1
|
Revencu N, Coulie J, Boon LM, Vikkula M. Genetic aspects of vascular malformations. Presse Med 2025:104295. [PMID: 40449790 DOI: 10.1016/j.lpm.2025.104295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 05/09/2025] [Indexed: 06/03/2025] Open
Abstract
Vascular anomalies are a heterogenous group of diseases that include vascular tumours and vascular malformations. Over the past two decades, significant progress has been made in elucidating the genetic basis of these anomalies, leading to improved classification, management and genetic counselling. Major signalling pathways, such as RAS/MAPK/ERK, PI3K/AKT/mTOR, and G protein-coupled receptor pathways, have been identified as central to the pathogenesis of vascular anomalies. This article reviews the major types of vascular malformations, addresses the challenges associated with genetic testing and counselling, and explores the emerging potential for precision medicine in the treatment of these diseases.
Collapse
Affiliation(s)
- Nicole Revencu
- Center for Human Genetics, Cliniques universitaires Saint-Luc, University of Louvain, Brussels, Belgium; VASCERN VASCA European Reference Centre.
| | - Julien Coulie
- Center for Vascular Anomalies, Division of Plastic Surgery, Cliniques universitaires Saint-Luc, University of Louvain, Brussels, Belgium; VASCERN VASCA European Reference Centre; Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| | - Laurence M Boon
- Center for Vascular Anomalies, Division of Plastic Surgery, Cliniques universitaires Saint-Luc, University of Louvain, Brussels, Belgium; VASCERN VASCA European Reference Centre; Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium; WELBIO department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
2
|
El Sissy FN, Bisdorff A, Perrier A, Guillerm E, Denis J, Favre L, Aubertin M, Eyries M, Coulet F. Identification of Somatic Genetic Variants in Superficial Vascular Malformations by Liquid Biopsy in a Cohort of 88 Patients from a French Hospital. Mol Diagn Ther 2025; 29:367-380. [PMID: 39899252 DOI: 10.1007/s40291-025-00770-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND AND OBJECTIVE Superficial vascular anomalies are complex disorders characterized by abnormal vascular growth. Next-generation sequencing has recently identified somatic genetic alterations associated with these malformations, offering new insights for targeted treatments. However, tissue biopsies for genetic testing can be invasive and difficult to obtain, especially in arteriovenous malformations (AVM) with hemorrhagic risks. A liquid biopsy, a non-invasive approach, offers a promising solution by detecting genetic mutations in cell-free DNA. This pilot study aimed to evaluate the feasibility of using a liquid biopsy for the genetic analysis of patients with superficial vascular anomalies through cell-free DNA sampling. Additionally, it explored whether specific sampling sites, such as the afferent artery, nidus, and efferent vein, could enhance the sensitivity of detecting pathogenic variants in patients with AVM. METHODS A total of 88 patients were enrolled, including 55 with AVM and 33 with lymphatic malformations. For patients with AVM, cell-free DNA samples were collected from peripheral blood, efferent veins, afferent arteries, and the AVM nidus. In patients with lymphatic malformations, cystic lymphatic fluid was collected by a direct puncture during diagnostic procedures. A molecular analysis was performed using a targeted gene panel relevant to somatic alterations in solid tumors. Pathogenic variants were validated by digital polymerase chain reaction for patients with lymphatic malformations. RESULTS Pathogenic variants were identified in 23.6% of patients with AVM, predominantly in MAP2K1 and KRAS genes, with higher sensitivity near the AVM nidus. In addition, pathogenic variants were identified in 27.3% of patients with lymphatic malformations, all involving the PIK3CA gene. Despite the lower sensitivity of a cell-free DNA analysis compared with a tissue biopsy, especially in patients with AVM, the detection rate suggests the utility for a cell-free DNA analysis, particularly when a tissue biopsy is not feasible. CONCLUSIONS This study confirms the feasibility of using a cell-free DNA liquid biopsy for genotyping patients with superficial vascular anomalies, although a tissue biopsy remains the gold standard for comprehensive genetic profiling because of its higher sensitivity. A liquid biopsy offers a non-invasive option for molecular analysis that is useful as a preliminary or alternative approach when direct tissue sampling is not possible. Importantly, the sensitivity of cell-free DNA sampling in AVM appeared highest when obtained close to the nidus, indicating an optimal sampling location for future studies. Further research is needed to improve detection sensitivity, especially for samples taken near the nidus, to validate and strengthen these findings. Although our study focused on superficial/extra-cranial AVM, further research should assess the applicability of this approach to cerebral AVM, where a tissue biopsy is particularly risky.
Collapse
Affiliation(s)
- Franck Neil El Sissy
- Department of Pathology, Lariboisière Hospital, Assistance Publique Hôpitaux de Paris, 2 rue Ambroise Paré, 75010, Paris, France.
- University of Paris Cité, Faculty of Medicine, Paris, France.
| | - Annouk Bisdorff
- Department of Neuroradiology, Lariboisière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Alexandre Perrier
- Department of Neuroradiology, Lariboisière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Erell Guillerm
- Department of Neuroradiology, Lariboisière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jérôme Denis
- Department of Neuroradiology, Lariboisière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Löetitia Favre
- Department of Neuroradiology, Lariboisière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Mathilde Aubertin
- Department of Neuroradiology, Lariboisière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Mélanie Eyries
- Department of Neuroradiology, Lariboisière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Florence Coulet
- Department of Genetics, La Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
- University of Sorbonne, Faculty of Medicine, Paris, France
| |
Collapse
|
3
|
Li GX, Sebaratnam DF, Pham JP. Targeted Therapies for Slow-Flow Vascular Malformations. Australas J Dermatol 2025; 66:142-151. [PMID: 40095204 PMCID: PMC12062736 DOI: 10.1111/ajd.14451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Advances in genetic sequencing technologies have enabled the identification of key activating somatic variants in cellular signalling pathways involved in the pathogenesis of vascular malformations. Given that these genetic variants are also implicated in the pathogenesis of several cancers, the repurposing of targeted therapies developed in oncology has been increasingly investigated for treating vascular malformations. This review provides an update on the current evidence for targeted therapies in slow-flow vascular malformations, particularly in the context of gain-of-function variants in the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Grace X. Li
- Faculty of Medicine and HealthUniversity of New South WalesKensingtonNew South WalesAustralia
- Department of DermatologyLiverpool HospitalLiverpoolNew South WalesAustralia
| | - Deshan F. Sebaratnam
- Faculty of Medicine and HealthUniversity of New South WalesKensingtonNew South WalesAustralia
- Department of DermatologyLiverpool HospitalLiverpoolNew South WalesAustralia
| | - James P. Pham
- Faculty of Medicine and HealthUniversity of New South WalesKensingtonNew South WalesAustralia
- Department of DermatologyLiverpool HospitalLiverpoolNew South WalesAustralia
| |
Collapse
|
4
|
Bonilla-Velez J, Heike CL, Kessler LG, Wang X, Wenger TL, Ramsey BW, Perkins JA. Incidence and Factors Associated With Spontaneous Regression in Head and Neck Lymphatic Malformations. JAMA Otolaryngol Head Neck Surg 2025; 151:503-512. [PMID: 40146172 PMCID: PMC11950974 DOI: 10.1001/jamaoto.2025.0104] [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: 06/26/2024] [Accepted: 01/17/2025] [Indexed: 03/28/2025]
Abstract
Importance Head and neck lymphatic malformations (HNLMs) demonstrate considerable variability in their natural history. While some malformations cause chronic severe functional impairment, others are asymptomatic and spontaneously regress. Understanding the frequency and features associated with regression will aid clinicians and families in making informed treatment choices and avoid unnecessary risks of intervention for a subset of patients. Objective To assess the incidence, timing, and factors associated with spontaneous regression of HNLMs. Design, Setting, and Participants This retrospective cohort study across a single regional pediatric tertiary care academic center included patients aged 0 to 21 years with HNLM who were evaluated in the vascular anomalies clinic and prospectively enrolled in an institutional quality improvement database between 2003 and 2022. Observation was offered to patients without symptoms or functional compromise. Treatment decisions were made during routine clinical care. Exposures Age of HNLM onset and HNLM location, distribution, cystic structure, grade, and de Serres staging. Main Outcome and Measures The incidence of complete spontaneous regression was estimated. The strength of the association between exposures and regression was assessed using risk ratios, Cliff Δ, and Cramer V. A Kaplan-Meier curve was used to estimate the probability of spontaneous regression over time. The association of the exposures on the likelihood of spontaneous regression were assessed using 2-sided log-rank tests and Cox proportional hazards models. Results Of 298 patients with HNLMs, 173 (58.1%) were male, and most HNLMs were diagnosed prenatally or at birth (137 patients [46.0%]). Among HNLMs, 189 (63.4%) had a focal distribution, 228 (76.5%) were de Serres stage I or II, and 128 (43.0%) were macrocystic. Complete spontaneous regression occurred in 27 patients (9.1%) at a median (IQR) time of 12.0 (6.7-27.4) months from onset. Factors associated with a large difference in the rate of complete spontaneous regression included macrocystic structure, neck location, focal distribution, and grade 1 or de Serres I stage. HNLMs of the upper face or midface, with mixed or microcystic composition, or with extensive unilateral or bilateral involvement did not regress. Conclusions and Relevance In this cohort study, HNLMs that were macrocystic, of limited extent, and/or in the neck were more likely to completely spontaneously regress. A 1-year observation period for asymptomatic HNLM, particularly when favorable features are present, should be considered, as nonintervention may be curative. Future studies will examine interaction among these factors. This work contributes to a deeper understanding of HNLM natural history that can directly inform clinical decision-making, decrease treatment risk, and optimize patient outcomes.
Collapse
Affiliation(s)
- Juliana Bonilla-Velez
- Division of Pediatric Otolaryngology–Head and Neck Surgery, Seattle Children’s Hospital, Seattle, Washington
- Department of Otolaryngology–Head and Neck Surgery, University of Washington School of Medicine, Seattle
- Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, Washington
| | - Carrie L. Heike
- Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, Washington
- Department of Pediatrics, University of Washington School of Medicine, Seattle
- Craniofacial Center, Seattle Children’s Hospital, Seattle, Washington
| | - Larry G. Kessler
- Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle
| | - Xing Wang
- Biostatistics Epidemiology and Analytics in Research (BEAR), Seattle Children’s Research Institute, Seattle, Washington
| | - Tara L. Wenger
- Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, Washington
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle
| | - Bonnie W. Ramsey
- Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, Washington
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle Children’s Hospital, Seattle
| | - Jonathan A. Perkins
- Division of Pediatric Otolaryngology–Head and Neck Surgery, Seattle Children’s Hospital, Seattle, Washington
- Department of Otolaryngology–Head and Neck Surgery, University of Washington School of Medicine, Seattle
- Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, Washington
- Craniofacial Center, Seattle Children’s Hospital, Seattle, Washington
| |
Collapse
|
5
|
He Q, Huo R, Sun Y, Zheng Z, Xu H, Zhao S, Ni Y, Yu Q, Jiao Y, Zhang W, Zhao J, Cao Y. Cerebral vascular malformations: pathogenesis and therapy. MedComm (Beijing) 2024; 5:e70027. [PMID: 39654683 PMCID: PMC11625509 DOI: 10.1002/mco2.70027] [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] [Received: 08/04/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Cerebral vascular malformations (CVMs), particularly cerebral cavernous malformations and cerebral arteriovenous malformations, pose significant neurological challenges due to their complex etiologies and clinical implications. Traditionally viewed as congenital conditions with structural abnormalities, CVMs have been treated primarily through resection, embolization, and stereotactic radiosurgery. While these approaches offer some efficacy, they often pose risks to neurological integrity due to their invasive nature. Advances in next-generation sequencing, particularly high-depth whole-exome sequencing and bioinformatics, have facilitated the identification of gene variants from neurosurgically resected CVMs samples. These advancements have deepened our understanding of CVM pathogenesis. Somatic mutations in key mechanistic pathways have been identified as causative factors, leading to a paradigm shift in CVM treatment. Additionally, recent progress in noninvasive and minimally invasive techniques, including gene imaging genomics, liquid biopsy, or endovascular biopsies (endovascular sampling of blood vessel lumens), has enabled the identification of gene variants associated with CVMs. These methods, in conjunction with clinical data, offer potential for early detection, dynamic monitoring, and targeted therapies that could be used as monotherapy or adjuncts to surgery. This review highlights advancements in CVM pathogenesis and precision therapies, outlining the future potential of precision medicine in CVM management.
Collapse
Affiliation(s)
- Qiheng He
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Ran Huo
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yingfan Sun
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Zhiyao Zheng
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Research Unit of Accurate DiagnosisTreatment, and Translational Medicine of Brain Tumors Chinese Academy of Medical Sciences and Peking Union Medical College Beijing ChinaBeijingChina
- Department of Neurosurgery Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical College Beijing ChinaBeijingChina
| | - Hongyuan Xu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Shaozhi Zhao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yang Ni
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Qifeng Yu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yuming Jiao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Wenqian Zhang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jizong Zhao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yong Cao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
- Collaborative Innovation CenterBeijing Institute of Brain DisordersBeijingChina
| |
Collapse
|
6
|
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
|
7
|
Helmijr J, Motta G, Jongbloed L, de Weerd V, van Bergen L, Verschoor N, Stella S, Beaufort C, Vigneri P, Martens JWM, Wilting SM, Jansen MPHM. A Multiplex Assay for Fast PIK3CA Hotspot Mutation Characterization in a Single Specimen by 3-Color Digital PCR Analysis. J Appl Lab Med 2024; 9:913-925. [PMID: 39012846 DOI: 10.1093/jalm/jfae064] [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: 11/23/2023] [Accepted: 05/01/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Activating mutations in the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) gene have been detected often in solid tumors. Targeted therapy for mutant PIK3CA is now available in the clinic, making molecular diagnostics pivotal. Our aim was to design a multiplex digital PCR (dPCR) assay to evaluate the 4 most common PIK3CA hotspot mutations simultaneously to characterize and quantify these in liquid biopsies. METHODS A multiplex assay was developed to detect exon 9 p.E542K and p.E545K mutations, and exon 20 p.H1047L and p.H1047R mutations using the Stilla 3-color dPCR Naica system. The assay was evaluated on stock and pre-amplified DNA from cell lines with the above mutations as single and pooled samples, and on cell-free DNA (cfDNA) from healthy blood donors (HBDs) and breast cancer patients, to determine detection thresholds and diagnostic accuracy. RESULTS The assay distinguished all 4 PIK3CA mutations in (cf)DNA, and also when dual mutations were present. Detection thresholds of stock and pre-amplified cfDNA samples were 0.11 and 0.40 copies/uL (cp/uL) for mutant copies concentration, and 0.003% and 0.68% for variant allele frequencies (VAFs), respectively. The assay confirmed the PIK3CA (mutation) status as defined by targeted next-generation sequencing (NGS) in 82 out of 96 patients that were mutant for PIK3CA, and in 11 out of 12 patients with wild-type PIK3CA. CONCLUSIONS Our designed multiplex dPCR assay detected PIK3CA mutations with high accuracy in stock and pre-amplified cfDNA. Furthermore, it is affordable and demands less cfDNA input when compared to available uniplex dPCR assays and NGS analyses.
Collapse
Affiliation(s)
- Jean Helmijr
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Gianmarco Motta
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
- Department of Clinical and Experimental Medicine, Center for Experimental Oncology and Hematology, University of Catania, Catania, Italy
| | - Lisa Jongbloed
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Vanja de Weerd
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Lotte van Bergen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Noortje Verschoor
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Stefania Stella
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
- Department of Clinical and Experimental Medicine, Center for Experimental Oncology and Hematology, University of Catania, Catania, Italy
| | - Corine Beaufort
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, Center for Experimental Oncology and Hematology, University of Catania, Catania, Italy
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Saskia M Wilting
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Maurice P H M Jansen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
8
|
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
|
9
|
Abstract
This review delves into the rapidly evolving landscape of liquid biopsy technologies based on cell-free DNA (cfDNA) and cell-free RNA (cfRNA) and their increasingly prominent role in precision medicine. With the advent of high-throughput DNA sequencing, the use of cfDNA and cfRNA has revolutionized noninvasive clinical testing. Here, we explore the physical characteristics of cfDNA and cfRNA, present an overview of the essential engineering tools used by the field, and highlight clinical applications, including noninvasive prenatal testing, cancer testing, organ transplantation surveillance, and infectious disease testing. Finally, we discuss emerging technologies and the broadening scope of liquid biopsies to new areas of diagnostic medicine.
Collapse
Affiliation(s)
- Conor Loy
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Lauren Ahmann
- Department of Pathology, Stanford University, Stanford, California, USA;
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Wei Gu
- Department of Pathology, Stanford University, Stanford, California, USA;
| |
Collapse
|
10
|
Revencu N, Eijkelenboom A, Bracquemart C, Alhopuro P, Armstrong J, Baselga E, Cesario C, Dentici ML, Eyries M, Frisk S, Karstensen HG, Gene-Olaciregui N, Kivirikko S, Lavarino C, Mero IL, Michiels R, Pisaneschi E, Schönewolf-Greulich B, Wieland I, Zenker M, Vikkula M. Assessment of gene-disease associations and recommendations for genetic testing for somatic variants in vascular anomalies by VASCERN-VASCA. Orphanet J Rare Dis 2024; 19:213. [PMID: 38778413 PMCID: PMC11110196 DOI: 10.1186/s13023-024-03196-9] [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: 12/12/2023] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Vascular anomalies caused by somatic (postzygotic) variants are clinically and genetically heterogeneous diseases with overlapping or distinct entities. The genetic knowledge in this field is rapidly growing, and genetic testing is now part of the diagnostic workup alongside the clinical, radiological and histopathological data. Nonetheless, access to genetic testing is still limited, and there is significant heterogeneity across the approaches used by the diagnostic laboratories, with direct consequences on test sensitivity and accuracy. The clinical utility of genetic testing is expected to increase progressively with improved theragnostics, which will be based on information about the efficacy and safety of the emerging drugs and future molecules. The aim of this study was to make recommendations for optimising and guiding the diagnostic genetic testing for somatic variants in patients with vascular malformations. RESULTS Physicians and lab specialists from 11 multidisciplinary European centres for vascular anomalies reviewed the genes identified to date as being involved in non-hereditary vascular malformations, evaluated gene-disease associations, and made recommendations about the technical aspects for identification of low-level mosaicism and variant interpretation. A core list of 24 genes were selected based on the current practices in the participating laboratories, the ISSVA classification and the literature. In total 45 gene-phenotype associations were evaluated: 16 were considered definitive, 16 strong, 3 moderate, 7 limited and 3 with no evidence. CONCLUSIONS This work provides a detailed evidence-based view of the gene-disease associations in the field of vascular malformations caused by somatic variants. Knowing both the gene-phenotype relationships and the strength of the associations greatly help laboratories in data interpretation and eventually in the clinical diagnosis. This study reflects the state of knowledge as of mid-2023 and will be regularly updated on the VASCERN-VASCA website (VASCERN-VASCA, https://vascern.eu/groupe/vascular-anomalies/ ).
Collapse
Affiliation(s)
- Nicole Revencu
- Center for Human Genetics, Cliniques universitaires Saint-Luc, University of Louvain, VASCERN VASCA European Reference Centre, Brussels, Belgium
| | - Astrid Eijkelenboom
- Department of Pathology, Radboud University Medical Center, VASCERN VASCA European Reference Centre, PO Box 9101, 6500, HB, Nijmegen, the Netherlands
| | - Claire Bracquemart
- Normandie Univ, UNICAEN, Service de Génétique, CHU Caen Normandie, BIOTARGEN EA 7450, VASCERN VASCA European Reference Centre, Caen, 14000, France
| | - Pia Alhopuro
- HUS Diagnostic Center, Laboratory of Genetics, University of Helsinki and Helsinki University Hospital, VASCERN VASCA European Reference Centre, Helsinki, Finland
| | - Judith Armstrong
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, and Genomic Unit, Molecular and Genetic Medicine Section, Hospital Sant Joan de Déu, VASCERN VASCA European Reference Centre, Barcelona, Spain
| | - Eulalia Baselga
- Department of Dermatology, Hospital Sant Joan de Deu, VASCERN VASCA European Reference Centre, Barcelona, Spain
| | - Claudia Cesario
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital and Research Institute, IRCCS, VASCERN VASCA European Reference Centre, Rome, Italy
| | - Maria Lisa Dentici
- Medical Genetics Unit, Bambino Gesù Children's Hospital, IRCCS, VASCERN VASCA European Reference Centre, 00165, Rome, Italy
| | - Melanie Eyries
- Sorbonne Université, Département de Génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, VASCERN VASCA European Reference Centre, Paris, France
| | - Sofia Frisk
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Clinical Genetics, Karolinska University Hospital, VASCERN VASCA European Reference Centre, Stockholm, Sweden
| | - Helena Gásdal Karstensen
- Department of Genetics, Center of Diagnostics, Copenhagen University Hospital - Rigshospitalet, VASCERN VASCA European Reference Centre, Copenhagen, Denmark
| | - Nagore Gene-Olaciregui
- Laboratory of Molecular Oncology, Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, VASCERN VASCA European Reference Centre, Barcelona, Spain
| | - Sirpa Kivirikko
- Department of Clinical Genetics, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, VASCERN VASCA European Reference Centre, Helsinki, Finland
| | - Cinzia Lavarino
- Laboratory of Molecular Oncology, Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, VASCERN VASCA European Reference Centre, Barcelona, Spain
| | - Inger-Lise Mero
- Department of Medical Genetics, Oslo University Hospital, VASCERN VASCA European Reference Centre, Oslo, Norway
| | - Rodolphe Michiels
- Center for Human Genetics, Cliniques universitaires Saint-Luc, University of Louvain, VASCERN VASCA European Reference Centre, Brussels, Belgium
| | - Elisa Pisaneschi
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital and Research Institute, IRCCS, VASCERN VASCA European Reference Centre, Rome, Italy
| | - Bitten Schönewolf-Greulich
- Department of Genetics, Center of Diagnostics, Copenhagen University Hospital - Rigshospitalet, VASCERN VASCA European Reference Centre, Copenhagen, Denmark
| | - Ilse Wieland
- Institute of Human Genetics, University Hospital Otto-Von-Guericke-University, Magdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Otto-Von-Guericke-University, Magdeburg, Germany
| | - Miikka Vikkula
- Center for Vascular Anomalies, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
- Human Molecular Genetics , de Duve Institute, University of Louvain, VASCERN VASCA European Reference Centre, Brussels, Belgium.
- WELBIO Department, WEL Research Institute, Avenue Pasteur, 6, 1300, Wavre, Belgium.
| |
Collapse
|
11
|
Petkova M, Ferby I, Mäkinen T. Lymphatic malformations: mechanistic insights and evolving therapeutic frontiers. J Clin Invest 2024; 134:e172844. [PMID: 38488007 PMCID: PMC10940090 DOI: 10.1172/jci172844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2024] Open
Abstract
The lymphatic vascular system is gaining recognition for its multifaceted role and broad pathological significance. Once perceived as a mere conduit for interstitial fluid and immune cell transport, recent research has unveiled its active involvement in critical physiological processes and common diseases, including inflammation, autoimmune diseases, and atherosclerosis. Consequently, abnormal development or functionality of lymphatic vessels can result in serious health complications. Here, we discuss lymphatic malformations (LMs), which are localized lesions that manifest as fluid-filled cysts or extensive infiltrative lymphatic vessel overgrowth, often associated with debilitating, even life-threatening, consequences. Genetic causes of LMs have been uncovered, and several promising drug-based therapies are currently under investigation and will be discussed.
Collapse
Affiliation(s)
- Milena Petkova
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ingvar Ferby
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| |
Collapse
|
12
|
Woodis KM, Garlisi Torales LD, Wolf A, Britt A, Sheppard SE. Updates in Genetic Testing for Head and Neck Vascular Anomalies. Oral Maxillofac Surg Clin North Am 2024; 36:1-17. [PMID: 37867039 PMCID: PMC11092895 DOI: 10.1016/j.coms.2023.09.001] [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] [Indexed: 10/24/2023]
Abstract
Vascular anomalies include benign or malignant tumors or benign malformations of the arteries, veins, capillaries, or lymphatic vasculature. The genetic etiology of the lesion is essential to define the lesion and can help navigate choice of therapy. . In the United States, about 1.2% of the population has a vascular anomaly, which may be underestimating the true prevalence as genetic testing for these conditions continues to evolve.
Collapse
Affiliation(s)
- Kristina M Woodis
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute for Child Health and Human Development, 10 Center Drive, MSC 1103, Bethesda, MD 20892-1103, USA
| | - Luciana Daniela Garlisi Torales
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute for Child Health and Human Development, 10 Center Drive, MSC 1103, Bethesda, MD 20892-1103, USA
| | - Alejandro Wolf
- Department of Pathology and ARUP Laboratories, University of Utah, 2000 Circle of Hope, Room 3100, Salt Lake City, UT 84112, USA
| | - Allison Britt
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sarah E Sheppard
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute for Child Health and Human Development, 10 Center Drive, MSC 1103, Bethesda, MD 20892-1103, USA.
| |
Collapse
|
13
|
Mansur A, Radovanovic I. The expansion of liquid biopsies to vascular care: an overview of existing principles, techniques and potential applications to vascular malformation diagnostics. Front Genet 2024; 15:1348096. [PMID: 38304336 PMCID: PMC10832994 DOI: 10.3389/fgene.2024.1348096] [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] [Received: 12/01/2023] [Accepted: 01/08/2024] [Indexed: 02/03/2024] Open
Abstract
Vascular malformations are congenital lesions that occur due to mutations in major cellular signalling pathways which govern angiogenesis, cell proliferation, motility, and cell death. These pathways have been widely studied in oncology and are substrates for various small molecule inhibitors. Given their common molecular biology, there is now a potential to repurpose these cancer drugs for vascular malformation care; however, a molecular diagnosis is required in order to tailour specific drugs to the individual patient's mutational profile. Liquid biopsies (LBs), emerging as a transformative tool in the field of oncology, hold significant promise in this feat. This paper explores the principles and technologies underlying LBs and evaluates their potential to revolutionize the management of vascular malformations. The review begins by delineating the fundamental principles of LBs, focusing on the detection and analysis of circulating biomarkers such as cell-free DNA, circulating tumor cells, and extracellular vesicles. Subsequently, an in-depth analysis of the technological advancements driving LB platforms is presented. Lastly, the paper highlights the current state of research in applying LBs to various vascular malformations, and uses the aforementioned principles and techniques to conceptualize a liquid biopsy framework that is unique to vascular malformation research and clinical care.
Collapse
Affiliation(s)
- Ann Mansur
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, School of Graduate Studies, University of Toronto, Toronto, ON, Canada
| | - Ivan Radovanovic
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
14
|
Sun Y, Su L, Wang D, Fan X, Cai R. Verrucous venous malformation and lymphatic malformation, former similar phenotypes where cell-free DNA could clarify in genotype. J Am Acad Dermatol 2024; 90:e27-e28. [PMID: 37717726 DOI: 10.1016/j.jaad.2023.07.1043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 09/19/2023]
Affiliation(s)
- Yi Sun
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lixin Su
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Deming Wang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xindong Fan
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ren Cai
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
15
|
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
|
16
|
Li D, Sheppard SE, March ME, Battig MR, Surrey LF, Srinivasan AS, Matsuoka LS, Tian L, Wang F, Seiler C, Dayneka J, Borst AJ, Matos MC, Paulissen SM, Krishnamurthy G, Nriagu B, Sikder T, Casey M, Williams L, Rangu S, O'Connor N, Thomas A, Pinto E, Hou C, Nguyen K, Pellegrino da Silva R, Chehimi SN, Kao C, Biroc L, Britt AD, Queenan M, Reid JR, Napoli JA, Low DM, Vatsky S, Treat J, Smith CL, Cahill AM, Snyder KM, Adams DM, Dori Y, Hakonarson H. Genomic profiling informs diagnoses and treatment in vascular anomalies. Nat Med 2023; 29:1530-1539. [PMID: 37264205 PMCID: PMC11184491 DOI: 10.1038/s41591-023-02364-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 04/24/2023] [Indexed: 06/03/2023]
Abstract
Vascular anomalies are malformations or tumors of the blood or lymphatic vasculature and can be life-threatening. Although molecularly targeted therapies can be life-saving, identification of the molecular etiology is often impeded by lack of accessibility to affected tissue samples, mosaicism or insufficient sequencing depth. In a cohort of 356 participants with vascular anomalies, including 104 with primary complex lymphatic anomalies (pCLAs), DNA from CD31+ cells isolated from lymphatic fluid or cell-free DNA from lymphatic fluid or plasma underwent ultra-deep sequencing thereby uncovering pathogenic somatic variants down to a variant allele fraction of 0.15%. A molecular diagnosis, including previously undescribed genetic causes, was obtained in 41% of participants with pCLAs and 72% of participants with other vascular malformations, leading to a new medical therapy for 63% (43/69) of participants and resulting in improvement in 63% (35/55) of participants on therapy. Taken together, these data support the development of liquid biopsy-based diagnostic techniques to identify previously undescribed genotype-phenotype associations and guide medical therapy in individuals with vascular anomalies.
Collapse
Affiliation(s)
- Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Sarah E Sheppard
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michael E March
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mark R Battig
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lea F Surrey
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Abhay S Srinivasan
- Division of Interventional Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Leticia S Matsuoka
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lifeng Tian
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fengxiang Wang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christoph Seiler
- Zebrafish Core, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jill Dayneka
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alexandra J Borst
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mary C Matos
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott M Paulissen
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Ganesh Krishnamurthy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bede Nriagu
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tamjeed Sikder
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Melissa Casey
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lydia Williams
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sneha Rangu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nora O'Connor
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alexandria Thomas
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Erin Pinto
- Jill and Mark Fishman Center for Lymphatic Disorders, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Cuiping Hou
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kenny Nguyen
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Samar N Chehimi
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Charlly Kao
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lauren Biroc
- Jill and Mark Fishman Center for Lymphatic Disorders, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Allison D Britt
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maria Queenan
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Janet R Reid
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joseph A Napoli
- Division of Plastic, Reconstructive, and Oral Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David M Low
- Division of Plastic, Reconstructive, and Oral Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Seth Vatsky
- Division of Interventional Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - James Treat
- Section of Dermatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christopher L Smith
- Jill and Mark Fishman Center for Lymphatic Disorders, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anne Marie Cahill
- Division of Interventional Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kristen M Snyder
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Denise M Adams
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yoav Dori
- Jill and Mark Fishman Center for Lymphatic Disorders, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Mansur A, Radovanovic I. Vascular malformations: An overview of their molecular pathways, detection of mutational profiles and subsequent targets for drug therapy. Front Neurol 2023; 14:1099328. [PMID: 36846125 PMCID: PMC9950274 DOI: 10.3389/fneur.2023.1099328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/25/2023] [Indexed: 02/12/2023] Open
Abstract
Vascular malformations are anomalies in vascular development that portend a significant risk of hemorrhage, morbidity and mortality. Conventional treatments with surgery, radiosurgery and/or endovascular approaches are often insufficient for cure, thereby presenting an ongoing challenge for physicians and their patients. In the last two decades, we have learned that each type of vascular malformation harbors inherited germline and somatic mutations in two well-known cellular pathways that are also implicated in cancer biology: the PI3K/AKT/mTOR and RAS/RAF/MEK pathways. This knowledge has led to recent efforts in: (1) identifying reliable mechanisms to detect a patient's mutational burden in a minimally-invasive manner, and then (2) understand how cancer drugs that target these mutations can be repurposed for vascular malformation care. The idea of precision medicine for vascular pathologies is growing in potential and will be critical in expanding the clinician's therapeutic armamentarium.
Collapse
Affiliation(s)
- Ann Mansur
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, School of Graduate Studies, University of Toronto, Toronto, ON, Canada
| | - Ivan Radovanovic
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
18
|
Richardson CM, Perkins JN, Zenner K, Bull C, Lutsky E, Jensen DM, Dmyterko V, Bennett JT, Wenger TL, Dahl JP, Bonilla-Velez J, Bly RA, Geddis AE, Perkins JA. Primary targeted medical therapy for management of bilateral head and neck lymphatic malformations in infants. Int J Pediatr Otorhinolaryngol 2023; 164:111371. [PMID: 36459725 PMCID: PMC10243723 DOI: 10.1016/j.ijporl.2022.111371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Patients born with bilateral head and neck lymphatic malformations (BHNLMs) often require multiple invasive treatments, including tracheostomy. We hypothesized that primary targeted medical therapy (pTMT) with diagnostic needle aspiration reduces the need for invasive therapy such as surgical resection and/or sclerotherapy. METHODS Retrospective case review was performed of infants with BHNLMs (Grade 2 or De Serres stage IV and V) treated only at our institution from 2000 to 2021. Patients were divided into two cohorts: those managed with pTMT and those managed with observation, sclerotherapy, or surgical intervention (non-pTMT). Data regarding interventions, clinical outcomes, morbidity, and mortality were analyzed with descriptive statistics. RESULTS Nine children with BHNLMs met inclusion criteria. Three (33%) were in the pTMT cohort and six (66%) were non-pTMT. Eight (89%) malformations were genotyped, and all demonstrated hotspot PIK3CA variants. All pTMT patients had sirolimus initiated in the first month of life and underwent needle aspiration of malformation cyst fluid for cell-free DNA samples. All pTMT patients tolerated medical therapy. For the non-pTMT cohort, primary treatment included none (deceased, n = 1, 17%), observation with needle aspiration (n = 1, 17%), surgical resection (n = 2, 33%), or combination surgery and sclerotherapy (n = 2, 33%). Intubation duration, intensive care and initial hospital length of stay were not different between cohorts. Four non-pTMT patients (67%) required tracheostomy, and two (33%) died prior to discharge. All pTMT patients survived and none required tracheostomy. Non-pTMT patients required a median of two invasive therapies prior to discharge (IQR 1-4) and a mean total of 13 over the course of their lifetime (IQR 1-16), compared to the pTMT group who did not require any lifetime invasive therapy, even after initial pTMT and discharge home. CONCLUSION This study compares patients with BHNLMs (Grade 2) treated with pTMT versus those treated with observation or invasive therapy. Patients treated with pTMT required no surgical or invasive procedural treatment of their malformations, no tracheostomy placement, no unplanned readmissions after discharge, and had no mortalities. Needle aspiration was useful as a therapeutic adjunct for cell-free DNA diagnosis of PIK3CA variants, which guided TMT.
Collapse
Affiliation(s)
- Clare M Richardson
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, Seattle Children's Hospital, MS OA.9.220, PO Box 5371, Seattle, WA, 98145, USA; Department of Otolaryngology - Head and Neck Surgery, University of Washington, 1959 NE Pacific St, Box 356515, Seattle, WA, 98195, USA.
| | - Jonathan N Perkins
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, Seattle Children's Hospital, MS OA.9.220, PO Box 5371, Seattle, WA, 98145, USA; Department of Otolaryngology - Head and Neck Surgery, University of Washington, 1959 NE Pacific St, Box 356515, Seattle, WA, 98195, USA.
| | - Kaitlyn Zenner
- Department of Otolaryngology - Head and Neck Surgery, University of Washington, 1959 NE Pacific St, Box 356515, Seattle, WA, 98195, USA.
| | - Catherine Bull
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, Seattle Children's Hospital, MS OA.9.220, PO Box 5371, Seattle, WA, 98145, USA.
| | - Erika Lutsky
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, Seattle Children's Hospital, MS OA.9.220, PO Box 5371, Seattle, WA, 98145, USA.
| | - Dana M Jensen
- Center for Clinical and Translational Research, Seattle Children's Hospita, USAl.
| | - Victoria Dmyterko
- Center for Clinical and Translational Research, Seattle Children's Hospita, USAl.
| | - James T Bennett
- Center for Clinical and Translational Research, Seattle Children's Hospita, USAl; Division of Genetic Medicine, Department of Pediatrics, Seattle Children's Hospital, USA.
| | - Tara L Wenger
- Division of Genetic Medicine, Department of Pediatrics, Seattle Children's Hospital, USA.
| | - John P Dahl
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, Seattle Children's Hospital, MS OA.9.220, PO Box 5371, Seattle, WA, 98145, USA; Department of Otolaryngology - Head and Neck Surgery, University of Washington, 1959 NE Pacific St, Box 356515, Seattle, WA, 98195, USA; Center for Clinical and Translational Research, Seattle Children's Hospita, USAl.
| | - Juliana Bonilla-Velez
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, Seattle Children's Hospital, MS OA.9.220, PO Box 5371, Seattle, WA, 98145, USA; Department of Otolaryngology - Head and Neck Surgery, University of Washington, 1959 NE Pacific St, Box 356515, Seattle, WA, 98195, USA; Center for Clinical and Translational Research, Seattle Children's Hospita, USAl.
| | - Randall A Bly
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, Seattle Children's Hospital, MS OA.9.220, PO Box 5371, Seattle, WA, 98145, USA; Department of Otolaryngology - Head and Neck Surgery, University of Washington, 1959 NE Pacific St, Box 356515, Seattle, WA, 98195, USA.
| | - Amy E Geddis
- Center for Clinical and Translational Research, Seattle Children's Hospita, USAl; Division of Hematology-Oncology, Department of Pediatrics, Seattle Children's Hospital, USA.
| | - Jonathan A Perkins
- Division of Otolaryngology - Head and Neck Surgery, Department of Surgery, Seattle Children's Hospital, MS OA.9.220, PO Box 5371, Seattle, WA, 98145, USA; Department of Otolaryngology - Head and Neck Surgery, University of Washington, 1959 NE Pacific St, Box 356515, Seattle, WA, 98195, USA; Center for Clinical and Translational Research, Seattle Children's Hospita, USAl.
| |
Collapse
|
19
|
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
|
20
|
Rose AL, Cathey SS. Genetic Causes of Vascular Malformations and Common Signaling Pathways Involved in Their Formation. Dermatol Clin 2022; 40:449-459. [DOI: 10.1016/j.det.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Bronkhorst AJ, Ungerer V, Oberhofer A, Holdenrieder S. The rising tide of cell-free DNA profiling: from snapshot to temporal genome analysis. J LAB MED 2022; 46:207-224. [DOI: 10.1515/labmed-2022-0030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Abstract
Genomes of diverse origins are continuously shed into human body fluids in the form of fragmented cell-free DNA (cfDNA). These molecules maintain the genetic and epigenetic codes of their originating source, and often carry additional layers of unique information in newly discovered physico-chemical features. Characterization of cfDNA thus presents the opportunity to non-invasively reconstruct major parts of the host- and metagenome in silico. Data from a single specimen can be leveraged to detect a broad range of disease-specific signatures and has already enabled the development of many pioneering diagnostic tests. Moreover, data from serial sampling may allow unparalleled mapping of the scantily explored landscape of temporal genomic changes as it relates to various changes in different physiological and pathological states of individuals. In this review, we explore how this vast dimension of biological information accessible through cfDNA analysis is being tapped towards the development of increasingly powerful molecular assays and how it is shaping emerging technologies. We also discuss how this departure from traditional paradigms of snapshot genetic testing may pave the way for an onrush of new and exciting discoveries in human biology.
Collapse
Affiliation(s)
- Abel Jacobus Bronkhorst
- Munich Biomarker Research Center , Institute of Laboratory Medicine , German Heart Centre Munich , Technical University Munich , Munich , Germany
| | - Vida Ungerer
- Munich Biomarker Research Center , Institute of Laboratory Medicine , German Heart Centre Munich , Technical University Munich , Munich , Germany
| | - Angela Oberhofer
- Munich Biomarker Research Center , Institute of Laboratory Medicine , German Heart Centre Munich , Technical University Munich , Munich , Germany
| | - Stefan Holdenrieder
- Munich Biomarker Research Center , Institute of Laboratory Medicine , German Heart Centre Munich , Technical University Munich , Munich , Germany
| |
Collapse
|
22
|
Setty BA, Wusik K, Hammill AM. How we approach genetics in the diagnosis and management of vascular anomalies. Pediatr Blood Cancer 2022; 69 Suppl 3:e29320. [PMID: 36070212 DOI: 10.1002/pbc.29320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 01/04/2023]
Abstract
Vascular anomalies are a heterogeneous group of disorders that are currently classified based on their clinical and histological characteristics. Over the past decade, there have been significant advances in molecular genetics that have led to identification of genetic alterations associated with vascular tumors, vascular malformations, and syndromes. Here, we describe known genetic alterations in vascular anomalies, discuss when and how to test, and examine how identification of causative genetic mutations provides for better management of these disorders through improved understanding of their pathogenesis and increasing use of targeted therapeutic agents in order to achieve better outcomes for our patients.
Collapse
Affiliation(s)
- Bhuvana A Setty
- Division of Hematology/Oncology/BMT, Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Katie Wusik
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - 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, Ohio, USA
| |
Collapse
|
23
|
Wasilewska K, Gambin T, Rydzanicz M, Szczałuba K, Płoski R. Postzygotic mutations and where to find them - Recent advances and future implications in the field of non-neoplastic somatic mosaicism. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 790:108426. [PMID: 35690331 DOI: 10.1016/j.mrrev.2022.108426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/05/2022] [Accepted: 06/03/2022] [Indexed: 01/01/2023]
Abstract
The technological progress of massively parallel sequencing (MPS) has triggered a remarkable development in the research on postzygotic mutations. Although the overwhelming majority of studies in the field focus on oncogenesis, non-neoplastic diseases are attracting more and more attention. The aim of this review was to summarize some of the most recent findings in the field of somatic mosaicism in diseases other than neoplastic events. We discuss the abundance and role of postzygotic mutations, with a special emphasis on disorders which occur only in a mosaic form (obligatory mosaic diseases; OMDs). Based on the list of OMDs compiled from the published literature and three databases (OMIM, Orphanet and MosaicBase), we demonstrate the prevalence of cancer-related genes across OMDs and suggest other sources to further explore OMDs and OMD-related genes. Additionally, we comment on some practical aspects related to mosaic diseases, such as approaches to tissue sampling, the MPS coverage required to detect variants at a very low frequency, as well as on bioinformatic and molecular tools dedicated to detect somatic mutations in MPS data.
Collapse
Affiliation(s)
- Krystyna Wasilewska
- Department of Medical Genetics, Medical University of Warsaw, ul. Pawińskiego 3c, 02-106 Warsaw, Poland
| | - Tomasz Gambin
- Institute of Computer Science, Warsaw University of Technology, Nowowiejska 15/19, 00-665 Warsaw, Poland
| | - Małgorzata Rydzanicz
- Department of Medical Genetics, Medical University of Warsaw, ul. Pawińskiego 3c, 02-106 Warsaw, Poland
| | - Krzysztof Szczałuba
- Department of Medical Genetics, Medical University of Warsaw, ul. Pawińskiego 3c, 02-106 Warsaw, Poland
| | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, ul. Pawińskiego 3c, 02-106 Warsaw, Poland.
| |
Collapse
|
24
|
Wiggins JM, Ali S, Polsky D. Cell-Free DNA in Dermatology Research. J Invest Dermatol 2022; 142:1523-1528.e1. [PMID: 35598899 PMCID: PMC10038729 DOI: 10.1016/j.jid.2022.02.021] [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: 10/18/2021] [Revised: 01/19/2022] [Accepted: 02/27/2022] [Indexed: 11/29/2022]
Abstract
In various diseases, particularly cancer, cell-free DNA (cfDNA) has been widely studied as a marker of disease prognosis or to facilitate the detection of therapeutic targets. In dermatology, most studies have focused on melanoma; other skin diseases such as vascular malformations and psoriasis have also been examined. Genetic alterations unique to the tissue of origin such as sequence variations, copy number alterations, chromosomal rearrangements, differential DNA methylation patterns, and fragmentation patterns can be identified in circulation providing information on patient disease status. These alterations can be detected either by PCR-based methods or next-generation sequencing depending on the target of interest. In this article, we discuss the origins of cfDNA, the most common methods of detection, current studies assessing cfDNA as a biomarker, and cfDNA's potential clinical applications in melanoma and other skin diseases. In addition, we provide important factors to consider during blood processing and DNA extraction as well as limitations for each assay.
Collapse
Affiliation(s)
- Jennifer M Wiggins
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, NYU Langone Health, New York, New York, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - Saim Ali
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, NYU Langone Health, New York, New York, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - David Polsky
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, NYU Langone Health, New York, New York, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA; Department of Pathology, NYU Grossman School of Medicine, NYU Langone Health, New York, New York, USA.
| |
Collapse
|
25
|
Zenner K, Jensen DM, Dmyterko V, Shivaram GM, Myers CT, Paschal CR, Rudzinski ER, Pham MHM, Cheng VC, Manning SC, Bly RA, Ganti S, Perkins JA, Bennett JT. Somatic activating BRAF variants cause isolated lymphatic malformations. HGG ADVANCES 2022; 3:100101. [PMID: 35373151 PMCID: PMC8972000 DOI: 10.1016/j.xhgg.2022.100101] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/10/2022] [Indexed: 11/20/2022] Open
Abstract
Somatic activating variants in PIK3CA, the gene that encodes the p110α catalytic subunit of phosphatidylinositol 3-kinase (PI3K), have been previously detected in ∼80% of lymphatic malformations (LMs).1 , 2 We report the presence of somatic activating variants in BRAF in individuals with LMs that do not possess pathogenic PIK3CA variants. The BRAF substitution p.Val600Glu (c.1799T>A), one of the most common driver mutations in cancer, was detected in multiple individuals with LMs. Histology revealed abnormal lymphatic channels with immunopositivity for BRAFV600E in endothelial cells that was otherwise indistinguishable from PIK3CA-positive LM. The finding that BRAF variants contribute to low-flow LMs increases the complexity of prior models associating low-flow vascular malformations (LM and venous malformations) with mutations in the PI3K-AKT-MTOR and high-flow vascular malformations (arteriovenous malformations) with mutations in the RAS-mitogen-activated protein kinase (MAPK) pathway.3 In addition, this work highlights the importance of genetic diagnosis prior to initiating medical therapy as more studies examine therapeutics for individuals with vascular malformations.
Collapse
Affiliation(s)
- Kaitlyn Zenner
- Seattle Children’s Hospital, Division of Pediatric Otolaryngology, Department of Otolaryngology/Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
- Vascular Anomalies Program, Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Dana M. Jensen
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Victoria Dmyterko
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Giridhar M. Shivaram
- Department of Radiology, Division of Interventional Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Candace T. Myers
- Department of Laboratories, Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Cate R. Paschal
- Department of Laboratories, Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Erin R. Rudzinski
- Department of Laboratories, Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Minh-Hang M. Pham
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - V. Chi Cheng
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Scott C. Manning
- Seattle Children’s Hospital, Division of Pediatric Otolaryngology, Department of Otolaryngology/Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
| | - Randall A. Bly
- Seattle Children’s Hospital, Division of Pediatric Otolaryngology, Department of Otolaryngology/Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
- Vascular Anomalies Program, Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Sheila Ganti
- Seattle Children’s Hospital, Division of Pediatric Otolaryngology, Department of Otolaryngology/Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
- Vascular Anomalies Program, Seattle Children’s Hospital, Seattle, WA 98105, USA
- Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Jonathan A. Perkins
- Seattle Children’s Hospital, Division of Pediatric Otolaryngology, Department of Otolaryngology/Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
- Vascular Anomalies Program, Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - James T. Bennett
- Vascular Anomalies Program, Seattle Children’s Hospital, Seattle, WA 98105, USA
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Seattle Children’s Hospital, Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
26
|
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
|
27
|
El Sissy FN, Wassef M, Faucon B, Salvan D, Nadaud S, Coulet F, Adle-Biassette H, Soubrier F, Bisdorff A, Eyries M. Somatic Mutational Landscape of Extracranial Arteriovenous Malformations and Phenotypic Correlations. J Eur Acad Dermatol Venereol 2022; 36:905-912. [PMID: 35238086 DOI: 10.1111/jdv.18046] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/15/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Somatic genetic variants may be the cause of extracranial arteriovenous malformations, but few studies have explored these genetic anomalies, and no genotype-phenotype correlations have been identified. OBJECTIVES To characterize the somatic genetic landscape of extracranial arteriovenous malformations and correlate these findings with the phenotypic characteristics of these lesions. METHODS This study included twenty-three patients with extracranial arteriovenous malformations that were confirmed clinically and treated by surgical resection, and for whom frozen tissue samples were available. Targeted next-generation sequencing analysis of tissues was performed using a gene panel that included vascular disease-related genes and tumor-related genes. RESULTS We identified a pathogenic variant in 18 out of 23 samples (78.3%). Pathogenic variants were mainly located in MAP2K1 (n=7) and KRAS (n=6), and more rarely in BRAF (n=2) and RASA1 (n=3). KRAS variants were significantly (p<0.005) associated with severe extended facial arteriovenous malformations, for which relapse after surgical resection is frequently observed, while MAP2K1 variants were significantly (p<0.005) associated with less severe, limited arteriovenous malformations located on the lips. CONCLUSIONS Our study highlights a high prevalence of pathogenic somatic variants, predominantly in MAP2K1 and KRAS, in extracranial arteriovenous malformations. In addition, our study identifies for the first time a correlation between the genotype, clinical severity and angiographic characteristics of extracranial arteriovenous malformations. The RAS/MAPK variants identified in this study are known to be associated with malignant tumors for which targeted therapies have already been developed. Thus, identification of these somatic variants could lead to new therapeutic options to improve the management of patients with extracranial arteriovenous malformations.
Collapse
Affiliation(s)
- F N El Sissy
- Sorbonne Université, Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Department of Pathology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris, Faculty of Medicine, Paris, France
| | - M Wassef
- Department of Pathology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris, Faculty of Medicine, Paris, France
| | - B Faucon
- Department of Otorhinolaryngology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - D Salvan
- Department of Otorhinolaryngology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - S Nadaud
- Sorbonne Université, INSERM, UMR_S1166, Unité de recherche sur les maladies cardiovasculaires, ICAN, le métabolisme et la nutrition, Paris, France
| | - F Coulet
- Sorbonne Université, Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - H Adle-Biassette
- Department of Pathology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris, Faculty of Medicine, Paris, France
| | - F Soubrier
- Sorbonne Université, Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, INSERM, UMR_S1166, Unité de recherche sur les maladies cardiovasculaires, ICAN, le métabolisme et la nutrition, Paris, France
| | - A Bisdorff
- Department of Neuroradiology, Lariboisère Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - M Eyries
- Sorbonne Université, Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, INSERM, UMR_S1166, Unité de recherche sur les maladies cardiovasculaires, ICAN, le métabolisme et la nutrition, Paris, France
| |
Collapse
|
28
|
Sun Y, Fan X, Rao Y, Wang Z, Wang D, Yang X, Zheng L, Wen M, Cai R, Su L. Cell-free DNA from plasma as a promising alternative for detection of gene mutations in patients with Maffucci syndrome. Hereditas 2022; 159:4. [PMID: 35042566 PMCID: PMC8764769 DOI: 10.1186/s41065-022-00223-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/29/2021] [Indexed: 11/10/2022] Open
Abstract
Maffucci syndrome (MS, OMIM 166000) is an extremely unusual, nonhereditary, multisystemic disorder that is characterized with multiple enchondromas and vascular lesions, most of which are spindle cell hemangiomas. Complications of MS, such as bone deformities and dysfunction caused by enchondromas, usually increase during childhood and adolescence. Malignant transformation of enchondromas and other malignancies are the most severe complications. MS is caused by somatic mosaic IDH1/2 mutations, 65% of which are the IDH1 p.Arg132Cys variant. Due to its rarity, there is no international consensus for the most appropriate treatment option of MS. Here, we report a case of a female patient presenting with multiple enchondromas and spindle cell hemangiomas (SCHs) on bilateral hand and feet diagnosed as MS. A detailed clinical, pathological and genetic diagnosis of MS was rendered. Integrative Genomics Viewer (IGV) visualization of next-generation sequencing (NGS) data revealed the consistent detection of the low-frequency somatic IDH1 p.Arg132Cys mutation between SCH tissue and cystic blood-derived cfDNA. This is the first successful molecular diagnosis of MS complicated with SCH utilizing minimally invasive cfDNA techniques. We suggest that cfDNA sequencing could potentially be used as an alternative, reliable and sensitive method to identify molecular information for genetic diagnosis and for future targeted therapies of MS.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Yamin Rao
- Department of pathology, 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
| | - 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.
| | - Lixin 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.
| |
Collapse
|
29
|
Sheppard SE, Sanders VR, Srinivasan A, Finn LS, Adams D, Elton A, Amlie-Lefond C, Nelson Z, Dmyterko V, Jensen D, Zenner K, Perkins J, Bennett JT. Cerebrofacial vascular metameric syndrome is caused by somatic pathogenic variants in PIK3CA. Cold Spring Harb Mol Case Stud 2021; 7:mcs.a006147. [PMID: 34887309 PMCID: PMC8751408 DOI: 10.1101/mcs.a006147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/30/2021] [Indexed: 12/26/2022] Open
Abstract
Disorganized morphogenesis of arteries, veins, capillaries, and lymphatic vessels results in vascular malformations. Most individuals with isolated vascular malformations have postzygotic (mosaic), activating pathogenic variants in a handful of oncogenes within the PI3K–RAS–MAPK pathway (Padia et al., Laryngoscope Investig Otolaryngol 4: 170–173 [2019]). Activating pathogenic variants in the gene PIK3CA, which encodes for the catalytic subunit of phosphatidylinositol 3-kinase, are present in both lymphatic and venous malformations as well as arteriovenous malformations in other complex disorders such as CLOVES syndrome (congenital, lipomatous, overgrowth, vascular malformations, epidermal anevi, scoliosis) (Luks et al., Pediatr Dev Pathol 16: 51 [2013]; Luks et al., J Pediatr 166: 1048–1054.e1–5 [2015]; Al-Olabi et al., J Clin Invest 128: 1496–1508 [2018]). These vascular malformations are part of the PIK3CA-related overgrowth spectrum, a spectrum of entities that have regionalized disordered growth due to the presence of tissue-restricted postzygotic PIK3CA pathogenic variants (Keppler-Noreuil et al., Am J Med Genet A 167A: 287–295 [2015]). Cerebrofacial vascular metameric syndrome (CVMS; also described as cerebrofacial arteriovenous metameric syndrome, Bonnet–Dechaume–Blanc syndrome, and Wyburn–Mason syndrome) is the association of retinal, facial, and cerebral vascular malformations (Bhattacharya et al., Interv Neuroradiol 7: 5–17 [2001]; Krings et al., Neuroimaging Clin N Am 17: 245–258 [2007]). The segmental distribution, the presence of tissue overgrowth, and the absence of familial recurrence are all consistent with CVMS being caused by a postzygotic mutation, which has been hypothesized by previous authors (Brinjiki et al., Am J Neuroradiol 39: 2103–2107 [2018]). However, the genetic cause of CVMS has not yet been described. Here, we present three individuals with CVMS and mosaic activating pathogenic variants within the gene PIK3CA. We propose that CVMS be recognized as part of the PIK3CA-related overgrowth spectrum, providing justification for future trials using pharmacologic PIK3CA inhibitors (e.g., alpelisib) for these difficult-to-treat patients.
Collapse
Affiliation(s)
- Sarah E Sheppard
- Division of Human Genetics, Department of Pediatrics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Comprehensive Vascular Anomaly Program, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Victoria R Sanders
- Division of Human Genetics, Department of Pediatrics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Comprehensive Vascular Anomaly Program, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Abhay Srinivasan
- Comprehensive Vascular Anomaly Program, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Division of Interventional Radiology, Department of Radiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Laura S Finn
- Department of Pathology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Denise Adams
- Comprehensive Vascular Anomaly Program, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Andrew Elton
- University of Minnesota School of Medicine, Minneapolis, Minnesota 55455, USA
| | | | - Zoe Nelson
- Seattle Children's Hospital, Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA
| | - Victoria Dmyterko
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington 98101, USA
| | - Dana Jensen
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington 98101, USA
| | - Kaitlyn Zenner
- Seattle Children's Hospital, Division of Pediatric Otolaryngology, Department of Otolaryngology/Head and Neck Surgery, University of Washington, Seattle, Washington 98105, USA.,Vascular Anomalies Program, Seattle Children's Hospital, Seattle, Washington 98105, USA
| | - Jonathan Perkins
- Seattle Children's Hospital, Division of Pediatric Otolaryngology, Department of Otolaryngology/Head and Neck Surgery, University of Washington, Seattle, Washington 98105, USA.,Vascular Anomalies Program, Seattle Children's Hospital, Seattle, Washington 98105, USA
| | - James T Bennett
- Seattle Children's Hospital, Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA.,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington 98101, USA.,Vascular Anomalies Program, Seattle Children's Hospital, Seattle, Washington 98105, USA
| |
Collapse
|
30
|
Schimmel K, Ali MK, Tan SY, Teng J, Do HM, Steinberg GK, Stevenson DA, Spiekerkoetter E. Arteriovenous Malformations-Current Understanding of the Pathogenesis with Implications for Treatment. Int J Mol Sci 2021; 22:ijms22169037. [PMID: 34445743 PMCID: PMC8396465 DOI: 10.3390/ijms22169037] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/18/2022] Open
Abstract
Arteriovenous malformations are a vascular anomaly typically present at birth, characterized by an abnormal connection between an artery and a vein (bypassing the capillaries). These high flow lesions can vary in size and location. Therapeutic approaches are limited, and AVMs can cause significant morbidity and mortality. Here, we describe our current understanding of the pathogenesis of arteriovenous malformations based on preclinical and clinical findings. We discuss past and present accomplishments and challenges in the field and identify research gaps that need to be filled for the successful development of therapeutic strategies in the future.
Collapse
Affiliation(s)
- Katharina Schimmel
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (K.S.); (M.K.A.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA 94305, USA
| | - Md Khadem Ali
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (K.S.); (M.K.A.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA 94305, USA
| | - Serena Y. Tan
- Department of Pathology, Stanford University, Stanford, CA 94305, USA;
| | - Joyce Teng
- Department of Dermatology, Lucile Packard Children’s Hospital, Stanford University, Stanford, CA 94305, USA;
| | - Huy M. Do
- Department of Radiology (Neuroimaging and Neurointervention), Stanford University, Stanford, CA 94305, USA;
- Department of Neurosurgery and Stanford Stroke Center, Stanford University, Stanford, CA 94305, USA;
| | - Gary K. Steinberg
- Department of Neurosurgery and Stanford Stroke Center, Stanford University, Stanford, CA 94305, USA;
| | - David A. Stevenson
- Department of Pediatrics, Division of Medical Genetics, Stanford University, Stanford, CA 94305, USA;
| | - Edda Spiekerkoetter
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (K.S.); (M.K.A.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA 94305, USA
- Correspondence: ; Tel.: +1-(650)-739-5031
| |
Collapse
|
31
|
Abstract
Lymphatic vessels maintain tissue fluid homeostasis by returning to blood circulation interstitial fluid that has extravasated from the blood capillaries. They provide a trafficking route for cells of the immune system, thus critically contributing to immune surveillance. Developmental or functional defects in the lymphatic vessels, their obstruction or damage, lead to accumulation of fluid in tissues, resulting in lymphedema. Here we discuss developmental lymphatic anomalies called lymphatic malformations and complex lymphatic anomalies that manifest as localized or multifocal lesions of the lymphatic vasculature, respectively. They are rare diseases that are caused mostly by somatic mutations and can present with variable symptoms based upon the size and location of the lesions composed of fluid-filled cisterns or channels. Substantial progress has been made recently in understanding the molecular basis of their pathogenesis through the identification of their genetic causes, combined with the elucidation of the underlying mechanisms in animal disease models and patient-derived lymphatic endothelial cells. Most of the solitary somatic mutations that cause lymphatic malformations and complex lymphatic anomalies occur in genes that encode components of oncogenic growth factor signal transduction pathways. This has led to successful repurposing of some targeted cancer therapeutics to the treatment of lymphatic malformations and complex lymphatic anomalies. Apart from the mutations that act as lymphatic endothelial cell-autonomous drivers of these anomalies, current evidence points to superimposed paracrine mechanisms that critically contribute to disease pathogenesis and thus provide additional targets for therapeutic intervention. Here, we review these advances and discuss new treatment strategies that are based on the recently identified molecular pathways.
Collapse
Affiliation(s)
- Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Sweden (T.M.)
| | - Laurence M Boon
- Division of Plastic Surgery, Center for Vascular Anomalies, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium (L.M.B.).,Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium (L.M.B., M.V.)
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium (L.M.B., M.V.).,Walloon Excellence in Lifesciences and Biotechnology, University of Louvain, Brussels, Belgium (M.V.)
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum, University of Helsinki, Finland (K.A.)
| |
Collapse
|
32
|
Doan RN, Miller MB, Kim SN, Rodin RE, Ganz J, Bizzotto S, Morillo KS, Huang AY, Digumarthy R, Zemmel Z, Walsh CA. MIPP-Seq: ultra-sensitive rapid detection and validation of low-frequency mosaic mutations. BMC Med Genomics 2021; 14:47. [PMID: 33579278 PMCID: PMC7881461 DOI: 10.1186/s12920-021-00893-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/03/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Mosaic mutations contribute to numerous human disorders. As such, the identification and precise quantification of mosaic mutations is essential for a wide range of research applications, clinical diagnoses, and early detection of cancers. Currently, the low-throughput nature of single allele assays (e.g., allele-specific ddPCR) commonly used for genotyping known mutations at very low alternate allelic fractions (AAFs) have limited the integration of low-level mosaic analyses into clinical and research applications. The growing importance of mosaic mutations requires a more rapid, low-cost solution for mutation detection and validation. METHODS To overcome these limitations, we developed Multiple Independent Primer PCR Sequencing (MIPP-Seq) which combines the power of ultra-deep sequencing and truly independent assays. The accuracy of MIPP-seq to quantifiable detect and measure extremely low allelic fractions was assessed using a combination of SNVs, insertions, and deletions at known allelic fractions in blood and brain derived DNA samples. RESULTS The Independent amplicon analyses of MIPP-Seq markedly reduce the impact of allelic dropout, amplification bias, PCR-induced, and sequencing artifacts. Using low DNA inputs of either 25 ng or 50 ng of DNA, MIPP-Seq provides sensitive and quantitative assessments of AAFs as low as 0.025% for SNVs, insertion, and deletions. CONCLUSIONS MIPP-Seq provides an ultra-sensitive, low-cost approach for detecting and validating known and novel mutations in a highly scalable system with broad utility spanning both research and clinical diagnostic testing applications. The scalability of MIPP-Seq allows for multiplexing mutations and samples, which dramatically reduce costs of variant validation when compared to methods like ddPCR. By leveraging the power of individual analyses of multiple unique and independent reactions, MIPP-Seq can validate and precisely quantitate extremely low AAFs across multiple tissues and mutational categories including both indels and SNVs. Furthermore, using Illumina sequencing technology, MIPP-seq provides a robust method for accurate detection of novel mutations at an extremely low AAF.
Collapse
Affiliation(s)
- Ryan N Doan
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA.
- Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA.
| | - Michael B Miller
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sonia N Kim
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
- Program in Biological and Biomedical Sciences, Harvard University, Boston, MA, USA
| | - Rachel E Rodin
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
| | - Javier Ganz
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
| | - Sara Bizzotto
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
| | - Katherine S Morillo
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
| | - August Yue Huang
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
| | - Reethika Digumarthy
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
| | - Zachary Zemmel
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Center for Life Sciences 15062, 300 Longwood Avenue, BCH3150, Boston, MA, 02115, USA.
- Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA.
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|