manchester, UK
| | - Julieta O'Flaherty
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, University of Manchester, UK
| | | | - Sylvie Tordjman
- Pôle Hospitalo-Unuversitaire de Psychiatrie de l'Enfant et de l'Adolescent, Université de Rennes 1, Centre Hospitalier Guilllaume Régnier, 35703, Rennes, France; Integrative Neuroscience and Cognition Center (INCC), CNRS UMR 8002, Université de Paris, France
| | - Jonathan Green
- Division of Neuroscience and Experimental Psychology, Faculty of Biological Medical & Health Sciences, University of Manchester, Oxford Road, M13 9PL, UK; Dept of Child and Adolescent Mental Health, Manchester University NHS Foundation Trust, Manchester, UK; Manchester Academic Health Sciences Centre, Manchester, M13 9PL, UK
| | - Grace Vassallo
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, M13 9PL, UK
| | - Shruti Garg
- Division of Neuroscience and Experimental Psychology, Faculty of Biological Medical & Health Sciences, University of Manchester, Oxford Road, M13 9PL, UK; Dept of Child and Adolescent Mental Health, Manchester University NHS Foundation Trust, Manchester, UK; Manchester Academic Health Sciences Centre, Manchester, M13 9PL, UK.
| |
Collapse
|
256
|
Solares I, Viñal D, Morales-Conejo M, Rodriguez-Salas N, Feliu J. Novel molecular targeted therapies for patients with neurofibromatosis type 1 with inoperable plexiform neurofibromas: a comprehensive review. ESMO Open 2021; 6:100223. [PMID: 34388689 PMCID: PMC8363824 DOI: 10.1016/j.esmoop.2021.100223] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 11/30/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a genetic disorder that carries a higher risk of tumor development. Plexiform neurofibromas (PNs) are present in 50% of NF1 and cause significant morbidity when surgery is not feasible. Systemic therapies had not succeeded to reduce PN tumor volume until 2016 when the first trial with an MAPK/extracellular-signal-regulated kinase (MEK) inhibitor was published. We performed a systematic research on novel targeted therapies for patients with NF1 and PNs in PubMed, EMBASE, and conference abstracts with the last update in February 2021. Since 2016, seven trials have reported positive results with MEK inhibitors and other molecular targeted therapies (cabozantinib). Selumetinib has shown an overall response rate of 68% in children with NF1 and symptomatic inoperable PNs, and was associated with pain improvement and a manageable adverse events profile. This led to Food and Drug Administration (FDA) approval of selumetinib in May 2020. Recently, cabozantinib and mirdametinib have also proven their efficacy in adult population. Other MEK inhibitors such as trametinib and binimetinib have also communicated promising preliminary results. Ongoing trials in different populations and with intermittent dosing strategies are underway.
Collapse
Affiliation(s)
- I Solares
- Department of Internal Medicine, Reference Center for Inherited Metabolic Disease - MetabERN, University Hospital 12 de Octubre, UCM Madrid, Madrid, Spain
| | - D Viñal
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain.
| | - M Morales-Conejo
- Department of Internal Medicine, Reference Center for Inherited Metabolic Disease - MetabERN, University Hospital 12 de Octubre, UCM Madrid, Madrid, Spain; Grupo de Enfermedades Mitocondriales y Neuromusculares, Instituto de Investigación Hospital 12 de Octubre (i+12), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - N Rodriguez-Salas
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain; Translational Oncology Group, IdiPAZ, Madrid, Spain; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC, Madrid, Spain
| | - J Feliu
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain; Translational Oncology Group, IdiPAZ, Madrid, Spain; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC, Madrid, Spain
| |
Collapse
|
257
|
Casey D, Demko S, Sinha A, Mishra-Kalyani PS, Shen YL, Khasar S, Goheer MA, Helms WS, Pan L, Xu Y, Fan J, Leong R, Liu J, Yang Y, Windsor K, Ou M, Stephens O, Oh B, Reaman GH, Nair A, Shord SS, Bhatnagar V, Daniels SR, Sickafuse S, Goldberg KB, Theoret MR, Pazdur R, Singh H. FDA Approval Summary: Selumetinib for Plexiform Neurofibroma. Clin Cancer Res 2021; 27:4142-4146. [PMID: 33712511 DOI: 10.1158/1078-0432.ccr-20-5032] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/10/2021] [Accepted: 03/10/2021] [Indexed: 11/16/2022]
Abstract
On April 10, 2020, the FDA approved selumetinib (KOSELUGO, AstraZeneca) for the treatment of pediatric patients 2 years of age and older with neurofibromatosis type 1 who have symptomatic, inoperable plexiform neurofibromas. Approval was based on demonstration of a durable overall response rate per Response Evaluation in Neurofibromatosis and Schwannomatosis criteria and supported by observed clinical improvements in plexiform neurofibroma-related symptoms and functional impairments in 50 pediatric patients with inoperable plexiform neurofibromas in a single-arm, multicenter trial. The overall reponse rate per NCI investigator assessment was 66% (95% confidence interval, 51-79) with at least 12 months of follow-up. The median duration of response was not reached, and 82% of responding patients experienced duration of response ≥12 months. Clinical outcome assessment endpoints provided supportive efficacy data. Risks of selumetinib are consistent with MAPK (MEK) inhibitor class effects, including ocular, cardiac, musculoskeletal, gastrointestinal, and dermatologic toxicities. Safety was assessed across a pooled database of 74 pediatric patients with plexiform neurofibromas and supported by adult and pediatric selumetinib clinical trial data in cancer indications. The benefit-risk assessment for selumetinib in patients with inoperable plexiform neurofibromas was considered favorable.
Collapse
Affiliation(s)
- Denise Casey
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland
| | - Suzanne Demko
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland
| | - Arup Sinha
- Office of Biostatistics, FDA, Silver Spring, Maryland
| | | | - Yuan-Li Shen
- Office of Biostatistics, FDA, Silver Spring, Maryland
| | - Sachia Khasar
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland
| | - M Anwar Goheer
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland
| | | | - Lili Pan
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Yuan Xu
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Jianghong Fan
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Ruby Leong
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Jiang Liu
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Yuching Yang
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | | | - Mei Ou
- Office of Pharmaceutical Quality, FDA, Silver Spring, Maryland
| | - Olen Stephens
- Office of Pharmaceutical Quality, FDA, Silver Spring, Maryland
| | - Byeongtaek Oh
- Office of Pharmaceutical Quality, FDA, Silver Spring, Maryland
| | | | - Abhilasha Nair
- Oncology Center of Excellence, FDA, Silver Spring, Maryland
| | - Stacy S Shord
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | | | - Selena R Daniels
- Division of Clinical Outcome Assessment, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland
| | | | | | - Marc R Theoret
- Oncology Center of Excellence, FDA, Silver Spring, Maryland
| | - Richard Pazdur
- Oncology Center of Excellence, FDA, Silver Spring, Maryland
| | - Harpreet Singh
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland. .,Oncology Center of Excellence, FDA, Silver Spring, Maryland
| |
Collapse
|
258
|
Kenborg L, Andersen EW, Duun-Henriksen AK, Jepsen JRM, Doser K, Dalton SO, Bidstrup PE, Krøyer A, Frederiksen LE, Johansen C, Østergaard JR, Hove H, Sørensen SA, Riccardi VM, Mulvihill JJ, Winther JF. Psychiatric disorders in individuals with neurofibromatosis 1 in Denmark: A nationwide register-based cohort study. Am J Med Genet A 2021; 185:3706-3716. [PMID: 34327813 DOI: 10.1002/ajmg.a.62436] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/01/2021] [Accepted: 07/10/2021] [Indexed: 12/19/2022]
Abstract
The aim of this study was to assess the risks of psychiatric disorders in a large cohort of 905 individuals with NF1 and 7614 population comparisons matched on sex and year of birth. The cohort was linked to the Danish Psychiatric Central Research Register to ascertain information on hospital contacts for psychiatric disorders based on the International Classification of Diseases version 8 and 10. The hazard ratio (HR) for a first psychiatric hospital contact was higher in girls (4.19, 95% confidence interval [CI] 1.81-9.69) and boys with NF1 (5.02, 95% CI 3.27-7.69) <7 years of age than in the population comparisons. Both sexes had increased HRs for developmental disorders, including attention deficit/hyperactivity disorders, autism spectrum disorders, and intellectual disabilities in childhood. Females with NF1 had also increased HRs for unipolar depression, other emotional and behavioral disorders, and severe stress reaction and adjustment disorders in early adulthood. The HRs for psychoses, schizophrenia, bipolar disorders, and substance abuse were similar in individuals with NF1 and the population comparisons. Finally, the cumulative incidence of a first hospital contact due to any psychiatric disorder by age 30 years was 35% (95% CI 29-41) in females and 28% (95% CI 19-37) in males with NF1. Thus, screening for psychiatric disorders may be important for early diagnosis and facilitation of appropriate and effective treatment in individuals with NF1.
Collapse
Affiliation(s)
- Line Kenborg
- Childhood Cancer Research Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elisabeth W Andersen
- Statistics and Data Analysis, Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Jens R M Jepsen
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research and Center for Neuropsychiatric Schizophrenia Research, Mental Health Service Capital Region, University of Copenhagen, Copenhagen, Denmark.,Child and Adolescent Mental Health Centre, Mental Health Services Capital Region, University of Copenhagen, Copenhagen, Denmark
| | - Karoline Doser
- Childhood Cancer Research Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Susanne O Dalton
- Survivorship and Inequality in Cancer, Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Clinical Oncology and Palliative Care, Zealand University Hospital, Naestved, Denmark
| | - Pernille E Bidstrup
- Psychological Aspects of Cancer, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anja Krøyer
- Childhood Cancer Research Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Line E Frederiksen
- Childhood Cancer Research Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Christoffer Johansen
- Psychological Aspects of Cancer, Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - John R Østergaard
- Department of Pediatrics, Centre for Rare Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Hanne Hove
- Department of Pediatrics, Centre for Rare Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,The RareDis Database, Section of Rare Diseases, Department of Clinical Genetics and Pediatrics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sven Asger Sørensen
- Department of Neurogenetics, Institute of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | - John J Mulvihill
- Department of Pediatrics, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Jeanette F Winther
- Childhood Cancer Research Group, Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University and University Hospital, Aarhus, Denmark
| |
Collapse
|
259
|
Kallionpää RA, Valtanen M, Auranen K, Uusitalo E, Rinne JO, Peltonen S, Peltonen J. Increased risk for dementia in neurofibromatosis type 1. Genet Med 2021; 23:2219-2222. [PMID: 34257422 PMCID: PMC8553610 DOI: 10.1038/s41436-021-01261-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To determine the risk for dementia in neurofibromatosis type 1 (NF1) using a Finnish nationwide cohort of individuals with NF1, and data from national registries. METHODS A Finnish cohort of 1,349 individuals with confirmed NF1 according to the US National Institutes of Health (NIH) diagnostic criteria was compared with a control cohort of 13,870 individuals matched for age, sex, and area of residence. Dementia-related hospital visits were retrieved from the Finnish Care Register for Health Care using International Classification of Diseases, 10th revision (ICD-10) diagnosis codes G30 and F00-F03. Purchases of antidementia drugs were queried with Anatomical Therapeutic Chemical (ATC) classification code N06D from the drug reimbursement register maintained by the Social Insurance Institution of Finland. The follow-up spanned 1998-2014. RESULTS Totals of 16 and 165 individuals with at least two dementia-related diagnoses or drug purchases were identified in the NF1 and control cohorts, respectively. The hazard ratio for dementia in NF1 was 1.67 (95% confidence interval [CI] 1.00-2.80, P = 0.050). In an analysis stratified by the type of dementia, the risk for Alzheimer disease was increased in NF1 compared to controls with a hazard ratio of 2.88 (95% CI 1.47-5.66, P = 0.002). CONCLUSION Dementia and especially Alzheimer disease are previously unrecognized neurological complications of NF1.
Collapse
Affiliation(s)
| | - Mikko Valtanen
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Dermatology and Venereology, University of Turku, Turku, Finland.,Department of Dermatology, Turku University Hospital, Turku, Finland.,Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Kari Auranen
- Department of Mathematics and Statistics, University of Turku, Turku, Finland.,Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Elina Uusitalo
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Juha O Rinne
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - Sirkku Peltonen
- Department of Dermatology and Venereology, University of Turku, Turku, Finland.,Department of Dermatology, Turku University Hospital, Turku, Finland.,Department of Dermatology and Venereology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Dermatology and Venereology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Juha Peltonen
- Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
260
|
Toro G, Santoro C, Ambrosio D, Landi G, Scilipoti M, Moretti A, Paoletta M, Liguori S, Schiavone Panni A, Picariello S, Iolascon G. Natural History of Scoliosis in Children with NF1: An Observation Study. Healthcare (Basel) 2021; 9:healthcare9070881. [PMID: 34356257 PMCID: PMC8303287 DOI: 10.3390/healthcare9070881] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 02/05/2023] Open
Abstract
(1) Background. Scoliosis is the most common musculoskeletal manifestation of Neurofibromatosis type 1 (NF1), and it might be dystrophic (D) or non-dystrophic (ND) depending on the presence of dysplastic changes of the spine. The aim of our study was to describe the characteristics and natural history of patients with NF1 and scoliosis. (2) Methods. We retrospectively reviewed records from patients with NF1 and scoliosis. Scoliosis was classified as D if at least two dystrophic changes were documented at imaging. (3) Results. Of the 438 patients reviewed, 43 fulfilled inclusion criteria; 17 were classified in D group and 26 in ND. The groups did not differ in age and localization of scoliosis curvature. Surgery was needed more often in D group, but the between-group difference was not significant. Male-to-female ratios of 3:1 and 4:1 were reported in surgically treated NF1 patients with ND and D scoliosis, respectively. (4) Conclusions. Our data suggests independently by the presence of dysplastic changes affecting the spine that males with NF1 are more often affected by scoliosis that requires surgery.
Collapse
Affiliation(s)
- Giuseppe Toro
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.T.); (D.A.); (G.L.); (A.M.); (S.L.); (A.S.P.); (G.I.)
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Claudia Santoro
- Department of Woman, Child and of General and Specialised Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.S.); (M.S.); (S.P.)
- Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Daniele Ambrosio
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.T.); (D.A.); (G.L.); (A.M.); (S.L.); (A.S.P.); (G.I.)
| | - Giovanni Landi
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.T.); (D.A.); (G.L.); (A.M.); (S.L.); (A.S.P.); (G.I.)
| | - Martina Scilipoti
- Department of Woman, Child and of General and Specialised Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.S.); (M.S.); (S.P.)
| | - Antimo Moretti
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.T.); (D.A.); (G.L.); (A.M.); (S.L.); (A.S.P.); (G.I.)
| | - Marco Paoletta
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.T.); (D.A.); (G.L.); (A.M.); (S.L.); (A.S.P.); (G.I.)
- Correspondence:
| | - Sara Liguori
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.T.); (D.A.); (G.L.); (A.M.); (S.L.); (A.S.P.); (G.I.)
| | - Alfredo Schiavone Panni
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.T.); (D.A.); (G.L.); (A.M.); (S.L.); (A.S.P.); (G.I.)
| | - Stefania Picariello
- Department of Woman, Child and of General and Specialised Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.S.); (M.S.); (S.P.)
| | - Giovanni Iolascon
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.T.); (D.A.); (G.L.); (A.M.); (S.L.); (A.S.P.); (G.I.)
| |
Collapse
|
261
|
Heyde CE, Völker A, von der Höh NH, Glasmacher S, Koller H. [Spinal deformity in neurofibromatosis type 1]. DER ORTHOPADE 2021; 50:650-656. [PMID: 34236453 DOI: 10.1007/s00132-021-04130-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND With a prevalence of up to 60%, spinal deformity represents the most common skeletal manifestation of neurofibromatosis type 1. The deformity can occur as a non-dystrophic or as a less common dystrophic type. This distinction is of great relevance because the therapeutic strategy is completely different in each case. NON-DYSTROPHIC TYPE The non-dystrophic type can be treated like idiopathic scoliosis due to the comparable behavior of both entities. However, care must be taken regarding the so-called modulation. Modulation describes the formation of dysplasias of the spine. This will result in a progression behavior as known from the dystrophic type. DYSTROPHIC TYPE For the dystrophic type, different spinal dysplastic changes are typical. These lead to a rapid progression of deformity and a lack of response to conservative treatment. If untreated, severe and grotesque deformities can arise. This type of deformity requires early surgical intervention, even in childhood. The knowledge about the peculiarities of this disease in general, as well as the typical changes of the spine are prerequisites to managing these often-challenging situations.
Collapse
Affiliation(s)
- Christoph-E Heyde
- Klinik und Poliklinik für Orthopädie, Unfallchirurgie u. Plastische Chirurgie, Bereich Wirbelsäulenchirurgie, Universitätsklinikum Leipzig AöR, Liebigstraße 20, 04103, Leipzig, Deutschland.
| | - A Völker
- Klinik und Poliklinik für Orthopädie, Unfallchirurgie u. Plastische Chirurgie, Bereich Wirbelsäulenchirurgie, Universitätsklinikum Leipzig AöR, Liebigstraße 20, 04103, Leipzig, Deutschland
| | - N H von der Höh
- Klinik und Poliklinik für Orthopädie, Unfallchirurgie u. Plastische Chirurgie, Bereich Wirbelsäulenchirurgie, Universitätsklinikum Leipzig AöR, Liebigstraße 20, 04103, Leipzig, Deutschland
| | - S Glasmacher
- Klinik und Poliklinik für Orthopädie, Unfallchirurgie u. Plastische Chirurgie, Bereich Wirbelsäulenchirurgie, Universitätsklinikum Leipzig AöR, Liebigstraße 20, 04103, Leipzig, Deutschland
| | - H Koller
- Klinik und Poliklinik für Neurochirurgie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
| |
Collapse
|
262
|
Akshintala S, Khalil N, Yohay K, Muzikansky A, Allen J, Yaffe A, Gross AM, Fisher MJ, Blakeley JO, Oberlander B, Pudel M, Engelson C, Obletz J, Mitchell C, Widemann BC, Stevenson DA, Plotkin SR. Reliability of Handheld Dynamometry to Measure Focal Muscle Weakness in Neurofibromatosis Types 1 and 2. Neurology 2021; 97:S99-S110. [PMID: 34230196 DOI: 10.1212/wnl.0000000000012439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To determine a suitable outcome measure for assessing muscle strength in neurofibromatosis (NF) type 1 and NF2 clinical trials, we evaluated the intraobserver reliability of handheld dynamometry (HHD) and developed consensus recommendations for its use in NF clinical trials. METHODS Patients ≥5 years of age with weakness in at least 1 muscle group by manual muscle testing (MMT) were eligible. Maximal isometric muscle strength of a weak muscle group and the biceps of the dominant arm was measured by HHD. An average of 3 repetitions per session was used as an observation, and 3 sessions with rest period between each were performed on the same day by a single observer. Intrasession and intersession intraclass correlation coefficients (ICCs) and coefficients of variation (CVs) were calculated to assess reliability and measurement error. RESULTS Twenty patients with NF1 and 13 with NF2 were enrolled; median age was 12 years (interquartile range [IQR] 9-17 years) and 29 years (IQR 22-38 years), respectively. By MMT, weak muscle strength ranged from 2-/5 to 4+/5. Biceps strength was 5/5 in all patients. Intersession ICCs for the weak muscles were 0.98 and 0.99 in the NF1 and NF2 cohorts, respectively, and for biceps were 0.97 and 0.97, respectively. The median CVs for average session strength were 5.4% (IQR 2.6%-7.3%) and 2.9% (IQR 2.0%-6.2%) for weak muscles and biceps, respectively. CONCLUSION HHD performed by a trained examiner with a well-defined protocol is a reliable technique to measure muscle strength in NF1 and NF2. Recommendations for strength testing in NF1 and NF2 trials are provided.
Collapse
Affiliation(s)
- Srivandana Akshintala
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston.
| | - Nashwa Khalil
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Kaleb Yohay
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Alona Muzikansky
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Jeffrey Allen
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Anna Yaffe
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Andrea M Gross
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Michael J Fisher
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Jaishri O Blakeley
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Beverly Oberlander
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Miriam Pudel
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Celia Engelson
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Jaime Obletz
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Carole Mitchell
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Brigitte C Widemann
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - David A Stevenson
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | - Scott R Plotkin
- From New York University (NYU) School of Medicine and NYU Langone Health (S.A., N.K., K.Y., J.A., A.Y., M.P., C.E., J.O., C.M.), New York; Pediatric Oncology Branch (S.A., A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; Massachusetts General Hospital (A.M.), Boston; Division of Oncology (M.J.F.), The Children's Hospital of Philadelphia, PA; Department of Neurology (J.O.B.), Johns Hopkins University, Baltimore, MD; Neurofibromatosis Network (B.O.); Department of Pediatrics (D.A.S.), Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA; and Cancer Center and Department of Neurology (S.R.P.), Massachusetts General Hospital, Boston
| | | |
Collapse
|
263
|
RASopathies: from germline mutations to somatic and multigenic diseases. Biomed J 2021; 44:422-432. [PMID: 34175492 PMCID: PMC8514848 DOI: 10.1016/j.bj.2021.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
The RAS-RAF-MEK-ERK signaling pathway is vital for different cellular mechanisms including cell proliferation, differentiation and apoptosis. This importance is highlighted by the high prevalence of mutations in RAS or related proteins of the pathway in cancers. More recently, development abnormalities have been linked to various germline mutations in this pathway and called RASopathies. Interestingly, rare disorders such as RAS-associated leukoproliferative diseases and histiocytosis have also been recently linked to multiple mutations in the same pathway, sometimes with the same mutation. This review will focus on germline RASopathies and rare somatic RASopathies and focus on how gain-of-function mutations in the same pathway can lead to various diseases.
Collapse
|
264
|
Vetrano IG, Acerbi F, Falco J, D'Ammando A, Devigili G, Nazzi V. High-Definition 4K 3D Exoscope (ORBEYETM) in Peripheral Nerve Sheath Tumor Surgery: A Preliminary, Explorative, Pilot Study. Oper Neurosurg (Hagerstown) 2021; 19:480-488. [PMID: 32357216 DOI: 10.1093/ons/opaa090] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/10/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Surgery for peripheral nerve sheath tumors aims to preserve functional fascicles achieving gross-total resection. Increasing the visualization of anatomic details helps to identify the different layers and the tumor-nerve interface. The traditional microscope can present some limitations in this type of surgery, such as its physical obstruction. OBJECTIVE To present a proof-of-concept study about exoscope-guided surgery for schwannomas of the lower limbs, to analyze the advantages and disadvantages of the 4K, high-quality, 3-dimensional (3D) imaging. METHODS We analyzed 2 consecutive surgical cases of suspected schwannomas of the lower limbs using the ORBEYE™ exoscope (Olympus). A standard operative microscope was also available in the operating room. All procedures were performed with neurophysiological monitoring, to identify functioning nerves and to localize the tumor capsule safest entry point. The cases are reported according to the PROCESS guidelines. RESULTS In both cases, we achieved a gross total resection of the schwannomas; the exoscope provided an excellent view of the anatomic details at tumor-nerve interface, as visible in intraoperative images and in the 3D-4K video supporting these findings. The surgeon's position was comfortable in both cases, although if the co-surgeon positioned himself in front of the first surgeon, the comfort was slightly reduced. The 4K monitor allowed a realistic, nontiring 3D vision for all the team. CONCLUSION The ORBEYETM, after an adequate learning curve, can represent a feasible and comfortable instrument for nerve tumor surgery, which is usually performed in a single horizontal plane. Further and wider clinical series are necessary to confirm this first impression.
Collapse
Affiliation(s)
- Ignazio G Vetrano
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesco Acerbi
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Jacopo Falco
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Antonio D'Ammando
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Grazia Devigili
- Neurological Unit 1, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Vittoria Nazzi
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
265
|
Prabhu VC, Pappu S, Borys E, Ormston L, Lomasney LM. Commentary: Machine-Learning Approach to Differentiation of Benign and Malignant Peripheral Nerve Sheath Tumors: A Multicenter Study. Neurosurgery 2021; 89:E156-E157. [PMID: 34131751 DOI: 10.1093/neuros/nyab224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Vikram C Prabhu
- Department of Neurological Surgery, Loyola University Medical Center, Maywood, Illinois, USA
| | - Suguna Pappu
- Section of Neurosurgery, Department of Surgery, Edward Hines Veterans Administration Hospital, Hines, Illinois, USA
| | - Ewa Borys
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois, USA
| | - Leighanne Ormston
- Department of Oncology, Loyola University Medical Center, Maywood, Illinois, USA
| | - Laurie M Lomasney
- Department of Radiology, Loyola University Medical Center, Maywood, Illinois, USA
| |
Collapse
|
266
|
Pacot L, Vidaud D, Sabbagh A, Laurendeau I, Briand-Suleau A, Coustier A, Maillard T, Barbance C, Morice-Picard F, Sigaudy S, Glazunova OO, Damaj L, Layet V, Quelin C, Gilbert-Dussardier B, Audic F, Dollfus H, Guerrot AM, Lespinasse J, Julia S, Vantyghem MC, Drouard M, Lackmy M, Leheup B, Alembik Y, Lemaire A, Nitschké P, Petit F, Dieux Coeslier A, Mutez E, Taieb A, Fradin M, Capri Y, Nasser H, Ruaud L, Dauriat B, Bourthoumieu S, Geneviève D, Audebert-Bellanger S, Nizon M, Stoeva R, Hickman G, Nicolas G, Mazereeuw-Hautier J, Jannic A, Ferkal S, Parfait B, Vidaud M, Wolkenstein P, Pasmant E. Severe Phenotype in Patients with Large Deletions of NF1. Cancers (Basel) 2021; 13:2963. [PMID: 34199217 PMCID: PMC8231977 DOI: 10.3390/cancers13122963] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/11/2021] [Indexed: 12/18/2022] Open
Abstract
Complete deletion of the NF1 gene is identified in 5-10% of patients with neurofibromatosis type 1 (NF1). Several studies have previously described particularly severe forms of the disease in NF1 patients with deletion of the NF1 locus, but comprehensive descriptions of large cohorts are still missing to fully characterize this contiguous gene syndrome. NF1-deleted patients were enrolled and phenotypically characterized with a standardized questionnaire between 2005 and 2020 from a large French NF1 cohort. Statistical analyses for main NF1-associated symptoms were performed versus an NF1 reference population. A deletion of the NF1 gene was detected in 4% (139/3479) of molecularly confirmed NF1 index cases. The median age of the group at clinical investigations was 21 years old. A comprehensive clinical assessment showed that 93% (116/126) of NF1-deleted patients fulfilled the NIH criteria for NF1. More than half had café-au-lait spots, skinfold freckling, Lisch nodules, neurofibromas, neurological abnormalities, and cognitive impairment or learning disabilities. Comparison with previously described "classic" NF1 cohorts showed a significantly higher proportion of symptomatic spinal neurofibromas, dysmorphism, learning disabilities, malignancies, and skeletal and cardiovascular abnormalities in the NF1-deleted group. We described the largest NF1-deleted cohort to date and clarified the more severe phenotype observed in these patients.
Collapse
Affiliation(s)
- Laurence Pacot
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
- Inserm U1016—CNRS UMR8104, Institut Cochin, Université de Paris, CARPEM, F-75014 Paris, France;
| | - Dominique Vidaud
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
- Inserm U1016—CNRS UMR8104, Institut Cochin, Université de Paris, CARPEM, F-75014 Paris, France;
| | - Audrey Sabbagh
- UMR 261, Laboratoire MERIT, IRD, Faculté de Pharmacie de Paris, Université de Paris, F-75006 Paris, France;
| | - Ingrid Laurendeau
- Inserm U1016—CNRS UMR8104, Institut Cochin, Université de Paris, CARPEM, F-75014 Paris, France;
| | - Audrey Briand-Suleau
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
- Inserm U1016—CNRS UMR8104, Institut Cochin, Université de Paris, CARPEM, F-75014 Paris, France;
| | - Audrey Coustier
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
| | - Théodora Maillard
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
| | - Cécile Barbance
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
| | - Fanny Morice-Picard
- Inserm U1211, Service de Génétique Médicale, CHU de Bordeaux, F-33000 Bordeaux, France;
| | - Sabine Sigaudy
- Department of Medical Genetics, Children’s Hospital La Timone, Assistance Publique des Hôpitaux de Marseille, F-13000 Marseille, France;
| | - Olga O. Glazunova
- Centre de Référence des Anomalies du Développement et Syndromes Malformatifs (UF 2970), CHU Timone, Assistance Publique des Hôpitaux de Marseille, F-13000 Marseille, France;
| | - Lena Damaj
- Department of Pediatrics, Competence Center of Inherited Metabolic Disorders, Rennes Hospital, F-35000 Rennes, France;
| | - Valérie Layet
- Consultations de Génétique, Groupe Hospitalier du Havre, F-76600 Le Havre, France;
| | - Chloé Quelin
- Service de Génétique Clinique, CLAD Ouest, CHU Rennes, Hôpital Sud, F-35000 Rennes, France; (C.Q.); (M.F.)
| | | | - Frédérique Audic
- Service de Neurologie Pédiatrique, CHU Timone Enfants, F-13000 Marseille, France;
| | - Hélène Dollfus
- Centre de Référence Pour les Affections Rares en Génétique Ophtalmologique, CARGO, Filière SENSGENE, Hôpitaux Universitaires de Strasbourg, F-67000 Strasbourg, France;
- Medical Genetics Laboratory, INSERM U1112, Institute of Medical Genetics of Alsace, Strasbourg Medical School, University of Strasbourg, F-67000 Strasbourg, France
| | | | - James Lespinasse
- Service de Génétique Clinique, CH de Chambéry, F-73000 Chambéry, France;
| | - Sophie Julia
- Service de Génétique Médicale, CHU de Toulouse, Hôpital Purpan, F-31000 Toulouse, France;
| | - Marie-Christine Vantyghem
- Endocrinology, Diabetology, Metabolism and Nutrition Department, Inserm 1190, Lille University Hospital EGID, F-59000 Lille, France;
| | - Magali Drouard
- Dermatology Department, CHU Lille, University of Lille, F-59000 Lille, France;
| | - Marilyn Lackmy
- Unité de Génétique Clinique, Centre de Compétences Maladies Rares Anomalies du Développement, CHRU de Pointe à Pitre, F-97110 Guadeloupe, France;
| | - Bruno Leheup
- Service de Génétique Médicale, Hôpitaux de Brabois, CHRU de Nancy, F-54500 Vandoeuvre-lès-Nancy, France;
| | - Yves Alembik
- Department of Medical Genetics, Strasbourg-Hautepierre Hospital, F-67000 Strasbourg, France; (Y.A.); (A.L.)
| | - Alexia Lemaire
- Department of Medical Genetics, Strasbourg-Hautepierre Hospital, F-67000 Strasbourg, France; (Y.A.); (A.L.)
| | - Patrick Nitschké
- Bioinformatics Platform, Imagine Institute, INSERM UMR 1163, Université de Paris, F-75015 Paris, France;
| | - Florence Petit
- CHU Lille, Clinique de Génétique, Centre de Référence Anomalies du Développement, F-59000 Lille, France; (F.P.); (A.D.C.)
| | - Anne Dieux Coeslier
- CHU Lille, Clinique de Génétique, Centre de Référence Anomalies du Développement, F-59000 Lille, France; (F.P.); (A.D.C.)
| | - Eugénie Mutez
- Lille University, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France;
| | - Alain Taieb
- Department of Dermatology and Pediatric Dermatology, Bordeaux University Hospital, F-33000 Bordeaux, France;
| | - Mélanie Fradin
- Service de Génétique Clinique, CLAD Ouest, CHU Rennes, Hôpital Sud, F-35000 Rennes, France; (C.Q.); (M.F.)
| | - Yline Capri
- Département de Génétique, APHP Nord, Hôpital Robert Debré, F-75019 Paris, France; (Y.C.); (H.N.); (L.R.)
| | - Hala Nasser
- Département de Génétique, APHP Nord, Hôpital Robert Debré, F-75019 Paris, France; (Y.C.); (H.N.); (L.R.)
| | - Lyse Ruaud
- Département de Génétique, APHP Nord, Hôpital Robert Debré, F-75019 Paris, France; (Y.C.); (H.N.); (L.R.)
- UMR 1141, NEURODIDEROT, INSERM, Université de Paris, F-75019 Paris, France
| | - Benjamin Dauriat
- Department of Cytogenetics and Clinical Genetics, Limoges University Hospital, F-87000 Limoges, France;
| | - Sylvie Bourthoumieu
- Service de Cytogénétique et Génétique Médicale, CHU Limoges, F-87000 Limoges, France;
| | - David Geneviève
- Department of Genetics, Arnaud de Villeneuve University Hospital, F-34000 Montpellier, France;
| | - Séverine Audebert-Bellanger
- Département de Génétique Médicale et Biologie de la Reproduction, CHU Brest, Hôpital Morvan, F-29200 Brest, France;
| | - Mathilde Nizon
- Genetic Medical Department, CHU Nantes, F-44000 Nantes, France;
| | - Radka Stoeva
- Service de Cytogénétique, Centre Hospitalier Universitaire du Mans, F-72000 Le Mans, France;
| | - Geoffroy Hickman
- Department of Dermatology, Reference Center for Rare Skin Diseases MAGEC, Saint Louis Hospital AP-HP, F-75010 Paris, France;
| | - Gaël Nicolas
- Department of Genetics, FHU G4 Génomique, Normandie University, UNIROUEN, CHU Rouen, Inserm U1245, F-76000 Rouen, France;
| | - Juliette Mazereeuw-Hautier
- Département de Dermatologie, Centre de Référence des Maladies Rares de la Peau, CHU de Toulouse, F-31000 Toulouse, France;
| | - Arnaud Jannic
- Département de Dermatologie, AP-HP and UPEC, Hôpital Henri-Mondor, F-94000 Créteil, France; (A.J.); (S.F.); (P.W.)
| | - Salah Ferkal
- Département de Dermatologie, AP-HP and UPEC, Hôpital Henri-Mondor, F-94000 Créteil, France; (A.J.); (S.F.); (P.W.)
- INSERM, Centre d’Investigation Clinique 1430, F-94000 Créteil, France
| | - Béatrice Parfait
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
- Inserm U1016—CNRS UMR8104, Institut Cochin, Université de Paris, CARPEM, F-75014 Paris, France;
| | - Michel Vidaud
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
- Inserm U1016—CNRS UMR8104, Institut Cochin, Université de Paris, CARPEM, F-75014 Paris, France;
| | | | - Pierre Wolkenstein
- Département de Dermatologie, AP-HP and UPEC, Hôpital Henri-Mondor, F-94000 Créteil, France; (A.J.); (S.F.); (P.W.)
| | - Eric Pasmant
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université de Paris, F-75014 Paris, France; (L.P.); (D.V.); (A.B.-S.); (A.C.); (T.M.); (C.B.); (B.P.); (M.V.)
- Inserm U1016—CNRS UMR8104, Institut Cochin, Université de Paris, CARPEM, F-75014 Paris, France;
| |
Collapse
|
267
|
Abstract
Cowden syndrome (CS) is an autosomal dominant condition caused by mutations in the phosphatase and tensin homolog (PTEN) gene, and is characterized by multiple hamartomas and a predisposition to malignant tumors. Characteristic skin lesions include trichilemmomas, acral keratosis, mucocutaneous neuromas, oral papillomas, and penile macules, and are often the first clues to the underlying diagnosis. Here, we discuss the mucocutaneous manifestations of CS, differential diagnoses of genetic causes of each cutaneous finding, genetic analyses for patients with skin manifestations, management of patients with CS, and potential new targeted therapies for CS.
Collapse
Affiliation(s)
- Agnes Lim
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Joanne Ngeow
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
268
|
Deng T, Gong Y, Liao X, Wang X, Zhou X, Zhu G, Mo L. Integrative Analysis of a Novel Eleven-Small Nucleolar RNA Prognostic Signature in Patients With Lower Grade Glioma. Front Oncol 2021; 11:650828. [PMID: 34164339 PMCID: PMC8215672 DOI: 10.3389/fonc.2021.650828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/12/2021] [Indexed: 12/13/2022] Open
Abstract
Objective The present study used the RNA sequencing (RNA-seq) dataset to identify prognostic snoRNAs and construct a prognostic signature of The Cancer Genome Atla (TCGA) lower grade glioma (LGG) cohort, and comprehensive analysis of this signature. Methods RNA-seq dataset of 488 patients from TCGA LGG cohort were included in this study. Comprehensive analysis including function enrichment, gene set enrichment analysis (GSEA), immune infiltration, cancer immune microenvironment, and connectivity map (CMap) were used to evaluate the snoRNAs prognostic signature. Results We identified 21 LGG prognostic snoRNAs and constructed a novel eleven-snoRNA prognostic signature for LGG patients. Survival analysis suggests that this signature is an independent prognostic risk factor for LGG, and the prognosis of LGG patients with a high-risk phenotype is poor (adjusted P = 0.003, adjusted hazard ratio = 2.076, 95% confidence interval = 1.290–3.340). GSEA and functional enrichment analysis suggest that this signature may be involved in the following biological processes and signaling pathways: such as cell cycle, Wnt, mitogen-activated protein kinase, janus kinase/signal transducer and activator of tran-ions, T cell receptor, nuclear factor-kappa B signaling pathway. CMap analysis screened out ten targeted therapy drugs for this signature: 15-delta prostaglandin J2, MG-262, vorinostat, 5155877, puromycin, anisomycin, withaferin A, ciclopirox, chloropyrazine and megestrol. We also found that high- and low-risk score phenotypes of LGG patients have significant differences in immune infiltration and cancer immune microenvironment. Conclusions The present study identified a novel eleven-snoRNA prognostic signature of LGG and performed a integrative analysis of its molecular mechanisms and relationship with tumor immunity.
Collapse
Affiliation(s)
- Teng Deng
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yizhen Gong
- Evidence-based Medicine Teaching and Research Section, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ligen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
269
|
Payne JM, Haebich KM, MacKenzie R, Walsh KS, Hearps SJC, Coghill D, Barton B, Pride NA, Ullrich NJ, Tonsgard JH, Viskochil D, Schorry EK, Klesse L, Fisher MJ, Gutmann DH, Rosser T, Packer RJ, Korf B, Acosta MT, Bellgrove MA, North KN. Cognition, ADHD Symptoms, and Functional Impairment in Children and Adolescents With Neurofibromatosis Type 1. J Atten Disord 2021; 25:1177-1186. [PMID: 31838937 DOI: 10.1177/1087054719894384] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective: We examined the contribution of attention and executive cognitive processes to ADHD symptomatology in NF1, as well as the relationships between cognition and ADHD symptoms with functional outcomes. Methods: The study sample consisted of 141 children and adolescents with NF1. Children were administered neuropsychological tests that assessed attention and executive function, from which latent cognitive variables were derived. ADHD symptomatology, adaptive skills, and quality of life (QoL) were assessed using parent-rated questionnaires. Path analyses were conducted to test relationships among cognitive functioning, ADHD symptomatology, and functional outcomes. Results: Significant deficits were observed on all outcome variables. Cognitive variables did not predict ADHD symptomatology. Neither did they predict functional outcomes. However, elevated ADHD symptomatology significantly predicted functional outcomes. Conclusion: Irrespective of cognitive deficits, elevated ADHD symptoms in children with NF1 negatively impact daily functioning and emphasize the importance of interventions aimed at minimizing ADHD symptoms in NF1.
Collapse
Affiliation(s)
- Jonathan M Payne
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,University of Melbourne, Parkville, Victoria, Australia
| | - Kristina M Haebich
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | | | - Karin S Walsh
- Children's National Health System, Washington, DC, USA
| | - Stephen J C Hearps
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - David Coghill
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,University of Melbourne, Parkville, Victoria, Australia
| | - Belinda Barton
- The Children's Hospital at Westmead, New South Wales, Australia.,The University of Sydney, New South Wales, Australia
| | - Natalie A Pride
- The Children's Hospital at Westmead, New South Wales, Australia.,The University of Sydney, New South Wales, Australia
| | | | - James H Tonsgard
- The University of Chicago Medicine Comer Children's Hospital, IL, USA
| | | | | | - Laura Klesse
- University of Texas Southwestern Medical Center, Dallas, USA
| | | | | | | | | | - Bruce Korf
- The University of Alabama at Birmingham, USA
| | | | | | - Kathryn N North
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
270
|
Jalabert M, Ferkal S, Souberbielle JC, Sbidian E, Mageau A, Eymard F, Le Corvoisier P, Allanore L, Chevalier X, Wolkenstein P, Guignard S. Bone Status According to Neurofibromatosis Type 1 Phenotype: A Descriptive Study of 60 Women in France. Calcif Tissue Int 2021; 108:738-745. [PMID: 33558959 DOI: 10.1007/s00223-021-00807-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022]
Abstract
There is an increased risk of osteoporosis and an abnormal bone turn over in neurofibromatosis 1 (NF1). Our objective is to evaluate bone status in NF1 and to look for associations with cutaneous phenotype. We conducted a descriptive, monocentric study. We included 60 NF1 women, 18-51 years old, non-menopausal, divided in 2 groups: «at risk phenotype» (ARP) composed by 30 patients with at least 2 subcutaneous neurofibromas (SC-NF) and «classical phenotype» (CP) composed by 30 patients with none or 1 SC-NF. We evaluated low bone mineral density (BMD) risk factors and measured BMD, calcium and phosphorus homeostasis and bone turnover markers. Before 50 years old, Z-score has to be used to assess BMD. Z-score < - 2 is below expected range and represents 2.5% of the population. There was no difference between the two groups. Overall, Z-scores were low and 5 patients had a Z-score < - 2 (8.3%), which is 3 times general population low BMD frequency. 10 fragility fractures occurred in 8 patients, among which 2 were vertebral fractures. 85% had low calcium intake. 12 patients had hypophosphoremia, 25 elevated PTH. Vitamin D levels were low for 86.4%. 41 patients (69.5%) had at least one abnormal bone turnover markers. Low BMD is 3.3 times more frequent in NF1 than in general population, with high fracture risk, regardless of the skin phenotype, classical or at risk, because of high bone turn over and secondary hyperparathyroidism due to vitamin D deficiency and poor calcium intake.
Collapse
Affiliation(s)
- Maud Jalabert
- Rhumatologie, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France.
| | - Salah Ferkal
- Centre d'investigation clinique, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | | | - Emilie Sbidian
- Dermatologie, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Arthur Mageau
- Médecine Interne, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Florent Eymard
- Rhumatologie, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Philippe Le Corvoisier
- Centre d'investigation clinique, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Laurence Allanore
- Dermatologie, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Xavier Chevalier
- Rhumatologie, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Pierre Wolkenstein
- Dermatologie, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Sandra Guignard
- Rhumatologie, Hôpital Henri Mondor, 51 Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| |
Collapse
|
271
|
Florou C, Aissopou E, Chalkiadaki E, Andreanos K, Koutsandrea C, Papaconstantinou D, Georgalas I. Corneal endothelial cells and central corneal thickness in patients with neurofibromatosis type 1. Indian J Ophthalmol 2021; 69:1522-1526. [PMID: 34011734 PMCID: PMC8302278 DOI: 10.4103/ijo.ijo_1967_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose: The aim of this study was to evaluate the morphological properties of corneal endothelial cells and central corneal thickness (CCT) in patients with neurofibromatosis type 1 (NF1) and to compare them with age-matched healthy controls. Methods: Nineteen NF1 patients and 38 healthy individuals were recruited. All participants underwent complete ophthalmological examination as well as noncontact specular microscopy to measure endothelial cell density (ECD), average cell area (AVG), coefficient of variation of cell area (CV), the percentage of hexagonal cells, and CCT. Eyes with previous ocular trauma, inflammation or surgery, and preexisting corneal and ocular surface diseases were excluded. Results: NF1 patients had higher ECD compared to healthy controls of the same age (2764.2 ± 270.4 versus 2570.4 ± 449.2 cells/mm, respectively), although at a borderline level (P = 0.051). Patients with NF1 presented significantly lower CV and AVG when compared to controls (32.9 ± 4.6 versus 37.8 ± 9.5%, P = 0.011 and 364.9 ± 34.4 versus 406.0 ± 107.4 µm, P = 0.038, respectively). The NF1 group had significantly higher hexagonality in comparison with controls (55.7 ± 6.5 versus 50.5 ± 9.9%, P = 0.025). CCT was similar between the two groups (P = 0.955). Conclusion: Our results show that corneal endothelium has more favorable morphological characteristics in NF1 patients compared to healthy individuals of the same age.
Collapse
Affiliation(s)
- Chrysoula Florou
- First Department of Ophthalmology, National and Kapodistrian University of Athens, General Hospital "G. Gennimatas", Athens, Greece
| | - Evaggelia Aissopou
- Ophthalmologist in Private Office, Papadiamantopoulou 186, Athens, Greece
| | - Evangelia Chalkiadaki
- First Department of Ophthalmology, National and Kapodistrian University of Athens, General Hospital "G. Gennimatas", Athens, Greece
| | | | - Chrysanthi Koutsandrea
- First Department of Ophthalmology, National and Kapodistrian University of Athens, General Hospital "G. Gennimatas", Athens, Greece
| | - Dimitrios Papaconstantinou
- First Department of Ophthalmology, National and Kapodistrian University of Athens, General Hospital "G. Gennimatas", Athens, Greece
| | - Ilias Georgalas
- First Department of Ophthalmology, National and Kapodistrian University of Athens, General Hospital "G. Gennimatas", Athens, Greece
| |
Collapse
|
272
|
Revised diagnostic criteria for neurofibromatosis type 1 and Legius syndrome: an international consensus recommendation. Genet Med 2021; 23:1506-1513. [PMID: 34012067 PMCID: PMC8354850 DOI: 10.1038/s41436-021-01170-5] [Citation(s) in RCA: 333] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 01/08/2023] Open
Abstract
Purpose By incorporating major developments in genetics, ophthalmology, dermatology, and neuroimaging, to revise the diagnostic criteria for neurofibromatosis type 1 (NF1) and to establish diagnostic criteria for Legius syndrome (LGSS). Methods We used a multistep process, beginning with a Delphi method involving global experts and subsequently involving non-NF experts, patients, and foundations/patient advocacy groups. Results We reached consensus on the minimal clinical and genetic criteria for diagnosing and differentiating NF1 and LGSS, which have phenotypic overlap in young patients with pigmentary findings. Criteria for the mosaic forms of these conditions are also recommended. Conclusion The revised criteria for NF1 incorporate new clinical features and genetic testing, whereas the criteria for LGSS were created to differentiate the two conditions. It is likely that continued refinement of these new criteria will be necessary as investigators (1) study the diagnostic properties of the revised criteria, (2) reconsider criteria not included in this process, and (3) identify new clinical and other features of these conditions. For this reason, we propose an initiative to update periodically the diagnostic criteria for NF1 and LGSS. ![]()
Collapse
|
273
|
Bergqvist C, Hemery F, Jannic A, Ferkal S, Wolkenstein P. Lymphoproliferative malignancies in patients with neurofibromatosis 1. Orphanet J Rare Dis 2021; 16:230. [PMID: 34011343 PMCID: PMC8136208 DOI: 10.1186/s13023-021-01856-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
Neurofibromatosis 1 (NF1) is an inherited, autosomal-dominant, tumor predisposition syndrome with a birth incidence as high as 1:2000. A patient with NF1 is four to five times more likely to develop a malignancy as compared to the general population. The number of epidemiologic studies on lymphoproliferative malignancies in patients with NF1 is limited. The aim of this study was to determine the incidence rate of lymphoproliferative malignancies (lymphoma and leukemia) in NF1 patients followed in our referral center for neurofibromatoses. We used the Informatics for Integrated Biology and the Bedside (i2b2) platform to extract information from the hospital’s electronic health records. We performed a keyword search on clinical notes generated between Jan/01/2014 and May/11/2020 for patients aged 18 years or older. A total of 1507 patients with confirmed NF1 patients aged 18 years and above were identified (mean age 39.2 years; 57% women). The total number of person-years in follow-up was 57,736 (men, 24,327 years; women, 33,409 years). Mean length of follow-up was 38.3 years (median, 36 years). A total of 13 patients had a medical history of either lymphoma or leukemia, yielding an overall incidence rate of 22.5 per 100,000 (0.000225, 95% confidence interval (CI) 0.000223–0.000227). This incidence is similar to that of the general population in France (standardized incidence ratio 1.07, 95% CI 0.60–1.79). Four patients had a medical history leukemia and 9 patients had a medical history of lymphoma of which 7 had non-Hodgkin lymphoma, and 2 had Hodgkin lymphoma. Our results show that adults with NF1 do not have an increased tendency to develop lymphoproliferative malignancies, in contrast to the general increased risk of malignancy. While our results are consistent with the recent population-based study in Finland, they are in contrast with the larger population-based study in England whereby NF1 individuals were found to be 3 times more likely to develop both non-Hodgkin lymphoma and lymphocytic leukemia. Large-scale epidemiological studies based on nationwide data sets are thus needed to confirm our findings.
Collapse
Affiliation(s)
- Christina Bergqvist
- Faculty of Medicine, Université Paris-Est Creteil (UPEC), 94010, Créteil, France.,Service de Dermatologie, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, 94010, Créteil, France
| | - François Hemery
- Department of Medical Informatics, Hôpital Henri-Mondor, AP-HP, 94010, Créteil, France
| | - Arnaud Jannic
- Service de Dermatologie, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, 94010, Créteil, France
| | - Salah Ferkal
- Service de Dermatologie, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, 94010, Créteil, France.,INSERM, Centre d'Investigation Clinique 006, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, Referral Center of Neurofibromatoses, 94010, Créteil, France
| | - Pierre Wolkenstein
- Faculty of Medicine, Université Paris-Est Creteil (UPEC), 94010, Créteil, France. .,Service de Dermatologie, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, 94010, Créteil, France. .,INSERM, Centre d'Investigation Clinique 006, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, Referral Center of Neurofibromatoses, 94010, Créteil, France. .,Faculty of Medicine of Paris East Créteil, Department of Dermatology, French Referral Center for Neurofibromatoses, Henri-Mondor Hospital, APHP, University Paris Est Créteil, 94010, Créteil Cedex, France.
| |
Collapse
|
274
|
Kazarian AG, West JM, Brown JA, Erickson BA, Gellhaus PT. Large para-testicular intra-scrotal malignant peripheral nerve sheath tumor managed with radical penectomy: A case report. Urol Case Rep 2021; 38:101695. [PMID: 33996500 PMCID: PMC8102162 DOI: 10.1016/j.eucr.2021.101695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 11/27/2022] Open
Abstract
Neurofibromatosis 1 is a relatively rare genetic disease characterized by widespread neurofibromas originating from the peripheral nervous system. Most growths are benign, but some carry a risk of transformation to malignant peripheral nerve sheath tumors. Although these growths can be found anywhere in the body, they are rarely found in the male external genitalia. This report discusses a case of a 25-year-old male patient with neurofibromatosis 1 presenting with a scrotal mass found to have a very large para-testicular intra-scrotal malignant peripheral nerve sheath tumor that required testicle-sparing radical penectomy.
Collapse
Affiliation(s)
| | - Jeremy M. West
- Department of Urology, University of Iowa, Iowa City, IA, USA
| | - James A. Brown
- Department of Urology, University of Iowa, Iowa City, IA, USA
| | | | - Paul T. Gellhaus
- Department of Urology, University of Iowa, Iowa City, IA, USA
- Corresponding author. Department of Urology, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA, 52242, USA.
| |
Collapse
|
275
|
Chevalier B, Dupuis H, Jannin A, Lemaitre M, Do Cao C, Cardot-Bauters C, Espiard S, Vantyghem MC. Phakomatoses and Endocrine Gland Tumors: Noteworthy and (Not so) Rare Associations. Front Endocrinol (Lausanne) 2021; 12:678869. [PMID: 34025587 PMCID: PMC8134657 DOI: 10.3389/fendo.2021.678869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/15/2021] [Indexed: 11/13/2022] Open
Abstract
Phakomatoses encompass a group of rare genetic diseases, such as von Hippel-Lindau syndrome (VHL), neurofibromatosis type 1 (NF1), tuberous sclerosis complex (TSC) and Cowden syndrome (CS). These disorders are due to molecular abnormalities on the RAS-PI3K-Akt-mTOR pathway for NF1, TSC and CS, and to hypoxia sensing for VHL. Phakomatoses share some phenotypic traits such as neurological, ophthalmological and cutaneous features. Patients with these diseases are also predisposed to developing multiple endocrine tissue tumors, e.g., pheochromocytomas/paragangliomas are frequent in VHL and NF1. All forms of phakomatoses except CS may be associated with digestive neuroendocrine tumors. More rarely, thyroid cancer and pituitary or parathyroid adenomas have been reported. These susceptibilities are noteworthy, because their occurrence rate, prognosis and management differ slightly from the sporadic forms. The aim of this review is to summarize current knowledge on endocrine glands tumors associated with VHL, NF1, TSC, and CS, especially neuroendocrine tumors and pheochromocytomas/paragangliomas. We particularly detail recent advances concerning prognosis and management, especially parenchyma-sparing surgery and medical targeted therapies such as mTOR, MEK and HIF-2 α inhibitors, which have shown truly encouraging results.
Collapse
Affiliation(s)
- Benjamin Chevalier
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
- University of Lille, Lille, France
| | - Hippolyte Dupuis
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
| | - Arnaud Jannin
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
- University of Lille, Lille, France
| | - Madleen Lemaitre
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
- University of Lille, Lille, France
| | - Christine Do Cao
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
| | - Catherine Cardot-Bauters
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
| | - Stéphanie Espiard
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
- University of Lille, Lille, France
- INSERM U1190, European Genomic Institute for Diabetes, Lille, France
| | - Marie Christine Vantyghem
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
- University of Lille, Lille, France
- INSERM U1190, European Genomic Institute for Diabetes, Lille, France
| |
Collapse
|
276
|
Konig MF, Sathiyakumar V, Phan CM, Schulman SP, Gelber AC. Inside and Out. N Engl J Med 2021; 384:1753-1760. [PMID: 33951365 PMCID: PMC9055568 DOI: 10.1056/nejmcps2034360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Maximilian F Konig
- From the Department of Medicine (M.F.K., V.S., S.P.S., A.C.G.), the Division of Rheumatology (M.F.K., A.C.G.), the Division of Cardiology (V.S., S.P.S.), the Division of Dermatopathology (C.M.P.), and the Department of Dermatology (C.M.P.), Johns Hopkins University School of Medicine, Baltimore
| | - Vasanth Sathiyakumar
- From the Department of Medicine (M.F.K., V.S., S.P.S., A.C.G.), the Division of Rheumatology (M.F.K., A.C.G.), the Division of Cardiology (V.S., S.P.S.), the Division of Dermatopathology (C.M.P.), and the Department of Dermatology (C.M.P.), Johns Hopkins University School of Medicine, Baltimore
| | - Casey M Phan
- From the Department of Medicine (M.F.K., V.S., S.P.S., A.C.G.), the Division of Rheumatology (M.F.K., A.C.G.), the Division of Cardiology (V.S., S.P.S.), the Division of Dermatopathology (C.M.P.), and the Department of Dermatology (C.M.P.), Johns Hopkins University School of Medicine, Baltimore
| | - Steven P Schulman
- From the Department of Medicine (M.F.K., V.S., S.P.S., A.C.G.), the Division of Rheumatology (M.F.K., A.C.G.), the Division of Cardiology (V.S., S.P.S.), the Division of Dermatopathology (C.M.P.), and the Department of Dermatology (C.M.P.), Johns Hopkins University School of Medicine, Baltimore
| | - Allan C Gelber
- From the Department of Medicine (M.F.K., V.S., S.P.S., A.C.G.), the Division of Rheumatology (M.F.K., A.C.G.), the Division of Cardiology (V.S., S.P.S.), the Division of Dermatopathology (C.M.P.), and the Department of Dermatology (C.M.P.), Johns Hopkins University School of Medicine, Baltimore
| |
Collapse
|
277
|
Belakhoua SM, Rodriguez FJ. Diagnostic Pathology of Tumors of Peripheral Nerve. Neurosurgery 2021; 88:443-456. [PMID: 33588442 DOI: 10.1093/neuros/nyab021] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Neoplasms of the peripheral nervous system represent a heterogenous group with a wide spectrum of morphological features and biological potential. They range from benign and curable by complete excision (schwannoma and soft tissue perineurioma) to benign but potentially aggressive at the local level (plexiform neurofibroma) to the highly malignant (malignant peripheral nerve sheath tumors [MPNST]). In this review, we discuss the diagnostic and pathologic features of common peripheral nerve sheath tumors, particularly those that may be encountered in the intracranial compartment or in the spine and paraspinal region. The discussion will cover schwannoma, neurofibroma, atypical neurofibromatous neoplasms of uncertain biological potential, intraneural and soft tissue perineurioma, hybrid nerve sheath tumors, MPNST, and the recently renamed enigmatic tumor, malignant melanotic nerve sheath tumor, formerly referred to as melanotic schwannoma. We also discuss the diagnostic relevance of these neoplasms to specific genetic and familial syndromes of nerve, including neurofibromatosis 1, neurofibromatosis 2, and schwannomatosis. In addition, we discuss updates in our understanding of the molecular alterations that represent key drivers of these neoplasms, including neurofibromatosis type 1 and type 2, SMARCB1, LZTR1, and PRKAR1A loss, as well as the acquisition of CDKN2A/B mutations and alterations in the polycomb repressor complex members (SUZ12 and EED) in the malignant progression to MPNST. In summary, this review covers practical aspects of pathologic diagnosis with updates relevant to neurosurgical practice.
Collapse
Affiliation(s)
- Sarra M Belakhoua
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- School of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Fausto J Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sydney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
278
|
Exploring associations between constipation, severity of neurofibromatosis type 1 and NF1 mutational spectrum. Sci Rep 2021; 11:9179. [PMID: 33911094 PMCID: PMC8080678 DOI: 10.1038/s41598-021-87686-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is inherited in an autosomal dominant manner and is a rather common rare disease. Until recently, studies on gastrointestinal symptoms in patients with NF1 have been few and mostly described as case reports. In three previously published studies, the frequency of constipation in patients with NF1 has been found to be as high as 30%. In this study, associations between the frequency of constipation and NF1 disease severity and NF1 mutational spectrum were investigated. Among 277 patients with NF1, 49 had constipation. The highest rate of constipation was found among patients with a high perception of NF1 illness burden, and patients with constipation had a significantly higher NF1 illness burden when comparing the “not bothered” and the “very bothered” (p = 0.013). We found no significant association between constipation and the remaining measures on severity of NF1, nor between constipation and genetic variants. When observing the NF1 mutational spectrum, one variant (c.1013A>G (p.Asp338Gly/p.?) was identified in three patients with constipation of which two patients were related. The variant c.2970_2972delAAT (p.Met992del) associated with a mild NF1 phenotype was identified in two related patients with constipation. This study is the first to explore the association between symptoms of constipation, NF1 severity, and NF1 mutational spectrum. The results suggest an association between constipation and a high degree of illness burden. Awareness of this association among physicians could lead to more patients with NF1 being diagnosed with constipation. Constipation impacts on quality of life, hence a timely diagnosis and treatment will improve quality of life.
Collapse
|
279
|
Laurent D, Smith AE, Bessler WK, Mendonca M, Chin-Sinex H, Descovich M, Horvai AE, Clapp DW, Nakamura JL. Irradiation of Nf1 mutant mouse models of spinal plexiform neurofibromas drives pathologic progression and decreases survival. Neurooncol Adv 2021; 3:vdab063. [PMID: 34131650 PMCID: PMC8193912 DOI: 10.1093/noajnl/vdab063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Genetically susceptible individuals can develop malignancies after irradiation of normal tissues. In the context of therapeutic irradiation, it is not known whether irradiating benign neoplasms in susceptible individuals promotes neoplastic transformation and worse clinical outcomes. Individuals with Neurofibromatosis 1 (NF1) are susceptible to both radiation-induced second malignancies and spontaneous progression of plexiform neurofibromas (PNs) to malignant peripheral nerve sheath tumors (MPNSTs). The role of radiotherapy in the treatment of benign neoplasms such as PNs is unclear. Methods To test whether radiotherapy promotes neoplastic progression of PNs and reduces overall survival, we administered spinal irradiation (SI) to conditional knockout mouse models of NF1-associated PNs in 2 germline contexts: Nf1fllfl; PostnCre+ and Nf1fl/-; PostnCre+. Both genotypes develop extensive Nf1 null spinal PNs, modeling PNs in NF1 patients. A total of 101 mice were randomized to 0 Gy, 15 Gy (3 Gy × 5), or 30 Gy (3 Gy × 10) of spine-focused, fractionated SI and aged until signs of illness. Results SI decreased survival in both Nf1fllfl mice and Nf1fl/- mice, with the worst overall survival occurring in Nf1fl/- mice receiving 30 Gy. SI was also associated with increasing worrisome histologic features along the PN-MPNST continuum in PNs irradiated to higher radiation doses. Conclusions This preclinical study provides experimental evidence that irradiation of pre-existing PNs reduces survival and may shift PNs to higher grade neoplasms.
Collapse
Affiliation(s)
- Danny Laurent
- Department of Radiation Oncology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Abbi E Smith
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Waylan K Bessler
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marc Mendonca
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Helen Chin-Sinex
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Martina Descovich
- Department of Radiation Oncology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Andrew E Horvai
- Department of Pathology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - D Wade Clapp
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jean L Nakamura
- Department of Radiation Oncology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
280
|
Scala M, Schiavetti I, Madia F, Chelleri C, Piccolo G, Accogli A, Riva A, Salpietro V, Bocciardi R, Morcaldi G, Di Duca M, Caroli F, Verrico A, Milanaccio C, Viglizzo G, Traverso M, Baldassari S, Scudieri P, Iacomino M, Piatelli G, Minetti C, Striano P, Garrè ML, De Marco P, Diana MC, Capra V, Pavanello M, Zara F. Genotype-Phenotype Correlations in Neurofibromatosis Type 1: A Single-Center Cohort Study. Cancers (Basel) 2021; 13:cancers13081879. [PMID: 33919865 PMCID: PMC8070780 DOI: 10.3390/cancers13081879] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Neurofibromatosis type 1 (NF1) is a complex disorder characterized by a multisystem involvement and cancer predisposition. It is caused by genetic variants in NF1, a large tumor suppressor gene encoding a cytoplasmatic protein (neurofibromin) with a regulatory role in essential cellular processes. Genotype–phenotype correlations in NF1 patients are so far elusive. We retrospectively reviewed clinical, radiological, and genetic data of 583 individuals with at least 1 National Institutes of Health (NIH) criterion for NF1 diagnosis, including 365 subjects fulfilling criteria for the diagnosis. Novel genotype–phenotype correlations were identified through uni- and multivariate statistical analysis. Missense variants negatively correlated with neurofibromas. Skeletal abnormalities were associated with frameshift variants and whole gene deletions. The c.3721C>T; p.(R1241*) variant positively correlated with structural brain alterations, whereas the c.6855C>A; p.(Y2285*) variant was associated with a higher prevalence of Lisch nodules and endocrinological disorders. These novel NF1 genotype–phenotype correlations may have a relevant role in the implementation of patients’ care. Abstract Neurofibromatosis type 1 (NF1) is a proteiform genetic condition caused by pathogenic variants in NF1 and characterized by a heterogeneous phenotypic presentation. Relevant genotype–phenotype correlations have recently emerged, but only few pertinent studies are available. We retrospectively reviewed clinical, instrumental, and genetic data from a cohort of 583 individuals meeting at least 1 diagnostic National Institutes of Health (NIH) criterion for NF1. Of these, 365 subjects fulfilled ≥2 NIH criteria, including 235 pediatric patients. Genetic testing was performed through cDNA-based sequencing, Next Generation Sequencing (NGS), and Multiplex Ligation-dependent Probe Amplification (MLPA). Uni- and multivariate statistical analysis was used to investigate genotype–phenotype correlations. Among patients fulfilling ≥ 2 NIH criteria, causative single nucleotide variants (SNVs) and copy number variations (CNVs) were detected in 267/365 (73.2%) and 20/365 (5.5%) cases. Missense variants negatively correlated with neurofibromas (p = 0.005). Skeletal abnormalities were associated with whole gene deletions (p = 0.05) and frameshift variants (p = 0.006). The c.3721C>T; p.(R1241*) variant positively correlated with structural brain alterations (p = 0.031), whereas Lisch nodules (p = 0.05) and endocrinological disorders (p = 0.043) were associated with the c.6855C>A; p.(Y2285*) variant. We identified novel NF1 genotype–phenotype correlations and provided an overview of known associations, supporting their potential relevance in the implementation of patient management.
Collapse
Affiliation(s)
- Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Irene Schiavetti
- Department of Health Sciences, Section of Biostatistics, University of Genova, 16132 Genoa, Italy;
| | - Francesca Madia
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Cristina Chelleri
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Gianluca Piccolo
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Andrea Accogli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
| | - Antonella Riva
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Vincenzo Salpietro
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Renata Bocciardi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Guido Morcaldi
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Marco Di Duca
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Francesco Caroli
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Antonio Verrico
- Neuro-Oncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (A.V.); (C.M.); (M.L.G.)
| | - Claudia Milanaccio
- Neuro-Oncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (A.V.); (C.M.); (M.L.G.)
| | | | - Monica Traverso
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Simona Baldassari
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Paolo Scudieri
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Michele Iacomino
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Gianluca Piatelli
- Neurosurgery Department, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy;
| | - Carlo Minetti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Maria Luisa Garrè
- Neuro-Oncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (A.V.); (C.M.); (M.L.G.)
| | - Patrizia De Marco
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Maria Cristina Diana
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (G.P.); (G.M.); (M.T.); (M.C.D.)
| | - Valeria Capra
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| | - Marco Pavanello
- Neurosurgery Department, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy;
- Correspondence:
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy; (M.S.); (C.C.); (A.A.); (A.R.); (V.S.); (R.B.); (P.S.); (C.M.); (P.S.); (F.Z.)
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, University of Genoa, 16147 Genoa, Italy; (F.M.); (M.D.D.); (F.C.); (S.B.); (M.I.); (P.D.M.); (V.C.)
| |
Collapse
|
281
|
Clinical features and disease severity in patients with mosaic neurofibromatosis type 1: a single-center study and literature review. Orphanet J Rare Dis 2021; 16:180. [PMID: 33853649 PMCID: PMC8048193 DOI: 10.1186/s13023-021-01796-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/30/2021] [Indexed: 02/08/2023] Open
Abstract
Background The mosaic form of neurofibromatosis type 1 (NF1) is called mosaic NF1 (MNF1). No specific MNF1 follow-up guidelines exist. It is debatable if patients with MNF1 should be clinically examined and undergo follow-up in accordance with the standard NF1 guidelines, as MNF1 patients more often may develop more benign phenotypes and thereby less disease-associated complications including cognitive impairment. We discussed the need for a specific MNF1 follow-up guideline with focus on frequency of plexiform neurofibromas and NF1-associated complications. Method A systematic retrospective data collection in a MNF1 cohort from one of two Danish national centers of NF1 Expertise was completed. Data collected included demographics, clinical features including NF1 diagnostic criteria and NF1-associated complications. Recent literature in the field was reviewed. Results We identified 17 patients with MNF1 with a median age of 37 years [4; 66]. Eleven (65%) were females. Five patients (30%) had a plexiform neurofibroma. The median age at detection of plexiform neurofibroma was 30 years [14; 60]. Nine (53%) had at least one NF1-related complication; scoliosis, hypertension, ADHD, learning disability, language delay, autism and delay in gross and fine motor function development. We reviewed nine articles. In total, 126 cases were described within three case-series. Nineteen (15%) had a plexiform neurofibroma and in total, 23 NF1-associated complications were reported including language delay, learning disability and skeletal abnormalities. Furthermore, from the literature it was evident that the diagnosing of MNF1 varies among physicians and across countries. Conclusion Patients with MNF1 present with plexiform neurofibromas and other NF1-related complications with a frequency requiring that follow-up of MNF1 patients should be in accordance with the standard NF1 guideline in both childhood and adulthood. Physicians should be aware of cognitive impairment as a complication to MNF1. To develop a specific MNF1 follow-up guideline, there is a need for an international consensus on the diagnostic criteria for MNF1 and a follow-up study conducted in a larger MNF1 cohort.
Collapse
|
282
|
Vasudevan HN, Lucas CHG, Villanueva-Meyer JE, Theodosopoulos PV, Raleigh DR. Genetic Events and Signaling Mechanisms Underlying Schwann Cell Fate in Development and Cancer. Neurosurgery 2021; 88:234-245. [PMID: 33094349 DOI: 10.1093/neuros/nyaa455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/08/2020] [Indexed: 01/08/2023] Open
Abstract
In this review, we describe Schwann cell development from embryonic neural crest cells to terminally differentiated myelinated and nonmyelinated mature Schwann cells. We focus on the genetic drivers and signaling mechanisms mediating decisions to proliferate versus differentiate during Schwann cell development, highlighting pathways that overlap with Schwann cell development and are dysregulated in tumorigenesis. We conclude by considering how our knowledge of the events underlying Schwann cell development and mouse models of schwannoma, neurofibroma, and malignant peripheral nerve sheath tumor can inform novel therapeutic strategies for patients with cancers derived from Schwann cell lineages.
Collapse
Affiliation(s)
- Harish N Vasudevan
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Calixto-Hope G Lucas
- Department of Anatomic Pathology, University of California, San Francisco, San Francisco, California
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Philip V Theodosopoulos
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - David R Raleigh
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
283
|
Bowers DC, Verbruggen LC, Kremer LCM, Hudson MM, Skinner R, Constine LS, Sabin ND, Bhangoo R, Haupt R, Hawkins MM, Jenkinson H, Khan RB, Klimo P, Pretorius P, Ng A, Reulen RC, Ronckers CM, Sadighi Z, Scheinemann K, Schouten-van Meeteren N, Sugden E, Teepen JC, Ullrich NJ, Walter A, Wallace WH, Oeffinger KC, Armstrong GT, van der Pal HJH, Mulder RL. Surveillance for subsequent neoplasms of the CNS for childhood, adolescent, and young adult cancer survivors: a systematic review and recommendations from the International Late Effects of Childhood Cancer Guideline Harmonization Group. Lancet Oncol 2021; 22:e196-e206. [PMID: 33845037 DOI: 10.1016/s1470-2045(20)30688-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/28/2020] [Accepted: 11/06/2020] [Indexed: 11/17/2022]
Abstract
Exposure to cranial radiotherapy is associated with an increased risk of subsequent CNS neoplasms among childhood, adolescent, and young adult (CAYA) cancer survivors. Surveillance for subsequent neoplasms can translate into early diagnoses and interventions that could improve cancer survivors' health and quality of life. The practice guideline presented here by the International Late Effects of Childhood Cancer Guideline Harmonization Group was developed with an evidence-based method that entailed the gathering and appraisal of published evidence associated with subsequent CNS neoplasms among CAYA cancer survivors. The preparation of these guidelines showed a paucity of high-quality evidence and highlighted the need for additional research to inform survivorship care. The recommendations are based on careful consideration of the evidence supporting the benefits, risks, and harms of the surveillance interventions, clinical judgment regarding individual patient circumstances, and the need to maintain flexibility of application across different health-care systems. Currently, there is insufficient evidence to establish whether early detection of subsequent CNS neoplasms reduces morbidity and mortality, and therefore no recommendation can be formulated for or against routine MRI surveillance. The decision to start surveillance should be made by the CAYA cancer survivor and health-care provider after careful consideration of the potential harms and benefits of surveillance for CNS neoplasms, including meningioma.
Collapse
Affiliation(s)
- Daniel C Bowers
- Division of Pediatric Hematology/Oncology, Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | | | | - Melissa M Hudson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Roderick Skinner
- Department of Paediatric and Adolescent Haematology and Oncology, Great North Children's Hospital, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Louis S Constine
- Department of Radiation Oncology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Noah D Sabin
- Department of Diagnostic Imaging, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ranjeev Bhangoo
- Neurosurgical Department, King's College Hospital Foundation Trust, London, UK
| | - Riccardo Haupt
- Epidemiology and Biostatistics Unit and DOPO Clinic, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Mike M Hawkins
- Centre for Childhood Cancer Survivor Studies, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Helen Jenkinson
- Department of Paediatric Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Raja B Khan
- Department of Pediatrics, Division of Neurology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul Klimo
- Department of Neurosurgery, University of Tennessee, Memphis, TN, USA
| | - Pieter Pretorius
- Department of Neuroradiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Antony Ng
- Department of Paediatric Oncology, Royal Hospital for Children, Bristol, UK
| | - Raoul C Reulen
- Centre for Childhood Cancer Survivor Studies, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Cécile M Ronckers
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands; Institute for Biostatistics and Registry Research, Brandenburg Medical School, Neuruppin, Germany
| | - Zsila Sadighi
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katrin Scheinemann
- Division of Oncology-Hematology, Department of Pediatrics, Kantonsspital Aarau, Switzerland; Division of Hematology & Oncology, University Children's Hospital Basel, University of Basel, Switzerland; Department of Pediatrics, McMaster Children's Hospital, McMaster University, Hamilton, ON, Canada
| | | | | | - Jop C Teepen
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Nicole J Ullrich
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Walter
- Department of Pediatrics, Sidney Kimmel Medical College, Thomas Jefferson University, Wilmington, DE, USA; Department of Pediatrics, A I duPont Hospital for Children, Wilmington, DE, USA
| | - W Hamish Wallace
- Department of Paediatric Oncology, Royal Hospital for Sick Children, Edinburgh, UK
| | - Kevin C Oeffinger
- Department of Community and Family Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Gregory T Armstrong
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA; Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Renée L Mulder
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
284
|
Pajavand AM, Sharifi G, Anvari A, Bidari-Zerehpoosh F, Shamsi MA, Nateghinia S, Meybodi TE. Case Report: Chemotherapy Indication in a Case of Neurofibromatosis Type 1 Presenting Optic Pathway Glioma: A One-Year Clinical Case Study Using Differential Tractography Approach. Front Hum Neurosci 2021; 15:620439. [PMID: 33994974 PMCID: PMC8115022 DOI: 10.3389/fnhum.2021.620439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/01/2021] [Indexed: 11/30/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is associated with peripheral and central nervous system tumors. It is noteworthy that the regions in which these tumors frequently arise are the optic pathways (OPs) and the brainstem. Thus, we decided to trace the procedure of diffusion Magnetic Resonance Imaging (dMRI) alterations along with Short-Wavelength Automated Perimetry (SWAP) examinations of the OPs after surgery and chemotherapy over 1 year, which enabled us to evaluate chemotherapy's efficacy in an NF1 patient with an OP tumor. In this study, a 25-year-old woman with NF1 and left optic radiation (OR) glioma underwent surgery to remove the glioma. Immunohistochemistry (IHC) revealed a Pilocytic Astrocytoma (PA) WHO grade I. Post-operation chemotherapy done using nine treatment cycles of administering Temozolomide (TMZ) for 5 days every 4 weeks. Applying the region of interest (ROI) differential tractography method and SWAP four times every 3 months allowed us to follow the patient's visual acuity alterations longitudinally. The differential deterministic tractography method and statistical analyses enabled us to discover the white matter (WM) tracts anisotropy alterations over time. Furthermore, statistical analyses on the SWAP results along time illustrated possible alterations in visual acuity. Then, we could compare and associate the findings with the SWAP examinations and patient symptoms longitudinally. Statistical analyses of SWAP tests revealed a significant improvement in visual fields, and longitudinal differential tractography showed myelination and dense axonal packing in the left OR after 1 year of treatment. In this study, we examined an old hypothesis suggesting that chemotherapy is more effective than radiotherapy for NF1 patients with OP gliomas (OPGs) because of the radiation side effects on the visual field, cognition, and cerebrovascular complications. Our longitudinal clinical case study involving dMRI and SWAP on a single NF1-OPG patient showed that chemotherapy did not suppress the OP myelination over time. However, it should be noted that this is a clinical case study, and, therefore, the generalization of results is limited. Future investigations might focus on genetic-based imaging, particularly in more cases. Further, meta-analyses are recommended for giving a proper Field Of View (FOV) to researchers as a subtle clue regarding precision medicine.
Collapse
Affiliation(s)
- Amir Mohammad Pajavand
- Institute for Cognitive and Brain Sciences, Shahid Beheshti University Government College University, Tehran, Iran.,Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Guive Sharifi
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Anvari
- Department of Radiation Oncology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farahnaz Bidari-Zerehpoosh
- Department of Pathology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad A Shamsi
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeedeh Nateghinia
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tohid Emami Meybodi
- Functional Neurosurgery Research Center, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
285
|
Cohen PR. Chronic Lymphocytic Leukemia in Neurofibromatosis Type 1 Patients: Case Report and Literature Review of a Rare Occurrence. Cureus 2021; 13:e14258. [PMID: 33954070 PMCID: PMC8088774 DOI: 10.7759/cureus.14258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant genodermatosis that may also occur as the result of a spontaneous mutation. The diagnosis can be established by the presence of two of the seven National Institutes of Health (NIH) diagnostic criteria; several dermatologic manifestations are NIH criteria used to establish the diagnosis: axillary and inguinal freckling, café-au-lait macules, and neurofibromas. Mucosal evaluation of the eyes may detect a fourth criteria: pigmented iris hamartomas (Lisch nodules). The remaining NIH criteria include optic path glioma, distinctive osseus lesions, and a positive family history of the condition. A breast cancer 2 (BRCA2) positive woman with NF1 and chronic lymphocytic leukemia is described. Patients with NF1 have an increased lifetime risk to develop breast cancer, gastrointestinal stromal tumor, malignant glioma, malignant peripheral nerve sheath tumor, and rhabdomyosarcoma. Chronic lymphocytic leukemia occurring in NF1 patients is rare; including my female patient reported in this paper, chronic lymphocytic leukemia has only been reported in three individuals with NF1--two women and one man. The man and the other woman presented with advanced chronic lymphocytic leukemia and treatment with antineoplastic therapy at diagnosis; the man achieved clinical remission and the woman passed away from complications associated with therapy-refractory progression of her leukemia. My female patient required treatment 41 months after diagnosis and had a good clinical response; she has been without significant disease progression for 34 months. Similar to NF1, breast cancer 1 (BRCA1) and BRCA2 mutations are associated with an increased lifetime risk of developing cancer--particularly breast and ovarian carcinoma. An increased risk of chronic lymphocytic leukemia has also been demonstrated in patients with mutations of either BRCA1 or BRCA2. Also, albeit uncommon, either BRCA1 or BRCA2 mutation has been detected in women with NF1 who develop breast cancer. In conclusion, the development of chronic lymphocytic leukemia in NF1 patients may be coincidental and not associated with the underlying genodermatosis; however, the occurrence of chronic lymphocytic leukemia in my patient with NF1, in part, may be related to her BRCA2 positivity.
Collapse
Affiliation(s)
- Philip R Cohen
- Dermatology, San Diego Family Dermatology, National City, USA
| |
Collapse
|
286
|
Maranduba CP, Souza GT, do Carmo AMR, de Campos JMS, Raposo NRB, de Olivera Santos M, da Costa Maranduba CM, de Sá Silva F. Effects of resveratrol on the proliferation and osteogenic differentiation of deciduous dental pulp stem cells from neurofibromatosis type 1 patient. Childs Nerv Syst 2021; 37:1095-1101. [PMID: 33216171 DOI: 10.1007/s00381-020-04968-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE We aimed at verifying whether resveratrol can decrease cell proliferation and change osteogenic differentiation of cells obtained from patients with type 1 neurofibromatosis (NF1). METHODS Deciduous dental pulp derived stem cells were isolated from NF1 patient and healthy volunteer. These cells were subjected to increasing concentrations of resveratrol and evaluated for proliferation and mineralization of osteogenic differentiation. RESULTS The results showed that resveratrol reduced the difference in proliferation between CNT and NF1 cells in a dose-dependent manner and this property was more prominent in affected cells than in healthy cells. Resveratrol showed no statistically significant changes in mineralization in osteogenic differentiation of NF1 cells, at low doses tested. CONCLUSIONS In conclusion, in a dose-dependent manner, resveratrol displays interesting properties that could be applied in a possible treatment aimed at decreasing cellular proliferation in neurofibromatosis. Furthermore, it is selective concerning healthy cells and not affecting cell differentiation. Further research to cell selectivity, differentiation to other tissue types, and cell cytotoxicity are needed.
Collapse
Affiliation(s)
- Claudinéia Pereira Maranduba
- Laboratory of Human Genetics and Cell Therapy, Biology Department, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, 36036-900, Brazil
| | - Gustavo Torres Souza
- Laboratory of Human Genetics and Cell Therapy, Biology Department, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, 36036-900, Brazil
| | | | - José Marcelo Sallabert de Campos
- Laboratory of Genetics, Biology Department, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, 36036-900, Brazil
| | - Nádia Rezende Barbosa Raposo
- Center of Research and Innovation in Health Sciences (NUPICS), Federal University of Juiz de Fora, Juiz de Fora, 36036-900, Brazil
| | - Marcelo de Olivera Santos
- Basic Life Sciences Department, Federal University of Juiz de Fora, Governador Valadares, 35020-670, Brazil
| | - Carlos Magno da Costa Maranduba
- Laboratory of Genetics, Biology Department, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, 36036-900, Brazil.
| | - Fernando de Sá Silva
- Basic Life Sciences Department, Federal University of Juiz de Fora, Governador Valadares, 35020-670, Brazil
| |
Collapse
|
287
|
Lorentzen T, Madsen H, Lausten-Thomsen MJZ, Bygum A. Spontaneous pneumothorax as a clinical manifestation of neurofibromatosis type 1. BMJ Case Rep 2021; 14:14/3/e238694. [PMID: 33737277 PMCID: PMC7978331 DOI: 10.1136/bcr-2020-238694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a genetic disorder affecting the skin, nervous system, eyes and bones. Pulmonary involvement is unknown to many physicians. Yet, patients may be affected by lung bullae and cysts, which represent an increased risk for secondary spontaneous pneumothorax (SSP). We present a 56-year-old patient with a pathogenic variant of the NF1 gene, who suffered from NF1 with lung manifestations and recurrent SSP. It is essential to identify the patients having an increased risk of developing SSP as preventive surgery seem to decrease the risk of new events. Pneumothorax can be a clinical manifestation of NF1 but is not yet widely acknowledged as such.
Collapse
Affiliation(s)
- Tine Lorentzen
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Hanne Madsen
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark.,Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark
| | | | - Anette Bygum
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
288
|
Oliveira LB, Geller M, Cunha KS, Santos A, Bernacchi A, Rubenstein AE, Takirambudde S, Mezitis S, de Almeida Ito Brum C, Darrigo LG, Ribeiro MG. Clinical assessment of the use of topical liquid diclofenac following laser microporation of cutaneous neurofibromas in individuals with neurofibromatosis type 1. Heliyon 2021; 7:e06518. [PMID: 33817379 PMCID: PMC8010391 DOI: 10.1016/j.heliyon.2021.e06518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/16/2020] [Accepted: 03/11/2021] [Indexed: 11/03/2022] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disorder with a prevalence of 1:3000 births and a wide variety of clinical manifestations. Cutaneous neurofibromas (cNF) are among the most common visible manifestations of NF1 and present a major clinical burden for patients. NF1 patients with cNF often report decreased quality of life, emotional well-being and physical comfort. Developing effective medical therapies for cNF has been identified as a priority for the majority of adults with NF1. Methods The study was an open, controlled and prospective proof-of-concept clinical trial. The topical treatment consisted of two steps: cNF microporation using a laser device followed by topical application of one drop of diclofenac 25 mg/mL on the surface of the cNF (T neurofibroma = treatment) or physiological saline (C neurofibroma = control) and reapplied twice daily for 3 days. Neurofibroma assessments included visual and dermatoscopy observations noting color and presence of necrosis, presence of flaccidity, measurements in two dimensions, photographs, and histopathology after excision. The primary efficacy variable was the presence of tissue necrosis. The primary safety variable was the occurrence of treatment-related adverse events. Results Six patients were included in the study. The treatment resulted in transitory topical changes (healing of the microporation grid with formation of scintillating tissue layer, hyperemia and desquamation), with no statistically significant variation in the dimensions of the T and C neurofibromas in relation to pretreatment measurements. There was no necrosis in the T or C neurofibromas. In the histopathological analysis, there was no significant difference in the distribution of chronic (lymphocytic) inflammatory infiltrate in the papillary reticular dermis (subepithelial), type of infiltrate (diffuse, perivascular, or both), presence of fibrosis, and presence of atrophy among the T and C neurofibromas. No adverse events attributable to the use of diclofenac were reported during the treatment period. Conclusions Treatment did not result in significant alterations in terms of presence of tissue necrosis, size, or histopathological features in the T neurofibromas or in comparison to the C neurofibromas. Topical diclofenac with laser microporation was well-tolerated, with no adverse events attributable to diclofenac reported. Whether these observations are due to minimal systemic and neurofibroma exposure remain to be explored in dosage studies with larger patient groups. Trial registration ClinicalTrials.gov (NCT03090971) retrospectively registered March 27, 2017.
Collapse
Affiliation(s)
- Lisa Brauer Oliveira
- Postgraduate Program in Clinical Medicine, Universidade Federal do Rio de Janeiro (UFRJ) - Rio de Janeiro, Brazil
| | - Mauro Geller
- Medical Genetics Service, Instituto de Puericultura e Pediatria Martagão Gesteira Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.,Department of Immunology and Microbiology, Centro Universitário Serra dos Órgãos (UNIFESO), Teresópolis, Brazil.,Hospital Israelita Albert Einstein, São Paulo, Brazil.,Department of Clinical Immunology, Instituto de Pós-Graduação Médica Carlos Chagas - Rio de Janeiro, Brazil
| | - Karin Soares Cunha
- Postgraduate Program in Pathology, Universidade Federal Fluminense (UFF) Medical School, Niterói, Brazil.,Pathology Department, Faculdade de Medicina - Universidade Federal Fluminense (UFF), Niterói, Brazil
| | - Alessandra Santos
- Postgraduate Program in Clinical Medicine, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Allan Bernacchi
- Department of Plastic Surgery, Serviço Prof. Pitanguy, Santa Casa da Misericórdia do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Allan E Rubenstein
- Department of Neurology and Pediatrics, NYU Langone Medical Center, New York, NY, USA
| | - Sanyu Takirambudde
- Department of Pediatrics, New York University Langone Medical Center, New York, NY, USA
| | - Spyros Mezitis
- Department of Endocrinology/Clinical Medicine, Weill Medical College of Cornell University, New York, NY, USA.,New York Presbyterian Hospital/Cornell Medicine, Lenox Hill Hospital/Northwell Health, New York, NY, USA
| | | | - Luiz Guilherme Darrigo
- Bone Marrow Transplant Unit, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marcia Gonçalves Ribeiro
- Department of Pediatrics, Federal University of Rio de Janeiro Medical School (UFRJ), Rio de Janeiro, Brazil.,Medical Genetics Service, Instituto de Puericultura e Pediatria Martagão Gesteira, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
289
|
Zhang Y, Cai H, Lv G, Li Y. A giant posterior mediastinal malignant peripheral nerve sheath tumor and benign neurofibroma in body surface: a case report. BMC Surg 2021; 21:128. [PMID: 33691671 PMCID: PMC7945373 DOI: 10.1186/s12893-021-01122-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 02/24/2021] [Indexed: 11/10/2022] Open
Abstract
Background Neurofibromatosis comprises neurofibromatosis type 1 (NF1) and type 2 (NF2). Major tumor type of NF1 are neurofibroma recognized as benign peripheral nerve tumor, malignant peripheral nerve sheath tumor (MPNST), and glioma. Case presentation We report a woman with a special condition, whose tumors in body surfaces were benign neurofibroma and tumors in posterior mediastinum are MPNST. The chest-enhanced CT suggested a round soft tissue density in posteriormediastium. The diagnosis was established by pathology and immunohistochemistry. A single-stage thoracoscopic mediastinal mass resection was performed. The whole operation went smoothly and the CT scan of lungs did not show relapse of tumor three months later. Conclusions The appearance of neurofibroma should draw particular attention to the possibility of developing MPNST. More careful imaging examinations should be carried out, and pathological examination could diagnose it.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Hongfei Cai
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Guangchao Lv
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yang Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
290
|
Chiara L, Marta T, Dario T, Tommaso V, Roberta O, Cesare C, Elisabetta F, Alessandro DL, Marco T, Donato R, Giuseppe Z. Enlarged spinal nerve roots in RASopathies: Report of two cases. Eur J Med Genet 2021; 64:104187. [PMID: 33676063 DOI: 10.1016/j.ejmg.2021.104187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/26/2021] [Accepted: 02/27/2021] [Indexed: 11/15/2022]
Abstract
RASopathies are a group of genetic conditions caused by germline variants in genes encoding signal transducers and modulators of the RAS-MAPK cascade. These disorders are multisystem diseases with considerable clinical overlap, even though distinct hallmarks are recognizable for each specific syndrome. Here we report on the presence of enlarged spinal nerve roots resembling neurofibromas, a typical neuroradiological finding of neurofibromatosis type 1, in two patients with a molecularly confirmed diagnosis of Noonan syndrome and cardio-facio-cutaneous syndrome, respectively. This evidence add enlarged spinal nerve roots as features shared among RASopathies. Future studies aiming to a better understanding of the molecular mechanisms leading to neurogenic tumor development in these patients are necessary to define their biological nature, evolution, prognosis and possible treatments.
Collapse
Affiliation(s)
- Leoni Chiara
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy.
| | - Tedesco Marta
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Talloa Dario
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Verdolotti Tommaso
- UOC Radiologia e Neuroradiologia, Dipartimento di diagnostica per immagini, Oncologica ed ematologia, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Onesimo Roberta
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Colosimo Cesare
- UOC Radiologia e Neuroradiologia, Dipartimento di diagnostica per immagini, Oncologica ed ematologia, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - Flex Elisabetta
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - De Luca Alessandro
- Medical Genetics Division, IRCCS Fondazione Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Tartaglia Marco
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Rigante Donato
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - Zampino Giuseppe
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
291
|
Affiliation(s)
- Thierry Alcindor
- Gerald Bronfman Department of Oncology, McGill University, Cedars Cancer Center, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
292
|
Zheng K, Liu T, Zhao J, Meng P, Bian Y, Ni C, Wang H, Pan Y, Wu S, Jiang H, Jin G. Mutational landscape and potential therapeutic targets for sporadic pancreatic neuroendocrine tumors based on target next-generation sequencing. Exp Ther Med 2021; 21:415. [PMID: 33747156 PMCID: PMC7967861 DOI: 10.3892/etm.2021.9859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic neuroendocrine tumor (PNET), a heterogenous type of neoplasm with limited treatment options, is relatively rare and to date, the genetic background has remained to be fully elucidated. The present study aimed to determine the mutational landscape of PNET with and without liver metastasis, as well as its clinical application value for treatment. Fresh tumor tissues were collected from 14 patients with PNET following surgery, 4 of whom had developed liver metastasis. Subsequently, targeted next-generation sequencing of 612 cancer-associated genes and comprehensive analysis were performed on the tumor tissues. The results identified 63 somatic mutations in 53 genes in the 14 patients with PNET, amongst which menin 1 was identified as the most recurrently mutated gene. The analysis also identified several novel recurrently mutated genes, including adrenoceptor alpha 2B, ARVCF delta catenin family member, carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase and neuregulin 1. Among the 53 mutated genes, 11 were enriched in the PI3K/AKT signaling pathway (adjusted P=7.12x10-5). In addition, 4 patients with PNET with liver metastasis had distinctly different mutational profiles compared with those without liver metastasis; 13 genes were discovered to be exclusively mutated in the liver metastasis group of the patients with PNET, including ATRX chromatin remodeler, thioredoxin reductase 2, anus kinase 3, ARVCF delta catenin family member, integrin subunit alpha V and RAD50 double strand break repair protein. In addition, two potentially actionable alterations in BRCA2 DNA repair-associated (p.Q548Q) and neurofibromin 1 (p.Q1188X) were identified using the OncoKB database. In conclusion, the present study generated a comprehensive mutational profile of 14 patients with PNET and further described the features of patients with liver metastasis, which highlights potential targets for drug development of PNET.
Collapse
Affiliation(s)
- Kailian Zheng
- Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Tao Liu
- Department of Emergency, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Jiangman Zhao
- Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, P.R. China.,Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 201204, P.R. China
| | - Peng Meng
- Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, P.R. China.,Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 201204, P.R. China
| | - Yun Bian
- Department of Imaging, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Chenming Ni
- Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Huan Wang
- Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yaqi Pan
- Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Shouxin Wu
- Shanghai Zhangjiang Institute of Medical Innovation, Shanghai 201204, P.R. China.,Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 201204, P.R. China
| | - Hui Jiang
- Department of Pathology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Gang Jin
- Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
293
|
Gu YH, Cui XW, Ren JY, Long MM, Wang W, Wei CJ, Aimaier R, Li YH, Chung MH, Gu B, Li QF, Wang ZC. Selection of internal references for RT-qPCR assays in Neurofibromatosis type 1 (NF1) related Schwann cell lines. PLoS One 2021; 16:e0241821. [PMID: 33630851 PMCID: PMC7906369 DOI: 10.1371/journal.pone.0241821] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/02/2021] [Indexed: 11/20/2022] Open
Abstract
Real-time quantitative PCR (RT-qPCR) has been widely applied in uncovering disease mechanisms and screening potential biomarkers. Internal reference gene selection determines the accuracy and reproducibility of data analyses. The aim of this study was to identify the optimal reference genes for the relative quantitative analysis of RT-qPCR in fourteen NF1 related cell lines, including non-tumor, benign and malignant Schwann cell lines. The expression characteristics of eleven candidate reference genes (RPS18, ACTB, B2M, GAPDH, PPIA, HPRT1, TBP, UBC, RPLP0, TFRC and RPL32) were screened and analyzed by four software programs: geNorm, NormFinder, BestKeeper and RefFinder. Results showed that GAPDH, the most frequently used internal reference gene, was significantly unstable between various cell lines. The combinational use of two reference genes (PPIA and TBP) was optimal in malignant Schwann cell lines and the use of single reference genes (PPIA or PRLP0) alone or in combination was optimal in benign Schwann cell lines. These recommended internal reference gene selections may improve the accuracy and reproducibility of RT-qPCR in gene expression analyses of NF1 related tumors.
Collapse
Affiliation(s)
- Yi-Hui Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi-Wei Cui
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie-Yi Ren
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man-Mei Long
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-Jiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rehanguli Aimaier
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue-Hua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man-Hon Chung
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (QFL); (ZCW)
| | - Zhi-Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (QFL); (ZCW)
| |
Collapse
|
294
|
Next-Generation Sequencing-Based Preimplantation Genetic Testing for De Novo NF1 Mutations. BIOCHIP JOURNAL 2021. [DOI: 10.1007/s13206-021-00006-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
295
|
Osum SH, Watson AL, Largaespada DA. Spontaneous and Engineered Large Animal Models of Neurofibromatosis Type 1. Int J Mol Sci 2021; 22:1954. [PMID: 33669386 PMCID: PMC7920315 DOI: 10.3390/ijms22041954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Animal models are crucial to understanding human disease biology and developing new therapies. By far the most common animal used to investigate prevailing questions about human disease is the mouse. Mouse models are powerful tools for research as their small size, limited lifespan, and defined genetic background allow researchers to easily manipulate their genome and maintain large numbers of animals in general laboratory spaces. However, it is precisely these attributes that make them so different from humans and explains, in part, why these models do not accurately predict drug responses in human patients. This is particularly true of the neurofibromatoses (NFs), a group of genetic diseases that predispose individuals to tumors of the nervous system, the most common of which is Neurofibromatosis type 1 (NF1). Despite years of research, there are still many unanswered questions and few effective treatments for NF1. Genetically engineered mice have drastically improved our understanding of many aspects of NF1, but they do not exemplify the overall complexity of the disease and some findings do not translate well to humans due to differences in body size and physiology. Moreover, NF1 mouse models are heavily reliant on the Cre-Lox system, which does not accurately reflect the molecular mechanism of spontaneous loss of heterozygosity that accompanies human tumor development. Spontaneous and genetically engineered large animal models may provide a valuable supplement to rodent studies for NF1. Naturally occurring comparative models of disease are an attractive prospect because they occur on heterogeneous genetic backgrounds and are due to spontaneous rather than engineered mutations. The use of animals with naturally occurring disease has been effective for studying osteosarcoma, lymphoma, and diabetes. Spontaneous NF-like symptoms including neurofibromas and malignant peripheral nerve sheath tumors (MPNST) have been documented in several large animal species and share biological and clinical similarities with human NF1. These animals could provide additional insight into the complex biology of NF1 and potentially provide a platform for pre-clinical trials. Additionally, genetically engineered porcine models of NF1 have recently been developed and display a variety of clinical features similar to those seen in NF1 patients. Their large size and relatively long lifespan allow for longitudinal imaging studies and evaluation of innovative surgical techniques using human equipment. Greater genetic, anatomic, and physiologic similarities to humans enable the engineering of precise disease alleles found in human patients and make them ideal for preclinical pharmacokinetic and pharmacodynamic studies of small molecule, cellular, and gene therapies prior to clinical trials in patients. Comparative genomic studies between humans and animals with naturally occurring disease, as well as preclinical studies in large animal disease models, may help identify new targets for therapeutic intervention and expedite the translation of new therapies. In this review, we discuss new genetically engineered large animal models of NF1 and cases of spontaneous NF-like manifestations in large animals, with a special emphasis on how these comparative models could act as a crucial translational intermediary between specialized murine models and NF1 patients.
Collapse
Affiliation(s)
- Sara H. Osum
- Masonic Cancer Center, Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - David A. Largaespada
- Masonic Cancer Center, Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
296
|
Pennanen P, Kallionpää RA, Peltonen S, Nissinen L, Kähäri VM, Heervä E, Peltonen J. Signaling pathways in human osteoclasts differentiation: ERK1/2 as a key player. Mol Biol Rep 2021; 48:1243-1254. [PMID: 33486672 PMCID: PMC7925492 DOI: 10.1007/s11033-020-06128-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023]
Abstract
Little is known about the signaling pathways involved in the differentiation of human osteoclasts. The present study evaluated the roles of the Ras/PI3K/Akt/mTOR, Ras/Raf/MEK1/2/ERK1/2, calcium-PKC, and p38 signaling pathways in human osteoclast differentiation. Mononuclear cells were isolated from the peripheral blood of control persons and patients with neurofibromatosis 1 (NF1), and the cells were differentiated into osteoclasts in the presence of signaling pathway inhibitors. Osteoclast differentiation was assessed using tartrate-resistant acid phosphatase 5B. Inhibition of most signaling pathways with chemical inhibitors decreased the number of human osteoclasts and disrupted F-actin ring formation, while the inhibition of p38 resulted in an increased number of osteoclasts, which is a finding contradictory to previous murine studies. However, the p38 inhibition did not increase the bone resorption capacity of the cells. Ras-inhibitor FTS increased osteoclastogenesis in samples from control persons, but an inhibitory effect was observed in NF1 samples. Inhibition of MEK, PI3K, and mTOR reduced markedly the number of NF1-deficient osteoclasts, but no effect was observed in control samples. Western blot analyses showed that the changes in the phosphorylation of ERK1/2 correlated with the number of osteoclasts. Our results highlight the fact that osteoclastogenesis is regulated by multiple interacting signaling pathways and emphasize that murine and human findings related to osteoclastogenesis are not necessarily equivalent.
Collapse
Affiliation(s)
- Paula Pennanen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Roope A Kallionpää
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Sirkku Peltonen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
- Department of Dermatology and Venereology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Dermatology and Venereology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
- MediCity Research Laboratory, University of Turku and Cancer Research Laboratory FICAN West, University of Turku and Turku University Hospital, Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
- MediCity Research Laboratory, University of Turku and Cancer Research Laboratory FICAN West, University of Turku and Turku University Hospital, Turku, Finland
| | - Eetu Heervä
- Department of Oncology, University of Turku and Turku University Hospital, Turku, Finland
| | - Juha Peltonen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland.
| |
Collapse
|
297
|
Tritz R, Benson T, Harris V, Hudson FZ, Mintz J, Zhang H, Kennard S, Chen W, Stepp DW, Csanyi G, Belin de Chantemèle EJ, Weintraub NL, Stansfield BK. Nf1 heterozygous mice recapitulate the anthropometric and metabolic features of human neurofibromatosis type 1. Transl Res 2021; 228:52-63. [PMID: 32781282 PMCID: PMC7779711 DOI: 10.1016/j.trsl.2020.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/26/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a heritable cancer predisposition syndrome resulting from mutations in the NF1 tumor suppressor gene. Genotype-phenotype correlations for NF1 are rare due to the large number of NF1 mutations and role of modifier genes in manifestations of NF1; however, emerging reports suggest that persons with NF1 display a distinct anthropometric and metabolic phenotype featuring short stature, low body mass index, increased insulin sensitivity, and protection from diabetes. Nf1 heterozygous (Nf1+/-) mice accurately reflect the dominant inheritance of NF1 and are regularly employed as a model of NF1. Here, we sought to identify whether Nf1+/- mice recapitulate the anthropometric and metabolic features identified in persons with NF1. Littermate 16-20 week-old male wildtype (WT) and Nf1+/- C57B/6J mice underwent nuclear magnetic resonance (NMR), indirect calorimetry, and glucose/insulin/pyruvate tolerance testing. In some experiments, tissues were harvested for NMR and histologic characterization. Nf1+/- mice are leaner with significantly reduced visceral and subcutaneous fat mass, which corresponds with an increased density of small adipocytes and reduced leptin levels. Additionally, Nf1+/- mice are highly reliant on carbohydrates as an energy substrate and display increased glucose clearance and insulin sensitivity, but normal response to pyruvate suggesting enhanced glucose utilization and preserved gluconeogenesis. Finally, WT and Nf1+/- mice subjected to high glucose diet were protected from diet-induced obesity and hyperglycemia. Our data suggest that Nf1+/- mice closely recapitulate the anthropometric and metabolic phenotype identified in persons with NF1, which will impact the interpretation of previous and future translational studies of NF1.
Collapse
Affiliation(s)
- Rebekah Tritz
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Tyler Benson
- Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Valerie Harris
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Farlyn Z Hudson
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - James Mintz
- Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Hanfang Zhang
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Simone Kennard
- Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Weiqin Chen
- Vascular Biology Center, Augusta University, Augusta, Georgia
| | - David W Stepp
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Gabor Csanyi
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Neal L Weintraub
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Brian K Stansfield
- Vascular Biology Center, Augusta University, Augusta, Georgia; Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, Georgia.
| |
Collapse
|
298
|
Ozarslan B, Russo T, Argenziano G, Santoro C, Piccolo V. Cutaneous Findings in Neurofibromatosis Type 1. Cancers (Basel) 2021; 13:463. [PMID: 33530415 PMCID: PMC7865571 DOI: 10.3390/cancers13030463] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/02/2021] [Accepted: 01/14/2021] [Indexed: 12/15/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a complex autosomal dominant disorder associated with germline mutations in the NF1 tumor suppressor gene. NF1 belongs to a class of congenital anomaly syndromes called RASopathies, a group of rare genetic conditions caused by mutations in the Ras/mitogen-activated protein kinase pathway. Generally, NF1 patients present with dermatologic manifestations. In this review the main features of café-au-lait macules, freckling, neurofibromas, juvenile xanthogranuloma, nevus anemicus and other cutaneous findings will be discussed.
Collapse
Affiliation(s)
| | - Teresa Russo
- Dermatology Unit, University of Campania Luigi Vanvitelli, 80100 Naples, Italy; (T.R.); (G.A.)
| | - Giuseppe Argenziano
- Dermatology Unit, University of Campania Luigi Vanvitelli, 80100 Naples, Italy; (T.R.); (G.A.)
| | - Claudia Santoro
- Department of Woman, Neurofibromatosis Referral Centre, Child and of General and Specialised Surgery, University of Campania Luigi Vanvitelli, 80100 Naples, Italy;
| | - Vincenzo Piccolo
- Dermatology Unit, University of Campania Luigi Vanvitelli, 80100 Naples, Italy; (T.R.); (G.A.)
| |
Collapse
|
299
|
Kallionpää RA, Ahramo K, Aaltonen M, Pennanen P, Peltonen J, Peltonen S. Circulating free DNA in the plasma of individuals with neurofibromatosis type 1. Am J Med Genet A 2021; 185:1098-1104. [PMID: 33484105 DOI: 10.1002/ajmg.a.62081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant syndrome whose characteristic manifestations include benign neurofibromas, yet NF1 is also associated with a high risk of cancer. Measurements of circulating free plasma DNA (cfDNA) are gaining wider applicability in cancer diagnostics, targeting of therapy, and monitoring of therapeutic response. Individuals with NF1 are likely to be followed up using this method, but the effects of NF1 and neurofibromas on cfDNA levels are not known. We studied peripheral blood samples from 19 adults with NF1 and 12 healthy controls. The cfDNA was isolated from plasma with QIAamp Circulating Nucleic Acid Kit and quantified using the Qubit 2.0 Fluorometer. The cfDNA concentration of each sample was normalized relative to the plasma protein concentration. The normalized median concentration of cfDNA in plasma was 19.3 ng/ml (range 6.6-78.6) among individuals with NF1 and 15.9 ng/ml (range 4.8-47.0) among controls (p = .369). Individuals with NF1 who also had plexiform neurofibroma (pNF) showed non-significantly elevated cfDNA concentration compared to individuals with NF1 and without known pNF (median 25.4 vs. 18.8 ng/ml, p = .122). The effect of NF1 on cfDNA seems to be relatively small and NF1 is therefore unlikely to hamper the use of cfDNA-based assays.
Collapse
Affiliation(s)
- Roope A Kallionpää
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kaisa Ahramo
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Marianna Aaltonen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku University of Applied Sciences, Turku, Finland
| | - Paula Pennanen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Juha Peltonen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sirkku Peltonen
- Department of Dermatology and Venereology, University of Turku, Turku, Finland.,Department of Dermatology, Turku University Hospital, Turku, Finland.,Department of Dermatology and Venereology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Dermatology and Venereology, Region Västra Götaland Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
300
|
Leoni C, Romeo DM, Pelliccioni M, Di Già M, Onesimo R, Giorgio V, Flex E, Tedesco M, Tartaglia M, Rigante D, Valassina A, Zampino G. Musculo-skeletal phenotype of Costello syndrome and cardio-facio-cutaneous syndrome: insights on the functional assessment status. Orphanet J Rare Dis 2021; 16:43. [PMID: 33482860 PMCID: PMC7821553 DOI: 10.1186/s13023-021-01674-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
Background Costello syndrome (CS)
and cardio-facio-cutaneous syndrome (CFCS) belong to the RASopathies, a group of neurodevelopmental disorders with skeletal anomalies. Due to their rarity, the characterization of the musculo-skeletal phenotype in both disorders has been poorly characterized. Patients and methods Herein we reported data on orthopedic findings and functional status of a large sample of CS and CFCS patients. Thirty-four patients (CS = 17 and CFCS = 17) were recruited. Functional and disability evaluations were performed by assessing the 6-min walking test (6MWT) and Pediatric Outcomes Data Collection Instrument (PODCI). Genotype/phenotype correlation was also provided. Results Orthopedic manifestations are highly prevalent in CS and CFCS and overlap in the two disorders. Overall, patients with CS harboring the recurrent HRAS Gly12Ser substitution show a more severe skeletal phenotype compared to patients carrying the Gly12Ala and Gly13Cys variants. Among CFCS patients, those with the MAP2K1/2 variant show different skeletal characteristics compared to BRAF variants, with a higher prevalence of orthopedic abnormalities. Functional assessment showed that patients with CS and CFCS reached lower values compared to the general population, with CFCS patients displaying the lowest scores. Conclusions Orthopedic manifestations appear universal features of CS and CFCS and they can evolve across patients’ life. Longitudinal assessment of disability status by using 6MWT and PODCI could be useful to evaluate the functional impact of orthopedic manifestations on patients’ outcome and help planning a tailored treatment of these comorbidities.
Collapse
Affiliation(s)
- Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Gemelli 8, 00168, Rome, Italy.
| | - Domenico Marco Romeo
- Pediatric Neurology Unit, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Michele Pelliccioni
- Center for Rare Diseases and Birth Defects, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Gemelli 8, 00168, Rome, Italy
| | - Mariangela Di Già
- Center for Rare Diseases and Birth Defects, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Gemelli 8, 00168, Rome, Italy
| | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Gemelli 8, 00168, Rome, Italy
| | - Valentina Giorgio
- Center for Rare Diseases and Birth Defects, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Gemelli 8, 00168, Rome, Italy
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Marta Tedesco
- Center for Rare Diseases and Birth Defects, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Gemelli 8, 00168, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Donato Rigante
- Center for Rare Diseases and Birth Defects, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Gemelli 8, 00168, Rome, Italy.,Università Cattolica Sacro Cuore, Rome, Italy
| | - Antonio Valassina
- Università Cattolica Sacro Cuore, Rome, Italy.,Unit of Neurophysiopathology and Sleep Medicine, Neurosciences and Orthopedics, Department of Geriatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Gemelli 8, 00168, Rome, Italy.,Università Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|