1
|
El Masri R, Iannuzzo A, Kuentz P, Tacine R, Vincent M, Barbarot S, Morice-Picard F, Boralevi F, Oillarburu N, Mazereeuw-Hautier J, Duffourd Y, Faivre L, Sorlin A, Vabres P, Delon J. A postzygotic GNA13 variant upregulates the RHOA/ROCK pathway and alters melanocyte function in a mosaic skin hypopigmentation syndrome. Nat Commun 2025; 16:1751. [PMID: 39966435 DOI: 10.1038/s41467-025-56995-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
The genetic bases of mosaic pigmentation disorders have increasingly been identified, but these conditions remain poorly characterised, and their pathophysiology is unclear. Here, we report in four unrelated patients that a recurrent postzygotic mutation in GNA13 is responsible for a recognizable syndrome with hypomelanosis of Ito associated with developmental anomalies. GNA13 encodes Gα13, a subunit of αβγ heterotrimeric G proteins coupled to specific transmembrane receptors known as G-protein coupled receptors. In-depth functional investigations revealed that this R200K mutation provides a gain of function to Gα13. Mechanistically, we show that this variant hyperactivates the RHOA/ROCK signalling pathway that consequently increases actin polymerisation and myosin light chains phosphorylation, and promotes melanocytes rounding. Our results also indicate that R200K Gα13 hyperactivates the YAP signalling pathway. All these changes appear to affect cell migration and adhesion but not the proliferation. Our results suggest that hypopigmentation can result from a defect in melanosome transfer to keratinocytes due to cell shape alterations. These findings highlight the interaction between heterotrimeric G proteins and the RHOA pathway, and their role in melanocyte function.
Collapse
Affiliation(s)
- Rana El Masri
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
- Department of Cell Physiology & Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alberto Iannuzzo
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Paul Kuentz
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France
- Oncobiologie Génétique Bioinformatique, PCBio, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Rachida Tacine
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Marie Vincent
- Unité de Génétique clinique, Service de génétique médicale, CHU de Nantes - Hôpital Mère-Enfant, Nantes, France
| | - Sébastien Barbarot
- Nantes Université, Department of Dermatology, CHU Nantes, INRAE, UMR 1280, PhAN, Nantes, France
| | - Fanny Morice-Picard
- MAGEC Reference Centre for Rare Genetic Skin Diseases, Paediatric Dermatology Unit, Department of Dermatology, CHU de Bordeaux - GH Pellegrin, Bordeaux, France
| | - Franck Boralevi
- MAGEC Reference Centre for Rare Genetic Skin Diseases, Paediatric Dermatology Unit, Department of Dermatology, CHU de Bordeaux - GH Pellegrin, Bordeaux, France
| | - Naia Oillarburu
- Service de dermatologie, CHU de Toulouse - Hôpital Larrey, Toulouse, France
| | | | - Yannis Duffourd
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France
- CHU Dijon, Unité Fonctionnelle "Innovation diagnostique dans les maladies rares", FHU-TRANSLAD & Institut GIMI, Dijon, France
| | - Laurence Faivre
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France
- CHU Dijon, Centre de Génétique et Centres de référence Anomalies du Développement et Déficience Intellectuelle, FHU-TRANSLAD & Institut GIMI, Dijon, France
| | - Arthur Sorlin
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France
- MAGEC Reference Centre for Rare Genetic Skin Diseases and Paediatric Dermatology Unit, Department of Paediatrics, University Hospital Dijon-Bourgogne, FHU-TRANSLAD & Institut GIMI, Dijon, France
- National Center of Genetics (NCG), Laboratoire national de santé (LNS), 1 Rue Louis Rech, Dudelange, Luxembourg
| | - Pierre Vabres
- UFR des Sciences de Santé, Inserm - Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHU-TRANSLAD, Dijon, France.
- MAGEC Reference Centre for Rare Genetic Skin Diseases and Paediatric Dermatology Unit, Department of Paediatrics, University Hospital Dijon-Bourgogne, FHU-TRANSLAD & Institut GIMI, Dijon, France.
- Rare Disease Collaborative Network (RDCN) Adult Mosaic Disorders Clinic, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom.
| | - Jérôme Delon
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France.
| |
Collapse
|
2
|
Luu M, Vabres P, Espitalier A, Maurer A, Garde A, Racine C, Carpentier M, Rega A, Loffroy R, Hadouiri N, Boddaert N, Curie A, Guibaud L, Chebbi M, Charligny J, Kuentz P, Canaud G, Bahi-Buisson N, Fleck C, Cransac A, Bardou M, Faivre L. A phase II double-blind multicentre, placebo-controlled trial to assess the efficacy and safety of alpelisib (BYL719) in paediatric and adult patients with Megalencephaly-CApillary malformation Polymicrogyria syndrome (MCAP): the SESAM study protocol. BMJ Open 2024; 14:e084614. [PMID: 39806603 PMCID: PMC11667470 DOI: 10.1136/bmjopen-2024-084614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 11/22/2024] [Indexed: 01/30/2025] Open
Abstract
INTRODUCTION The megalencephaly capillary malformation polymicrogyria (MCAP syndrome) results from mosaic gain-of-function PIK3CA variants. The main clinical features are macrocephaly, somatic overgrowth, neurodevelopmental delay and brain anomalies. Alpelisib (Vijoice) is a recently FDA-approved PI3Kα-specific inhibitor for patients with PIK3CA-related overgrowth spectrum (PROS). During its development, in patients with the MCAP subgroup of PROS, there was no specific, standardised evaluation of the effect on neuro-cognitive functioning. Moreover, it remains unknown if the molecule crosses the blood-brain barrier. Our objective is to evaluate the efficacy of a 24 month treatment with alpelisib on adaptive behaviour in patients with MCAP syndrome. METHODS AND ANALYSIS SESAM is an industry-sponsored two-period multicentre French academic phase II trial, with a 6-month double-blind, placebo-controlled period followed by an open-label period. The primary endpoint is a ≥4-point improvement in the Vineland II Adaptive Behaviour Scale (VABS), 24 months after treatment initiation. Secondary objectives are safety, VABS improvement at 6 months, impact on the quality of life, epilepsy and hypotonia. 20 patients aged 2 to 40 years with an MCAP diagnosis and neurodevelopmental disorders of various degrees, will be followed monthly in local centres, centrally assessed (clinical, biological, neuropsychological and functional evaluation) at baseline and every 6 months. Patients will be evaluated by volumetric MRI at baseline and at 24 months. An optional lumbar puncture will be performed to investigate blood-brain barrier crossing. Inclusions were completed by April 2024, with the end of follow-up in November 2026.Given the efficacy of alpelisib in patients with PROS, if the drug crosses the blood-brain barrier, we can expect a clinical benefit for patients with neurocognitive disorders. ETHICS AND DISSEMINATION Ethical approval was given by CPP Sud-Ouest et Outre-Mer I (reference: 2022-500197-34-01). Findings from this study will be disseminated via publication, reports and conference presentations. TRIAL REGISTRATION NUMBER NCT05577754.
Collapse
Affiliation(s)
- Maxime Luu
- Centre d’investigation clinique – module plurithématique (CIC-P) INSERM 1432, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
- INSERM UMR1231 Génétique des Anomalies du Développement (GAD), Université de Bourgogne, Dijon, France
| | - Pierre Vabres
- Dermatology, Centre référence MAGEC, Dijon, France
- St John's Institute of Dermatology, London, UK
| | - Aurélie Espitalier
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Agnès Maurer
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Aurore Garde
- INSERM UMR1231 Génétique des Anomalies du Développement (GAD), Université de Bourgogne, Dijon, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Caroline Racine
- INSERM UMR1231 Génétique des Anomalies du Développement (GAD), Université de Bourgogne, Dijon, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Maud Carpentier
- Direction de la Recherche Clinique, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Adélaide Rega
- Département de Radiologie et Imagerie Diagnostique et Thérapeutique, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Romaric Loffroy
- Département de Radiologie et Imagerie Diagnostique et Thérapeutique, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Nawale Hadouiri
- Département de Médecine Physique et de Réadaptation, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Nathalie Boddaert
- INSERM UMR-1163 Institut Imagine, Hôpital Universitaire Necker-Enfants Malades, Paris, France
- Département de Radiologie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique - Hopitaux de Paris, Paris, Île-de-France, France
| | - Aurore Curie
- Centre de référence Déficience Intellectuelle de causes rares, Service de neuropédiatrie, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Laurent Guibaud
- Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Mouna Chebbi
- Service de Pharmacologie périnatale, pédiatrique et adulte (site HEGP), Recherche Clinique Entrepôts de Données et Pharmacologie, GHU Paris. Université Paris Cité, Assistance Publique - Hopitaux de Paris, Paris, Île-de-France, France
| | - Julie Charligny
- Centre d’investigation clinique – module plurithématique (CIC-P) INSERM 1432, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Paul Kuentz
- INSERM UMR1231 Génétique des Anomalies du Développement (GAD), Université de Bourgogne, Dijon, France
- Oncobiologie Génétique Bioinformatique, FHU-TRANSLAD et Institut GIMI, Centre Hospitalier Universitaire de Besancon, Besancon, Bourgogne-Franche-Comté, France
| | - Guillaume Canaud
- INSERM UMR-1163 Institut Imagine, Hôpital Universitaire Necker-Enfants Malades, Paris, France
- INSERM U1151, Unité de médecine translationnelle et thérapies ciblées, Hôpital Necker-Enfants Malades, Université Paris Cité, Paris, Île-de-France, France
| | - Nadia Bahi-Buisson
- Département de Radiologie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique - Hopitaux de Paris, Paris, Île-de-France, France
- Service de Neurologie Pédiatrique, DMU MICADO, Hôpital Necker Enfants Malades, Assistance Publique - Hopitaux de Paris, Paris, Île-de-France, France
| | - Camille Fleck
- Direction de la Recherche Clinique, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| | - Amelie Cransac
- Département de Pharmacie, Centre Hospitalier Universitaire Dijon Bourgogne, Dijon, Bourgogne-Franche-Comté, France
- INSERM LNC-UMR1231, Université de Bourgogne, Dijon, Bourgogne-Franche-Comté, France
| | - Marc Bardou
- Centre d’investigation clinique – module plurithématique (CIC-P) INSERM 1432, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
- INSERM UMR1231 Génétique des Anomalies du Développement (GAD), Université de Bourgogne, Dijon, France
| | - Laurence Faivre
- INSERM UMR1231 Génétique des Anomalies du Développement (GAD), Université de Bourgogne, Dijon, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, Centre Hospitalier Universitaire de Dijon, Dijon, Bourgogne-Franche-Comté, France
| |
Collapse
|
3
|
Horta E, Dahlen E, Engel C, Piard J, Thauvin-Robinet C, Faivre L, Vabres P, Kuentz P. Correspondence on "The ClinGen Brain Malformation Variant Curation Expert Panel: Rules for somatic variants in AKT3, MTOR, PIK3CA, and PIK3R2" by Lai et al. Genet Med 2024; 26:101214. [PMID: 39011768 DOI: 10.1016/j.gim.2024.101214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024] Open
Affiliation(s)
- Edgar Horta
- Université de Franche-Comté, CHU Besançon, Oncobiologie Génétique Bioinformatique, FHU-TRANSLAD et Institut GIMI, Besançon, France
| | - Eric Dahlen
- Université de Franche-Comté, CHU Besançon, Oncobiologie Génétique Bioinformatique, FHU-TRANSLAD et Institut GIMI, Besançon, France
| | - Camille Engel
- Université de Franche-Comté, CHU Besançon, Centre de Génétique Humaine, Besançon, France; Université de Bourgogne, INSERM UMR1231 GAD "Génétique des Anomalies du Développement," Dijon, France
| | - Juliette Piard
- Université de Franche-Comté, CHU Besançon, Centre de Génétique Humaine, Besançon, France; Université de Bourgogne, INSERM UMR1231 GAD "Génétique des Anomalies du Développement," Dijon, France
| | - Christel Thauvin-Robinet
- Université de Bourgogne, INSERM UMR1231 GAD "Génétique des Anomalies du Développement," Dijon, France; CHU Dijon, Unité Fonctionnelle "Innovation diagnostique dans les maladies rares," FHU TRANSLAD et Institut GIMI, Dijon, France
| | - Laurence Faivre
- Université de Bourgogne, INSERM UMR1231 GAD "Génétique des Anomalies du Développement," Dijon, France; CHU Dijon, Centre de Génétique et Centres de référence Anomalies du Développement et Déficience Intellectuelle, FHU-TRANSLAD et Institut GIMI, Dijon, France
| | - Pierre Vabres
- CHU Dijon, Centre de référence MAGEC Nord "Maladies rares de la peau et des muqueuses d'origine Génétique à Expression Cutanée, "FHU-TRANSLAD et Institut GIMI, Dijon, France; Reference centre for rare diseases, St John's Institute of Dermatology, St Thomas' Hospital, London, United Kingdom
| | - Paul Kuentz
- Université de Franche-Comté, CHU Besançon, Oncobiologie Génétique Bioinformatique, FHU-TRANSLAD et Institut GIMI, Besançon, France; Université de Bourgogne, INSERM UMR1231 GAD "Génétique des Anomalies du Développement," Dijon, France.
| |
Collapse
|
4
|
Wedemeyer MA, Ding T, Garfinkle EAR, Westfall JJ, Navarro JB, Hernandez Gonzalez ME, Varga EA, Witman P, Mardis ER, Cottrell CE, Miller AR, Miller KE. Defining the transcriptome of PIK3CA-altered cells in a human capillary malformation using single cell long-read sequencing. Sci Rep 2024; 14:25440. [PMID: 39455600 PMCID: PMC11512043 DOI: 10.1038/s41598-024-72167-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/04/2024] [Indexed: 10/28/2024] Open
Abstract
PIK3CA-related overgrowth spectrum (PROS) disorders are caused by somatic mosaic variants that result in constitutive activation of the phosphatidylinositol-3-kinase/AKT/mTOR pathway. Promising responses to molecularly targeted therapy have been reported, although identification of an appropriate agent can be hampered by the mosaic nature and corresponding low variant allele frequency of the causal variant. Moreover, our understanding of the molecular consequences of these variants-for example how they affect gene expression profiles-remains limited. Here we describe in vitro expansion of a human capillary malformation followed by molecular characterization using exome sequencing, single cell gene expression, and targeted long-read single cell RNA-sequencing in a patient with clinical features consistent with Megalencephaly-Capillary Malformation Syndrome (MCAP, a PROS condition). These approaches identified a targetable PIK3CA variant with expression restricted to PAX3+ fibroblast and undifferentiated keratinocyte populations. This study highlights the innovative combination of next-generation single cell sequencing methods to better understand unique transcriptomic profiles and cell types associated with MCAP, revealing molecular intricacies of this genetic syndrome.
Collapse
Affiliation(s)
- Michelle A Wedemeyer
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Neurosurgery, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Tianli Ding
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Elizabeth A R Garfinkle
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Jesse J Westfall
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Jaye B Navarro
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Maria Elena Hernandez Gonzalez
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Elizabeth A Varga
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Patricia Witman
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Division of Dermatology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Elaine R Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Catherine E Cottrell
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Anthony R Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Katherine E Miller
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
5
|
Ondič O, Michalová K, Švajdler M, Presl J, Kosťun J, Hájková V, Martínek P, Michal M. Molecular substratification of endometrial carcinomas with no special molecular profile (NSMP) by using a limited NGS custom panel may facilitate effective patient selection for the PIK3CA-targeted therapy. Virchows Arch 2024:10.1007/s00428-024-03905-6. [PMID: 39235514 DOI: 10.1007/s00428-024-03905-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/07/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024]
Abstract
Endometrial carcinomas (EC) of no special molecular profile (NSMP) represent the largest molecular category of EC, comprising a mixture of tumors with different histology and molecular profiles. These facts likely point to different tumor biology, clinical outcomes, and targeted therapy responses within this molecular category. The PIK3CA is currently the only targetable kinase oncoprotein directly implicated in EC carcinogenesis. Investigating a unique single-institution cohort, we attempted to stratify NSMP ECs based on the presence of the PIK3CA pathogenic mutation. Those cases were further analyzed for other well-established-associated oncogenic driver gene mutations. Histological and clinical variables were also correlated in each case. Altogether, 175 ECs were prospectively tested by a limited custom NGS panel containing ARID1A, BCOR, BRCA1, BRCA2, CTNNB1, KRAS, MLH1, MSH2, MSH6, NRAS, PIK3CA, PMS2, POLD1, POLE, PTEN,and TP53 genes. We identified 24 PIK3CA mutated cases in the group of 80 NSMP ECs, with another co-occurring mutation in at least one oncogenic driver gene (CTNNB1, PTEN, ARID1A, KRAS, BCOR, PMS2) in 19 cases. In conclusion, a limited NGS panel can effectively test EC tissue for specific pathogenetically relevant oncogene mutations. The NSMP EC category contains 30% of the PIK3CA mutated cases. Of those, 21% contain the PIK3CA mutation as a sole EC-associated oncogene mutation, while 79% harbor at least one more mutated gene. These findings may inform future healthcare planning and improve the effectiveness of EC patient selection for the PIK3CA-targeted therapy.
Collapse
Affiliation(s)
- Ondrej Ondič
- Department of Pathology, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic.
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic.
| | - Květoslava Michalová
- Department of Pathology, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic
| | - Marián Švajdler
- Department of Pathology, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic
| | - Jiří Presl
- Department of Gynecology and Obstetrics, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
| | - Jan Kosťun
- Department of Gynecology and Obstetrics, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
| | - Veronika Hájková
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic
| | - Petr Martínek
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic
| | - Michal Michal
- Department of Pathology, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
| |
Collapse
|
6
|
Nozawa A, Abe T, Niihori T, Ozeki M, Aoki Y, Ohnishi H. Lymphatic endothelial cell-specific NRAS p.Q61R mutant embryos show abnormal lymphatic vessel morphogenesis. Hum Mol Genet 2024; 33:1420-1428. [PMID: 38743908 DOI: 10.1093/hmg/ddae080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/14/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
Generalized lymphatic anomaly (GLA) and kaposiform lymphangiomatosis (KLA) are rare congenital disorders that arise through anomalous embryogenesis of the lymphatic system. A somatic activating NRAS p.Q61R variant has been recently detected in GLA and KLA tissues, suggesting that the NRAS p.Q61R variant plays an important role in the development of these diseases. To address this role, we studied the effect of the NRAS p.Q61R variant in lymphatic endothelial cells (LECs) on the structure of the lymphatics during embryonic and postnatal lymphangiogenesis applying inducible, LEC-specific NRAS p.Q61R variant in mice. Lox-stop-Lox NrasQ61R mice were crossed with Prox1-CreERT2 mice expressing tamoxifen-inducible Cre recombinase specifically in LECs. Whole-mount immunostaining of embryonic back skin using an antibody against the LEC surface marker VEGFR3 showed considerably greater lymphatic vessel width in LEC-specific NRAS p.Q61R mutant embryos than in littermate controls. These mutant embryos also showed a significant reduction in the number of lymphatic vessel branches. Furthermore, immunofluorescence staining of whole-mount embryonic back skin using an antibody against the LEC-specific nuclear marker Prox1 showed a large increase in the number of LECs in LEC-specific NRAS p.Q61R mutants. In contrast, postnatal induction of the NRAS p.Q61R variant in LECs did not cause abnormal lymphatic vessel morphogenesis. These results suggest that the NRAS p.Q61R variant in LECs plays a role in development of lymphatic anomalies. While this model does not directly reflect the human pathology of GLA and KLA, there are overlapping features, suggesting that further study of this model may help in studying GLA and KLA mechanisms.
Collapse
Affiliation(s)
- Akifumi Nozawa
- Department of Pediatrics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Taiki Abe
- Department of Medical Genetics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Sendai 980-8574, Japan
| | - Tetsuya Niihori
- Department of Medical Genetics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Sendai 980-8574, Japan
| | - Michio Ozeki
- Department of Pediatrics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, 1-1 Yanagido,Gifu 501-1194, Japan
| | - Yoko Aoki
- Department of Medical Genetics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Sendai 980-8574, Japan
| | - Hidenori Ohnishi
- Department of Pediatrics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, 1-1 Yanagido,Gifu 501-1194, Japan
- Clinical Genetics Center, Gifu University Hospital, 1-1 Yanagido,Gifu501-1194, Japan
| |
Collapse
|
7
|
Morin GM, Zerbib L, Kaltenbach S, Fraissenon A, Balducci E, Asnafi V, Canaud G. PIK3CA-Related Disorders: From Disease Mechanism to Evidence-Based Treatments. Annu Rev Genomics Hum Genet 2024; 25:211-237. [PMID: 38316164 DOI: 10.1146/annurev-genom-121222-114518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Recent advances in genetic sequencing are transforming our approach to rare-disease care. Initially identified in cancer, gain-of-function mutations of the PIK3CA gene are also detected in malformation mosaic diseases categorized as PIK3CA-related disorders (PRDs). Over the past decade, new approaches have enabled researchers to elucidate the pathophysiology of PRDs and uncover novel therapeutic options. In just a few years, owing to vigorous global research efforts, PRDs have been transformed from incurable diseases to chronic disorders accessible to targeted therapy. However, new challenges for both medical practitioners and researchers have emerged. Areas of uncertainty remain in our comprehension of PRDs, especially regarding the relationship between genotype and phenotype, the mechanisms underlying mosaicism, and the processes involved in intercellular communication. As the clinical and biological landscape of PRDs is constantly evolving, this review aims to summarize current knowledge regarding PIK3CA and its role in nonmalignant human disease, from molecular mechanisms to evidence-based treatments.
Collapse
Affiliation(s)
- Gabriel M Morin
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- UFR de Médecine, Site Necker, Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Lola Zerbib
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- UFR de Médecine, Site Necker, Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sophie Kaltenbach
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Antoine Fraissenon
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- CREATIS, CNRS UMR 5220, Villeurbanne, France
- Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne, France
- Service d'Imagerie Pédiatrique, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Estelle Balducci
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vahid Asnafi
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- UFR de Médecine, Site Necker, Université Paris Cité, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Guillaume Canaud
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France;
- UFR de Médecine, Site Necker, Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
8
|
Kuentz P, Engel C, Laeng M, Chevarin M, Duffourd Y, Martel J, Piard J, Morice-Picard F, Aubert H, Bessis D, Guerrot AM, Maruani A, Boccara O, Mazereeuw-Hautier J, Ott H, Phan A, Puzenat E, Quelin C, Thauvin-Robinet C, Faivre L, Vabres P. Clinical phenotype of the PIK3R1-related vascular overgrowth syndrome. Br J Dermatol 2024; 191:303-305. [PMID: 38623710 DOI: 10.1093/bjd/ljae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/25/2024] [Accepted: 04/18/2024] [Indexed: 04/17/2024]
Abstract
Here we report 19 additional patients with PIK3R1 mosaic variants with clinical phenotyping, showing that the PIK3R1 phenotype is indistinguishable from the PIK3CA-related phenotypes, although the megalencephaly-capillary malformation phenotype is consistently absent in patients with PIK3R1 variants. We also report novel PIK3R1 variants. We consider that the meaning of PROS should shift from ‘PIK3CA-related overgrowth spectrum’ to ‘PI3-kinase-related overgrowth spectrum’.
Collapse
Affiliation(s)
- Paul Kuentz
- Université de Franche-Comté, CHU Besançon, Oncobiologie Génétique Bioinformatique, FHU-TRANSLAD et Institut GIMI, F-25000 Besançon, France
| | | | - Mathieu Laeng
- Université de Franche-Comté, CHU Besançon, Oncobiologie Génétique Bioinformatique, FHU-TRANSLAD et Institut GIMI, F-25000 Besançon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Chen H, Sun B, Liu H, Gao W, Qiu Y, Hua C, Lin X. Delineation of the phenotypes and genotypes of PIK3CA-related overgrowth spectrum in East asians. Mol Genet Genomics 2024; 299:66. [PMID: 38980418 DOI: 10.1007/s00438-024-02159-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024]
Abstract
PIK3CA-related overgrowth spectrum (PROS) is an umbrella term to describe a diverse range of developmental disorders. Research to date has predominantly emerged from Europe and North America, resulting in a notable scarcity of studies focusing on East Asian populations. Currently, the prevalence and distribution of PIK3CA variants across various genetic loci and their correlation with distinct phenotypes in East Asian populations remain unclear. This study aims to elucidate the phenotype-genotype correlations of PROS in East Asian populations. We presented the phenotypes and genotypes of 82 Chinese patients. Among our cohort, 67 individuals carried PIK3CA variants, including missense, frameshift, and splice variants. Six patients presented with both PIK3CA and an additional variant. Seven PIK3CA-negative patients exhibited overlapping PROS manifestations with variants in GNAQ, AKT1, PTEN, MAP3K3, GNA11, or KRAS. An integrative review of the literature pertaining to East Asian populations revealed that specific variants are uniquely associated with certain PROS phenotypes. Some rare variants were exclusively identified in cases of megalencephaly and diffuse capillary malformation with overgrowth. Non-hotspot variants with undefined oncogenicity were more common in CNS phenotypes. Diseases with vascular malformation were more likely to have variants in the helical domain, whereas phenotypes involving adipose/muscle overgrowth without vascular abnormalities predominantly presented variants in the C2 domain. Our findings underscore the unique phenotype-genotype patterns within the East Asian PROS population, highlighting the necessity for an expanded cohort to further elucidate these correlations. Such endeavors would significantly facilitate the development of PI3Kα selective inhibitors tailored for the East Asian population in the future.
Collapse
Affiliation(s)
- Hongrui Chen
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Bin Sun
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Hongyuan Liu
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Wei Gao
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Yajing Qiu
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Chen Hua
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China.
| | - Xiaoxi Lin
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China.
| |
Collapse
|
10
|
Giżewska-Kacprzak K, Śliwiński M, Nicieja K, Babiak-Choroszczak L, Walaszek I. Macrodactyly. CHILDREN (BASEL, SWITZERLAND) 2024; 11:753. [PMID: 39062202 PMCID: PMC11274991 DOI: 10.3390/children11070753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024]
Abstract
Macrodactyly is a rare congenital limb difference manifesting as an overgrowth of one or more fingers or toes. The pathological process affects all tissues of the ray in the hand or foot. The enlargement can significantly alter the limb's appearance and impair its function. The role of a pediatrician is to distinguish isolated macrodactyly from syndromic conditions (including PIK3CA-Related Overgrowth Spectrum) or mimicking conditions to enable early interdisciplinary consultation and treatment planning. The psychological stigma associated with this often disfiguring condition necessitates support for patients and their family. We present a practical guide for physicians who might be the first to raise suspicion of macrodactyly and initiate further diagnostics to achieve adequate treatment and support for children and caregivers.
Collapse
Affiliation(s)
- Kaja Giżewska-Kacprzak
- Department of Pediatric and Oncological Surgery, Urology and Hand Surgery, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland; (M.Ś.)
| | - Maximilian Śliwiński
- Department of Pediatric and Oncological Surgery, Urology and Hand Surgery, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland; (M.Ś.)
| | - Karol Nicieja
- Department of Pediatric and Oncological Surgery, Urology and Hand Surgery, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland; (M.Ś.)
| | - Lidia Babiak-Choroszczak
- Department of Pediatric and Oncological Surgery, Urology and Hand Surgery, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland; (M.Ś.)
| | - Ireneusz Walaszek
- Department of Pediatric and Oncological Surgery, Urology and Hand Surgery, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland; (M.Ś.)
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 48 Żołnierska St., 71-210 Szczecin, Poland
| |
Collapse
|
11
|
Abdelilah-Seyfried S, Ola R. Shear stress and pathophysiological PI3K involvement in vascular malformations. J Clin Invest 2024; 134:e172843. [PMID: 38747293 PMCID: PMC11093608 DOI: 10.1172/jci172843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024] Open
Abstract
Molecular characterization of vascular anomalies has revealed that affected endothelial cells (ECs) harbor gain-of-function (GOF) mutations in the gene encoding the catalytic α subunit of PI3Kα (PIK3CA). These PIK3CA mutations are known to cause solid cancers when occurring in other tissues. PIK3CA-related vascular anomalies, or "PIKopathies," range from simple, i.e., restricted to a particular form of malformation, to complex, i.e., presenting with a range of hyperplasia phenotypes, including the PIK3CA-related overgrowth spectrum. Interestingly, development of PIKopathies is affected by fluid shear stress (FSS), a physiological stimulus caused by blood or lymph flow. These findings implicate PI3K in mediating physiological EC responses to FSS conditions characteristic of lymphatic and capillary vessel beds. Consistent with this hypothesis, increased PI3K signaling also contributes to cerebral cavernous malformations, a vascular disorder that affects low-perfused brain venous capillaries. Because the GOF activity of PI3K and its signaling partners are excellent drug targets, understanding PIK3CA's role in the development of vascular anomalies may inform therapeutic strategies to normalize EC responses in the diseased state. This Review focuses on PIK3CA's role in mediating EC responses to FSS and discusses current understanding of PIK3CA dysregulation in a range of vascular anomalies that particularly affect low-perfused regions of the vasculature. We also discuss recent surprising findings linking increased PI3K signaling to fast-flow arteriovenous malformations in hereditary hemorrhagic telangiectasias.
Collapse
Affiliation(s)
| | - Roxana Ola
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
12
|
Engel C, Chevarin M, Piard J, Abad M, Thomas Q, Carmignac V, Duffourd Y, Lemesle-Martin M, Tarris G, Thauvin-Robinet C, Vabres P, Faivre L, Kuentz P. Allelic heterogeneity in a patient with postzygotic MTOR-related hypomelanosis of Ito with neurodevelopmental abnormalities. Clin Genet 2024; 105:581-583. [PMID: 38379111 DOI: 10.1111/cge.14511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/25/2024] [Accepted: 02/12/2024] [Indexed: 02/22/2024]
Abstract
A case of mosaic MTOR-associated hemimegalencephaly and hypomelanosis of Ito, died at 33 probably because of sudden unexpected death in epilepsy. Assessment of the variant allele fraction (VAF) in different tissues postmortem showed high variability not correlated with clinical features, representing the most detailed assessment of VAFs in different tissues to date.
Collapse
Affiliation(s)
- Camille Engel
- Centre de Génétique Humaine, CHU Besançon, Besançon, France
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
| | - Martin Chevarin
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
- Unité Fonctionnelle Innovation Diagnostique dans les Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Juliette Piard
- Centre de Génétique Humaine, CHU Besançon, Besançon, France
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
| | - Marine Abad
- Service d'Anatomie Pathologique, CHU Besançon, Besançon, France
| | - Quentin Thomas
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
- Service de Neurologie, CHU Dijon Bourgogne, Dijon, France
| | - Virginie Carmignac
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
| | - Yannis Duffourd
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
- Unité Fonctionnelle Innovation Diagnostique dans les Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | | | - Georges Tarris
- Service de Neurologie, CHU Dijon Bourgogne, Dijon, France
| | - Christel Thauvin-Robinet
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
- Unité Fonctionnelle Innovation Diagnostique dans les Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
- Centre de Génétique et Centres de référence Anomalies du Développement et Déficience Intellectuelle, FHU-TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Pierre Vabres
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
- Centre de référence MAGEC "Maladies Génétiques à Expression Cutanée", FHU-TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Laurence Faivre
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
- Centre de Génétique et Centres de référence Anomalies du Développement et Déficience Intellectuelle, FHU-TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Paul Kuentz
- INSERM UMR1231 GAD "Génétique des Anomalies du Développement", Université de Bourgogne Franche-Comté, Dijon, France
- Oncobiologie Génétique Bioinformatique, FHU-TRANSLAD et Institut GIMI, CHU Besançon, Besançon, France
| |
Collapse
|
13
|
Laval N, Kleiber N, Soucy JF, Dubois J, Assaad MA. Atypical Presentation and Evolution of Necrotizing Enterocolitis as a PIK3CA Pathological Variant. Cureus 2024; 16:e59243. [PMID: 38813336 PMCID: PMC11134116 DOI: 10.7759/cureus.59243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2024] [Indexed: 05/31/2024] Open
Abstract
Activating mutation of PIK3CA is linked with cases of overgrowth syndromes and belongs to the PIK3CA-related overgrowth spectrum (PROS). Mutations in this gene are associated with vascular malformations, brain abnormalities, and an increased risk for certain tumors. We report the case of a newborn girl, preterm at 34 weeks of gestation, referred to our center for atypical necrotizing enterocolitis (NEC). At laparotomy, the appearance of the intestinal tract was described as puffy, cauliflower-like with a dark purplish coloration. Subsequently, the colostomy was described as having a consistent proliferative appearance. Medical treatment with sirolimus resulted in minimal improvement. There are no reported cases in the literature of association between NEC and PIK3CA mutation. It is possible that PIK3CA mutation, including the related vascular anomalies, plays a role in the pathogenesis of NEC with this condition.
Collapse
Affiliation(s)
- Nancy Laval
- Neonatology, Centre Hospitalier Universitaire Sainte-Justine, Montréal, CAN
- Neonatology, Centre Hospitalier Chrétien MontLégia, Liège, BEL
| | - Niina Kleiber
- Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Montréal, CAN
| | - Jean-François Soucy
- Medical Genetics, Centre Hospitalier Universitaire Sainte-Justine, Montréal, CAN
| | - Josée Dubois
- Radiology, Centre Hospitalier Universitaire Sainte-Justine, Montréal, CAN
| | | |
Collapse
|
14
|
de Kock L, Cuillerier A, Gillespie M, Couse M, Hartley T, Mears W, Bernier FP, Chudley AE, Frosk P, Nikkel SM, Innes AM, Lauzon J, Thomas M, Guerin A, Armour CM, Weksberg R, Scott JN, Watkins D, Harvey S, Cytrynbaum C, Kernohan KD, Boycott KM. Molecular characterization of 13 patients with PIK3CA-related overgrowth spectrum using a targeted deep sequencing approach. Am J Med Genet A 2024; 194:e63466. [PMID: 37949664 DOI: 10.1002/ajmg.a.63466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
Activating variants in the PIK3CA gene cause a heterogeneous spectrum of disorders that involve congenital or early-onset segmental/focal overgrowth, now referred to as PIK3CA-related overgrowth spectrum (PROS). Historically, the clinical diagnoses of patients with PROS included a range of distinct syndromes, including CLOVES syndrome, dysplastic megalencephaly, hemimegalencephaly, focal cortical dysplasia, Klippel-Trenaunay syndrome, CLAPO syndrome, fibroadipose hyperplasia or overgrowth, hemihyperplasia multiple lipomatosis, and megalencephaly capillary malformation-polymicrogyria (MCAP) syndrome. MCAP is a sporadic overgrowth disorder that exhibits core features of progressive megalencephaly, vascular malformations, distal limb malformations, cortical brain malformations, and connective tissue dysplasia. In 2012, our research group contributed to the identification of predominantly mosaic, gain-of-function variants in PIK3CA as an underlying genetic cause of the syndrome. Mosaic variants are technically more difficult to detect and require implementation of more sensitive sequencing technologies and less stringent variant calling algorithms. In this study, we demonstrated the utility of deep sequencing using the Illumina TruSight Oncology 500 (TSO500) sequencing panel in identifying variants with low allele fractions in a series of patients with PROS and suspected mosaicism: pathogenic, mosaic PIK3CA variants were identified in all 13 individuals, including 6 positive controls. This study highlights the importance of screening for low-level mosaic variants in PROS patients. The use of targeted panels with deep sequencing in clinical genetic testing laboratories would improve diagnostic yield and accuracy within this patient population.
Collapse
Affiliation(s)
- Leanne de Kock
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Alexanne Cuillerier
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Meredith Gillespie
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Madeline Couse
- The Centre for Computational Medicine, the Hospital for Sick Children (SickKids) Research Institute, Toronto, Ontario, Canada
| | - Taila Hartley
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Wendy Mears
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Francois P Bernier
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Albert E Chudley
- Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Patrick Frosk
- Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sarah M Nikkel
- Provincial Medical Genetics Program, BC Women's Hospital, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - A Micheil Innes
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Julie Lauzon
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Maryann Thomas
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Andrea Guerin
- Division of Medical Genetics, Department of Pediatrics, Queen's University, Kingston, Ontario, Canada
| | - Christine M Armour
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Rosanna Weksberg
- Division of Clinical and Metabolic Genetics, Department of Paediatrics and Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - James N Scott
- Departments of Diagnostic Imaging and Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Debra Watkins
- Northeastern Ontario Medical Genetics Program, Health Sciences North, Greater Sudbury, Ontario, Canada
| | - Shirley Harvey
- Program of Genetics and Metabolism, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Cheryl Cytrynbaum
- Division of Clinical and Metabolic Genetics, Department of Genetic Counselling and Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kristin D Kernohan
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
- Newborn Screening Ontario, Ottawa, Ontario, Canada
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
15
|
Wataya-Kaneda M, Maeda S, Nakamura A, Hayashi M, Fujimoto M. Verification of the efficacy of topical sirolimus gel for systemic rare vascular malformations: a pilot study. J Dermatol 2023; 50:1619-1624. [PMID: 37649426 DOI: 10.1111/1346-8138.16930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023]
Abstract
Numerous clinical trials of sirolimus, an inhibitor of mechanistic/mammalian target of rapamycin complex 1, for the treatment of vascular malformations have been conducted. However, aside from lymphatic malformations, the efficacy of sirolimus for venous and capillary malformations has not been established. Moreover, no generalized venous or capillary malformations have been treated with topical sirolimus. To evaluate the safety and efficacy of topical sirolimus for venous and capillary malformations and to compare the efficacy of topical and systemic sirolimus therapy, an open-label single-arm pilot study with 0.2% sirolimus gel was conducted from July 19, 2019, to January 30, 2020, in four patients diagnosed with different vascular malformations (blue rubber bleb nevus syndrome, common venous malformation, phakomatosis pigmentovascularis type IVb, and angiokeratoma in Fabry disease). The primary endpoint was the safety evaluation of sirolimus gel. The main secondary endpoint was the improvement rate evaluated by the Central Judgment Committee at 12 weeks using photographs. No adverse events were observed. Blood sirolimus was not detected in any patient. Two patients (50%) had mild improvement, and the remaining two patients (50%) showed no change after 12 weeks of treatment. Blue rubber bleb nevus syndrome, a generalized venous malformation, showed the greatest response. In conclusion, 0.2% sirolimus gel was found to be as clinically effective as systemic sirolimus treatment in patients with venous and capillary malformations and more effective for early active lesions, even systemic venous malformations.
Collapse
Affiliation(s)
- Mari Wataya-Kaneda
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Neurocutaneous Medicine, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shinichirou Maeda
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Ayumi Nakamura
- Department of Pharmacy, Osaka University Hospital, Osaka, Japan
| | - Misa Hayashi
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
16
|
Gazzin A, Leoni C, Viscogliosi G, Borgini F, Perri L, Iacoviello M, Piglionica M, De Pellegrin M, Ferrero GB, Bartuli A, Zampino G, Buonuomo PS, Resta N, Mussa A. Work-Up and Treatment Strategies for Individuals with PIK3CA-Related Disorders: A Consensus of Experts from the Scientific Committee of the Italian Macrodactyly and PROS Association. Genes (Basel) 2023; 14:2134. [PMID: 38136956 PMCID: PMC10742781 DOI: 10.3390/genes14122134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 11/23/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
PIK3CA-related disorders encompass many rare and ultra-rare conditions caused by somatic genetic variants that hyperactivate the PI3K-AKT-mTOR signaling pathway, which is essential for cell cycle control. PIK3CA-related disorders include PIK3CA-related overgrowth spectrum (PROS), PIK3CA-related vascular malformations and PIK3CA-related non-vascular lesions. Phenotypes are extremely heterogeneous and overlapping. Therefore, diagnosis and management frequently involve various health specialists. Given the rarity of these disorders and the limited number of centers offering optimal care, the Scientific Committee of the Italian Macrodactyly and PROS Association has proposed a revision of the most recent recommendations for the diagnosis, molecular testing, clinical management, follow-up, and treatment strategies. These recommendations give insight on molecular diagnosis, eligible samples, preferable sequencing, and validation methods and management of negative results. The purpose of this paper is to promote collaboration between health care centers and clinicians with a joint shared approach. Finally, we suggest the direction of present and future research studies, including new systemic target therapies, which are currently under evaluation in several clinical trials, such as specific inhibitors that can be employed to downregulate the signaling pathway.
Collapse
Affiliation(s)
- Andrea Gazzin
- Clinical Pediatric Genetics Unit, Department of Public Health and Pediatrics, University of Torino, Regina Margherita Children’s Hospital, 10126 Torino, Italy; (A.G.); (A.M.)
- Postgraduate School of Pediatrics, University of Torino, 10126 Torino, Italy
| | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (G.V.); (L.P.); (G.Z.)
| | - Germana Viscogliosi
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (G.V.); (L.P.); (G.Z.)
| | - Federica Borgini
- Italian Macrodactyly and PROS Association, 27010 San Genesio ed Uniti (PV), Italy;
| | - Lucrezia Perri
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (G.V.); (L.P.); (G.Z.)
| | - Matteo Iacoviello
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.I.); (M.P.); (N.R.)
| | - Marilidia Piglionica
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.I.); (M.P.); (N.R.)
| | - Maurizio De Pellegrin
- Pediatric Orthopedic Unit, Piccole Figlie Hospital, 43125 Parma, Italy
- Department of Orthopedics, ASST Ospedale Papa Giovanni XXIII, 24127 Bergamo, Italy
| | | | - Andrea Bartuli
- Rare Disease and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (A.B.); (P.S.B.)
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (G.V.); (L.P.); (G.Z.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Paola Sabrina Buonuomo
- Rare Disease and Medical Genetics Unit, Bambino Gesù Children’s Hospital, IRCCS, 00168 Rome, Italy; (A.B.); (P.S.B.)
| | - Nicoletta Resta
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.I.); (M.P.); (N.R.)
| | - Alessandro Mussa
- Clinical Pediatric Genetics Unit, Department of Public Health and Pediatrics, University of Torino, Regina Margherita Children’s Hospital, 10126 Torino, Italy; (A.G.); (A.M.)
| |
Collapse
|
17
|
Welters A, Leiter SM, Bachmann N, Bergmann C, Hoermann H, Korsch E, Meissner T, Payne F, Williams R, Hussain K, Semple RK, Kummer S. An expanded clinical spectrum of hypoinsulinaemic hypoketotic hypoglycaemia. Orphanet J Rare Dis 2023; 18:360. [PMID: 37974153 PMCID: PMC10652530 DOI: 10.1186/s13023-023-02954-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Hypoketotic hypoglycaemia with suppressed plasma fatty acids and detectable insulin suggests congenital hyperinsulinism (CHI). Severe hypoketotic hypoglycaemia mimicking hyperinsulinism but without detectable insulin has recently been described in syndromic individuals with mosaic genetic activation of post-receptor insulin signalling. We set out to expand understanding of this entity focusing on metabolic phenotypes. METHODS Metabolic profiling, candidate gene and exome sequencing were performed in six infants with hypoketotic, hypoinsulinaemic hypoglycaemia, with or without syndromic features. Additional signalling studies were carried out in dermal fibroblasts from two individuals. RESULTS Two infants had no syndromic features. One was mistakenly diagnosed with CHI. One had mild features of megalencephaly-capillary malformation-polymicrogyria (MCAP) syndrome, one had non-specific macrosomia, and two had complex syndromes. All required intensive treatment to maintain euglycaemia, with CHI-directed therapies being ineffective. Pathogenic PIK3CA variants were found in two individuals - de novo germline c.323G>A (p.Arg108His) in one non-syndromic infant and postzygotic mosaic c.2740G>A (p.Gly914Arg) in the infant with MCAP. No causal variants were proven in the other individuals despite extensive investigation, although rare variants in mTORC components were identified in one. No increased PI3K signalling in fibroblasts of two individuals was seen. CONCLUSIONS We expand the spectrum of PI3K-related hypoinsulinaemic hypoketotic hypoglycaemia. We demonstrate that pathogenic germline variants activating post-insulin-receptor signalling may cause non-syndromic hypoinsulinaemic hypoketotic hypoglycaemia closely resembling CHI. This distinct biochemical footprint should be sought and differentiated from CHI in infantile hypoglycaemia. To facilitate adoption of this differential diagnosis, we propose the term "pseudohyperinsulinism".
Collapse
Affiliation(s)
- Alena Welters
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Children's Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Sarah M Leiter
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Nadine Bachmann
- Medizinische Genetik Mainz, Limbach Genetics, Mainz, Germany
| | | | - Henrike Hoermann
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Children's Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Eckhard Korsch
- Paediatric Endocrinology, Children's Hospital, Amsterdamer Straße 59, Cologne, Germany
| | - Thomas Meissner
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Children's Hospital, Heinrich-Heine University, Düsseldorf, Germany
| | - Felicity Payne
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Rachel Williams
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Khalid Hussain
- Department of Paediatric Medicine, Division of Endocrinology and Diabetes, Sidra Medicine, Education City North Campus, Doha, Qatar
| | - Robert K Semple
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Sebastian Kummer
- Department of General Paediatrics, Neonatology and Paediatric Cardiology, Medical Faculty, University Children's Hospital, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
18
|
Reynolds G, Cardaropoli S, Carli D, Luca M, Gazzin A, Coppo P, La Selva R, Piglionica M, Bagnulo R, Turchiano A, Ranieri C, Resta N, Mussa A. Epidemiology of the disorders of the Pik3ca-related overgrowth spectrum (Pros). Eur J Hum Genet 2023; 31:1333-1336. [PMID: 37365400 PMCID: PMC10620148 DOI: 10.1038/s41431-023-01414-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/18/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
PIK3CA pathogenic variants are responsible for a group of overgrowth syndromes, collectively known as PIK3CA-Related Overgrowth Spectrum (PROS). These gain-of-function variants arise postzygotically, and, according to time of onset, kind of embryonal tissue affected and regional body extension, give rise to heterogeneous phenotypes. PROS rarity and heterogeneity hamper the correct estimation of its epidemiology. Our work represents the first attempt to define the prevalence of PROS according to the established diagnostic criteria and molecular analysis and based on solid demographic data. We assessed the prevalence in Piedmont Region (Italy), including in the study all participants diagnosed with PROS born there from 1998 to 2021. The search identified 37 cases of PROS born across the 25-year period, providing a prevalence of 1:22,313 live births. Molecular analysis was positive in 81.0% of participants. Taking into account the cases with a detected variant in PIK3CA (n = 30), prevalence of molecularly positive PROS was 1:27,519.
Collapse
Affiliation(s)
- Giuseppe Reynolds
- Department of Public Health and Pediatric Sciences, School of Medicine, University of Torino, Torino, Italy
| | - Simona Cardaropoli
- Department of Public Health and Pediatric Sciences, School of Medicine, University of Torino, Torino, Italy
| | - Diana Carli
- Department of Medical Science, University of Torino, Torino, Italy
| | - Maria Luca
- Department of Medical Science, University of Torino, Torino, Italy
| | - Andrea Gazzin
- Department of Public Health and Pediatric Sciences, School of Medicine, University of Torino, Torino, Italy
| | - Paola Coppo
- Pediatric Dermatology, Regina Margherita Children's Hospital, Città della Salute e della Scienza di Torino, Torino, Italy
| | - Roberta La Selva
- Pediatric Dermatology, Regina Margherita Children's Hospital, Città della Salute e della Scienza di Torino, Torino, Italy
| | - Marilidia Piglionica
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J) University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Rosanna Bagnulo
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J) University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Antonella Turchiano
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J) University of Bari "Aldo Moro", 70124, Bari, Italy
| | | | - Nicoletta Resta
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J) University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Alessandro Mussa
- Department of Public Health and Pediatric Sciences, School of Medicine, University of Torino, Torino, Italy.
| |
Collapse
|
19
|
Chen H, Sun B, Gao W, Jia H, Zhou L, Hua C, Lin X. Facial infiltrating lipomatosis with hemimegalencephaly and lymphatic malformations caused by nonhotspot phosphatidylinositol 3-kinase catalytic subunit alpha mutation. Pediatr Dermatol 2023; 40:1115-1119. [PMID: 37190882 DOI: 10.1111/pde.15346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/28/2023] [Indexed: 05/17/2023]
Abstract
We report an unusual case of facial infiltrating lipomatosis with hemimegalencephaly and lymphatic malformations. In addition to the clinical data and imaging findings, detection of a heterozygous PIK3CA nonhotspot known pathogenic variant C420R in a facial epidermal nevus provided novel insight into the pathogenic effect of somatic PIK3CA mutations.
Collapse
Affiliation(s)
- Hongrui Chen
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bin Sun
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wei Gao
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Hechen Jia
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Lucia Zhou
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Chen Hua
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoxi Lin
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
20
|
Faivre L, Crépin JC, Réda M, Nambot S, Carmignac V, Abadie C, Mirault T, Faure-Conter C, Mazereeuw-Hautier J, Maza A, Puzenat E, Collonge-Rame MA, Bursztejn AC, Philippe C, Thauvin-Robinet C, Chevarin M, Abasq-Thomas C, Amiel J, Arpin S, Barbarot S, Baujat G, Bessis D, Bourrat E, Boute O, Chassaing N, Coubes C, Demeer B, Edery P, El Chehadeh S, Goldenberg A, Hadj-Rabia S, Haye D, Isidor B, Jacquemont ML, Van Kien PK, Lacombe D, Lehalle D, Lambert L, Martin L, Maruani A, Morice-Picard F, Petit F, Phan A, Pinson L, Rossi M, Touraine R, Vanlerberghe C, Vincent M, Vincent-Delorme C, Whalen S, Willems M, Marle N, Verkarre V, Devalland C, Devouassoux-Shisheboran M, Abad M, Rioux-Leclercq N, Bonniaud B, Duffourd Y, Martel J, Binquet C, Kuentz P, Vabres P. Low risk of embryonic and other cancers in PIK3CA-related overgrowth spectrum: Impact on screening recommendations. Clin Genet 2023; 104:554-563. [PMID: 37580112 DOI: 10.1111/cge.14410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 07/07/2023] [Accepted: 07/19/2023] [Indexed: 08/16/2023]
Abstract
The PIK3CA-related overgrowth spectrum (PROS) encompasses various conditions caused by mosaic activating PIK3CA variants. PIK3CA somatic variants are also involved in various cancer types. Some generalized overgrowth syndromes are associated with an increased risk of Wilms tumor (WT). In PROS, abdominal ultrasound surveillance has been advocated to detect WT. We aimed to determine the risk of embryonic and other types of tumors in patients with PROS in order to evaluate surveillance relevance. We searched the clinical charts from 267 PROS patients for the diagnosis of cancer, and reviewed the medical literature for the risk of cancer. In our cohort, six patients developed a cancer (2.2%), and Kaplan Meier analyses estimated cumulative probabilities of cancer occurrence at 45 years of age was 5.6% (95% CI = 1.35%-21.8%). The presence of the PIK3CA variant was only confirmed in two out of four tumor samples. In the literature and our cohort, six cases of Wilms tumor/nephrogenic rests (0.12%) and four cases of other cancers have been reported out of 483 proven PIK3CA patients, in particular the p.(His1047Leu/Arg) variant. The risk of WT in PROS being lower than 5%, this is insufficient evidence to recommend routine abdominal imaging. Long-term follow-up studies are needed to evaluate the risk of other cancer types, as well as the relationship with the extent of tissue mosaicism and the presence or not of the variant in the tumor samples.
Collapse
Affiliation(s)
- Laurence Faivre
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Jean-Charles Crépin
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Service de Dermatologie, CHU Dijon Bourgogne, Dijon, France
- Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), CHU Dijon, Dijon, France
| | - Manon Réda
- Oncogénétique, Centre de lutte contre le cancer Georges François Leclerc, Dijon, France
| | - Sophie Nambot
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, CHU Dijon Bourgogne, Dijon, France
- Oncogénétique, Centre de lutte contre le cancer Georges François Leclerc, Dijon, France
| | - Virginie Carmignac
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), CHU Dijon, Dijon, France
| | | | - Tristan Mirault
- Université Paris Cité, PARCC INSERM U970, Centre de référence des maladies vasculaires rares, Hôpital européen Georges-Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | | | - Aude Maza
- Service de Dermatologie, CHU Toulouse, Toulouse, France
| | - Eve Puzenat
- Service de Dermatologie, CHU Besançon, Besançon, France
| | | | | | - Christophe Philippe
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- UF6254 Innovation en Diagnostic Génomique des Maladies Rares, Plate-forme de Biologie Hospitalo-Universitaire, CHU Dijon-Bourgogne, Dijon, France
| | - Christel Thauvin-Robinet
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Déficiences Intellectuelles de Causes Rares, CHU Dijon Bourgogne, Dijon, France
| | - Martin Chevarin
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- UF6254 Innovation en Diagnostic Génomique des Maladies Rares, Plate-forme de Biologie Hospitalo-Universitaire, CHU Dijon-Bourgogne, Dijon, France
| | - Claire Abasq-Thomas
- Département de Pédiatrie et Génétique Médicale, CHU Brest Morvan, Brest, France
| | - Jeanne Amiel
- Service de Médecine Génomique des Maladies Rares et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Stéphanie Arpin
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Tours, Tours, France
| | | | - Geneviève Baujat
- Service de Médecine Génomique des Maladies Rares et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Didier Bessis
- Département de Dermatologie, CHRU de Montpellier, Montpellier, France
| | - Emmanuelle Bourrat
- Service de dermatologie, centre de référence maladies génétiques à expression cutanée MAGEC, CHU St-Louis, Service de pédiatrie générale, CHU Robert Debré, Paris, France
| | - Odile Boute
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Lille, Lille, France
| | - Nicolas Chassaing
- Service de Génétique Médicale et Centre de Compétence Anomalies du Développement et Syndromes Malformatifs, CHU Toulouse, Toulouse, France
| | - Christine Coubes
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Montpellier, Montpellier, France
| | - Bénédicte Demeer
- Centre d'Activité de Génétique Clinique et Oncogénétique, CHU d'Amiens, Amiens, France
| | - Patrick Edery
- Service de génétique, Centre de Référence Anomalies du Développement, Hospices Civils de Lyon, Bron, France
- INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Université Claude Bernard Lyon 1, Bron, France
| | - Salima El Chehadeh
- Service de Génétique Médicale, Centre de Référence Déficiences Intellectuelles de Causes Rares, Institut de Génétique Médicale d'Alsace (IGMA), CHRU de Strasbourg, Strasbourg, France
| | - Alice Goldenberg
- Service de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Rouen et Centre Normand de Génomique Médicale et Médecine Personnalisée, Rouen, France
| | - Smail Hadj-Rabia
- Service de Dermatologie et Centre de Référence des Maladies Rares Génétiques à Expression Cutanée (MAGEC), Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Universitaire Necker Enfants Malades, Paris, France
| | - Damien Haye
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Tours, Tours, France
| | - Bertrand Isidor
- Service de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Nantes, Nantes, France
| | - Marie-Line Jacquemont
- Unité de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de la Réunion, Saint-Pierre, France
| | - Philippe Khau Van Kien
- Unité de Génétique Médicale et Cytogénétique, Centre de Compétence Anomalies du Développement et Syndromes Malformatifs, CHU de Nîmes, Nîmes, France
| | - Didier Lacombe
- Service de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Bordeaux, Bordeaux, France
| | - Daphné Lehalle
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
| | - Laetitia Lambert
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Nancy, Nancy, France
| | | | | | - Fanny Morice-Picard
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Nancy, Nancy, France
- Service de Dermatologie, CHU de Bordeaux, Bordeaux, France
| | - Florence Petit
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Lille, Lille, France
| | - Alice Phan
- Service de Dermatologie, CHU de Lyon, Lyon, France
| | - Lucile Pinson
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Montpellier, Montpellier, France
| | - Massimiliano Rossi
- Service de génétique, Centre de Référence Anomalies du Développement, Hospices Civils de Lyon, Bron, France
- INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Université Claude Bernard Lyon 1, Bron, France
| | - Renaud Touraine
- Service de Génétique Clinique et Centre de Compétence Anomalies du Développement et Syndromes Malformatifs, CHU de Saint-Etienne, Saint-Etienne, France
| | - Clémence Vanlerberghe
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Lille, Lille, France
| | - Marie Vincent
- Service de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Nantes, Nantes, France
| | - Catherine Vincent-Delorme
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Lille, Lille, France
| | - Sandra Whalen
- Unité Fonctionnelle de Génétique Clinique, Hôpital Armand-Trousseau, Paris, France
| | - Marjolaine Willems
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Montpellier, Montpellier, France
| | - Nathalie Marle
- UF6254 Innovation en Diagnostic Génomique des Maladies Rares, Plate-forme de Biologie Hospitalo-Universitaire, CHU Dijon-Bourgogne, Dijon, France
| | - Virginie Verkarre
- Service d'Anatomie Pathologique, Hôpital Européen Georges Pompidou, Paris, France et INSERM UMR 970, Equipe 13, PARCC Université de Paris Cité, Paris, France
| | - Christine Devalland
- Service d'Anatomie Pathologique, Hôpital Nord Franche Comté, Trevenans, France
| | | | - Marine Abad
- Service d'Anatomie Pathologique, CHU Besançon, Besançon, France
| | | | | | - Yannis Duffourd
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
| | - Jehanne Martel
- Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), CHU Dijon, Dijon, France
| | - Christine Binquet
- INSERM, Université de Bourgogne, CHU Dijon Bourgogne, CIC 1432, Module Épidémiologie Clinique, Dijon, France
| | - Paul Kuentz
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Oncobiologie Génétique Bioinformatique, PCBio, CHU Besançon, Besançon, France
| | - Pierre Vabres
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Service de Dermatologie, CHU Dijon Bourgogne, Dijon, France
- Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), CHU Dijon, Dijon, France
| |
Collapse
|
21
|
Navarro M, Allemang-Trivalle A, Leducq S, Jonville-Bera AP, Maurier A, Zejli T, Edée AE, Harchaoui E, Giraudeau B, Maruani A. Indication for a Pneumocystis Prophylaxis Therapy in Patients with Vascular Anomalies Treated with PIK3/AKT/mTOR Pathway Inhibitors: Experts' Opinion and Systematic Review from the Literature. Dermatology 2023; 239:942-951. [PMID: 37793356 DOI: 10.1159/000533675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 08/14/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Vascular anomalies (VAs) are increasingly being treated with PI3K/AKT/mTOR pathway inhibitors. These drugs have immunosuppressive properties and thus theoretically overexpose patients to opportunistic infections, especially Pneumocystis jirovecii pneumonia (PJP). PJP prophylaxis use lacks consensus. We aimed to investigate the prevalence of PJP in patients receiving mTOR/PI3K/AKT inhibitors for VAs and determine any indication for pneumocystis prophylaxis in this population. METHODS The study was conducted in 2 parts: (1) we sent a survey to a panel of international experts of VAs asking about their use of pneumocystis prophylaxis drugs and (2) we performed a systematic review of the literature of all published cases of patients receiving these drugs for VA to estimate the prevalence of PJP in this population. RESULTS Answers from 68 experts were analyzed: 21 (30.9%) answered they always add PJP prophylaxis when prescribing mTOR inhibitors, 20 (29.4%) case-by-case, and 27 (39.7%) never. For the systematic review, among 3,053 reports screened, 217 were included involving 1,189 patients (1,143 received sirolimus, 38 everolimus, 4 alpelisib, 4 miransertib). Among the 1,189 cases, 2 (0.2%) PJP were reported: one under sirolimus and one under everolimus. Thus, the prevalence of PJP was estimated at 0.88 cases/1,000 patients under sirolimus (95% CI: -0.84 to 2.59) and 26.31 cases/1,000 under everolimus (95% CI: -24.58 to 77.18). Patients with PJP never received prophylaxis drugs. We found no PJP cases under alpelisib and miransertib. PJP prophylaxis was given in 218 (18.3%) cases, more frequently for children (91.3 vs. 77.2% in the non-prophylaxis group, p = 0.012), mostly trimethoprim-sulfamethoxazole (186 patients, 85.3%). CONCLUSION Our study shows that even if PJP is a rare event, it may occur in patients with VAs treated with an mTOR inhibitor. Although our results cannot allow for revising guidelines, prophylaxis with TMP-SMX might be appropriate for a subgroup of patients with risk factors for PJP.
Collapse
Affiliation(s)
- Maxime Navarro
- CHRU Tours, Department of Dermatology, Unit of Pediatric dermatology, Tours, France
- Reference Center for Genodermatoses and Rare Skin Diseases (MAGEC-Tours), Tours, France
| | | | - Sophie Leducq
- CHRU Tours, Department of Dermatology, Unit of Pediatric dermatology, Tours, France
- Reference Center for Genodermatoses and Rare Skin Diseases (MAGEC-Tours), Tours, France
- University of Tours, University of Nantes, INSERM 1246-SPHERE, Tours, France
| | - Annie-Pierre Jonville-Bera
- University of Tours, University of Nantes, INSERM 1246-SPHERE, Tours, France
- CHRU Tours, Department of Clinical Pharmacology, Regional Pharmacovigilance Center, Tours, France
| | - Anaïs Maurier
- CHRU Tours, Department of Clinical Pharmacology, Regional Pharmacovigilance Center, Tours, France
| | - Tarik Zejli
- CHRU Tours, Clinical Investigation Center of Tours, INSERM 1415, Bretonneau Hospital, Tours, France
| | - Afi-Emiliène Edée
- Reference Center for Genodermatoses and Rare Skin Diseases (MAGEC-Tours), Tours, France
| | - Emilie Harchaoui
- CHRU Tours, Department of Dermatology, Unit of Pediatric dermatology, Tours, France
- University of Tours, University of Nantes, INSERM 1246-SPHERE, Tours, France
| | - Bruno Giraudeau
- University of Tours, University of Nantes, INSERM 1246-SPHERE, Tours, France
- CHRU Tours, Clinical Investigation Center of Tours, INSERM 1415, Bretonneau Hospital, Tours, France
| | - Annabel Maruani
- CHRU Tours, Department of Dermatology, Unit of Pediatric dermatology, Tours, France
- Reference Center for Genodermatoses and Rare Skin Diseases (MAGEC-Tours), Tours, France
- University of Tours, University of Nantes, INSERM 1246-SPHERE, Tours, France
| |
Collapse
|
22
|
Sasaki Y, Ishikawa K, Hatanaka KC, Oyamada Y, Sakuhara Y, Shimizu T, Saito T, Murao N, Onodera T, Miura T, Maeda T, Funayama E, Hatanaka Y, Yamamoto Y, Sasaki S. Targeted next-generation sequencing for detection of PIK3CA mutations in archival tissues from patients with Klippel-Trenaunay syndrome in an Asian population : List the full names and institutional addresses for all authors. Orphanet J Rare Dis 2023; 18:270. [PMID: 37667289 PMCID: PMC10478188 DOI: 10.1186/s13023-023-02893-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/26/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Klippel-Trenaunay syndrome (KTS) is a rare slow-flow combined vascular malformation with limb hypertrophy. KTS is thought to lie on the PIK3CA-related overgrowth spectrum, but reports are limited. PIK3CA encodes p110α, a catalytic subunit of phosphatidylinositol 3-kinase (PI3K) that plays an essential role in the PI3K/AKT/mammalian target of rapamycin (mTOR) signaling pathway. We aimed to demonstrate the clinical utility of targeted next-generation sequencing (NGS) in identifying PIK3CA mosaicism in archival formalin-fixed paraffin-embedded (FFPE) tissues from patients with KTS. RESULTS Participants were 9 female and 5 male patients with KTS diagnosed as capillaro-venous malformation (CVM) or capillaro-lymphatico-venous malformation (CLVM). Median age at resection was 14 years (range, 5-57 years). Median archival period before DNA extraction from FFPE tissues was 5.4 years (range, 3-7 years). NGS-based sequencing of PIK3CA achieved an amplicon mean coverage of 119,000x. PIK3CA missense mutations were found in 12 of 14 patients (85.7%; 6/8 CVM and 6/6 CLVM), with 8 patients showing the hotspot variants E542K, E545K, H1047R, and H1047L. The non-hotspot PIK3CA variants C420R, Q546K, and Q546R were identified in 4 patients. Overall, the mean variant allele frequency for identified PIK3CA variants was 6.9% (range, 1.6-17.4%). All patients with geographic capillary malformation, histopathological lymphatic malformation or macrodactyly of the foot had PIK3CA variants. No genotype-phenotype association between hotspot and non-hotspot PIK3CA variants was found. Histologically, the vessels and adipose tissues of the lesions showed phosphorylation of the proteins in the PI3K/AKT/mTOR signaling pathway, including p-AKT, p-mTOR, and p-4EBP1. CONCLUSIONS The PI3K/AKT/mTOR pathway in mesenchymal tissues was activated in patients with KTS. Amplicon-based targeted NGS could identify low-level mosaicism from low-input DNA extracted from FFPE tissues, potentially providing a diagnostic option for personalized medicine with inhibitors of the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuki Sasaki
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
- Center for Vascular Anomalies, Department of Plastic and Reconstructive Surgery, Tonan Hospital, Hokkaido, Japan
| | - Kosuke Ishikawa
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.
- Center for Vascular Anomalies, Department of Plastic and Reconstructive Surgery, Tonan Hospital, Hokkaido, Japan.
| | - Kanako C Hatanaka
- Center for Development of Advanced Diagnostics, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Hokkaido, Japan
| | - Yumiko Oyamada
- Department of Diagnostic Pathology, Tonan Hospital, Hokkaido, Japan
| | - Yusuke Sakuhara
- Department of Diagnostic and Interventional Radiology, Tonan Hospital, Hokkaido, Japan
| | - Tadashi Shimizu
- Department of Diagnostic and Interventional Radiology, Tonan Hospital, Hokkaido, Japan
| | - Tatsuro Saito
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Hokkaido, Japan
- Riken Genesis Co., Ltd, Tokyo, Japan
| | - Naoki Murao
- Center for Vascular Anomalies, Department of Plastic and Reconstructive Surgery, Tonan Hospital, Hokkaido, Japan
| | - Tomohiro Onodera
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Takahiro Miura
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Taku Maeda
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Emi Funayama
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Hokkaido, Japan
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Hokkaido, Japan
| | - Yuhei Yamamoto
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Satoru Sasaki
- Center for Vascular Anomalies, Department of Plastic and Reconstructive Surgery, Tonan Hospital, Hokkaido, Japan
| |
Collapse
|
23
|
Zhang B, He R, Xu Z, Sun Y, Wei L, Li L, Liu Y, Guo W, Song L, Wang H, Lin Z, Ma L. Somatic mutation spectrum of a Chinese cohort of pediatrics with vascular malformations. Orphanet J Rare Dis 2023; 18:261. [PMID: 37658401 PMCID: PMC10474751 DOI: 10.1186/s13023-023-02860-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/20/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Somatic mutations of cancer driver genes are found to be responsible for vascular malformations with clinical manifestations ranging from cutaneous birthmarks to life-threatening systemic anomalies. Till now, only a limited number of cases and mutations were reported in Chinese population. The purpose of this study was to describe the somatic mutation spectrum of a cohort of Chinese pediatrics with vascular malformations. METHODS Pediatrics diagnosed with various vascular malformations were collected between May 2019 and October 2020 from Beijing Children's Hospital. Genomic DNA of skin lesion of each patient was extracted and sequenced by whole-exome sequencing to identify pathogenic somatic mutations. Mutations with variant allele frequency less than 5% were validated by ultra-deep sequencing. RESULTS A total of 67 pediatrics (33 males, 34 females, age range: 0.1-14.8 years) were analyzed. Exome sequencing identified somatic mutations of corresponding genes in 53 patients, yielding a molecular diagnosis rate of 79.1%. Among 29 PIK3CA mutations, 17 were well-known hotspot p.E542K, p.E545K and p.H1047R/L. Non-hotspot mutations were prevalent in patients with PIK3CA-related overgrowth spectrum, accounting for 50.0% (11/22) of detected mutations. The hotspot GNAQ p.R183Q and TEK p.L914F mutations were responsible for the majority of port-wine stain/Sturge-Weber syndrome and venous malformation, respectively. In addition, we identified a novel AKT1 p.Q79K mutation in Proteus syndrome and MAP3K3 p.E387D mutation in verrucous venous malformation. CONCLUSIONS The somatic mutation spectrum of vascular malformations in Chinese population is similar to that reported in other populations, but non-hotspot PIK3CA mutations may also be prevalent. Molecular diagnosis may help the clinical diagnosis, treatment and management of these pediatric patients with vascular malformations.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University (National Center for Children's Health, China), No. 56 Nanlishi Road, Xicheng District, Beijing, 100045, China.
- Department of Dermatology, Zhengzhou University, Affiliated Children's Hospital, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450000, Henan, China.
| | - Rui He
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University (National Center for Children's Health, China), No. 56 Nanlishi Road, Xicheng District, Beijing, 100045, China
| | - Zigang Xu
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University (National Center for Children's Health, China), No. 56 Nanlishi Road, Xicheng District, Beijing, 100045, China
| | - Yujuan Sun
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University (National Center for Children's Health, China), No. 56 Nanlishi Road, Xicheng District, Beijing, 100045, China
| | - Li Wei
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University (National Center for Children's Health, China), No. 56 Nanlishi Road, Xicheng District, Beijing, 100045, China
| | - Li Li
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University (National Center for Children's Health, China), No. 56 Nanlishi Road, Xicheng District, Beijing, 100045, China
| | - Yuanxiang Liu
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University (National Center for Children's Health, China), No. 56 Nanlishi Road, Xicheng District, Beijing, 100045, China
| | - Wu Guo
- Department of Dermatology, Zhengzhou University, Affiliated Children's Hospital, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450000, Henan, China
| | - Li Song
- Department of Dermatology, Zhengzhou University, Affiliated Children's Hospital, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450000, Henan, China
| | - Huijun Wang
- Dermatology Hospital, Southern Medical University, No.2 Lujing Road, Guangzhou, 510091, China
| | - Zhimiao Lin
- Dermatology Hospital, Southern Medical University, No.2 Lujing Road, Guangzhou, 510091, China.
| | - Lin Ma
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University (National Center for Children's Health, China), No. 56 Nanlishi Road, Xicheng District, Beijing, 100045, China.
| |
Collapse
|
24
|
Chen H, Sun B, Gao W, Qiu Y, Hua C, Lin X. Delineation of the phenotypes and genotypes of facial infiltrating lipomatosis associated with PIK3CA mutations. Orphanet J Rare Dis 2023; 18:189. [PMID: 37452404 PMCID: PMC10347770 DOI: 10.1186/s13023-023-02786-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/18/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Facial infiltrating lipomatosis (FIL) is a rare congenital disorder characterized by unilateral facial swelling, for which surgery is the prevailing therapeutic option. Several studies have shown that the development of FIL is closely associated with PIK3CA mutations. This study aimed to further identify rare clinical features and underlying molecular variants in patients with FIL. RESULTS Eighteen patients were included in this study, and all patients presented with infiltrating adipose tissues confirmed by magnetic resonance imaging. Macrodactyly, polydactyly, hemimegalencephaly and hemihyperplasia were also observed in patients with FIL. In total, eight different PIK3CA mutations were detected in tissues obtained from sixteen patients, including the missense mutations p.His1047Arg (n = 4), p.Cys420Arg (n = 2), p.Glu453Lys (n = 2), p.Glu542Lys (n = 2), p.Glu418Lys (n = 1), p.Glu545Lys (n = 1), and p.His1047Tyr (n = 1) and the deletion mutation p.Glu110del (n = 3). Furthermore, the GNAQ mutation p.Arg183Gln was detected in the epidermal nevus tissue of one patient. Imaging revealed that several patients carrying hotspot mutations had more severe adipose infiltration and skeletal deformities. CONCLUSIONS The abundant clinical presentations and genetic profiles of FIL make it difficult to treat. PIK3CA mutations drive the pathogenesis of FIL, and PIK3CA hotspot mutations may lead to more extensive infiltration of lipomatosis. Understanding the molecular variant profile of FIL will facilitate the application of novel PI3K-targeted inhibitors.
Collapse
Affiliation(s)
- Hongrui Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Bin Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Wei Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Yajing Qiu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Chen Hua
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Xiaoxi Lin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
25
|
Ahakoud M, Daha Belghiti H, Ihlal H, Bouguenouch L. A Mosaic PIK3CA Mutation in a Moroccan Female: Exploring the Diagnostic Challenges of PIK3CA-Related Overgrowth Spectrum. Cureus 2023; 15:e36996. [PMID: 37139028 PMCID: PMC10150574 DOI: 10.7759/cureus.36996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2023] [Indexed: 04/04/2023] Open
Abstract
The PIK3CA-related overgrowth spectrum (PROS) encompasses a group of rare disorders characterized by the overgrowth of various body parts, driven by mutations in the PIK3CA gene. This study presents a case of a Moroccan female patient with PROS, demonstrating a phenotype associated with genetic mosaicism in the PIK3CA gene. A multidisciplinary approach, involving clinical examination, radiological assessment, and genetic and bioinformatic analyses, was employed for diagnosis and management. Next-generation sequencing and Sanger sequencing identified a rare variant, c.353G>A, in exon 3 of the PIK3CA gene, not detected in leukocyte DNA but confirmed in tissue biopsy samples. The comprehensive analysis of this case furthers our understanding of PROS and highlights the importance of a multidisciplinary approach to the diagnosis and management of this rare disorder.
Collapse
|
26
|
Di Rocco F, Licci ML, Garde A, Mottolese C, Thauvin-Robinet C, Chevarin M, Guibaud L, Vabres P, Kuentz P, Faivre L. Surgical management of Chiari malformation type 1 associated to MCAP syndrome and study of cerebellar and adjacent tissues for PIK3CA mosaicism. Eur J Med Genet 2023; 66:104678. [PMID: 36503153 DOI: 10.1016/j.ejmg.2022.104678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Subjects with Megalencephaly-Capillary Malformation-Polymicrogyria syndrome (MCAP) can present with a Chiari Malformation Type 1 and resulting alterations in cerebrospinal fluid (CSF) dynamics, which may require surgical treatment. The aim of this paper is to describe the features of children with MCAP who underwent surgical decompression for CM1, and to explore the PIK3CA variant allele frequency (VAF) identified in cerebellar parenchyma and other adjacent structures. METHODS This study reviewed two cases of children with CM1 and MCAP who underwent surgical decompression treatment. These two cases were part of a national cohort of 12 MCAP patients who had CM1, due to their surgical eligibility. Tissue samples were obtained from the cerebellar tonsils and adjacent anatomical structures during the surgical procedures. Samples were then subsequently analyzed for PIK3CA postzygotic variants. RESULTS In both cases, alterations in CSF dynamics, specifically hydrocephalus and syringomyelia, were observed and required surgical treatment. PIK3CA targeted sequencing determined the VAF of the postzygotic variant in both cerebellar and adjacent bone/connective tissues. DISCUSSION The recognition of a CM1 comorbidity in MCAP patients is of paramount importance when considering personalized treatment options, especially because these patients are at higher risk of developing complications during surgical decompression surgery. The variable PIK3CA VAF identified in the different analyzed tissues might help explain the heterogeneous nature and severity of anomalies observed in the volume of the posterior fossa structures in MCAP patients and associated CSF and venous disorders.
Collapse
Affiliation(s)
- Federico Di Rocco
- Service de Neurochirurgie Pédiatrique, Centre de Référence Craniosténoses-Lyon, HCL, Hôpital Femme Mère Enfant, Bron, France.
| | - Maria Lucia Licci
- Service de Neurochirurgie Pédiatrique, Centre de Référence Craniosténoses-Lyon, HCL, Hôpital Femme Mère Enfant, Bron, France
| | - Aurore Garde
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, Dijon, France; Centre de Référence Déficiences Intellectuelles de Causes Rares, FHU TRANSLAD, Hôpital d'Enfants, Dijon, France; Equipe GAD, INSERM UMR1231, Université de Bourgogne, Dijon, France
| | - Carmine Mottolese
- Service de Neurochirurgie Pédiatrique, Centre de Référence Craniosténoses-Lyon, HCL, Hôpital Femme Mère Enfant, Bron, France
| | - Christel Thauvin-Robinet
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, Dijon, France; Centre de Référence Déficiences Intellectuelles de Causes Rares, FHU TRANSLAD, Hôpital d'Enfants, Dijon, France; Equipe GAD, INSERM UMR1231, Université de Bourgogne, Dijon, France
| | - Martin Chevarin
- Equipe GAD, INSERM UMR1231, Université de Bourgogne, Dijon, France
| | - Laurent Guibaud
- Service de Radiologie Pédiatrique, HCL, Hôpital Femme Mère Enfant, Bron, France
| | - Pierre Vabres
- Centre de Référence des Maladies Rares de la Peau et des Muqueuses d'origine Génétique (MAGEC), FHU TRANSLAD, Service de Dermatologie, CHU Dijon, France
| | - Paul Kuentz
- Equipe GAD, INSERM UMR1231, Université de Bourgogne, Dijon, France; Centre de Référence des Maladies Rares de la Peau et des Muqueuses d'origine Génétique (MAGEC), FHU TRANSLAD, Service de Dermatologie, CHU Dijon, France; Oncobiologie Génétique Bioinformatique, CHU Besançon, F-25000, Besançon, France
| | - Laurence Faivre
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, Dijon, France; Centre de Référence Déficiences Intellectuelles de Causes Rares, FHU TRANSLAD, Hôpital d'Enfants, Dijon, France; Equipe GAD, INSERM UMR1231, Université de Bourgogne, Dijon, France
| |
Collapse
|
27
|
Mussa A, Leoni C, Iacoviello M, Carli D, Ranieri C, Pantaleo A, Buonuomo PS, Bagnulo R, Ferrero GB, Bartuli A, Melis D, Maitz S, Loconte DC, Turchiano A, Piglionica M, De Luisi A, Susca FC, Bukvic N, Forleo C, Selicorni A, Zampino G, Onesimo R, Cappuccio G, Garavelli L, Novelli C, Memo L, Morando C, Della Monica M, Accadia M, Capurso M, Piscopo C, Cereda A, Di Giacomo MC, Saletti V, Spinelli AM, Lastella P, Tenconi R, Dvorakova V, Irvine AD, Resta N. Genotypes and phenotypes heterogeneity in PIK3CA-related overgrowth spectrum and overlapping conditions: 150 novel patients and systematic review of 1007 patients with PIK3CA pathogenetic variants. J Med Genet 2023; 60:163-173. [PMID: 35256403 DOI: 10.1136/jmedgenet-2021-108093] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 02/18/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Postzygotic activating PIK3CA variants cause several phenotypes within the PIK3CA-related overgrowth spectrum (PROS). Variant strength, mosaicism level, specific tissue involvement and overlapping disorders are responsible for disease heterogeneity. We explored these factors in 150 novel patients and in an expanded cohort of 1007 PIK3CA-mutated patients, analysing our new data with previous literature to give a comprehensive picture. METHODS We performed ultradeep targeted next-generation sequencing (NGS) on DNA from skin biopsy, buccal swab or blood using a panel including phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin pathway genes and GNAQ, GNA11, RASA1 and TEK. Additionally, 914 patients previously reported were systematically reviewed. RESULTS 93 of our 150 patients had PIK3CA pathogenetic variants. The merged PROS cohort showed that PIK3CA variants span thorough all gene domains, some were exclusively associated with specific PROS phenotypes: weakly activating variants were associated with central nervous system (CNS) involvement, and strongly activating variants with extra-CNS phenotypes. Among the 57 with a wild-type PIK3CA allele, 11 patients with overgrowth and vascular malformations overlapping PROS had variants in GNAQ, GNA11, RASA1 or TEK. CONCLUSION We confirm that (1) molecular diagnostic yield increases when multiple tissues are tested and by enriching NGS panels with genes of overlapping 'vascular' phenotypes; (2) strongly activating PIK3CA variants are found in affected tissue, rarely in blood: conversely, weakly activating mutations more common in blood; (3) weakly activating variants correlate with CNS involvement, strong variants are more common in cases without; (4) patients with vascular malformations overlapping those of PROS can harbour variants in genes other than PIK3CA.
Collapse
Affiliation(s)
- Alessandro Mussa
- Department of Public Health and Pediatric Sciences, Università degli Studi di Torino, Torino, Italy.,Pediatric Clinical Genetics, Regina Margherita Children's Hospital, Hospital, Città della Salute e della Scienza di Torino, Torino, Italy
| | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Matteo Iacoviello
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Diana Carli
- Department of Public Health and Pediatric Sciences, Università degli Studi di Torino, Torino, Italy.,Pediatric Onco-Hematology, Stem Cell Transplantation and Cell Therapy Division, Regina Margherita Children's Hospital, Città Della Salute e Della Scienza di Torino, Torino, Italy
| | - Carlotta Ranieri
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Antonino Pantaleo
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Paola Sabrina Buonuomo
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children's Hospital IRCCS, Roma, Italy
| | - Rosanna Bagnulo
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | | | - Andrea Bartuli
- Rare Diseases and Medical Genetics Unit, Bambino Gesù Children's Hospital IRCCS, Roma, Italy
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Fisciano, Italy
| | - Silvia Maitz
- Clinical Pediatric Genetics Unit, MBBM Foundation, San Gerardo Hospital, Monza, Italy
| | - Daria Carmela Loconte
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Antonella Turchiano
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Marilidia Piglionica
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Annunziata De Luisi
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Francesco Claudio Susca
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Nenad Bukvic
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Cinzia Forleo
- Cardiology Unit, Department of Emergency and Organ Transplantation, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | | | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University Hospital, Napoli, Italy
| | - Livia Garavelli
- Medical Genetics Unit, Mother and Child Health Department, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Chiara Novelli
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milano, Italy
| | - Luigi Memo
- Department of Pediatrics, Neonatal Intensive Care Unit, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Carla Morando
- Department of Pediatrics, Neonatal Intensive Care Unit, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | | | - Maria Accadia
- Medical Genetics Unit, Hospital "Cardinale G. Panico", Tricase, Italy
| | - Martina Capurso
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Carmelo Piscopo
- Medical Genetics Unit, Cardarelli Hospital, Napoli, Italy, Italy
| | - Anna Cereda
- Pediatric Department, ASST Papa Giovanni XXIII, Bergamo, Italy
| | | | - Veronica Saletti
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | - Patrizia Lastella
- Centro Sovraziendale di Assistenza e Ricerca per le Malattie Rare, Internal Medicine Unit 'C. Frugoni', Ospedale Consorziale Policlinico di Bari, Bari, Italy
| | - Romano Tenconi
- Department of Pediatrics, Clinical Genetics, Universita degli Studi di Padova, Padova, Italy
| | - Veronika Dvorakova
- Dermatology Clinic, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Alan D Irvine
- Dermatology Clinic, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Nicoletta Resta
- Department of Biomedical Sciences and Human Oncology, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
28
|
Lai A, Soucy A, El Achkar CM, Barkovich AJ, Cao Y, DiStefano M, Evenson M, Guerrini R, Knight D, Lee YS, Mefford HC, Miller DT, Mirzaa G, Mochida G, Rodan LH, Patel M, Smith L, Spencer S, Walsh CA, Yang E, Yuskaitis CJ, Yu T, Poduri A. The ClinGen Brain Malformation Variant Curation Expert Panel: Rules for somatic variants in AKT3, MTOR, PIK3CA, and PIK3R2. Genet Med 2022; 24:2240-2248. [PMID: 35997716 PMCID: PMC9883838 DOI: 10.1016/j.gim.2022.07.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 01/30/2023] Open
Abstract
PURPOSE Postzygotic (somatic) variants in the mTOR pathway genes cause a spectrum of distinct developmental abnormalities. Accurate classification of somatic variants in this group of disorders is crucial for affected individuals and their families. METHODS The ClinGen Brain Malformation Variant Curation Expert Panel was formed to curate somatic variants associated with developmental brain malformations. We selected the genes AKT3, MTOR, PIK3CA, and PIK3R2 as the first set of genes to provide additional specifications to the 2015 American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) sequence variant interpretation guidelines, which currently focus solely on germline variants. RESULTS A total of 24 of the original 28 ACMG/AMP criteria required modification. Several modifications used could be applied to other genes and disorders in which somatic variants play a role: 1) using variant allele fraction differences as evidence that somatic mutagenesis occurred as a proxy for de novo variation, 2) incorporating both somatic and germline evidence, and 3) delineating phenotype on the basis of variable tissue expression. CONCLUSION We have established a framework for rigorous interpretation of somatic mosaic variants, addressing issues unique to somatic variants that will be applicable to many genes and conditions.
Collapse
Affiliation(s)
- Abbe Lai
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA; Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Aubrie Soucy
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Christelle Moufawad El Achkar
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA; Department of Neurology, Harvard Medical School, Boston, MA
| | | | - Yang Cao
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Marina DiStefano
- Broad Institute of MIT and Harvard, Cambridge, MA; Precision Health Program, Geisinger, Danville, PA
| | - Michael Evenson
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Department of Neuroscience, Meyer Children's University Hospital, University of Florence, Florence, Italy
| | - Devon Knight
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA
| | - Yi-Shan Lee
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Heather C Mefford
- Center for Pediatric Neurological Disease Research, St. Jude Hospital, Memphis, TN
| | - David T Miller
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Ghayda Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA; Department of Pediatrics, University of Washington, Seattle, WA
| | - Ganesh Mochida
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Lance H Rodan
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Department of Pediatrics, Harvard Medical School, Boston, MA; Department of Neurology, Harvard Medical School, Boston, MA
| | - Mayher Patel
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Lacey Smith
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA
| | - Sara Spencer
- Division of Reproductive Genetics, Northwestern Medicine, Chicago, IL
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Department of Pediatrics, Harvard Medical School, Boston, MA; Department of Neurology, Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA
| | - Edward Yang
- Department of Radiology, Boston Children's Hospital and Harvard Medical School, Boston, MA
| | - Christopher J Yuskaitis
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA; Department of Neurology, Harvard Medical School, Boston, MA
| | - Timothy Yu
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Department of Neurology, Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA.
| | - Annapurna Poduri
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA; Department of Neurology, Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA.
| |
Collapse
|
29
|
Becker J, Gross UC, Weber DM, Weibel L, Theiler M, Brandt S, Bode PK. PIK3CA Mutational Analysis in Patients With Macrodactyly. Pediatr Dev Pathol 2022; 25:624-634. [PMID: 36314082 DOI: 10.1177/10935266221080155] [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] [Indexed: 11/05/2022]
Abstract
BACKGROUND Somatic mosaicism for PIK3CA mutations causes various types of growth disorders, which have been summarized under the term PROS (PIK3CA related overgrowth spectrum). Targeted therapy with PI3K inhibitors seems to be a promising alternative for severe PROS cases. Therefore, PIK3CA testing may become more relevant in the future. METHODS We report on 14 PROS patients, who had surgery for macrodactyly in the majority of cases. Clinical data were retrieved from the patient's records. Macroscopic and microscopic findings were retrospectively reviewed. Mutational analysis was performed on formalin-fixed paraffin-embedded (FFPE) material. RESULTS Patient age ranged from 7 months to 35 years. Five patients showed additional anomalies. One patient had CLOVES syndrome. The majority of the specimens were ray resections characterized by hypertrophic fat tissue. Overall, microscopy was subtle. The abnormal adipose tissue showed lobules exhibiting at least focally fibrous septa. In each case, we could detect a PIK3CA mutation. CONCLUSION Histology of affected fat tissue in PROS patients is overall nonspecific. Therefore, mutational analysis represents the key to the diagnosis, especially in unclear clinical cases. We demonstrated that FFPE material is suitable for PIK3CA testing, which can be considered as basis for targeted therapy with PI3K inhibitors.
Collapse
Affiliation(s)
- Jakob Becker
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Ulrike Camenisch Gross
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Daniel M Weber
- Division of Hand Surgery, Department of Pediatric Surgery, 30995University Children's Hospital Zürich, Zürich, Switzerland
| | - Lisa Weibel
- Pediatric Skin Center, Department of Dermatology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Martin Theiler
- Pediatric Skin Center, Department of Dermatology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Simone Brandt
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland.,Institute of Pathology Medica, Zürich, Switzerland
| | - Peter K Bode
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
30
|
Wenger TL, Ganti S, Bull C, Lutsky E, Bennett JT, Zenner K, Jensen DM, Dmyterko V, Mercan E, Shivaram GM, Friedman SD, Bindschadler M, Drusin M, Perkins JN, Kong A, Bly RA, Dahl JP, Bonilla-Velez J, Perkins JA. Alpelisib for the treatment of PIK3CA-related head and neck lymphatic malformations and overgrowth. Genet Med 2022; 24:2318-2328. [PMID: 36066547 PMCID: PMC11091962 DOI: 10.1016/j.gim.2022.07.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE PIK3CA-related overgrowth spectrum (PROS) conditions of the head and neck are treatment challenges. Traditionally, these conditions require multiple invasive interventions, with incomplete malformation removal, disfigurement, and possible dysfunction. Use of the PI3K inhibitor alpelisib, previously shown to be effective in PROS, has not been reported in PIK3CA-associated head and neck lymphatic malformations (HNLMs) or facial infiltrating lipomatosis (FIL). We describe prospective treatment of 5 children with PIK3CA-associated HNLMs or head and neck FIL with alpelisib monotherapy. METHODS A total of 5 children with PIK3CA-associated HNLMs (n = 4) or FIL (n = 1) received alpelisib monotherapy (aged 2-12 years). Treatment response was determined by parental report, clinical evaluation, diary/questionnaire, and standardized clinical photography, measuring facial volume through 3-dimensional photos and magnetic resonance imaging. RESULTS All participants had reduction in the size of lesion, and all had improvement or resolution of malformation inflammation/pain/bleeding. Common invasive therapy was avoided (ie, tracheotomy). After 6 or more months of alpelisib therapy, facial volume was reduced (range 1%-20%) and magnetic resonance imaging anomaly volume (range 0%-23%) were reduced, and there was improvement in swallowing, upper airway patency, and speech clarity. CONCLUSION Individuals with head and neck PROS treated with alpelisib had decreased malformation size and locoregional overgrowth, improved function and symptoms, and fewer invasive procedures.
Collapse
Affiliation(s)
- Tara L Wenger
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA.
| | - Sheila Ganti
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA; Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - Catherine Bull
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA; Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - Erika Lutsky
- Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - James T Bennett
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital, Seattle, WA
| | - Kaitlyn Zenner
- Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - Dana M Jensen
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital, Seattle, WA
| | - Victoria Dmyterko
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital, Seattle, WA
| | - Ezgi Mercan
- Craniofacial Center, Seattle Children's Hospital, Seattle, WA
| | - Giri M Shivaram
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA; Interventional Radiology, Department of Radiology, Seattle Children's Hospital, Seattle, WA
| | - Seth D Friedman
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA
| | - Michael Bindschadler
- Division of Neurology, Department of Pediatrics, Seattle Children's Hospital, Seattle, WA
| | - Madeleine Drusin
- Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA; Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - Jonathan N Perkins
- Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA; Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - Ada Kong
- Investigational Drug Services, Seattle Children's Hospital, Seattle, WA
| | - Randall A Bly
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA; Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA; Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - John P Dahl
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA; Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA; Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - Juliana Bonilla-Velez
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA; Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA; Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| | - Jonathan A Perkins
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA; Division of Pediatric Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA; Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
31
|
Kim YM, Lee Y, Choi Y, Choi IH, Heo SH, Choi JM, Do HS, Jang JH, Yum MS, Yoo HW, Lee BH. Clinical and genetic analyses of patients with lateralized overgrowth. BMC Med Genomics 2022; 15:206. [PMID: 36175890 PMCID: PMC9524090 DOI: 10.1186/s12920-022-01362-1] [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: 06/01/2022] [Accepted: 09/20/2022] [Indexed: 11/14/2022] Open
Abstract
Background The genetic features and treatment strategies of lateralized overgrowth have been elusive. We performed this study to analyze the genetic characteristics and treatment results of propranolol- or alpelisib-treated patients with lateralized overgrowth. Methods Fifteen patients with lateralized overgrowth were involved. Clinical characteristics and whole-body magnetic resonance imaging (WB-MRI) findings were evaluated. Targeted exome sequencing with a gene panel of affected tissue and peripheral white blood cells was performed. Propranolol was administered and treatment results were evaluated. The PIK3CA inhibitor alpelisib was prescribed via a managed access program. Results The identified mutations were PIK3CA (n = 7), KRAS (n = 2), PTEN (n = 1), MAP2K3 (n = 1), GNAQ (n = 1), TBC1D4 (n = 1), and TEK (n = 1). Propranolol was prescribed in 12 patients, and 7 experienced mild improvement of symptoms. Alpelisib was prescribed in two patients with a PIK3CA mutation, and the reduction of proliferated masses after 1 year of treatment was proved by WB-MRI. Conclusions Targeted exome sequencing identified various genetic features of lateralized overgrowth. Propranolol could be applied as an adjuvant therapy for reducing vascular symptoms, but a PIK3CA inhibitor would be the primary therapeutic strategy for PIK3CA-related overgrowth syndrome. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01362-1.
Collapse
Affiliation(s)
- Yoon-Myung Kim
- Department of Pediatrics, Gangneung Asan Hospital, College of Medicine, University of Ulsan, Gangneung, South Korea
| | - Yena Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Yunha Choi
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine, University of Ulsan, Seoul, South Korea
| | - In Hee Choi
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Sun Hee Heo
- Asan Medical Center, Asan Institute for Life Sciences, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Jung Min Choi
- Asan Medical Center, Asan Institute for Life Sciences, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Hyo-Sang Do
- Asan Medical Center, Asan Institute for Life Sciences, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Ja-Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Mi-Sun Yum
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine, University of Ulsan, Seoul, South Korea.,Medical Genetics Center, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, College of Medicine, University of Ulsan, Seoul, South Korea. .,Medical Genetics Center, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea.
| |
Collapse
|
32
|
Angulo-Urarte A, Graupera M. When, where and which PIK3CA mutations are pathogenic in congenital disorders. NATURE CARDIOVASCULAR RESEARCH 2022; 1:700-714. [PMID: 39196083 DOI: 10.1038/s44161-022-00107-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/22/2022] [Indexed: 08/29/2024]
Abstract
PIK3CA encodes the class I PI3Kα isoform and is frequently mutated in cancer. Activating mutations in PIK3CA also cause a range of congenital disorders featuring asymmetric tissue overgrowth, known as the PIK3CA-related overgrowth spectrum (PROS), with frequent vascular involvement. In PROS, PIK3CA mutations arise postzygotically, during embryonic development, leading to a mosaic body pattern distribution resulting in a variety of phenotypic features. A clear skewed pattern of overgrowth favoring some mesoderm-derived and ectoderm-derived tissues is observed but not understood. Here, we summarize our current knowledge of the determinants of PIK3CA-related pathogenesis in PROS, including intrinsic factors such as cell lineage susceptibility and PIK3CA variant bias, and extrinsic factors, which refers to environmental modifiers. We also include a section on PIK3CA-related vascular malformations given that the vasculature is frequently affected in PROS. Increasing our biological understanding of PIK3CA mutations in PROS will contribute toward unraveling the onset and progression of these conditions and ultimately impact on their treatment. Given that PIK3CA mutations are similar in PROS and cancer, deeper insights into one will also inform about the other.
Collapse
Affiliation(s)
- Ana Angulo-Urarte
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain.
| | - Mariona Graupera
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
33
|
Diociaiuti A, Rotunno R, Pisaneschi E, Cesario C, Carnevale C, Condorelli AG, Rollo M, Di Cecca S, Quintarelli C, Novelli A, Zambruno G, El Hachem M. Clinical and Molecular Spectrum of Sporadic Vascular Malformations: A Single-Center Study. Biomedicines 2022; 10:biomedicines10061460. [PMID: 35740480 PMCID: PMC9220263 DOI: 10.3390/biomedicines10061460] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/22/2022] [Accepted: 06/16/2022] [Indexed: 01/04/2023] Open
Abstract
Sporadic vascular malformations (VMs) are a large group of disorders of the blood and lymphatic vessels caused by somatic mutations in several genes—mainly regulating the RAS/MAPK/ERK and PI3K/AKT/mTOR pathways. We performed a cross-sectional study of 43 patients affected with sporadic VMs, who had received molecular diagnosis by high-depth targeted next-generation sequencing in our center. Clinical and imaging features were correlated with the sequence variants identified in lesional tissues. Six of nine patients with capillary malformation and overgrowth (CMO) carried the recurrent GNAQ somatic mutation p.Arg183Gln, while two had PIK3CA mutations. Unexpectedly, 8 of 11 cases of diffuse CM with overgrowth (DCMO) carried known PIK3CA mutations, and the remaining 3 had pathogenic GNA11 variants. Recurrent PIK3CA mutations were identified in the patients with megalencephaly–CM–polymicrogyria (MCAP), CLOVES, and Klippel–Trenaunay syndrome. Interestingly, PIK3CA somatic mutations were associated with hand/foot anomalies not only in MCAP and CLOVES, but also in CMO and DCMO. Two patients with blue rubber bleb nevus syndrome carried double somatic TEK mutations, two of which were previously undescribed. In addition, a novel sporadic case of Parkes Weber syndrome (PWS) due to an RASA1 mosaic pathogenic variant was described. Finally, a girl with a mild PWS and another diagnosed with CMO carried pathogenic KRAS somatic variants, showing the variability of phenotypic features associated with KRAS mutations. Overall, our findings expand the clinical and molecular spectrum of sporadic VMs, and show the relevance of genetic testing for accurate diagnosis and emerging targeted therapies.
Collapse
Affiliation(s)
- Andrea Diociaiuti
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (R.R.); (C.C.); (M.E.H.)
- Correspondence: ; Tel.: +39-0668592509
| | - Roberta Rotunno
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (R.R.); (C.C.); (M.E.H.)
| | - Elisa Pisaneschi
- Translational Cytogenomics Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (E.P.); (C.C.); (A.N.)
| | - Claudia Cesario
- Translational Cytogenomics Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (E.P.); (C.C.); (A.N.)
| | - Claudia Carnevale
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (R.R.); (C.C.); (M.E.H.)
| | - Angelo Giuseppe Condorelli
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.C.); (G.Z.)
| | - Massimo Rollo
- Interventional Radiology Unit, Department of Imaging, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy;
| | - Stefano Di Cecca
- Department Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (S.D.C.); (C.Q.)
| | - Concetta Quintarelli
- Department Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (S.D.C.); (C.Q.)
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Antonio Novelli
- Translational Cytogenomics Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (E.P.); (C.C.); (A.N.)
| | - Giovanna Zambruno
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.C.); (G.Z.)
| | - May El Hachem
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (R.R.); (C.C.); (M.E.H.)
| |
Collapse
|
34
|
Rotunno R, Diociaiuti A, Pisaneschi E, Carnevale C, Dentici M, El Hachem M. PIK3CA-related overgrowth with an uncommon phenotype: case report. Ital J Pediatr 2022; 48:71. [PMID: 35551640 PMCID: PMC9097398 DOI: 10.1186/s13052-022-01268-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/02/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Megalencephaly-capillary malformation syndrome is a rare multiple-malformation syndrome secondary to somatic activating mutations in the PI3K-AKT-MTOR pathway. This is included in a heterogeneous group of disorders, now defined "PIK3CA-related overgrowth spectrum". CASE PRESENTATION We report a 22-months-old female presenting an uncommon phenotype associated with a genetic mosaicism in the PIK3CA gene, detected on DNA extracted from blood peripheral and tissue biopsy. CONCLUSIONS NGS is the preferred method for molecular diagnosis of PROS on affected skin and overgrown tissues as primary samples. The wide phenotypic variability is based on the distribution of mosaicism, in fact the same mutation can cause different PIK3CA related disorders. Continuous understanding of the clinical spectrum and of molecular basis of PROS and their overlap will lead to improve diagnosis, management and new treatment strategies.
Collapse
Affiliation(s)
- Roberta Rotunno
- Dermatology Unit, Bambino Gesù Children's Hospital-IRCCS, P.zza St. Onofrio 4, 00165, Rome, Italy.
- VASCERN VASCA and ERN-Skin European Reference Centre, Rome, Italy.
| | - Andrea Diociaiuti
- Dermatology Unit, Bambino Gesù Children's Hospital-IRCCS, P.zza St. Onofrio 4, 00165, Rome, Italy
- VASCERN VASCA and ERN-Skin European Reference Centre, Rome, Italy
| | - Elisa Pisaneschi
- Medical Genetics Laboratory, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - Claudia Carnevale
- Dermatology Unit, Bambino Gesù Children's Hospital-IRCCS, P.zza St. Onofrio 4, 00165, Rome, Italy
- VASCERN VASCA and ERN-Skin European Reference Centre, Rome, Italy
| | - Marialisa Dentici
- Unit of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - May El Hachem
- Dermatology Unit, Bambino Gesù Children's Hospital-IRCCS, P.zza St. Onofrio 4, 00165, Rome, Italy
- VASCERN VASCA and ERN-Skin European Reference Centre, Rome, Italy
| |
Collapse
|
35
|
Rodríguez-Laguna L, Davis K, Finger M, Aubel D, Vlamis R, Johnson C. Mapping the PIK3CA-related overgrowth spectrum (PROS) patient and caregiver journey using a patient-centered approach. Orphanet J Rare Dis 2022; 17:189. [PMID: 35526022 PMCID: PMC9077929 DOI: 10.1186/s13023-022-02338-1] [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: 11/05/2021] [Accepted: 04/26/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND PROS disorders are driven by somatic, gain-of-function mutations in PIK3CA that result in hyperactivation of the phosphatidylinositol-3-kinase (PI3K) signaling pathway. PROS encompasses a broad spectrum of overlapping phenotypes (including overgrowth and vascular malformations) that vary significantly in their severity; every case is unique, leading to different, complex experiences. Here, we aim to describe the PROS experience from the patients' and caregivers' points of view, from onset to diagnosis to treatment and support. RESULTS The PROS patient journey was developed using a literature review, an ethnography study, health care professional (HCP) research, and social listening. It was then validated with patients, caregivers, and patient advocates. Physician research included 94 PROS centers and other vascular anomaly centers throughout the United States and Europe. Ethnographic research included 24 patients, caregivers, and/or advocates; selected data from 223 patients were reviewed. Key priority areas of need were identified, along with barriers to and potential enablers of quality care. Visual mapping of the PROS patient and family journey was developed to identify key personal health and system issues, and opportunities for improvements throughout patients' lifespans. Maps were also developed for 3 specific conditions: Klippel-Trénaunay syndrome (K-T); congenital lipomatous overgrowth, vascular malformations, epidermal nevi, scoliosis/skeletal and spinal anomalies (CLOVES) syndrome; and megalencephaly-capillary malformation syndrome (M-CM). Overall, most patients with PROS conditions and their families struggle with a long path to diagnosis, access to genetic testing, and finding qualified specialists. Following diagnosis, patients and families are frequently challenged with major medical events, comorbidities, unpredictability, frequent hospitalization, impact on school and work, the need for multidisciplinary care, unwanted attention, adverse impact on mental and emotional health, and financial pressures. Lack of effective pain management emerged as a substantial issue. Challenges and barriers to quality care shift throughout patients' lifespans; transition from pediatric to adult care can be especially difficult. CONCLUSIONS This patient journey in PROS was created in collaboration with patients, caregivers, and advocates as key partners. This novel methodology, which could be applied elsewhere, can more accurately identify areas of unmet need, barriers to care, education topics, and assist HCPs to understand the patient and family perspective.
Collapse
Affiliation(s)
- Lara Rodríguez-Laguna
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, La Paz University Hospital, Paseo de La Castellana, 261, 28046, Madrid, Spain.
| | | | | | - Dawn Aubel
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Robin Vlamis
- Solstice HealthCommunications, Far Hills, NJ, USA
| | - Craig Johnson
- Interventional Radiology, Nemours Children's Hospital, Orlando, FL, USA
| |
Collapse
|
36
|
Öztürk Durmaz E, Demircioğlu D, Yalınay Dikmen P, Alanay Y, Alanay A, Demirkesen C, Tokat F, Karaarslan E. A Review on Cutaneous and Musculoskeletal Manifestations of CLOVES Syndrome. Clin Cosmet Investig Dermatol 2022; 15:621-630. [PMID: 35444443 PMCID: PMC9013705 DOI: 10.2147/ccid.s351637] [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: 11/29/2021] [Accepted: 03/17/2022] [Indexed: 01/19/2023]
Abstract
CLOVES syndrome is a novel sporadic mosaic segmental overgrowth syndrome, currently categorized under the canopy of PROS (PIK3CA-related overgrowth spectrum) disorders. All PROS disorders harbor heterozygous postzygotic activating somatic mutations involving the PIK3CA gene. As an upstream regulator of the PI3K/AKT/mTOR signal transduction pathway, activating mutations of PIK3CA gene commence in uncontrolled growth of cutaneous, vascular (capillaries, veins, and lymphatics), adipose, neural, and musculoskeletal tissues. The excessive growth is segmental, patchy, asymmetric, and confined to body parts affected by the mutation. The term ‘CLOVES’ is an acronym denoting congenital lipomatous overgrowth, vascular malformations, epidermal nevi and spinal (scoliosis) and/ or skeletal anomalies. The syndrome is characterized by an admixture of overgrown tissues, derived mainly from mesoderm and neuroectoderm. Among PROS disorders, CLOVES syndrome represents the extreme end of the spectrum with massive affection of almost the entire body. The syndrome might judiciously be treated with medications hampering with the PI3K/AKT/mTOR signal transduction pathway. This article aims at reviewing the cutaneous and musculoskeletal manifestations of CLOVES syndrome, as the paradigm for PROS disorders. CLOVES syndrome and other PROS disorders are still misdiagnosed, underdiagnosed, underreported, and undertreated by the dermatology community.
Collapse
Affiliation(s)
- Emel Öztürk Durmaz
- Department of Dermatology, Acıbadem Mehmet Ali Aydınlar University School of Medicine, İstanbul, Turkey
| | - Deniz Demircioğlu
- Department of Dermatology, Acıbadem Mehmet Ali Aydınlar University School of Medicine, İstanbul, Turkey
| | - Pınar Yalınay Dikmen
- Department of Neurology, Acıbadem Mehmet Ali Aydınlar University School of Medicine, İstanbul, Turkey
| | - Yasemin Alanay
- Department of Pediatrics, Acıbadem Mehmet Ali Aydınlar University School of Medicine, İstanbul, Turkey
| | - Ahmet Alanay
- Department of Orthopedics, Acıbadem Mehmet Ali Aydınlar University School of Medicine, İstanbul, Turkey
| | - Cüyan Demirkesen
- Department of Pathology, Acıbadem Mehmet Ali Aydınlar University School of Medicine, İstanbul, Turkey
| | - Fatma Tokat
- Department of Pathology, Acıbadem Mehmet Ali Aydınlar University School of Medicine, İstanbul, Turkey
| | - Ercan Karaarslan
- Department of Radiology, Acıbadem Mehmet Ali Aydınlar University School of Medicine, İstanbul, Turkey
| |
Collapse
|
37
|
Chen WL, Pao E, Owens J, Glass I, Pritchard C, Shirts BH, Lockwood C, Mirzaa GM. The utility of cerebrospinal fluid-derived cell-free DNA in molecular diagnostics for the PIK3CA-related megalencephaly-capillary malformation (MCAP) syndrome: a case report. Cold Spring Harb Mol Case Stud 2022; 8:a006188. [PMID: 35483878 PMCID: PMC9059787 DOI: 10.1101/mcs.a006188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
The megalencephaly-capillary malformation (MCAP) syndrome is an overgrowth disorder caused by mosaic gain-of-function variants in PIK3CA It is characterized by megalencephaly or hemimegalencephaly, vascular malformations, somatic overgrowth, among other features. Epilepsy is commonly associated with MCAP, and a subset of individuals have cortical malformations requiring resective epilepsy surgery. Like other mosaic disorders, establishing a molecular diagnosis is largely achieved by screening lesional tissues (such as brain or skin), with a low diagnostic yield from peripheral tissues (such as blood). Therefore, in individuals with MCAP in whom lesional tissues are scarce or unavailable or those ineligible for epilepsy surgery, establishing a molecular diagnosis can be challenging. Here we report on the utility of cerebrospinal fluid (CSF)-derived cfDNA for the molecular diagnosis of an individual with MCAP syndrome harboring a mosaic PIK3CA variant (c.3139C > T, p.His1047Tyr). The proband presented with asymmetric megalencephaly without significant dysgyria. He did not have refractory epilepsy and was therefore not a candidate for epilepsy surgery. However, he developed diffuse large B-cell lymphoma (DLBCL) in late childhood, with four CSF samples obtained via lumbar puncture for cancer staging during which one sample was collected for cfDNA extraction and sequencing. PIK3CA variant allele fractions in CSF cell-free DNA (cfDNA), skin fibroblasts, and peripheral blood were 3.08%, 37.31%, and 2.04%, respectively. This report illustrates the utility of CSF-derived cfDNA in MCAP syndrome. Minimally invasive-based molecular diagnostic approaches utilizing cfDNA not only facilitate accurate genetic diagnosis but also have important therapeutic implications for individuals with refractory epilepsy as repurposed PI3K-AKT-MTOR pathway-inhibitors become more widely available.
Collapse
Affiliation(s)
- Wei-Liang Chen
- School of Medicine, Division of Medical Genetics, University of Washington, Seattle, Washington 98195, USA
| | - Emily Pao
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101, USA
| | - James Owens
- Department of Neurology, University of Washington, Seattle, Washington 98195, USA
| | - Ian Glass
- Department of Pediatrics, University of Washington, Seattle, Washington 98195, USA
| | - Colin Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, USA
| | - Brain H Shirts
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, USA
| | - Christina Lockwood
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, USA
| | - Ghayda M Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101, USA
- Department of Pediatrics, University of Washington, Seattle, Washington 98195, USA
- The Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, USA
| |
Collapse
|
38
|
Bourgon N, Carmignac V, Sorlin A, Duffourd Y, Philippe C, Thauvin-Robinet C, Guibaud L, Faivre L, Vabres P, Kuentz P. Clinical and molecular data in cases of prenatal localized overgrowth disorder: major implication of genetic variants in PI3K-AKT-mTOR signaling pathway. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2022; 59:532-542. [PMID: 34170046 DOI: 10.1002/uog.23715] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVES To describe clinical and molecular findings in a French multicenter cohort of fetuses with prenatal diagnosis of congenital abnormality and suspicion of a localized overgrowth disorder (LOD) suggestive of genetic variants in the PI3K-AKT-mTOR signaling pathway. METHODS We analyzed retrospectively data obtained between 1 January 2013 and 1 May 2020 from fetuses with brain and/or limb overgrowth referred for molecular diagnosis of PI3K-AKT-mTOR pathway genes by next-generation sequencing (NGS) using pathological tissue obtained by fetal autopsy. We also assessed the diagnostic yield of amniotic fluid. RESULTS During the study period, 21 subjects with LOD suspected of being secondary to a genetic variant of the PI3K-AKT-mTOR pathway were referred for analysis. Of these, 17 fetuses had brain overgrowth, including six with isolated megalencephaly (MEG) and 11 with hemimegalencephaly (HMEG). Of the six with MEG, germline variants were identified in four cases, in either PIK3R2, AKT3 or MTOR, and a postzygotic PIK3R2 variant was found in the other two cases. Of the 11 with HMEG, a postzygotic PIK3CA variant was found in three fetuses with extracerebral features of PIK3CA-related overgrowth spectrum, and in seven fetuses with isolated HMEG. No pathogenic variant was identified in the 11th case with HMEG. Four fetuses with limb overgrowth also had one or more lymphatic malformations (LM) and harbored a postzygotic PIK3CA variant. NGS on cultured amniocytes performed in 10 cases, of which nine had been found positive on analysis of pathological fetal tissue, showed variants in four, in either PIK3CA, PIK3R2 or AKT3. CONCLUSIONS Isolated MEG or HMEG may lead to identification of genetic variants in the PI3K-AKT-mTOR signaling pathway. Cases of limb overgrowth and LM or isolated HMEG are likely associated with PIK3CA variants. © 2021 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- N Bourgon
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Service d'Obstétrique-Maternité, Chirurgie Médecine et Imagerie Fœtale, Hôpital Necker Enfants Malades, AP-HP, Paris, France
| | - V Carmignac
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Centre de Référence des Maladies Rares de la Peau et des Muqueuses d'Origine Génétique (MAGEC), Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
| | - A Sorlin
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Centre de Référence des Maladies Rares de la Peau et des Muqueuses d'Origine Génétique (MAGEC), Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Centre de Génétique et Centre de Référence 'Anomalies du Développement et Syndromes Malformatifs de l'Inter-région Est', Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- UF Innovation en Diagnostic Génomique des Maladies Rares, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
| | - Y Duffourd
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
| | - C Philippe
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- UF Innovation en Diagnostic Génomique des Maladies Rares, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
| | - C Thauvin-Robinet
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Centre de Génétique et Centre de Référence 'Anomalies du Développement et Syndromes Malformatifs de l'Inter-région Est', Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
| | - L Guibaud
- Service d'Imagerie Médicale, Hôpital Femme-Mère-Enfants, Hospices Civils de Lyon, Bron, France
| | - L Faivre
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Centre de Génétique et Centre de Référence 'Anomalies du Développement et Syndromes Malformatifs de l'Inter-région Est', Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
| | - P Vabres
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Centre de Référence des Maladies Rares de la Peau et des Muqueuses d'Origine Génétique (MAGEC), Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Service de Dermatologie, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
| | - P Kuentz
- INSERM UMR 1231, Equipe 'Génétique des Anomalies du Développement', Université de Bourgogne Franche-Comté, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement, Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Centre de Référence des Maladies Rares de la Peau et des Muqueuses d'Origine Génétique (MAGEC), Centre Hospitalier Universitaire de Dijon Bourgogne, Dijon, France
- Oncobiologie Génétique Bioinformatique, PCBio, Centre Hospitalier Universitaire de Besançon, Besançon, France
| |
Collapse
|
39
|
Su LX, Sun Y, Wang Z, Wang D, Yang X, Zheng L, Wen M, Fan X, Cai R. Complex vascular anomalies and tissue overgrowth of limbs associated with increased skin temperature and peripheral venous dilatation: parks weber syndrome or PROS? Hereditas 2022; 159:1. [PMID: 34980271 PMCID: PMC8725539 DOI: 10.1186/s41065-021-00217-6] [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: 10/25/2021] [Accepted: 12/09/2021] [Indexed: 01/19/2023] Open
Abstract
PIK3CA-related overgrowth spectrum (PROS) is a series of congenital, sporadic disorders that are associated with segmental overgrowth phenotypes and postzygotic, somatic gene mutations in the PIK3CA-ATK-mTOR pathway. The variability and overlapping phenotypes between PROS and other complex vascular malformations make the differential diagnosis confusing and challenging. PROS should be considered for the differential diagnosis with other complex vascular malformations and syndromes with a tissue overgrowth phenotype, such as Parkes-Weber syndrome (PWS). Herein, we diagnosed one unique clinically challenging case manifested as capillary malformation (CM), limb overgrowth, as well as increased skin temperature and peripheral venous dilatation of lower limb that indicated a potential fast-flow lesion. The patient was initially diagnosed with PWS. Contrary to the previous diagnosis, based on further MR imaging and digital subtraction angiography (DSA), which ruled out the existence of AVMs and AVFs, and molecular analysis with targeted next-generation sequencing (NGS) revealing a somatic PIK3CA mutation, we ultimately diagnosed that the patient had a unique form of PROS simulating PWS phenotypes. We suggest that it is important to propose the differential diagnosis of PWS and PROS, two diseases that share a common overgrowth phenotype. We recommended radiological diagnosis such as MRI, CT and DSA as well as further molecular diagnosis to provide more information for the assessment of vascular lesions and to further guide clinical treatment strategies.
Collapse
Affiliation(s)
- Li Xin Su
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yi Sun
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Zhenfeng Wang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Deming Wang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xitao Yang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lianzhou Zheng
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Mingzhe Wen
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xindong Fan
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| | - Ren Cai
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| |
Collapse
|
40
|
Douzgou S, Rawson M, Baselga E, Danielpour M, Faivre L, Kashanian A, Keppler-Noreuil KM, Kuentz P, Mancini GMS, Maniere MC, Martinez-Glez V, Parker VE, Semple RK, Srivastava S, Vabres P, de Wit MCY, Graham JM, Clayton-Smith J, Mirzaa GM, Biesecker LG. A standard of care for individuals with PIK3CA-related disorders: An international expert consensus statement. Clin Genet 2022; 101:32-47. [PMID: 34240408 PMCID: PMC8664971 DOI: 10.1111/cge.14027] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 01/19/2023]
Abstract
Growth promoting variants in PIK3CA cause a spectrum of developmental disorders, depending on the developmental timing of the mutation and tissues involved. These phenotypically heterogeneous entities have been grouped as PIK3CA-Related Overgrowth Spectrum disorders (PROS). Deep sequencing technologies have facilitated detection of low-level mosaic, often necessitating testing of tissues other than blood. Since clinical management practices vary considerably among healthcare professionals and services across different countries, a consensus on management guidelines is needed. Clinical heterogeneity within this spectrum leads to challenges in establishing management recommendations, which must be based on patient-specific considerations. Moreover, as most of these conditions are rare, affected families may lack access to the medical expertise that is needed to help address the multi-system and often complex medical issues seen with PROS. In March 2019, macrocephaly-capillary malformation (M-CM) patient organizations hosted an expert meeting in Manchester, United Kingdom, to help address these challenges with regards to M-CM syndrome. We have expanded the scope of this project to cover PROS and developed this consensus statement on the preferred approach for managing affected individuals based on our current knowledge.
Collapse
Affiliation(s)
- Sofia Douzgou
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, M13 9WL, United Kingdom
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Oxford Road, M13 9PL, United Kingdom
| | - Myfanwy Rawson
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, M13 9WL, United Kingdom
| | - Eulalia Baselga
- Department of Dermatology, Hospital Sant Joan de Déu, Passeig de Sant Joan de Déu, 2, 08950 Esplugues de Llobregat, Barcelona, Spain
| | - Moise Danielpour
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Centre, Los Angeles, CA 90048, USA; Department of Neurosurgery, Cedars-Sinai Medical Centre, Los Angeles, CA 90048, USA
| | - Laurence Faivre
- Department of Medical Genetics and Centre of Reference for Developmental Anomalies and Malformative syndromes, CHU de Dijon, 14 Rue Paul Gaffarel, 21000 Dijon, France
| | - Alon Kashanian
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Centre, Los Angeles, CA 90048, USA; Department of Neurosurgery, Cedars-Sinai Medical Centre, Los Angeles, CA 90048, USA
| | - Kim M Keppler-Noreuil
- Division of Genetics & Metabolism, Department of Paediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul Kuentz
- Oncobiologie Génétique Bioinformatique, PCBio, CHU Besançon, France
| | - Grazia MS Mancini
- Department of Clinical Genetics, Erasmus MC University Medical Centre, 3015, GD, Rotterdam, the Netherlands
| | - Marie-Cecile Maniere
- Centre de Référence, Maladies orales et dentaires rares, Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Victor Martinez-Glez
- IdiPAZ Research Institute, Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, Institute of Health Carlos III, Madrid, Spain
- Institute of Medical and Molecular Genetics (INGEMM), La Paz University Hospital, Madrid, Spain
| | - Victoria E Parker
- The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Robert K Semple
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pierre Vabres
- Department of Medical Genetics and Centre of Reference for Developmental Anomalies and Malformative syndromes, CHU de Dijon, 14 Rue Paul Gaffarel, 21000 Dijon, France
| | - Marie-Claire Y de Wit
- Department of Child Neurology, Sophia Children's hospital, Erasmus MC University Medical Centre Rotterdam, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - John M Graham
- Department of Paediatrics, Division of Medical Genetics, Cedars Sinai Medical Centre, David Geffen School of Medicine at UCLA, Los Angeles, CA 90048, USA
| | - Jill Clayton-Smith
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, M13 9WL, United Kingdom
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Oxford Road, M13 9PL, United Kingdom
| | - Ghayda M Mirzaa
- Genetic Medicine, Department of Paediatrics, University of Washington, Seattle, USA
| | - Leslie G Biesecker
- Centre for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
41
|
Martinez-Glez V, Rodriguez-Laguna L, Viana-Huete V, García Torrijos C, Hurtado B, Lapunzina P, Triana P, López-Gutiérrez JC. Segmental undergrowth is associated with pathogenic variants in vascular malformation genes: A retrospective case-series study. Clin Genet 2021; 101:296-306. [PMID: 34850385 DOI: 10.1111/cge.14095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/11/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022]
Abstract
Segmental overgrowth has been widely described in patients with congenital vascular anomalies. However, segmental undergrowth has been poorly characterized, and no large series of patients have been published. We present the clinical and molecular characteristics a cohort of 37 patients with vascular malformations and segmental undergrowth. True undergrowth was only considered when the musculoskeletal system was involved to avoid confusion with other causes of segmental reduction, as in lipodystrophy or the long-term osteopenia seen in patients with Servelle-Martorell syndrome. Deep high-throughput sequencing was performed in tissue samples from 20 patients using a custom panel. We identified three groups: undergrowth associated with (1) venous, (2) capillary-venous, and (3) lymphatic-capillary-venous malformations. Congenital or early childhood onset undergrowth can occur with or without associated overgrowth. Different likely pathogenic or pathogenic variants were detected in 13 of 20 (65%) tissue samples in the PIK3CA, TEK, GNAQ, or GNA11 genes. In conclusion, the eponymous Servelle-Martorell syndrome should not be used as a synonym for undergrowth. Segmental undergrowth should be considered a characteristic associated with vascular malformations. Patients with PIK3CA variants show all different combinations of overgrowth and undergrowth. Thus, the term PROS (PIK3CA-related overgrowth spectrum) does not cover the entire spectrum.
Collapse
Affiliation(s)
- Victor Martinez-Glez
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain.,Clinical Genetics Section, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Ithaca, European Reference Network, Hospital Universitario La Paz, Madrid, Spain
| | - Lara Rodriguez-Laguna
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Vanesa Viana-Huete
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Carolina García Torrijos
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Begoña Hurtado
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Pablo Lapunzina
- Clinical Genetics Section, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.,Ithaca, European Reference Network, Hospital Universitario La Paz, Madrid, Spain.,Overgrowth Syndromes Laboratory, Institute of Medical and Molecular Genetics, INGEMM-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Paloma Triana
- Vascular Anomalies Unit, Department of Pediatric Surgery, Hospital Universitario La Paz, Madrid, Spain
| | | |
Collapse
|
42
|
Maruani A, Tavernier E, Boccara O, Mazereeuw-Hautier J, Leducq S, Bessis D, Guibaud L, Vabres P, Carmignac V, Mallet S, Barbarot S, Chiaverini C, Droitcourt C, Bursztejn AC, Lengellé C, Woillard JB, Herbreteau D, Le Touze A, Joly A, Léauté-Labrèze C, Powell J, Bourgoin H, Gissot V, Giraudeau B, Morel B. Sirolimus (Rapamycin) for Slow-Flow Malformations in Children: The Observational-Phase Randomized Clinical PERFORMUS Trial. JAMA Dermatol 2021; 157:1289-1298. [PMID: 34524406 DOI: 10.1001/jamadermatol.2021.3459] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Sirolimus is increasingly being used to treat various vascular anomalies, although evidence of its efficacy is lacking. Objective To assess the efficacy and safety of sirolimus for children with slow-flow vascular malformations to better delineate the indications for treatment. Design, Setting and Participants This multicenter, open-label, observational-phase randomized clinical trial included 59 children aged 6 to 18 years with a slow-flow vascular malformation who were recruited between September 28, 2015, and March 22, 2018, in 11 French tertiary hospital centers. Statistical analysis was performed on an intent-to-treat basis from December 4, 2019, to November 10, 2020. Interventions Patients underwent an observational period, then switched to an interventional period when they received oral sirolimus (target serum levels, 4-12 ng/mL). The switch time was randomized from month 4 to month 8, and the whole study period lasted 12 months for each patient. Main Outcomes and Measures The primary outcome was change in the volume of vascular malformations detected on magnetic resonance imaging scan (with centralized interpretation) per unit of time (ie, between the interventional period and the observational period). Secondary outcomes included subjective end points: pain, bleeding, oozing, quality of life, and safety. Results Among the participants (35 girls [59.3%]; mean [SD] age, 11.6 [3.8] years), 22 (37.3%) had a pure venous malformation, 18 (30.5%) had a cystic lymphatic malformation, and 19 (32.2%) had a combined malformation, including syndromic forms. Variations in the volume of vascular malformations detected on magnetic resonance imaging scans associated with the duration period were not overall significantly different between the interventional period and the observational period (all vascular malformations: mean [SD] difference, -0.001 [0.007]; venous malformations: mean [SD] difference, 0.001 [0.004]; combined malformations: mean [SD] difference, 0.001 [0.009]). However, a significant decrease in volume was observed for children with pure lymphatic malformations (mean [SD] difference, -0.005 [0.005]). Overall, sirolimus had positive effects on pain, especially for combined malformations, and on bleeding, oozing, self-assessed efficacy, and quality of life. During sirolimus treatment, 56 patients experienced 231 adverse events (5 serious adverse events, none life-threatening). The most frequent adverse event was an oral ulcer (29 patients [49.2%]). Conclusions and Relevance This observational-phase randomized clinical trial allows for clarifying the goals of patients and families when starting sirolimus therapy for children older than 6 years. Pure lymphatic malformations seem to be the best indication for sirolimus therapy because evidence of decreasing lymphatic malformation volume per unit of time, oozing, and bleeding and increasing quality of life was found. In combined malformations, sirolimus significantly reduced pain, oozing, and bleeding. Benefits seemed lower for pure venous malformations than for the 2 other subgroups, also based on symptoms. Trial Registration ClinicalTrials.gov Identifier: NCT02509468; clinicaltrialsregister.eu Identifier: 2015-001096-43.
Collapse
Affiliation(s)
- Annabel Maruani
- University of Tours, University of Nantes, Institut National de la Santé et de la Recherche Médicale, SPHERE U1246, Tours, France.,Centre Hospitalier Régional Universitaire Tours, Department of Dermatology, Reference Center for Genodermatoses and Rare Skin Diseases (Maladies Génétiques rares à Expression Cutanée-Tours), Tours, France.,Centre Hospitalier Régional Universitaire Tours, Institut National de la Santé et de la Recherche Médicale Clinical Investigation Center 1415, Tours, France
| | - Elsa Tavernier
- University of Tours, University of Nantes, Institut National de la Santé et de la Recherche Médicale, SPHERE U1246, Tours, France.,Centre Hospitalier Régional Universitaire Tours, Institut National de la Santé et de la Recherche Médicale Clinical Investigation Center 1415, Tours, France
| | - Olivia Boccara
- Department of Dermatology and Reference Center for Genodermatoses and Rare Skin Diseases (Maladies Génétiques rares à Expression Cutanée-Necker), University Hospital Necker-Enfants Malades, Paris, France
| | | | - Sophie Leducq
- University of Tours, University of Nantes, Institut National de la Santé et de la Recherche Médicale, SPHERE U1246, Tours, France.,Centre Hospitalier Régional Universitaire Tours, Department of Dermatology, Reference Center for Genodermatoses and Rare Skin Diseases (Maladies Génétiques rares à Expression Cutanée-Tours), Tours, France
| | - Didier Bessis
- Department of Dermatology, University Hospital Center of Montpellier, Montpellier, France
| | - Laurent Guibaud
- University Hospital Center of Lyon, Consultation Multidisciplinaire Lyonnaise des Angiomes, Lyon, France
| | - Pierre Vabres
- Department of Dermatology, University Hospital Center of Dijon, Dijon, France
| | - Virginie Carmignac
- Department of Dermatology, University Hospital Center of Dijon, Dijon, France
| | - Stéphanie Mallet
- Department of Dermatology, University Hospital Center of Marseille, Marseille, France
| | - Sébastien Barbarot
- Department of Dermatology, University Hospital Center of Nantes, Nantes, France
| | | | | | | | - Céline Lengellé
- Centre Hospitalier Régional Universitaire Tours, Department of Clinical Pharmacology, Regional Pharmacovigilance Center, Tours, France
| | - Jean-Baptiste Woillard
- Centre Hospitalier Universitaire Limoges, Department of Pharmacology and Toxicology, University of Limoges, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 850, Limoges, France
| | - Denis Herbreteau
- University of Tours, Centre Hospitalier Régional Universitaire Tours, Department of Neuroradiology, Reference Center for Genodermatoses and Rare Skin Diseases (Maladies Génétiques rares à Expression Cutanée-Tours), Tours, France
| | - Anne Le Touze
- Centre Hospitalier Régional Universitaire Tours, Department of Pediatric Surgery, Reference Center for Genodermatoses and Rare Skin Diseases (Maladies Génétiques rares à Expression Cutanée-Tours), Tours, France
| | - Aline Joly
- Centre Hospitalier Régional Universitaire Tours, Department of Pediatric Maxillofacial Surgery, Reference Center for Genodermatoses and Rare Skin Diseases (Maladies Génétiques rares à Expression Cutanée-Tours), Tours, France
| | | | - Julie Powell
- Department of Pediatric Dermatology, Hospital Sainte-Justine, Montréal, Québec, Canada
| | - Hélène Bourgoin
- Centre Hospitalier Régional Universitaire Tours, Department of Pharmacy, Tours, France
| | - Valérie Gissot
- Centre Hospitalier Régional Universitaire Tours, Institut National de la Santé et de la Recherche Médicale Clinical Investigation Center 1415, Tours, France
| | - Bruno Giraudeau
- University of Tours, University of Nantes, Institut National de la Santé et de la Recherche Médicale, SPHERE U1246, Tours, France.,Centre Hospitalier Régional Universitaire Tours, Institut National de la Santé et de la Recherche Médicale Clinical Investigation Center 1415, Tours, France
| | - Baptiste Morel
- University of Tours, Centre Hospitalier Régional Universitaire Tours, Department of Pediatric Radiology, Tours, France
| |
Collapse
|
43
|
Madsen RR, Erickson EC, Rueda OM, Robin X, Caldas C, Toker A, Semple RK, Vanhaesebroeck B. Positive correlation between transcriptomic stemness and PI3K/AKT/mTOR signaling scores in breast cancer, and a counterintuitive relationship with PIK3CA genotype. PLoS Genet 2021; 17:e1009876. [PMID: 34762647 PMCID: PMC8584750 DOI: 10.1371/journal.pgen.1009876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
A PI3Kα-selective inhibitor has recently been approved for use in breast tumors harboring mutations in PIK3CA, the gene encoding p110α. Preclinical studies have suggested that the PI3K/AKT/mTOR signaling pathway influences stemness, a dedifferentiation-related cellular phenotype associated with aggressive cancer. However, to date, no direct evidence for such a correlation has been demonstrated in human tumors. In two independent human breast cancer cohorts, encompassing nearly 3,000 tumor samples, transcriptional footprint-based analysis uncovered a positive linear association between transcriptionally-inferred PI3K/AKT/mTOR signaling scores and stemness scores. Unexpectedly, stratification of tumors according to PIK3CA genotype revealed a "biphasic" relationship of mutant PIK3CA allele dosage with these scores. Relative to tumor samples without PIK3CA mutations, the presence of a single copy of a hotspot PIK3CA variant was associated with lower PI3K/AKT/mTOR signaling and stemness scores, whereas the presence of multiple copies of PIK3CA hotspot mutations correlated with higher PI3K/AKT/mTOR signaling and stemness scores. This observation was recapitulated in a human cell model of heterozygous and homozygous PIK3CAH1047R expression. Collectively, our analysis (1) provides evidence for a signaling strength-dependent PI3K-stemness relationship in human breast cancer; (2) supports evaluation of the potential benefit of patient stratification based on a combination of conventional PI3K pathway genetic information with transcriptomic indices of PI3K signaling activation.
Collapse
Affiliation(s)
- Ralitsa R. Madsen
- University College London Cancer Institute, Paul O’Gorman Building, University College London, London, United Kingdom
| | - Emily C. Erickson
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Oscar M. Rueda
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Cambridge Breast Unit, Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
- NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Xavier Robin
- SIB Swiss Institute of Bioinformatics, Biozentrum, University of Basel, Basel, Switzerland
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Cambridge Breast Unit, Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
- NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Alex Toker
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Robert K. Semple
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Bart Vanhaesebroeck
- University College London Cancer Institute, Paul O’Gorman Building, University College London, London, United Kingdom
| |
Collapse
|
44
|
Te Paske IBAW, Garcia-Pelaez J, Sommer AK, Matalonga L, Starzynska T, Jakubowska A, van der Post RS, Lubinski J, Oliveira C, Hoogerbrugge N, de Voer RM. A mosaic PIK3CA variant in a young adult with diffuse gastric cancer: case report. Eur J Hum Genet 2021; 29:1354-1358. [PMID: 34075207 PMCID: PMC8440670 DOI: 10.1038/s41431-021-00853-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/16/2021] [Accepted: 02/26/2021] [Indexed: 12/11/2022] Open
Abstract
Hereditary diffuse gastric cancer (HDGC) is associated with germline deleterious variants in CDH1 and CTNNA1. The majority of HDGC-suspected patients are still genetically unresolved, raising the need for identification of novel HDGC predisposing genes. Under the collaborative environment of the SOLVE-RD consortium, re-analysis of whole-exome sequencing data from unresolved gastric cancer cases (n = 83) identified a mosaic missense variant in PIK3CA in a 25-year-old female with diffuse gastric cancer (DGC) without familial history for cancer. The variant, c.3140A>G p.(His1047Arg), a known cancer-related somatic hotspot, was present at a low variant allele frequency (18%) in leukocyte-derived DNA. Somatic variants in PIK3CA are usually associated with overgrowth, a phenotype that was not observed in this patient. This report highlights mosaicism as a potential, and understudied, mechanism in the etiology of DGC.
Collapse
Grants
- EC | Horizon 2020 Framework Programme (EU Framework Programme for Research and Innovation H2020)
- Data was reanalysed using the RD‐Connect Genome‐Phenome Analysis Platform, which received funding from EU projects RD‐Connect, Solve-RD and EJP-RD (grant numbers FP7 305444, H2020 779257, H2020 825575), Instituto de Salud Carlos III (grant numbers PT13/0001/0044, PT17/0009/0019; Instituto Nacional de Bioinformática, INB) and ELIXIR Implementation Studies.
- the European Regional Development Fund (ERDF) through COMPETE 2020 - Operacional Programme for Competitiveness and Internationalisation (POCI), Portugal 2020, and by Portuguese funds through FCT/ Ministério da Ciência, Tecnologia e Inovação in the framework of the project "Institute for Research and Innovation in Health Sciences" (POCI-01-0145-FEDER-007274) and Project Ref. POCI-01-0145-FEDER-030164
Collapse
Affiliation(s)
- Iris B A W Te Paske
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - José Garcia-Pelaez
- i3S, Institute for Research and Innovation in Health of the University of Porto, Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- FMUP, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Anna K Sommer
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Leslie Matalonga
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Teresa Starzynska
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
- Independent Laboratory of Molecular Biology and Genetic Diagnostics, Pomeranian Medical University, Szczecin, Poland
| | - Rachel S van der Post
- Department of Pathology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Jan Lubinski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Carla Oliveira
- i3S, Institute for Research and Innovation in Health of the University of Porto, Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- FMUP, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Richarda M de Voer
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands.
| |
Collapse
|
45
|
Bernhard SM, Adam L, Atef H, Häberli D, Bramer WM, Minder B, Döring Y, Laine JE, Muka T, Rössler J, Baumgartner I. A systematic review of the safety and efficacy of currently used treatment modalities in the treatment of patients with PIK3CA-related overgrowth spectrum. J Vasc Surg Venous Lymphat Disord 2021; 10:527-538.e2. [PMID: 34358672 DOI: 10.1016/j.jvsv.2021.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/22/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND PIK3CA (activating mutations of the p110α subunit of phosphatidylinositol 3-kinases)-related overgrowth spectrums (PROS) include a variety of clinical presentations that are associated with hypertrophy of different parts of the body. We performed a systematic literature review to assess the current treatment options and their efficacy and safety for PROS. METHODS A literature search was performed in Embase, MEDLINE (Ovid), Web of Science Core Collection, Cochrane Central Register of Controlled Trials, ClinicalTrials.gov, and Google Scholar to retrieve studies on the treatment of hypertrophy in PROS. Randomized controlled trials, cohort studies, and case series with ≥10 patients were included in the present review. The titles, abstracts, and full text were assessed by two reviewers independently. The risk of bias was assessed using the Newcastle-Ottawa scale. RESULTS We included 16 studies of the treatment of hypertrophy in PROS patients, 13 (81.3%) from clinical retrospective studies and 3 (13.7%) from prospective cohort studies. The risk of bias grade was low for 2, medium for 12, and high for 2 studies. Of the 16 studies, 13 reported on surgical treatment and 3 reported pharmacologic treatment using phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway inhibitors in PROS patients. In 3 studies, PROS was defined by a mutation in the PIK3CA gene, and 13 studies relied on a clinical definition of PROS. Surgical therapy was beneficial for a specific subgroup of PROS (macrodactyly). However, little has been reported concerning surgery and the potential benefits for other PROS entities. The reported side effects after surgical therapy were mostly prolonged wound healing or scarring. PI3K/mTOR pathway inhibition was beneficial in patients with PROS by reducing hypertrophy and systemic symptoms. The adverse effects reported included infection, changes in blood count, liver enzymes, and metabolic measures. CONCLUSIONS Surgery is a locally limited treatment option for specific types of PROS. A promising treatment option for PROS is pharmacologic PIK3CA inhibition. However, the level of evidence on the treatment of overgrowth in PROS patients is limited.
Collapse
Affiliation(s)
- Sarah M Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Luise Adam
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Primary Health Care, University of Bern, Bern, Switzerland
| | - Hady Atef
- Faculty of Physical therapy, Cairo University, Cairo, Egypt; Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Dario Häberli
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Wichor M Bramer
- Medical Library, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Beatrice Minder
- Public Health and Primary Care Library, University Library of Bern, University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Jessica E Laine
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Taulant Muka
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Jochen Rössler
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
46
|
Canaud G, Hammill AM, Adams D, Vikkula M, Keppler-Noreuil KM. A review of mechanisms of disease across PIK3CA-related disorders with vascular manifestations. Orphanet J Rare Dis 2021; 16:306. [PMID: 34238334 PMCID: PMC8268514 DOI: 10.1186/s13023-021-01929-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/27/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND PIK3CA-related disorders include vascular malformations and overgrowth of various tissues that are caused by postzygotic, somatic variants in the gene encoding phosphatidylinositol-3-kinase (PI3K) catalytic subunit alpha. These mutations result in activation of the PI3K/AKT/mTOR signaling pathway. The goals of this review are to provide education on the underlying mechanism of disease for this group of rare conditions and to summarize recent advancements in the understanding of, as well as current and emerging treatment options for PIK3CA-related disorders. MAIN BODY PIK3CA-related disorders include PIK3CA-related overgrowth spectrum (PROS), PIK3CA-related vascular malformations, and PIK3CA-related nonvascular lesions. Somatic activating mutations (predominantly in hotspots in the helical and kinase domains of PIK3CA, but also in other domains), lead to hyperactivation of the PI3K signaling pathway, which results in abnormal tissue growth. Diagnosis is complicated by the variability and overlap in phenotypes associated with PIK3CA-related disorders and should be performed by clinicians with the required expertise along with coordinated care from a multidisciplinary team. Although tissue mosaicism presents challenges for confirmation of PIK3CA mutations, next-generation sequencing and tissue selection have improved detection. Clinical improvement, radiological response, and patient-reported outcomes are typically used to assess treatment response in clinical studies of patients with PIK3CA-related disorders, but objective assessment of treatment response is difficult using imaging (due to the heterogeneous nature of these disorders, superimposed upon patient growth and development). Despite their limitations, patient-reported outcome tools may be best suited to gauge patient improvement. New therapeutic options are needed to provide an alternative or supplement to standard approaches such as surgery and sclerotherapy. Currently, there are no systemic agents that have regulatory approval for these disorders, but the mTOR inhibitor sirolimus has been used for several years in clinical trials and off label to address symptoms. There are also other agents under investigation for PIK3CA-related disorders that act as inhibitors to target different components of the PI3K signaling pathway including AKT (miransertib) and PI3K alpha (alpelisib). CONCLUSION Management of patients with PIK3CA-related disorders requires a multidisciplinary approach. Further results from ongoing clinical studies of agents targeting the PI3K pathway are highly anticipated.
Collapse
Affiliation(s)
- Guillaume Canaud
- Overgrowth Syndrome and Vascular Anomalies Unit, Hôpital Necker Enfants Malades, INSERM U1151, Assistance Publique-Hôpitaux de Paris, Université de Paris, 149 rue de Sèvres, 75105, Paris, France.
| | - Adrienne M Hammill
- Division of Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Denise Adams
- Division of Oncology, Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Perelman School of Medicine and the University of Pennsylvania, Philadelphia, PA, USA
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium.,Center for Vascular Anomalies, Division of Plastic Surgery, Cliniques Universitaires Saint Luc, University of Louvain, Brussels, Belgium.,VASCERN VASCA European Reference Centre, Bichat-Claude Bernard Hospital, Paris, France.,Walloon Excellence in Lifesciences and Biotechnology (WELBIO), University of Louvain, Brussels, Belgium
| | - Kim M Keppler-Noreuil
- Division of Genetics and Metabolism, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
47
|
Sorlin A, Carmignac V, Amiel J, Boccara O, Fraitag S, Maruani A, Theiler M, Weibel L, Duffourd Y, Philippe C, Thauvin-Robinet C, Faivre L, Rivière JB, Vabres P, Kuentz P. Expanding the clinical spectrum of mosaic BRAF skin phenotypes. J Eur Acad Dermatol Venereol 2021; 35:e690-e693. [PMID: 34051131 DOI: 10.1111/jdv.17413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/19/2021] [Indexed: 11/28/2022]
Affiliation(s)
- A Sorlin
- Centre de Génétique et Centre de référence « Anomalies du Développement et Syndromes Malformatifs », Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon, Dijon, France.,UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France
| | - V Carmignac
- UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France
| | - J Amiel
- Service de Génétique Médicale, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - O Boccara
- Department of Dermatology and Reference Center for Genodermatoses and Rare Skin Diseases (MAGEC), Université Paris, Paris-Centre, Institut Imagine, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France
| | - S Fraitag
- Service d'Anatomie et de Cytologie Pathologiques, APHP, Hôpital Necker-Enfants Malades, Paris, France
| | - A Maruani
- Service de Dermatologie, Centre de Référence des Maladies Rares - MAGEC, Centre Hospitalier Universitaire de Tours, Université de Tours, SPHERE-INSERM 1246, Tours, France
| | - M Theiler
- Pediatric Skin Center, Department of Dermatology, University Children's Hospital Zurich, Zurich, Switzerland
| | - L Weibel
- Pediatric Skin Center, Department of Dermatology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Y Duffourd
- UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), Centre Hospitalier Universitaire de Dijon et Université de Bourgogne Franche-Comté, Dijon, France
| | - C Philippe
- UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), Centre Hospitalier Universitaire de Dijon et Université de Bourgogne Franche-Comté, Dijon, France.,UF Innovation en diagnostic génomique des maladies rares, CHU de Dijon Bourgogne, Dijon, France
| | - C Thauvin-Robinet
- Centre de Génétique et Centre de référence « Anomalies du Développement et Syndromes Malformatifs », Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon, Dijon, France.,UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), Centre Hospitalier Universitaire de Dijon et Université de Bourgogne Franche-Comté, Dijon, France
| | - L Faivre
- Centre de Génétique et Centre de référence « Anomalies du Développement et Syndromes Malformatifs », Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon, Dijon, France.,UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), Centre Hospitalier Universitaire de Dijon et Université de Bourgogne Franche-Comté, Dijon, France
| | - J-B Rivière
- UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), Centre Hospitalier Universitaire de Dijon et Université de Bourgogne Franche-Comté, Dijon, France.,UF Innovation en diagnostic génomique des maladies rares, CHU de Dijon Bourgogne, Dijon, France
| | - P Vabres
- UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), Centre Hospitalier Universitaire de Dijon et Université de Bourgogne Franche-Comté, Dijon, France.,Service de Dermatologie, Centre Hospitalier Universitaire de Dijon, Dijon, France
| | - P Kuentz
- UMR-Inserm 1231 GAD, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France.,Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), Centre Hospitalier Universitaire de Dijon et Université de Bourgogne Franche-Comté, Dijon, France.,Oncobiologie Génétique Bioinformatique, PCBio, Centre Hospitalier Universitaire de Besançon, Besançon, France
| |
Collapse
|
48
|
Chenbhanich J, Hu Y, Hetts S, Cooke D, Dowd C, Devine P, Russell B, Kang SHL, Chang VY, Abla AA, Cornett P, Yeh I, Lee H, Martinez-Agosto JA, Frieden IJ, Shieh JT. Segmental overgrowth and aneurysms due to mosaic PDGFRB p.(Tyr562Cys). Am J Med Genet A 2021; 185:1430-1436. [PMID: 33683022 DOI: 10.1002/ajmg.a.62126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/18/2020] [Accepted: 01/14/2021] [Indexed: 01/19/2023]
Abstract
Activating variants in the platelet-derived growth factor receptor β gene (PDGFRB) have been associated with Kosaki overgrowth syndrome, infantile myofibromatosis, and Penttinen premature aging syndrome. A recently described phenotype with fusiform aneurysm has been associated with mosaic PDGFRB c.1685A > G p.(Tyr562Cys) variant. Few reports however have examined the vascular phenotypes and mosaic effects of PDGFRB variants. We describe clinical characteristics of two patients with a recurrent mosaic PDGFRB p.(Tyr562Cys) variant identified via next-generation sequencing-based genetic testing. We observed intracranial fusiform aneurysm in one patient and found an additional eight patients with aneurysms and phenotypes associated with PDGFRB-activating variants through literature search. The conditions caused by PDGFRB-activating variants share overlapping features including overgrowth, premature aged skin, and vascular malformations including aneurysms. Aneurysms are progressive and can result in morbidities and mortalities in the absence of successful intervention. Germline and/or somatic testing for PDGFRB gene should be obtained when PDGFRB activating variant-related phenotypes are present. Whole-body imaging of the arterial tree and echocardiography are recommended after diagnosis. Repeating the imaging study within a 6- to 12-month period after detection is reasonable. Finally, further evaluation for the effectiveness and safety profile of kinase inhibitors in this patient population is warranted.
Collapse
Affiliation(s)
- Jirat Chenbhanich
- Division of Medical Genetics, Department of Pediatrics, University of California, San Francisco, California, USA
| | - Yan Hu
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Steven Hetts
- Division of Neurointerventional Radiology, Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Daniel Cooke
- Division of Neurointerventional Radiology, Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Christopher Dowd
- Division of Neurointerventional Radiology, Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Patrick Devine
- Department of Pathology and Laboratory Medicine, University of California, San Francisco, California, USA.,Institute of Human Genetics, University of California, San Francisco, California, USA
| | | | - Bianca Russell
- Department of Pediatrics, Division of Medical Genetics, University of California Los Angeles, Los Angeles, California, USA
| | - Sung Hae L Kang
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Vivian Y Chang
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Adib A Abla
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Patricia Cornett
- Department of Hematology and Oncology, University of California, San Francisco, California, USA
| | - Iwei Yeh
- Department of Pathology and Laboratory Medicine, University of California, San Francisco, California, USA.,Department of Dermatology, University of California, San Francisco, California, USA
| | - Hane Lee
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Human Genetics, University of California Los Angeles, Los Angeles, California, USA
| | - Julian A Martinez-Agosto
- Division of Neurointerventional Radiology, Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA.,Department of Human Genetics, University of California Los Angeles, Los Angeles, California, USA
| | - Ilona J Frieden
- Department of Dermatology, University of California, San Francisco, California, USA
| | - Joseph T Shieh
- Division of Medical Genetics, Department of Pediatrics, University of California, San Francisco, California, USA.,Institute of Human Genetics, University of California, San Francisco, California, USA
| |
Collapse
|
49
|
Garde A, Guibaud L, Goldenberg A, Petit F, Dard R, Roume J, Mazereeuw-Hautier J, Chassaing N, Lacombe D, Morice-Picard F, Toutain A, Arpin S, Boccara O, Touraine R, Blanchet P, Coubes C, Willems M, Pinson L, Van Kien PK, Chiaverini C, Giuliano F, Alessandri JL, Mathieu-Dramard M, Morin G, Bursztejn AC, Mignot C, Doummar D, Di Rocco F, Cornaton J, Nicolas C, Gautier E, Luu M, Bardou M, Sorlin A, Philippe C, Edery P, Rossi M, Carmignac V, Thauvin-Robinet C, Vabres P, Faivre L. Clinical and neuroimaging findings in 33 patients with MCAP syndrome: A survey to evaluate relevant endpoints for future clinical trials. Clin Genet 2021; 99:650-661. [PMID: 33415748 DOI: 10.1111/cge.13918] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/17/2020] [Accepted: 01/04/2021] [Indexed: 11/30/2022]
Abstract
Megalencephaly-CApillary malformation-Polymicrogyria (MCAP) syndrome results from somatic mosaic gain-of-function variants in PIK3CA. Main features are macrocephaly, somatic overgrowth, cutaneous vascular malformations, connective tissue dysplasia, neurodevelopmental delay, and brain anomalies. The objectives of this study were to describe the clinical and radiological features of MCAP, to suggest relevant clinical endpoints applicable in future trials of targeted drug therapy. Based on a French collaboration, we collected clinical features of 33 patients (21 females, 12 males, median age of 9.9 years) with MCAP carrying mosaic PIK3CA pathogenic variants. MRI images were reviewed for 21 patients. The main clinical features reported were macrocephaly at birth (20/31), postnatal macrocephaly (31/32), body/facial asymmetry (21/33), cutaneous capillary malformations (naevus flammeus 28/33, cutis marmorata 17/33). Intellectual disability was present in 15 patients. Among the MRI images reviewed, the neuroimaging findings were megalencephaly (20/21), thickening of corpus callosum (16/21), Chiari malformation (12/21), ventriculomegaly/hydrocephaly (10/21), cerebral asymmetry (6/21) and polymicrogyria (2/21). This study confirms the main known clinical features that defines MCAP syndrome. Taking into account the phenotypic heterogeneity in MCAP patients, in the context of emerging clinical trials, we suggest that patients should be evaluated based on the main neurocognitive expression on each patient.
Collapse
Affiliation(s)
- Aurore Garde
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Laurent Guibaud
- Service de Radiologie, Hôpital Femme-Mère-Enfant, Lyon, France
| | | | - Florence Petit
- Service de Génétique Clinique, Centre de Référence Anomalies du Développement CHU, Lille, France
| | - Rodolphe Dard
- Département de Génétique, CHI Poissy, St Germain-en-Laye, France
| | - Joelle Roume
- Département de Génétique, CHI Poissy, St Germain-en-Laye, France
| | - Juliette Mazereeuw-Hautier
- Département de Dermatologie, Centre de Référence des Maladies Rares de la Peau, CHU de Toulouse, Toulouse, France
| | - Nicolas Chassaing
- Service de Génétique Médicale, INSERM U543, Hôpital Purpan, CHU de Toulouse, Toulouse, France
| | - Didier Lacombe
- INSERM U1211, Service de Génétique Médicale, CHU de Bordeaux, Bordeaux, France
| | - Fanny Morice-Picard
- INSERM U1211, Service de Génétique Médicale, CHU de Bordeaux, Bordeaux, France
| | | | | | - Olivia Boccara
- Département de Dermatologie, Centre de Reference MAGEC, Hopital universitaire Necker-Enfants malades, Paris, France
| | - Renaud Touraine
- Service de Génétique Clinique, Chromosomique et Moléculaire, Centre de Référence des Anomalies du Développement, CHU, de Saint-Etienne, France
| | - Patricia Blanchet
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, CHRU de Montpellier, Montpellier, France
| | - Christine Coubes
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, CHRU de Montpellier, Montpellier, France
| | - Marjolaine Willems
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, CHRU de Montpellier, Montpellier, France
| | - Lucile Pinson
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, CHRU de Montpellier, Montpellier, France
| | | | | | | | | | | | - Gilles Morin
- Service de Génétique Clinique, CHU Amiens-Picardie, Amiens, France
| | | | - Cyril Mignot
- Département de Génétique and Centre de Référence Déficiences Intellectuelles de Causes Rares, AP-HP, Sorbonne Université, Paris, France
| | - Diane Doummar
- Service de Neurologie pédiatrique, Hôpital Armand Trousseau, AP-HP, Paris, France
| | - Frederico Di Rocco
- Service de neurochirurgie pédiatrique, Hôpital Femme-Mère-Enfant, Lyon, France
| | - Jenny Cornaton
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, Dijon, France
| | - Claire Nicolas
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, Dijon, France
| | - Elodie Gautier
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, Dijon, France
| | - Maxime Luu
- INSERM CIC 1432, Université de Bourgogne, Dijon, France
| | - Marc Bardou
- INSERM CIC 1432, Université de Bourgogne, Dijon, France
| | - Arthur Sorlin
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France
| | - Christophe Philippe
- Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France
| | - Patrick Edery
- Département de Génétique, Hospices Civils de Lyon et GENDEV, INSERM U1028, Lyon, France
| | - Massimiliano Rossi
- Département de Génétique, Hospices Civils de Lyon et GENDEV, INSERM U1028, Lyon, France
| | - Virginie Carmignac
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Centre de Référence MAGEC, Service de Dermatologie, Centre Hospitalier Universitaire Dijon Bourgogne, Dijon, France
| | - Christel Thauvin-Robinet
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France
| | - Pierre Vabres
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Centre de Référence MAGEC, Service de Dermatologie, Centre Hospitalier Universitaire Dijon Bourgogne, Dijon, France
| | - Laurence Faivre
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de Référence MAGEC, Service de Dermatologie, Centre Hospitalier Universitaire Dijon Bourgogne, Dijon, France
| |
Collapse
|
50
|
Tian W, Huang Y, Sun L, Guo Y, Zhao S, Lin M, Dong X, Zhong W, Yin Y, Chen Z, Zhang N, Zhang Y, Wang L, Lin J, Yan Z, Yang X, Zhao J, Qiu G, Zhang J, Wu Z, Wu N. Phenotypic and genetic spectrum of isolated macrodactyly: somatic mosaicism of PIK3CA and AKT1 oncogenic variants. Orphanet J Rare Dis 2020; 15:288. [PMID: 33054853 PMCID: PMC7556951 DOI: 10.1186/s13023-020-01572-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 10/05/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Isolated macrodactyly is a severe congenital hand anomaly with functional and physiological impact. Known causative genes include PIK3CA, AKT1 and PTEN. The aim of this study is to gain insights into the genetics basis of isolated macrodactyly. RESULTS We enrolled 24 patients with isolated macrodactyly. Four of them were diagnosed with Proteus syndrome based on skin presentations characteristic to this disease. Targeted next-generation sequencing was performed using patients' blood and affected tissues. Overall, 20 patients carry mosaic PIK3CA pathogenic variants, i.e. p.His1047Arg (N = 7), p.Glu542Lys (N = 6), p.Glu545Lys (N = 2), p.His1047Leu (N = 2), p.Glu453Lys (N = 1), p.Gln546Lys (N = 1) and p.His1047Tyr (N = 1). Four patients who met the diagnostic criteria of Proteus syndrome carry mosaic AKT1 p.Glu17Lys variant. Variant allele frequencies of these mosaic variants obtained through next-generation sequencing range from 10 to 33%. In genotype-phenotype correlation analysis of patients with PIK3CA variant, we found that patients with the macrodactyly of the lower limbs tend to carry PIK3CA variants located in the helical domain (P = 0.005). CONCLUSIONS Mosaic PIK3CA and AKT1 variants can be found in all of our samples with isolated macrodactyly. Insights into phenotypic and genetic spectrum of isolated macrodactyly may be helpful in perusing a more precise and effective management of isolated macrodactyly.
Collapse
Affiliation(s)
- Wen Tian
- Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Yingzhao Huang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Liying Sun
- Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Yang Guo
- Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Sen Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Mao Lin
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiying Dong
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Wenyao Zhong
- Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Yuehan Yin
- Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Zefu Chen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Nan Zhang
- Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Yuanqiang Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lianlei Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jiachen Lin
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zihui Yan
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinzhuang Yang
- Department of Central Laboratory, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Junhui Zhao
- Department of Hand Surgery, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Guixing Qiu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jianguo Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zhihong Wu
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China. .,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China. .,Department of Central Laboratory, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China.
| | - Nan Wu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Beijing, 100730, China. .,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China. .,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | | |
Collapse
|