1
|
Zhang H, Wang W, Zhou Q, Hou J, Ying W, Hui X, Sun J, Liu L, Liu L, Wang C, Zhang H, Sun B, Wang X. Characterization of the epidemiology, susceptibility genes and clinical features of viral infections among children with inborn immune errors: a retrospective study. Virol J 2025; 22:91. [PMID: 40176105 PMCID: PMC11963556 DOI: 10.1186/s12985-025-02697-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/06/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Although viral infections are one of the common clinical manifestations in patients with inborn errors of immunity (IEIs), little is known about the epidemiology, susceptibility genes, and clinical status of viral infections in patients with IEIs. METHODS The demographic information, clinical diagnoses, and laboratory findings of 931 IEI patients who underwent viral testing from January 2016 to December 2022 were collected and analyzed. RESULTS In total, 47.15% (439/931) patients with IEI tested positive for at least one virus during hospitalization. There were a total of 640 viral infections during the study period, mainly from EBV 131 (20.47%), HRV 102(15.94%), CMV 100(15.63%), and RV 84(13.13%). CMV and RV infections were more common in the combined immunodeficiencies (IEI_I) group during the infant stage, whereas EBV infection was more common in the immune dysregulation (IEI_IV) group during the preschool stage. Mutations in SH2D1A (57.14%), PIK3CD (56.41%) and LRBA (50%) make individuals susceptible to EBV infection; mutations in WAS (30%) make individuals susceptible to CMV infection; and mutations in IL2RG (56.52%) and RAG1 (37.5%) make individuals susceptible to RV infection. Joinpoint analysis revealed trends in viral positivity in different years. CONCLUSION These data suggest that it is possible to target the prevention, treatment, and management of IEI patients who are infected with a virus by accounting for the age at infection, type of IEI, and mutant genes, but special attention needs to be paid to viral infections in IEI_I and IEI_IV patients during the infant stage.
Collapse
Affiliation(s)
- Haiqiao Zhang
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Wenjie Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Qinhua Zhou
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jia Hou
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Wenjing Ying
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Xiaoying Hui
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jinqiao Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Lipin Liu
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Luyao Liu
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Chenhao Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Hai Zhang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Bijun Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China.
| | - Xiaochuan Wang
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China.
| |
Collapse
|
2
|
Toriello E, Maritato R, De Rosa A, Esposito MV, Damiano C, Rosano C, Cirillo E, Tarallo A, Abagnale C, Cillo F, Romano R, Grilli L, Comegna M, Blasio G, Parenti G, Surace EM, Castaldo G, Pignata C, Giardino G. Characterization of a WAS splice-site variant in a patient with Wiskott-Aldrich syndrome. Front Immunol 2025; 16:1517347. [PMID: 39917307 PMCID: PMC11798879 DOI: 10.3389/fimmu.2025.1517347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025] Open
Abstract
Wiskott-Aldrich syndrome (WAS) (MIM #301000) is a rare X-linked primary immunodeficiency due to mutations in the WAS gene, characterized by thrombocytopenia with small platelets, eczema, recurrent infections, and an increased incidence of autoimmunity and malignancies. A wide spectrum of mutations has been identified in the WAS gene responsible for a broad variety of clinical phenotypes. By using targeted next-generation sequencing (t-NGS), we identified in a 2-month-old boy with thrombocytopenia and immunological alterations a 4-nucleotide deletion from position +3 to +6 of intron 8 (c.777 + 3_777 + 6delGAGT) of WAS, currently classified on ClinVar as a variant of uncertain significance. The in-vitro characterization of the variant revealed the complete retention of intron 8 in the mature transcript, suggesting a splicing defect due to the loss of a splice donor site at the 5'-end of intron 8. By sequencing the polymerase chain reaction product, we identified a premature stop at codon 269; thus, consequently, no Wiskott-Aldrich syndrome protein (WASp) was detectable in peripheral blood mononuclear cells from the patient. Due to the total absence of a full-length WASp, it is expected that the patient will develop the severe form of the disease, although further monitoring is needed to better define his phenotype.
Collapse
Affiliation(s)
- Elisabetta Toriello
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Rosa Maritato
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Antonio De Rosa
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | | | - Carla Damiano
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Carmen Rosano
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Emilia Cirillo
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Antonietta Tarallo
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Cosimo Abagnale
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Francesca Cillo
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Roberta Romano
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Laura Grilli
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Marika Comegna
- CEINGE-Biotecnologie Avanzate Franco Salvatore S.c.a.r.l., Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University of Naples, Naples, Italy
| | - Giancarlo Blasio
- CEINGE-Biotecnologie Avanzate Franco Salvatore S.c.a.r.l., Naples, Italy
| | - Giancarlo Parenti
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Enrico Maria Surace
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie Avanzate Franco Salvatore S.c.a.r.l., Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University of Naples, Naples, Italy
| | - Claudio Pignata
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| | - Giuliana Giardino
- Department of Translational Medical Sciences, Pediatric Section, Federico II University of Naples, Naples, Italy
| |
Collapse
|
3
|
Hamanaka S, Uchiyama T, Kaname T, Matsui M, Yoshihashi H, Makimoto A, Yuza Y, Ishiguro A. X-linked Thrombocytopenia with Normal Wiskott-Aldrich Syndrome Protein Expression in Lymphocytes and a Novel Wiskott-Aldrich Syndrome Protein Gene Variant: A Case Report and Brief Review of the Literature. JOURNAL OF PEDIATRICS. CLINICAL PRACTICE 2024; 14:200128. [PMID: 39629200 PMCID: PMC11613185 DOI: 10.1016/j.jpedcp.2024.200128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024]
Abstract
We present a case of X-linked thrombocytopenia (XLT) with a novel WAS gene variant expressing a normal amount of Wiskott-Aldrich syndrome protein (WASp) in lymphocytes. XLT usually decreases WASp expression not only in platelets, but also in lymphocytes. However, there were cases, such as the present one, in which WASp was expressed normally in lymphocytes and absent only in platelets. Our finding suggests that it is of greater diagnostic sensitivity to perform an expression analysis of WASp in both platelets and lymphocytes when XLT is suspected.
Collapse
Affiliation(s)
- Serena Hamanaka
- Department of General Pediatrics, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Toru Uchiyama
- Department of Human Genetics, National Center for Child Health and Development, Tokyo, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Motohiro Matsui
- Department of Hematology/Oncology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Hiroshi Yoshihashi
- Department of Clinical Genetics, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Atsushi Makimoto
- Department of Hematology/Oncology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Yuki Yuza
- Department of Hematology/Oncology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Akira Ishiguro
- Center for Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
4
|
Sato D, Kirikae H, Nakano T, Katayama S, Yaoita H, Takayama J, Tamiya G, Kure S, Kikuchi A, Sasahara Y. Comprehensive genetic analysis for identification of monogenic disorders and selection of appropriate treatments in pediatric patients with persistent thrombocytopenia. Pediatr Hematol Oncol 2024; 41:541-556. [PMID: 39318204 DOI: 10.1080/08880018.2024.2395358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/27/2024] [Accepted: 08/18/2024] [Indexed: 09/26/2024]
Abstract
Persistent thrombocytopenia is caused by various diseases, including immune thrombocytopenia, inherited thrombocytopenia, and inherited bone marrow failure syndromes. Considering the large number of genes responsible for inherited disorders, comprehensive genetic analysis is required to diagnose monogenic disorders. In this study, we enrolled 53 pediatric patients with persistent thrombocytopenia exhibiting visually small or normal-sized platelets. We performed whole-exome sequencing, including 56 genes responsible for inherited thrombocytopenia, and evaluated clinical parameters according to disease type. Among 53 patients, 12 patients (22.6%) were diagnosed with monogenic disorders. Nine patients had a family history of thrombocytopenia. Pathogenic or novel variants of genes responsible for inherited thrombocytopenia were identified in three and six patients, respectively. The variants in genes for inherited thrombocytopenia with large or giant platelets were unexpectedly identified in six patients. Pathogenic variants in genes for inherited bone marrow failure syndromes with systemic features were identified in three patients with atypical symptoms. Since the definitive diagnostic methods for immune thrombocytopenia are limited, and a substantial number of patients with inherited thrombocytopenia are at a high risk of developing malignancies, comprehensive genetic analysis is indispensable for selecting appropriate therapies, avoidance of unnecessary treatments for immune thrombocytopenia, and long-term follow-up of patients with inherited thrombocytopenia.
Collapse
Affiliation(s)
- Daichi Sato
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hinako Kirikae
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Tomohiro Nakano
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Saori Katayama
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hisao Yaoita
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Jun Takayama
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of AI and Innovative Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Gen Tamiya
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of AI and Innovative Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Shigeo Kure
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Miyagi Children's Hospital, Miyagi, Japan
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yoji Sasahara
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
5
|
Fekrvand S, Abolhassani H, Esfahani ZH, Fard NNG, Amiri M, Salehi H, Almasi-Hashiani A, Saeedi-Boroujeni A, Fathi N, Mohtashami M, Razavi A, Heidari A, Azizi G, Khanmohammadi S, Ahangarzadeh M, Saleki K, Hassanpour G, Rezaei N, Yazdani R. Cancer Trends in Inborn Errors of Immunity: A Systematic Review and Meta-Analysis. J Clin Immunol 2024; 45:34. [PMID: 39466473 DOI: 10.1007/s10875-024-01810-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/16/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Patients with inborn errors of immunity (IEI) are susceptible to developing cancer due to defects in the immune system. The prevalence of cancer is higher in IEI patients compared to the immunocompetent population and cancers are considered as an important and common cause of death in IEI patients. OBJECTIVES To systematically review demographic, genetic and cancer-related data of IEI patients with a history of malignancy. Moreover, we performed a meta-analysis aiming to determine the frequency of cancer in patients with different types of IEI. METHODS We conducted electronic searches on Embase, Web of Science, PubMed, and Scopus (until September 2023) introducing terms related to IEI and cancer. Studies with human subjects with confirmed IEI who had developed at least one malignancy during their lifetime were included. RESULTS A total number of 4607 IEI patients with a cancer history were included in the present study. Common variable immunodeficiency (CVID) had the highest number of reported cases (1284 cases), mainly due to a higher relative proportion of patients with predominantly antibody deficiencies (PAD) and their increased life expectancy contributing to the higher detection and reporting of cancers among these patients. The most common malignancy was hematologic/blood cancers (3026 cases, mainly diffuse large B cell lymphoma). A total number of 1173 cases (55.6%) succumbed to cancer, with the highest rate of bone marrow failure (64.9%). Among the patients with monogenic defects in IEI-associated genes, the majority of cases had ATM deficiency (926 cases), but the highest cancer frequency rate belonged to NBS1 deficiency (50.5%). 1928 cases out of total 4607 eligible cases had detailed data to allow further statistical analysis that revealed BRCA2 deficiency had the earliest cancer development (~ 38 months), lowest cure frequency, and highest fatality rate (85%), while ATM deficiency had the lowest cure frequency and highest fatality rate (72%) among total cases reviewed with exclusion of Fanconi anemia. CONCLUSION The overall reported cancer frequency in the cases reviewed with and without exclusion of Fanconi anemia was 11.1% (95% confidence interval: 9.8-12.5%) and 12.0% (95% confidence interval: 10.6-13.5%), respectively. Our study revealed that the incidence of cancer is significantly dependent on the molecular and pathway defects in IEI patients, and individualized early screening and appropriate treatment, might improve the prognosis of these patients.
Collapse
Affiliation(s)
- Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Division of Clinical Immunology, Department of Biosciences and Nutrition, Karolinska Institutet, Karolinska University Hospital, Stockholm, Huddinge, Sweden
| | - Zahra Hamidi Esfahani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Najmeh Nameh Goshay Fard
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahboube Amiri
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Helia Salehi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Almasi-Hashiani
- Department of Epidemiology, School of Health, Arak University of Medical Sciences, Arak, Iran
| | - Ali Saeedi-Boroujeni
- Department of Basic Medical Sciences, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Nazanin Fathi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Mohtashami
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Azadehsadat Razavi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Arash Heidari
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gholamreza Azizi
- Noncommunicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shaghayegh Khanmohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Milad Ahangarzadeh
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Department of E-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of MedicalSciences (SBMU), Tehran, Iran
- USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Gholamreza Hassanpour
- Center for Research of Endemic Parasites of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
6
|
Vallée TC, Glasmacher JS, Buchner H, Arkwright PD, Behrends U, Bondarenko A, Browning MJ, Buchbinder D, Cattoni A, Chernyshova L, Ciznar P, Cole T, Czogała W, Dueckers G, Edgar JDM, Erbey F, Fasth A, Ferrua F, Formankova R, Gambineri E, Gennery AR, Goldman FD, Gonzalez-Granado LI, Heilmann C, Heiskanen-Kosma T, Juntti H, Kainulainen L, Kanegane H, Karaca NE, Kilic SS, Klein C, Kołtan S, Kondratenko I, Meyts I, Nasrullayeva GM, Notarangelo LD, Pasic S, Pellier I, Pignata C, Misbah S, Schulz A, Segundo GR, Shcherbina A, Slatter M, Sokolic R, Soler-Palacin P, Stepensky P, van Montfrans JM, Ryhänen S, Wolska-Kuśnierz B, Ziegler JB, Zhao X, Aiuti A, Ochs HD, Albert MH. Wiskott-Aldrich syndrome: a study of 577 patients defines the genotype as a biomarker for disease severity and survival. Blood 2024; 143:2504-2516. [PMID: 38579284 DOI: 10.1182/blood.2023021411] [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: 07/10/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 04/07/2024] Open
Abstract
ABSTRACT Wiskott-Aldrich syndrome (WAS) is a multifaceted monogenic disorder with a broad disease spectrum and variable disease severity and a variety of treatment options including allogeneic hematopoietic stem cell transplantation (HSCT) and gene therapy (GT). No reliable biomarker exists to predict disease course and outcome for individual patients. A total of 577 patients with a WAS variant from 26 countries and a median follow-up of 8.9 years (range, 0.3-71.1), totaling 6118 patient-years, were included in this international retrospective study. Overall survival (OS) of the cohort (censored at HSCT or GT) was 82% (95% confidence interval, 78-87) at age 15 years and 70% (61-80) at 30 years. The type of variant was predictive of outcome: patients with a missense variant in exons 1 or 2 or with the intronic hot spot variant c.559+5G>A (class I variants) had a 15-year OS of 93% (89-98) and a 30-year OS of 91% (86-97), compared with 71% (62-81) and 48% (34-68) in patients with any other variant (class II; P < .0001). The cumulative incidence rates of disease-related complications such as severe bleeding (P = .007), life-threatening infection (P < .0001), and autoimmunity (P = .004) occurred significantly later in patients with a class I variant. The cumulative incidence of malignancy (P = .6) was not different between classes I and II. It confirms the spectrum of disease severity and quantifies the risk for specific disease-related complications. The class of the variant is a biomarker to predict the outcome for patients with WAS.
Collapse
Affiliation(s)
- Tanja C Vallée
- Pediatric Hematology/Oncology, Dr von Hauner University Children's Hospital, Munich, Germany
| | - Jannik S Glasmacher
- Pediatric Hematology/Oncology, Dr von Hauner University Children's Hospital, Munich, Germany
| | | | - Peter D Arkwright
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester & Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Uta Behrends
- Children's Hospital, School of Medicine, Technical University Munich, Munich, Germany
| | - Anastasia Bondarenko
- Department of Pediatrics, Immunology, Infectious and Rare Diseases and Allergology, European Medical School, International European University, Kyiv, Ukraine
| | - Michael J Browning
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - David Buchbinder
- Department of Hematology, Children's Hospital of Orange County, Orange, CA
| | - Alessandro Cattoni
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Liudmyla Chernyshova
- Department of Pediatrics, Pediatric Infectious Diseases, Immunology and Allergology, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - Peter Ciznar
- Department of Pediatrics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Theresa Cole
- Department of Allergy and Immunology, The Royal Children's Hospital, Melbourne, Australia
| | - Wojciech Czogała
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Gregor Dueckers
- Helios Kliniken Krefeld, Children's Hospital, Krefeld, Germany
| | - John David M Edgar
- St James's Hospital & School of Medicine, Trinity College, Dublin, Ireland
| | - Fatih Erbey
- Department of Pediatric Hematology/Oncology, Koç University School of Medicine, İstanbul, Turkey
| | - Anders Fasth
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Francesca Ferrua
- Pediatric Immunohematology and Stem Cell Program, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Renata Formankova
- Department of Pediatric Hematology and Oncology, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Eleonora Gambineri
- Department of NEUROFARBA, Section of Child's Health, University of Florence, Florence, Italy
- Department of Haematology-Oncology, Anna Meyer University Children's Hospital (AOU Meyer IRCCS), Florence, Italy
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, and Paediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Frederick D Goldman
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL
| | - Luis I Gonzalez-Granado
- Department of Pediatrics, Primary Immunodeficiencies Unit, Research Institute, Hospital 12 Octubre, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Carsten Heilmann
- Department for Children and Adolescents, Pediatric Hematopoietic Stem Cell Transplantation and Immunodeficiency, Copenhagen University Hospital Rigshospitalet, København, Denmark
| | | | - Hanna Juntti
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital and Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Leena Kainulainen
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Neslihan E Karaca
- Division of Pediatric Immunology, Department of Pediatrics, Ege University, The Medical School, Izmir, Turkey
| | - Sara S Kilic
- Pediatric Immunology and Rheumatology, Bursa Uludag University School of Medicine, Bursa, Turkey
| | - Christoph Klein
- Pediatric Hematology/Oncology, Dr von Hauner University Children's Hospital, Munich, Germany
| | - Sylwia Kołtan
- Department of Paediatrics, Haematology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Irina Kondratenko
- Russian Children's Clinical Hospital, Pirogov National Research Medical University, Moscow, Russia
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Srdjan Pasic
- Department of Immunology, Mother and Child Health Care Institute of Serbia, Belgrade, Serbia
| | - Isabelle Pellier
- Centre de référence des déficits immunitaires primitifs CEREDIH, CHU d'Angers, Angers, France
| | - Claudio Pignata
- Department of Translational Medical Science, Section of Pediatrics, Federico II University, Napoli, Italy
| | - Siraj Misbah
- Clinical Immunology, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Gesmar R Segundo
- Allergy and Immunology Division, Pediatrics Department, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Anna Shcherbina
- Dmitry Rogachev National Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Mary Slatter
- Translational and Clinical Research Institute, Newcastle University, and Paediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Robert Sokolic
- Hematologic Malignancies Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| | - Pere Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Children's Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Polina Stepensky
- Bone Marrow Transplantation Department, Hadassah-Hebrew, University Medical Center, Jerusalem, Israel
| | - Joris M van Montfrans
- Department of Pediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Samppa Ryhänen
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Pediatric Research Center, Helsinki, Finland
| | | | - John B Ziegler
- School of Women's & Children's Health, University of New South Wales, Sydney, Australia
| | - Xiaodong Zhao
- Department of Rheumatism and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Aiuti
- Pediatric Immunohematology and Stem Cell Program, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Hans D Ochs
- University of Washington School of Medicine, Seattle, WA
| | - Michael H Albert
- Pediatric Hematology/Oncology, Dr von Hauner University Children's Hospital, Munich, Germany
| |
Collapse
|
7
|
Anantharachagan A, Loh SY, Burns SO, Laurence A, Tadros S, Tholouli E, Lwin Y, Martinez-Calle N, Vaitla P, Morris EC. Allogeneic hematopoietic stem cell transplantation outcome in oldest known surviving patients with Wiskott-Aldrich syndrome. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100191. [PMID: 38187865 PMCID: PMC10770606 DOI: 10.1016/j.jacig.2023.100191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 01/09/2024]
Abstract
Regardless of their age, adult patients with Wiskott-Aldrich syndrome should be considered for hematopoietic stem cell transplantation if clinically indicated.
Collapse
Affiliation(s)
- Ariharan Anantharachagan
- Department of Allergy and Clinical Immunology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom
- Department of Immunology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom
| | - Sook Yin Loh
- Department of Allergy and Clinical Immunology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom
| | - Siobhan O. Burns
- University College London Institute of Immunity and Transplantation, London, United Kingdom
- Department of Immunology, The Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Arian Laurence
- University College London Institute of Immunity and Transplantation, London, United Kingdom
- Department of Immunology, The Royal Free London NHS Foundation Trust, London, United Kingdom
- Department of Clinical Haematology, University College London NHS Foundation Trust, London, United Kingdom
| | - Susan Tadros
- Department of Immunology, The Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Eleni Tholouli
- Manchester University NHS Foundation Trust, Department of Haematology, Manchester, United Kingdom
| | - Yadanar Lwin
- Department of Haematology, Nottingham, United Kingdom
| | | | - P. Vaitla
- Department of Immunology Nottingham University Hospitals, NHS Trust, Nottingham, United Kingdom
| | - Emma C. Morris
- University College London Institute of Immunity and Transplantation, London, United Kingdom
- Department of Immunology, The Royal Free London NHS Foundation Trust, London, United Kingdom
- Department of Clinical Haematology, University College London NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
8
|
Sun Y, Song X, Pan H, Li X, Sun L, Song L, Ma F, Hao J. Wiskott-Aldrich syndrome: A new synonym mutation in the WAS gene. Intractable Rare Dis Res 2024; 13:69-72. [PMID: 38404734 PMCID: PMC10883844 DOI: 10.5582/irdr.2023.01102] [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: 11/07/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/27/2024] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is a rare X-linked recessive primary immunodeficiency disorder. Mutations in the WAS gene are considered to be the primary cause of WAS. In this work, we report a boy who presented with intracranial hemorrhage (ICH) as an initial symptom and detects a novel pathogenic synonymous mutation in his WAS gene. His mother was a carrier of the mutant gene. The mutation, located at position c.273 (c.273 G>A) in exon 2, is a synonym mutation and predicted to affect protein expression by disrupting gene splicing. This study summarizes the diagnosis and treatment process of the patient and expands the genetic spectrum of WAS.
Collapse
Affiliation(s)
- Yuxin Sun
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomin Song
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Hua Pan
- Department of Pediatric Intensive Care Unit, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lirong Sun
- Department of Pediatric Hematology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liang Song
- Department of Emergency Paediatrics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fei Ma
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junnan Hao
- Department of Emergency Paediatrics, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
R P, Shanmugam G, Rakshit S, Sarkar K. Role of Wiskott Aldrich syndrome protein in haematological malignancies: genetics, molecular mechanisms and therapeutic strategies. Pathol Res Pract 2024; 253:155026. [PMID: 38118219 DOI: 10.1016/j.prp.2023.155026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/22/2023]
Abstract
As patients continue to suffer from lymphoproliferative and myeloproliferative diseases known as haematopoietic malignancies can affect the bone marrow, blood, lymph nodes, and lymphatic and non-lymphatic organs. Despite advances in the current treatment, there is still a significant challenge for physicians to improve the therapy of HMs. WASp is an important regulator of actin polymerization and the involvement of WASp in transcription is thought to be linked to the DNA damage response and repair. In some studies, severe immunodeficiency and lymphoid malignancy are caused by WASp mutations or the absence of WASp and these mutations in WAS can alter the function and/or expression of the intracellular protein. Loss-of-function and Gain-of-function mutations in WASp have an impact on cancer malignancies' incidence and onset. Recent studies suggest that depending on the clinical or experimental situation, WASPs and WAVEs can operate as a suppressor or enhancers for cancer malignancy. These dual functions of WASPs and WAVEs in cancer likely arose from their multifaceted role in cells that could be targeted for anticancer drug development. The significant role and their association of WASp in Chronic myeloid leukaemia, Juvenile myelomonocytic leukaemia and T-cell lymphoma is discussed. In this review, we described the structure and function of WASp and its family mechanism, analysing major regulatory effectors and summarising the clinical relevance and drugs that specifically target WASp in disease treatment in various hematopoietic malignancies by different approaches.
Collapse
Affiliation(s)
- Pradeep R
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Geetha Shanmugam
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Sudeshna Rakshit
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India.
| |
Collapse
|
10
|
Chuong HQ, Xinh PT, Tram DB, Ha NTT, Nguyen TM, Anh PNL, Van ND, Anh NHM, Dung PC, Nghia H, Vu HA. Spectrum of WAS gene mutations in Vietnamese patients with Wiskott-Aldrich syndrome. Pediatr Int 2024; 66:e15770. [PMID: 38641933 DOI: 10.1111/ped.15770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 04/21/2024]
Abstract
BACKGROUND WAS gene mutational analysis is crucial to establish a definite diagnosis of Wiskott-Aldrich syndrome (WAS). Data on the genetic background of WAS in Vietnamese patients have not been reported. METHODS We recruited 97 male, unrelated patients with WAS and analyzed WAS gene mutation using Sanger sequencing technology. RESULTS We identified 36 distinct hemizygous pathogenic mutations, with 17 novel variants, from 38 patients in the entire cohort (39.2%). The mutational spectrum included 14 missense, 12 indel, five nonsense, four splicing, and one non-stop mutations. Most mutations appear only once, with the exception of c.37C>T (p.R13X) and c.374G>A (p.G125E) each of which occurs twice in unrelated patients. CONCLUSION Our data enrich the mutational spectrum of the WAS gene and are crucial for understanding the genetic background of WAS and for supporting genetic counseling.
Collapse
Affiliation(s)
- Ho Quoc Chuong
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Phan Thi Xinh
- Department of Hematology, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| | - Duong Bich Tram
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Ho Chi Minh City Open University, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Thanh Ha
- Department of Molecular Biology, Dai Phuoc Clinic, Ho Chi Minh City, Vietnam
| | - Tuan Minh Nguyen
- Department of Hematology, Children's Hospital 1, Ho Chi Minh City, Vietnam
| | | | - Nguyen Dinh Van
- Department of Oncology and Hematology, Children's Hospital 2, Ho Chi Minh City, Vietnam
| | | | - Phu Chi Dung
- Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| | - Huynh Nghia
- Department of Hematology, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| | - Hoang Anh Vu
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| |
Collapse
|
11
|
Labrosse R, Chu JI, Armant MA, Everett JK, Pellin D, Kareddy N, Frelinger AL, Henderson LA, O’Connell AE, Biswas A, Coenen-van der Spek J, Miggelbrink A, Fiorini C, Adhikari H, Berry CC, Cantu VA, Fong J, Jaroslavsky J, Karadeniz DF, Li QZ, Reddy S, Roche AM, Zhu C, Whangbo JS, Dansereau C, Mackinnon B, Morris E, Koo SM, London WB, Baris S, Ozen A, Karakoc-Aydiner E, Despotovic JM, Forbes Satter LR, Saitoh A, Aizawa Y, King A, Nguyen MAT, Vu VDU, Snapper SB, Galy A, Notarangelo LD, Bushman FD, Williams DA, Pai SY. Outcomes of hematopoietic stem cell gene therapy for Wiskott-Aldrich syndrome. Blood 2023; 142:1281-1296. [PMID: 37478401 PMCID: PMC10731922 DOI: 10.1182/blood.2022019117] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 06/16/2023] [Accepted: 06/17/2023] [Indexed: 07/23/2023] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is a rare X-linked disorder characterized by combined immunodeficiency, eczema, microthrombocytopenia, autoimmunity, and lymphoid malignancies. Gene therapy (GT) to modify autologous CD34+ cells is an emerging alternative treatment with advantages over standard allogeneic hematopoietic stem cell transplantation for patients who lack well-matched donors, avoiding graft-versus-host-disease. We report the outcomes of a phase 1/2 clinical trial in which 5 patients with severe WAS underwent GT using a self-inactivating lentiviral vector expressing the human WAS complementary DNA under the control of a 1.6-kB fragment of the autologous promoter after busulfan and fludarabine conditioning. All patients were alive and well with sustained multilineage vector gene marking (median follow-up: 7.6 years). Clinical improvement of eczema, infections, and bleeding diathesis was universal. Immune function was consistently improved despite subphysiologic levels of transgenic WAS protein expression. Improvements in platelet count and cytoskeletal function in myeloid cells were most prominent in patients with high vector copy number in the transduced product. Two patients with a history of autoimmunity had flares of autoimmunity after GT, despite similar percentages of WAS protein-expressing cells and gene marking to those without autoimmunity. Patients with flares of autoimmunity demonstrated poor numerical recovery of T cells and regulatory T cells (Tregs), interleukin-10-producing regulatory B cells (Bregs), and transitional B cells. Thus, recovery of the Breg compartment, along with Tregs appears to be protective against development of autoimmunity after GT. These results indicate that clinical and laboratory manifestations of WAS are improved with GT with an acceptable safety profile. This trial is registered at clinicaltrials.gov as #NCT01410825.
Collapse
Affiliation(s)
- Roxane Labrosse
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Division of Allergy and Immunology, Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montreal, QC, Canada
| | - Julia I. Chu
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
- Division of Pediatric Allergy, Immunology and Bone Marrow Transplantation, Benioff Children’s Hospital, University of California San Francisco, San Francisco, CA
| | - Myriam A. Armant
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - John K. Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Danilo Pellin
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Niharika Kareddy
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Andrew L. Frelinger
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | | | - Amy E. O’Connell
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
| | - Amlan Biswas
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Jet Coenen-van der Spek
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Alexandra Miggelbrink
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Claudia Fiorini
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Hriju Adhikari
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Charles C. Berry
- Division of Biostatistics and Bioinformatics, Herbert Wertheim School of Public Health, UC San Diego, La Jolla, CA
| | - Vito Adrian Cantu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Johnson Fong
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Jason Jaroslavsky
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Derin F. Karadeniz
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Quan-Zhen Li
- Department of Immunology, Microarray and Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, Dallas, TX
| | - Shantan Reddy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Aoife M. Roche
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Chengsong Zhu
- Department of Immunology, Microarray and Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jennifer S. Whangbo
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Colleen Dansereau
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Brenda Mackinnon
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Emily Morris
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Stephanie M. Koo
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Wendy B. London
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Safa Baris
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University, Istanbul, Turkey
- The Işıl Berat Barlan Center for Translational Medicine, Marmara University, Istanbul, Turkey
| | - Ahmet Ozen
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University, Istanbul, Turkey
- The Işıl Berat Barlan Center for Translational Medicine, Marmara University, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Department of Pediatrics, Division of Allergy and Immunology, Marmara University, Istanbul, Turkey
- The Işıl Berat Barlan Center for Translational Medicine, Marmara University, Istanbul, Turkey
| | - Jenny M. Despotovic
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | - Lisa R. Forbes Satter
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | - Akihiko Saitoh
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuta Aizawa
- Department of Pediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Alejandra King
- Hospital Luis Calvo Mackenna, Clínica Alemana De Santiago Universidad del Desarrollo, Santiago, Chile
| | - Mai Anh Thi Nguyen
- Department of Pediatrics, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Vy Do Uyen Vu
- Department of Pediatrics, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Scott B. Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Anne Galy
- Genethon, Évry, France
- University of Paris-Saclay, University of Évry, INSERM, Genethon, Integrare Research Unit UMR_S951, Évry, France
| | - Luigi D. Notarangelo
- Division of Immunology, Boston Children’s Hospital, Boston, MA
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Frederic D. Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David A. Williams
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Sung-Yun Pai
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
12
|
Bargehr C, Knöfler R, Streif W. Treatment of Inherited Platelet Disorders: Current Status and Future Options. Hamostaseologie 2023; 43:261-270. [PMID: 37611608 DOI: 10.1055/a-2080-6602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Inherited platelet disorders (IPDs) comprise a heterogeneous group of entities that manifest with variable bleeding tendencies. For successful treatment, the underlying platelet disorder, bleeding severity and location, age, and sex must be considered in the broader clinical context. Previous information from the AWMF S2K guideline #086-004 (www.awmf.org) is evaluated for validity and supplemented by information of new available and future treatment options and clinical scenarios that need specific measures. Special attention is given to the treatment of menorrhagia and risk management during pregnancy in women with IPDs. Established treatment options of IPDs include local hemostatic treatment, tranexamic acid, desmopressin, platelet concentrates, and recombinant activated factor VII. Hematopoietic stem cell therapy is a curative approach for selected patients. We also provide an outlook on promising new therapies. These include autologous hematopoietic stem cell gene therapy, artificial platelets and nanoparticles, and various other procoagulant treatments that are currently tested in clinical trials in the context of hemophilia.
Collapse
Affiliation(s)
- Caroline Bargehr
- Department of Paediatrics 1, Medical University of Innsbruck, Innsbruck, Austria
| | - Ralf Knöfler
- Department of Paediatric Haemostaseology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Werner Streif
- Department of Paediatrics 1, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
13
|
Espejo AG, Dols ST, Gestal MC. Síndrome de Wiskott-Aldrich en España: incidencia, mortalidad y sesgo de género durante 21 años. Rev Clin Esp 2023; 223:262-269. [PMID: 37929276 PMCID: PMC10621733 DOI: 10.1016/j.rce.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Antecedentes El síndrome de Wiskott-Aldrich (SWA) es un raro trastorno ligado al cromosoma X que se considera que afecta predominantemente a varones. Objetivo El objetivo de este estudio consistía en investigar la incidencia y la mortalidad intrahospitalaria del SWA en España, así como el sesgo de género. Métodos Se llevó a cabo un estudio epidemiológico retrospectivo poblacional en 97 pacientes con SWA diagnosticados en hospitales españoles entre 1997 y 2017, utilizando para ello datos del Sistema Nacional de Vigilancia de Datos Hospitalarios. Resultados Nuestros resultados revelaron que la incidencia anual media del SAW en España fue de 1,1 caso por cada 10 millones de habitantes (IC del 95 %, 0,45-2,33). El riesgo relativo fue mayor en los varones que en las mujeres (2,42). El diagnóstico de SWA se establece a una edad más avanzada en las mujeres (mediana de 47 años) que en los varones (mediana de 5,5 años). Únicamente los varones ingresaron en el hospital en al menos 10 ocasiones diferentes y todas las muertes se detectaron en varones. La tasa de mortalidad intrahospitalaria fue del 9,28 % en el SAW y la mayoría de las muertes se asociaron a hemorragia cerebral o infección. Conclusiones El SWA, una enfermedad rara, se diagnostica a una edad más avanzada en las mujeres y la mortalidad se observó exclusivamente en varones, asociada en la mayoría de los casos a hemorragia cerebral e infección.
Collapse
Affiliation(s)
- Antonio Guerrero Espejo
- Grupo de Investigación de Enfermedades Infecciosas, Facultad de Medicina y Odontología, Universidad Católica de Valencia “San Vicente Mártir”, Valencia, España
| | - Sofía Tomás Dols
- Grupo de Investigación de Enfermedades Infecciosas, Facultad de Medicina y Odontología, Universidad Católica de Valencia “San Vicente Mártir”, Valencia, España
| | - Mónica C. Gestal
- Servicio de Microbiología e Inmunología. LSU Health, 71103, Shreveport, LA, Estados Unidos
| |
Collapse
|
14
|
Hsu AP. Not too little, not too much: the impact of mutation types in Wiskott-Aldrich syndrome and RAC2 patients. Clin Exp Immunol 2023; 212:137-146. [PMID: 36617178 PMCID: PMC10128166 DOI: 10.1093/cei/uxad001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/23/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Primary immune deficiencies (PIDs) are genetic disorders impacting the appropriate development or functioning of any portion of the immune system. The broad adoption of high-throughput sequencing has driven discovery of new genes as well as expanded phenotypes associated with known genes. Beginning with the identification of WAS mutations in patients with severe Wiskott-Aldrich Syndrome, recognition of WAS mutations in additional patients has revealed phenotypes including isolated thrombocytopenia and X-linked neutropenia. Likewise RAC2 patients present with vastly different phenotypes depending on the mutation-ranging from reticular dysgenesis or severe neutrophil dysfunction with neonatal presentation to later onset common variable immune deficiency. This review examines genotype-phenotype correlations in patients with WAS (Wiskott-Aldrich Syndrome) and RAC2 mutations, highlighting functional protein domains, how mutations alter protein interactions, and how specific mutations can affect isolated functions of the protein leading to disparate phenotypes.
Collapse
Affiliation(s)
- Amy P Hsu
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Infections in Inborn Errors of Immunity with Combined Immune Deficiency: A Review. Pathogens 2023; 12:pathogens12020272. [PMID: 36839544 PMCID: PMC9958715 DOI: 10.3390/pathogens12020272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/13/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Enhanced susceptibility to microbes, often resulting in severe, intractable and frequent infections due to usually innocuous organisms at uncommon sites, is the most striking feature in individuals with an inborn error of immunity. In this narrative review, based on the International Union of Immunological Societies' 2022 (IUIS 2022) Update on phenotypic classification of human inborn errors of immunity, the focus is on commonly encountered Combined Immunodeficiency Disorders (CIDs) with susceptibility to infections. Combined immune deficiency disorders are usually commensurate with survival beyond infancy unlike Severe Combined Immune Deficiency (SCID) and are often associated with clinical features of a syndromic nature. Defective humoral and cellular immune responses result in susceptibility to a broad range of microbial infections. Although disease onset is usually in early childhood, mild defects may present in late childhood or even in adulthood. A precise diagnosis is imperative not only for determining management strategies, but also for providing accurate genetic counseling, including prenatal diagnosis, and also in deciding empiric treatment of infections upfront before investigation reports are available.
Collapse
|
16
|
Ji X, Hou X, Guo X, Sun Y, Ma F, Hao J. Identification of a novel WAS mutation and the non-splicing effect of a second-site mutation in a Chinese pedigree with Wiskott-Aldrich syndrome. Orphanet J Rare Dis 2022; 17:447. [PMID: 36550574 PMCID: PMC9783790 DOI: 10.1186/s13023-022-02589-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Wiskott-Aldrich syndrome (WAS) is a rare X-linked immunodeficiency disorder caused by abnormal expression of the WAS protein (WASp) due to mutations in the WAS gene, and is generally characterized by microthrombocytopenia, eczema, recurrent infections, and high susceptibility to autoimmune complications and hematological malignancies. RESULTS Herein, we identified a novel WAS mutation (c.158 T > C) using next-generation sequencing in a Chinese pedigree with WAS. The expression of WASp in the patients and their families was detected by flow cytometry and western blot analysis. To explore the exon-splicing effect of intron mutations and the correlation between the genotype and clinical phenotype, four groups of wild-type (WT), exon mutant, intron mutant, and combined mutant recombinant plasmids were transfected into COS-7 cells in vitro. The proband showed dramatically decreased WASp expression, while the female carriers showed a slightly lower level of WASp. The expression of products in the mutant and WT recombinant plasmids was detected by real-time fluorescence quantitative polymerase chain reaction (PCR), which showed a significant reduction in the combined mutant group than in the WT, exon mutant, and intron mutant groups. The length of the expression products in the four groups showed no differences, each containing 360 base pairs. Sequence analysis confirmed that the c.158 T > C mutation appeared in the exon mutant and combined mutant groups, whereas the intron variant c.273 + 14C > T caused no other sequence changes. CONCLUSION This study confirmed that the intron mutation did not affect the splicing of exons and excluded the influence of the double mutations at the transcription level on the severe clinical manifestations in the cousin. This in vitro study provided new insights into the pathogenesis of intronic mutations in WAS.
Collapse
Affiliation(s)
- Xin Ji
- grid.452702.60000 0004 1804 3009Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, 215#, Heping West Road, Shijiazhuang, Hebei Province, 050000 China
| | - Xuening Hou
- grid.452702.60000 0004 1804 3009Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, 215#, Heping West Road, Shijiazhuang, Hebei Province, 050000 China
| | - Xin Guo
- grid.452702.60000 0004 1804 3009Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, 215#, Heping West Road, Shijiazhuang, Hebei Province, 050000 China
| | - Yifeng Sun
- grid.452702.60000 0004 1804 3009Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, 215#, Heping West Road, Shijiazhuang, Hebei Province, 050000 China
| | - Futian Ma
- grid.470210.0Department of Hematology and Oncology, Children’s Hospital of Hebei Province, 133#, Jianhua South Street, Shijiazhuang, Hebei Province, 050031 China
| | - Jihong Hao
- grid.452702.60000 0004 1804 3009Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, 215#, Heping West Road, Shijiazhuang, Hebei Province, 050000 China
| |
Collapse
|
17
|
Abdulla M, Mohammed N. A Review on Inflammatory Bowel Diseases: Recent Molecular Pathophysiology Advances. Biologics 2022; 16:129-140. [PMID: 36118798 PMCID: PMC9481278 DOI: 10.2147/btt.s380027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/27/2022] [Indexed: 11/24/2022]
Abstract
Inflammatory bowel diseases are considered immune disorders with a complex genetic architecture involving constantly changing endogenous and exogenous factors. The rapid evolution of genomic technologies and the emergence of newly discovered molecular actors are compelling the research community to reevaluate the knowledge and molecular processes. The human intestinal tract contains intestinal human microbiota consisting of commensal, pathogenic, and symbiotic strains leading to immune responses that can contribute and lead to both systemic and intestinal disorders including IBD. In this review, we attempted to highlight some updates of the new IBD features related to genomics, microbiota, new emerging therapies and some major established IBD risk factors.
Collapse
Affiliation(s)
- Maheeba Abdulla
- Internal Medicine Department, Ibn AlNafees Hospital, Arabian Gulf University, Manama, Bahrain
- Correspondence: Maheeba Abdulla, Consultant Gastroenterologist, Internal Medicine Department, Ibn AlNafees Hospital, Arabian Gulf University, Manama, Bahrain, Email
| | | |
Collapse
|
18
|
Naseem A, Steinberg Z, Cavazza A. Genome editing for primary immunodeficiencies: A therapeutic perspective on Wiskott-Aldrich syndrome. Front Immunol 2022; 13:966084. [PMID: 36059471 PMCID: PMC9433875 DOI: 10.3389/fimmu.2022.966084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Primary immunodeficiency diseases (PIDs) are a group of rare inherited disorders affecting the immune system that can be conventionally treated with allogeneic hematopoietic stem cell transplantation and with experimental autologous gene therapy. With both approaches still facing important challenges, gene editing has recently emerged as a potential valuable alternative for the treatment of genetic disorders and within a relatively short period from its initial development, has already entered some landmark clinical trials aimed at tackling several life-threatening diseases. In this review, we discuss the progress made towards the development of gene editing-based therapeutic strategies for PIDs with a special focus on Wiskott - Aldrich syndrome and outline their main challenges as well as future directions with respect to already established treatments.
Collapse
|
19
|
Conditioning regimens for inborn errors of immunity: current perspectives and future strategies. Int J Hematol 2022; 116:7-15. [PMID: 35675025 DOI: 10.1007/s12185-022-03389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 10/18/2022]
Abstract
Inborn errors of immunity (IEI) are caused by germline genetic mutations, resulting in defects of innate or acquired immunity. Hematopoietic cell transplantation (HCT) is indicated for curative therapy especially in patients with IEI who develop fatal opportunistic infections or severe manifestations of immune dysregulation. The first successful HCT for severe combined immunodeficiency (SCID) was reported in 1968. Since then, the indications for HCT have expanded from SCID to various non-SCID IEI. In general, HCT for IEI differs from that for other hematological malignancies in that the goal is not to eradicate certain immune cells but to achieve immune reconstitution. European Society for Blood and Marrow Transplantation/European Society for Immunodeficiencies guidelines recommend reduced-intensity conditioning to avoid treatment-related toxicity, and the optimal conditioning regimen should be considered for each IEI. We review conditioning regimens for some representative IEI disorders in Japanese and worldwide cohort studies, and future strategies for treating IEI.
Collapse
|
20
|
WASp modulates RPA function on single-stranded DNA in response to replication stress and DNA damage. Nat Commun 2022; 13:3743. [PMID: 35768435 PMCID: PMC9243104 DOI: 10.1038/s41467-022-31415-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 06/08/2022] [Indexed: 02/07/2023] Open
Abstract
Perturbation in the replication-stress response (RSR) and DNA-damage response (DDR) causes genomic instability. Genomic instability occurs in Wiskott-Aldrich syndrome (WAS), a primary immunodeficiency disorder, yet the mechanism remains largely uncharacterized. Replication protein A (RPA), a single-strand DNA (ssDNA) binding protein, has key roles in the RSR and DDR. Here we show that human WAS-protein (WASp) modulates RPA functions at perturbed replication forks (RFs). Following genotoxic insult, WASp accumulates at RFs, associates with RPA, and promotes RPA:ssDNA complexation. WASp deficiency in human lymphocytes destabilizes RPA:ssDNA-complexes, impairs accumulation of RPA, ATR, ETAA1, and TOPBP1 at genotoxin-perturbed RFs, decreases CHK1 activation, and provokes global RF dysfunction. las17 (yeast WAS-homolog)-deficient S. cerevisiae also show decreased ScRPA accumulation at perturbed RFs, impaired DNA recombination, and increased frequency of DNA double-strand break (DSB)-induced single-strand annealing (SSA). Consequently, WASp (or Las17)-deficient cells show increased frequency of DSBs upon genotoxic insult. Our study reveals an evolutionarily conserved, essential role of WASp in the DNA stress-resolution pathway, such that WASp deficiency provokes RPA dysfunction-coupled genomic instability. Cancer develops in Wiskott-Aldrich syndrome (WAS). Here the authors identify a role for WAS-protein (WASp) in the DNA stress-resolution pathway by promoting the function of Replication Protein A at replication forks after DNA damage.
Collapse
|
21
|
Abstract
The new techniques of genetic analysis have made it possible to identify many new forms of inherited thrombocytopenias (IT) and study large series of patients. In recent years, this has changed the view of IT, highlighting the fact that, in contrast to previous belief, most patients have a modest bleeding diathesis. On the other hand, it has become evident that some of the mutations responsible for platelet deficiency predispose the patient to serious, potentially life-threatening diseases. Today's vision of IT is, therefore, very different from that of the past and the therapeutic approach must take these changes into account while also making use of the new therapies that have become available in the meantime. This review, the first devoted entirely to IT therapy, discusses how to prevent bleeding in those patients who are exposed to this risk, how to treat it if it occurs, and how to manage the serious illnesses to which patients with IT may be predisposed.
Collapse
|
22
|
Cavannaugh C, Ochs HD, Buchbinder D. Diagnosis and clinical management of Wiskott-Aldrich syndrome: current and emerging techniques. Expert Rev Clin Immunol 2022; 18:609-623. [PMID: 35533396 DOI: 10.1080/1744666x.2022.2074400] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Wiskott-Aldrich syndrome (WAS) serves as the prototype of how variants in a gene which encodes a protein central to actin cytoskeletal homeostasis can manifest clinically in a variety of ways including infection, atopy, autoimmunity, inflammation, bleeding, neutropenia, non-malignant lymphoproliferation, and malignancy. Despite the discovery of the WAS gene almost 30 years ago, our understanding of the pathophysiological mechanisms underlying WAS continues to unfold. AREAS COVERED This review will provide an overview of the approach to the diagnosis of WAS as well as the management of its associated complications. Advances in the use of allogeneic hematopoietic stem cell transplantation (HSCT) and gene therapy as well as the associated challenges unique to WAS will be discussed. EXPERT OPINION Basic research, combined with clinical research focusing on longitudinal analysis of WAS patients, will help clarify determinants that influence WAS pathogenesis as well as clinical complications and outcomes. Advances in curative approaches including the use of alternative donor HSCT for WAS continue to evolve. Gene therapy employing safer and more effective protocols ensuring full correction of WAS will provide life-saving benefit to WAS patients that are unable to undergo HSCT.
Collapse
Affiliation(s)
- Corey Cavannaugh
- Department of Pediatrics University of California at Irvine 333 The City Blvd. West Suite 800 Orange, CA 92868
| | - Hans D Ochs
- Department of Pediatrics University of Washington and Seattle Children's Research Institute Seattle, WA 98105
| | - David Buchbinder
- Division of Hematology Children's Hospital of Orange County 1201 La Veta Avenue Orange, CA 92868
| |
Collapse
|
23
|
Jindal AK, Rastogi P, Anjani G, Rikhi R, Rawat A, Ahluwalia J. An Autopsy Case of Wiskott-Aldrich Syndrome Revealing "FDC-Only Lymphoid Follicles" in Lymphoid Tissue: A Morphologic Correlate of Defective Immune Synapse. Pediatr Dev Pathol 2022; 25:345-350. [PMID: 35236172 DOI: 10.1177/10935266211058345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Wiskott-Aldrich Syndrome (WAS) is an inherited disorder characterized by the classical triad of eczema, micro-thrombocytopenia, and immune deficiency. This disease affects the hematopoietic cells to a variable extent. The spectrum of clinical and laboratory data for WAS has been well described in the literature though there is a paucity of its histopathologic and immunohistochemical correlates. The current case describes the autopsy findings of this rare entity in an 8-year old male child with specific recognition of altered histology noticed in the lymphoreticular tissues. The predominant morphological finding in lymphoid tissue was atretic hyalinized germinal centers labeled as "the follicular dendritic cell (FDC)-only lymphoid follicles." Immunohistochemistry revealed a reduction in germinal-center B-cells, T-follicular helper cells, attenuated mantle zone, FDC proliferation, and paracortical plasmacytosis. This case highlights the crippled immune cell population in WAS, ultimately leading to the morphology of atretic follicles rich in FDCs.
Collapse
Affiliation(s)
- Ankur K Jindal
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, RinggoldID:29751Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pulkit Rastogi
- Department of Histopathology, RinggoldID:29751Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gummadi Anjani
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, RinggoldID:29751Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rashmi Rikhi
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, RinggoldID:29751Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, RinggoldID:29751Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jasmina Ahluwalia
- Department of Hematology, RinggoldID:29751Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
24
|
Cavazzana M, Thrasher A. Gene therapy for Whiskott-Aldrich syndrome: The latest news. Clin Transl Med 2022; 12:e815. [PMID: 35437889 PMCID: PMC9016166 DOI: 10.1002/ctm2.815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Marina Cavazzana
- Department of Biotherapy, Hôpital Universitaire Necker-Enfants Malades, Groupe Hospitalier Paris Centre, Assistance Publique-Hopitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Paris Centre, Assistance Publique-Hôpitaux de Paris, Paris, France.,Imagine Institute, Université Paris, Paris, France
| | - Adrian Thrasher
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
25
|
A gain-of-function variant in the Wiskott-Aldrich syndrome gene is associated with a MYH9-related disease-like syndrome. Blood Adv 2022; 6:5279-5284. [PMID: 35404999 PMCID: PMC9631694 DOI: 10.1182/bloodadvances.2021006789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/27/2022] [Indexed: 11/21/2022] Open
Abstract
The gain-of-function p.I294T variant in WASp causes a disease combining neutropenia, macrothrombocytopenia, proteinuria, and renal failure. The expanded phenotypic spectrum associated with gain-of-function WAS variants supports renal function assessment in these patients. While loss-of-function variants in the WAS gene are associated with Wiskott-Aldrich syndrome and lead to microthrombocytopenia, gain-of-function variants of WAS are associated with X-linked neutropenia (XLN) and the absence of microthrombocytopenia. Only a few XLN families have been reported so far, and their platelet phenotype was not described in detail. To date, no renal involvement was described in XLN. In the present study, we report exome sequencing of individuals from 3 generations of a family with a dominant disease combining neutropenia, macrothrombocytopenia, and renal failure. We identified a heterozygous missense gain-of-function variant in the WAS gene (c.881T>C, p.I294T) that segregates with the disease and is already known to cause XLN. There was no pathogenic variant in MYH9, TUBB1, or ACTN1. This is the first report of a WAS gain-of-function variant associated with both the hematological phenotype of XLN (neutropenia, macrothrombocytopenia) and renal disease (proteinuria, renal failure) with glomerular tip lesion hyalinosis and actin condensations in effaced podocytes foot processes.
Collapse
|
26
|
Mohite RS, Furtado S, Kaur T, Mn V, Bhattad S. Dermatitis herpetiformis in a child with Wiskott-Aldrich syndrome: A rare occurrence. Pediatr Blood Cancer 2022; 69:e29459. [PMID: 34811870 DOI: 10.1002/pbc.29459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Rachna Shanbhag Mohite
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Aster CMI Hospital, Bangalore, Karnataka, India
| | - Shireen Furtado
- Department of Dermatology, Aster CMI Hospital, Bangalore, Karnataka, India
| | - Tripti Kaur
- Department of Pathology, Aster CMI Hospital, Bangalore, Karnataka, India
| | - Vidya Mn
- Department of Pathology, Aster CMI Hospital, Bangalore, Karnataka, India
| | - Sagar Bhattad
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Aster CMI Hospital, Bangalore, Karnataka, India
| |
Collapse
|
27
|
Towards a standard of care in transplant for WAS. Blood 2022; 139:1935-1936. [PMID: 35357483 DOI: 10.1182/blood.2022015612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/02/2022] [Indexed: 11/20/2022] Open
|
28
|
Acquired Thrombotic Thrombocytopenic Purpura in a 5-Year-old Child With Wiskott-Aldrich Syndrome. J Pediatr Hematol Oncol 2022; 44:e434-e437. [PMID: 33769392 DOI: 10.1097/mph.0000000000002150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/09/2021] [Indexed: 11/26/2022]
Abstract
Thrombocytopenia is often seen as a laboratory finding during childhood. A supposed idiopathic thrombocytopenic purpura patient who was later diagnosed as Wiskott-Aldrich syndrome (WAS) and developed acquired thrombotic thrombocytopenic purpura (aTTP). Although autoimmune manifestations in WAS described, aTTP was reported just once. Five-year-old-boy was initially brought with cough, bloody stool (diarrhea), oral mucosal bleeding at 12th months of age. Following diagnosed with idiopathic thrombocytopenic purpura and receiving intravenous immunoglobulin, platelet count raised from 20,000 to 50,000/µL. One year after WAS diagnosis by mutation analysis, he presented with complaints of resistant fever, epistaxis, and melena. Hemoglobin decreased from 10 to 5.9 g/dL. Schistocytes in peripheral blood smear and high anti-ADAMTS-13 antibody level indicated development of aTTP.
Collapse
|
29
|
Successful Allogeneic Peripheral Blood Stem Cell Transplantation in 4 Wiskott-Aldrich Syndrome Patients. J Pediatr Hematol Oncol 2022; 44:e324-e328. [PMID: 33828030 DOI: 10.1097/mph.0000000000002154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/24/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation is a potential curative treatment in Wiskott-Aldrich syndrome (WAS). Here, we analyzed the outcomes in 4 WAS patients who underwent this procedure with peripheral blood stem cell (PBSC) in our center. PATIENTS AND METHODS Four patients with severe WAS phenotype have received allogeneic hematopoietic stem cell transplantation between January 2014 and December 2019 from matched sibling donors with PBSC. Two different preparative conditioning regimens were provided: the first associated busulfan-cyclophosphamide (2 patients) and the second with busulfan-fludarabine administered to the others. Cyclosporine gave as preferred graft-versus-host disease prophylaxis with a short course of methotrexate. RESULTS All patients achieved engraftment after PBSC with a median CD34+ cell count: 13.6×106/kg (8 to 24.9×106/kg). Chronic graft-versus-host disease developed in 2 patients treated by cyclosporine-steroids with complete resolution. Chimerism for all the patients was fully donor (>95% donor). After a median follow-up of 41 months (8 to 74 mo), all patients (100%) are alive, healthy, with complete clinical, immunologic, and hematologic recovery, without signs of WAS. CONCLUSION This limited study with high-dose PBSC transplantation approach for WAS, demonstrated a safe and effective treatment option, with rapid engraftment, without complications, excellent long-term outcomes, independent of conditioning regimen.
Collapse
|
30
|
Hematopoietic stem cell transplantation for Wiskott-Aldrich syndrome: an EBMT inborn errors working party analysis. Blood 2022; 139:2066-2079. [PMID: 35100336 DOI: 10.1182/blood.2021014687] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/17/2022] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative treatment for patients affected by Wiskott-Aldrich syndrome (WAS). Reported HSCT outcomes have improved over time with respect to overall survival, but some studies have identified older age and HSCT from alternative donors as risk factors predicting poorer outcome. We analyzed 197 patients transplanted at EBMT centers between 2006 and 2017, who received conditioning as recommended by the inborn errors working party (IEWP): either busulfan (n=103) or treosulfan (n=94) combined with fludarabine ± thiotepa. After a median follow-up after HSCT of 44.9 months, 176 patients were alive, resulting in a 3-year overall survival of 88.7%, and chronic GVHD-free survival (CRFS; events: death, graft failure, severe chronic GVHD) of 81.7%. Overall survival and CRFS were not significantly impacted by conditioning regimen (busulfan- versus treosulfan-based), donor type (MSD/MFD vs MUD/MMUD vs. MMFD), and period of HSCT (2006-2013 vs. 2014-2017). Patients younger than 5 years at HSCT had a significantly better overall survival. The overall cumulative incidences of grade III-IV acute GVHD and extensive/moderate/severe chronic GVHD were 6.6% and 2.1%, respectively. Patients receiving treosulfan-based conditioning had a higher incidence of graft failure, mixed donor chimerism and more frequently received secondary procedures (2nd HSCT, unconditioned stem cell boost, donor lymphocyte infusion, or splenectomy). In summary, HSCT for WAS with conditioning regimens currently recommended by IEWP results in excellent survival and low rates of GVHD, regardless of donor or stem cell source, but age ≥5 years remains a risk factor for overall survival.
Collapse
|
31
|
Long-term safety and efficacy of lentiviral hematopoietic stem/progenitor cell gene therapy for Wiskott-Aldrich syndrome. Nat Med 2022; 28:71-80. [PMID: 35075289 PMCID: PMC8799465 DOI: 10.1038/s41591-021-01641-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Patients with Wiskott–Aldrich syndrome (WAS) lacking a human leukocyte antigen-matched donor may benefit from gene therapy through the provision of gene-corrected, autologous hematopoietic stem/progenitor cells. Here, we present comprehensive, long-term follow-up results (median follow-up, 7.6 years) (phase I/II trial no. NCT02333760) for eight patients with WAS having undergone phase I/II lentiviral vector-based gene therapy trials (nos. NCT01347346 and NCT01347242), with a focus on thrombocytopenia and autoimmunity. Primary outcomes of the long-term study were to establish clinical and biological safety, efficacy and tolerability by evaluating the incidence and type of serious adverse events and clinical status and biological parameters including lentiviral genomic integration sites in different cell subpopulations from 3 years to 15 years after gene therapy. Secondary outcomes included monitoring the need for additional treatment and T cell repertoire diversity. An interim analysis shows that the study meets the primary outcome criteria tested given that the gene-corrected cells engrafted stably, and no serious treatment-associated adverse events occurred. Overall, severe infections and eczema resolved. Autoimmune disorders and bleeding episodes were significantly less frequent, despite only partial correction of the platelet compartment. The results suggest that lentiviral gene therapy provides sustained clinical benefits for patients with WAS. Long-term monitoring of patients with Wiskott–Aldrich syndrome following lentiviral gene therapy shows a safe profile and a reduction in the frequency of autoimmune manifestations and bleeding events, despite incomplete platelet reconstitution.
Collapse
|
32
|
Hosahalli Vasanna S, Pereda MA, Dalal J. Clinical Features, Cancer Biology, Transplant Approach and Other Integrated Management Strategies for Wiskott-Aldrich Syndrome. J Multidiscip Healthc 2022; 14:3497-3512. [PMID: 34992377 PMCID: PMC8711845 DOI: 10.2147/jmdh.s295386] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/24/2021] [Indexed: 11/23/2022] Open
Abstract
Wiskott–Aldrich syndrome (WAS) is a rare X-linked recessive inborn error of immunity (IEI) first described in 1937. Classic WAS is characterized by the triad of thrombocytopenia with small platelets, recurrent infections due to combined immunodeficiency, and eczema. Hematopoietic stem cell transplantation (HSCT) was the only curative option available for five decades, with excellent outcomes reported for matched sibling donors (MSD) and matched unrelated donors (MUD). More recently, alternative donor transplants such as umbilical cord blood (UCB) and haploidentical transplant have emerged as viable options due to improvements in better graft selection, cell dosing, and effective allograft manipulation measures. Gene therapy is another potential curative option with promising results, yet currently is offered only as part of a clinical trial.
Collapse
Affiliation(s)
- Smitha Hosahalli Vasanna
- Department of Pediatrics, Division of Pediatric Hematology Oncology, Rainbow Babies and Children's Hospital, University Hospitals, Cleveland, OH, USA
| | - Maria A Pereda
- Department of Pediatrics, Division of Pediatric Hematology Oncology, Rainbow Babies and Children's Hospital, University Hospitals, Cleveland, OH, USA
| | - Jignesh Dalal
- Department of Pediatrics, Division of Pediatric Hematology Oncology, Rainbow Babies and Children's Hospital, University Hospitals, Cleveland, OH, USA
| |
Collapse
|
33
|
Jiang J, Zhou J, Wei M, Singh S, Nikuze L, Huang L, Li Y, Jiang J, Wei H. Clinical and molecular characteristics of Wiskott-Aldrich Syndrome in five unrelated Chinese families. Scand J Immunol 2021; 95:e13115. [PMID: 34758123 DOI: 10.1111/sji.13115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 11/26/2022]
Abstract
Wiskott-Aldrich syndrome (WAS) also called the eczema-thrombocytopenia-immunodeficiency syndrome is a primary immunodeficiency disease with X-linked recessive inheritance caused by mutations in the WAS protein (WASp) gene and characterized by thrombocytopenia with reduced platelet volume, eczema, immunodeficiency, and increased risk of malignant tumours. The mutations will lead to separate WAS severity which can be typical severe 'classical' WAS or less severe 'non-classical' WAS. This article will review and analyse clinical and immune characteristics of five unrelated Chinese families harbouring classical and non-classical WAS. The expression of WASp was detected in the peripheral blood monocytes (PBMC) by flow cytometry, and five mutations were found by WAS gene sequencing, one of which had not been reported in the literature, namely frameshift mutation c.1240_1247delCCACTCCC (p. P414Sfs*41).
Collapse
Affiliation(s)
- Jiali Jiang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Junli Zhou
- Department of Pediatrics, Xiamen Children's Hospital Affiliated to Fudan University, Xiamen, China
| | - Manlv Wei
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Sanjeev Singh
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lauriane Nikuze
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lifang Huang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuping Li
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinxia Jiang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hongying Wei
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
34
|
Okamoto K, Morio T. Inborn errors of immunity with eosinophilia. Allergol Int 2021; 70:415-420. [PMID: 34456137 DOI: 10.1016/j.alit.2021.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022] Open
Abstract
Monogenic diseases of the immune system, also known as inborn errors of immunity (IEIs), are caused by single-gene mutations and result in immune deficiency and dysregulation. More than 400 monogenic diseases have been described to date, and this number is rapidly expanding. The increasing availability of next-generation sequencing is now facilitating the diagnosis of IEIs. It is known that IEIs can predispose a person to not only infectious diseases but also cancer and immune disorders, such as inflammatory, autoimmune, and atopic diseases. IEIs with eosinophilia and atopic diseases can occur in several disorders. IEIs with eosinophilia have provided insights into human immunity and the pathogenesis of allergic diseases. Eosinophilia is not a rare finding in clinical practice, and it often poses problems in terms of etiologic research and differential diagnoses. Secondary eosinophilia is the most common form. The main underlying conditions are infectious diseases such as parasitic infections, allergic disorders, drug reactions, and of course IEIs. In clinical settings, the recognition of IEIs in the context of an allergic phenotype with eosinophilia is critical for prompt diagnosis and appropriate treatment aimed at modulating pathophysiological mechanisms and improving clinical symptoms.
Collapse
Affiliation(s)
- Keisuke Okamoto
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
35
|
Targeting the actin nucleation promoting factor WASp provides a therapeutic approach for hematopoietic malignancies. Nat Commun 2021; 12:5581. [PMID: 34552085 PMCID: PMC8458504 DOI: 10.1038/s41467-021-25842-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/03/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer cells depend on actin cytoskeleton rearrangement to carry out hallmark malignant functions including activation, proliferation, migration and invasiveness. Wiskott–Aldrich Syndrome protein (WASp) is an actin nucleation-promoting factor and is a key regulator of actin polymerization in hematopoietic cells. The involvement of WASp in malignancies is incompletely understood. Since WASp is exclusively expressed in hematopoietic cells, we performed in silico screening to identify small molecule compounds (SMCs) that bind WASp and promote its degradation. We describe here one such identified molecule; this WASp-targeting SMC inhibits key WASp-dependent actin processes in several types of hematopoietic malignancies in vitro and in vivo without affecting naïve healthy cells. This small molecule demonstrates limited toxicity and immunogenic effects, and thus, might serve as an effective strategy to treat specific hematopoietic malignancies in a safe and precisely targeted manner. Cancer cells proliferate and invade via cytoskeletal proteins such as WASp, exclusively expressed in hematopoietic cells. Here the authors show a specific small molecule compound inhibiting cancer cell activity by WASp degradation and demonstrating its therapeutic potential in vitro and in vivo.
Collapse
|
36
|
Sudhakar M, Rikhi R, Loganathan SK, Suri D, Singh S. Autoimmunity in Wiskott-Aldrich Syndrome: Updated Perspectives. Appl Clin Genet 2021; 14:363-388. [PMID: 34447261 PMCID: PMC8384432 DOI: 10.2147/tacg.s213920] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/18/2021] [Indexed: 11/23/2022] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is an uncommon X-linked combined-immunodeficiency disorder characterized by a triad of thrombocytopenia, eczema, and immunodeficiency. Patients with WAS are also predisposed to autoimmunity and malignancy. Autoimmune manifestations have been reported in 26%-72% of patients with WAS. Autoimmunity is an independent predictor of poor prognosis and predisposes to malignancy. Development of autoimmunity is also an early pointer of the need for hematopoietic stem-cell transplantation. In this manuscript, we have collated the published data and present a narrative review on autoimmune manifestations in WAS. A summary of currently proposed immunopathogenic mechanisms and genetic variants associated with development of autoimmunity in WAS is also included.
Collapse
Affiliation(s)
- Murugan Sudhakar
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rashmi Rikhi
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sathish Kumar Loganathan
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepti Suri
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Surjit Singh
- Department of Pediatrics, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
37
|
Tsai FD, Battinelli EM. Inherited Platelet Disorders. Hematol Oncol Clin North Am 2021; 35:1069-1084. [PMID: 34391603 DOI: 10.1016/j.hoc.2021.07.003] [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: 01/19/2023]
Abstract
Bleeding disorders due to platelet dysfunction are a common hematologic complication affecting patients, and typically present with mucocutaneous bleeding or hemorrhage. An inherited platelet disorder should be suspected in individuals with a suggestive family history and no identified secondary causes of bleeding. Genetic defects have been described at all levels of platelet activation, including receptor binding, signaling, granule release, cytoskeletal remodeling, and platelet hematopoiesis. Management of these disorders is typically supportive, with an emphasis on awareness, patient education, and anticipatory guidance to prevent future episodes of bleeding.
Collapse
Affiliation(s)
- Frederick D Tsai
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA; Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Elisabeth M Battinelli
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Mallhi KK, Petrovic A, Ochs HD. Hematopoietic Stem Cell Therapy for Wiskott-Aldrich Syndrome: Improved Outcome and Quality of Life. J Blood Med 2021; 12:435-447. [PMID: 34149291 PMCID: PMC8206065 DOI: 10.2147/jbm.s232650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/13/2021] [Indexed: 12/21/2022] Open
Abstract
The Wiskott-Aldrich syndrome (WAS) is an X-linked disorder caused by mutations in the WAS gene resulting in congenital thrombocytopenia, eczema, recurrent infections and an increased incidence of autoimmune diseases and malignancies. Without curative therapies, affected patients have diminished life expectancy and reduced quality of life. Since WAS protein (WASP) is constitutively expressed only in hematopoietic stem cell-derived lineages, hematopoietic stem cell transplantation (HSCT) and gene therapy (GT) are well suited to correct the hematologic and immunologic defects. Advances in high-resolution HLA typing, new techniques to prevent GvHD allowing the use of haploidentical donors, and the introduction of reduced intensity conditioning regimens with myeloablative features have increased overall survival (OS) to over 90%. The development of GT for WAS has provided basic knowledge into vector selection and random integration of various viral vectors into the genome, with the possibility of inducing leukemogenesis. After trials and errors, inactivating lentiviral vectors carrying the WAS gene were successfully evaluated in clinical trials, demonstrating cure of the disease except for insufficient resolution of the platelet defect. Thus, 50 years of clinical evaluation, genetic exploration and extensive clinical trials, a lethal syndrome has turned into a curable disorder.
Collapse
Affiliation(s)
- Kanwaldeep K Mallhi
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Division of Hematology and Oncology, Seattle Children’s Hospital, Seattle, WA, USA
| | - Aleksandra Petrovic
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Division of Immunology and Division of Hematology and Oncology, Seattle Children’s Hospital, Seattle, WA, USA
| | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Seattle Children’s Research Institute, Seattle, WA, USA
| |
Collapse
|
39
|
How I Treat: Allogeneic HSCT for adults with Inborn Errors of Immunity. Blood 2021; 138:1666-1676. [PMID: 34077952 DOI: 10.1182/blood.2020008187] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/10/2021] [Indexed: 11/20/2022] Open
Abstract
Inborn Errors of Immunity (IEI) are rare inherited disorders arising from monogenic germline mutations in genes that regulate the immune system. The majority of IEI are Primary Immunodeficiencies characterised by severe infection often associated with autoimmunity, autoinflammation and/or malignancy. Allogeneic hematopoietic stem cell transplant (HSCT) has been the corrective treatment of choice for many IEI presenting with severe disease in early childhood and experience has made this a successful and comparatively safe treatment in affected children. Early HSCT outcomes in adults were poor, resulting in extremely limited use worldwide. This is changing due to a combination of improved IEI diagnosis to inform patient selection, better understanding of the natural history of specific IEI and improvements in transplant practice. Recently published HSCT outcomes for adults with IEI have been comparable with pediatric data, making HSCT an important option for correction of clinically severe IEI in adulthood. Here we discuss our practice for patient selection, timing of HSCT, donor selection and conditioning, peri- and post HSCT management and our approach to long term follow up. We stress the importance of multidisciplinary involvement in the complex decision-making process that we believe is required for successful outcomes in this rapidly emerging area.
Collapse
|
40
|
Slatter MA, Gennery AR. Treosulfan-based conditioning for inborn errors of immunity. Ther Adv Hematol 2021; 12:20406207211013985. [PMID: 34094045 PMCID: PMC8141989 DOI: 10.1177/20406207211013985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/12/2021] [Indexed: 11/17/2022] Open
Abstract
Inborn errors of immunity (IEI) are inherited disorders that lead to defects in the development and/or function of the immune system. The number of disorders that can be treated by haematopoietic stem-cell transplantation (HSCT) has increased rapidly with the advent of next-generation sequencing. The methods used to transplant children with IEI have improved dramatically over the last 20 years. The introduction of reduced-toxicity conditioning is an important factor in the improved outcome of HSCT. Treosulfan has myeloablative and immunosuppressive properties, enabling engraftment with less toxicity than traditionally used doses of busulfan. It is firmly incorporated into the conditioning guidelines of the Inborn Errors Working Party of the European Society for Blood and Marrow Transplantation. Unlike busulfan, pharmacokinetically guided dosing of treosulfan is not part of routine practice, but data are emerging which indicate that further improvements in outcome may be possible, particularly in infants who have a decreased clearance of treosulfan. It is likely that individualized dosing, not just of treosulfan, but of all agents used in conditioning regimens, will be developed and implemented in the future. This will lead to a reduction in unwanted variability in drug exposure, leading to more predictable and adjustable exposure, and improved outcome of HSCT, with fewer late adverse effects and improved quality of life. Such conditioning regimens can be used as the basis to study the need for additional agents in certain disorders which are difficult to engraft or require high levels of donor chimerism, the dosing of individual cellular components within grafts, and effects of adjuvant cellular or immunotherapy post-transplant. This review documents the establishment of treosulfan worldwide, as a safe and effective agent for conditioning children with IEI prior to HSCT.
Collapse
Affiliation(s)
- Mary A Slatter
- Great North Children's Hospital, Clinical Resource Building, Floor 4, Block 2, Queen Victoria Road, Newcastle Upon Tyne NE1 4LP, UK
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
41
|
Kobar K, Collett K, Prykhozhij SV, Berman JN. Zebrafish Cancer Predisposition Models. Front Cell Dev Biol 2021; 9:660069. [PMID: 33987182 PMCID: PMC8112447 DOI: 10.3389/fcell.2021.660069] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer predisposition syndromes are rare, typically monogenic disorders that result from germline mutations that increase the likelihood of developing cancer. Although these disorders are individually rare, resulting cancers collectively represent 5-10% of all malignancies. In addition to a greater incidence of cancer, affected individuals have an earlier tumor onset and are frequently subjected to long-term multi-modal cancer screening protocols for earlier detection and initiation of treatment. In vivo models are needed to better understand tumor-driving mechanisms, tailor patient screening approaches and develop targeted therapies to improve patient care and disease prognosis. The zebrafish (Danio rerio) has emerged as a robust model for cancer research due to its high fecundity, time- and cost-efficient genetic manipulation and real-time high-resolution imaging. Tumors developing in zebrafish cancer models are histologically and molecularly similar to their human counterparts, confirming the validity of these models. The zebrafish platform supports both large-scale random mutagenesis screens to identify potential candidate/modifier genes and recently optimized genome editing strategies. These techniques have greatly increased our ability to investigate the impact of certain mutations and how these lesions impact tumorigenesis and disease phenotype. These unique characteristics position the zebrafish as a powerful in vivo tool to model cancer predisposition syndromes and as such, several have already been created, including those recapitulating Li-Fraumeni syndrome, familial adenomatous polyposis, RASopathies, inherited bone marrow failure syndromes, and several other pathogenic mutations in cancer predisposition genes. In addition, the zebrafish platform supports medium- to high-throughput preclinical drug screening to identify compounds that may represent novel treatment paradigms or even prevent cancer evolution. This review will highlight and synthesize the findings from zebrafish cancer predisposition models created to date. We will discuss emerging trends in how these zebrafish cancer models can improve our understanding of the genetic mechanisms driving cancer predisposition and their potential to discover therapeutic and/or preventative compounds that change the natural history of disease for these vulnerable children, youth and adults.
Collapse
Affiliation(s)
- Kim Kobar
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Keon Collett
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | | | - Jason N. Berman
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
42
|
Suri D, Rikhi R, Jindal AK, Rawat A, Sudhakar M, Vignesh P, Gupta A, Kaur A, Sharma J, Ahluwalia J, Bhatia P, Khadwal A, Raj R, Uppuluri R, Desai M, Taur P, Pandrowala AA, Gowri V, Madkaikar MR, Lashkari HP, Bhattad S, Kumar H, Verma S, Imai K, Nonoyama S, Ohara O, Chan KW, Lee PP, Lau YL, Singh S. Wiskott Aldrich Syndrome: A Multi-Institutional Experience From India. Front Immunol 2021; 12:627651. [PMID: 33936041 PMCID: PMC8086834 DOI: 10.3389/fimmu.2021.627651] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/18/2021] [Indexed: 12/21/2022] Open
Abstract
Background Wiskott Aldrich syndrome (WAS) is characterized by bleeding manifestations, recurrent infections, eczema, autoimmunity, and malignancy. Over the last decade, improved awareness and better in-house diagnostic facilities at several centers in India has resulted in increased recognition of WAS. This study reports collated data across major primary immunodeficiency diseases (PID) centers in India that are involved in care of children with WAS and highlights the varied clinical presentations, genetic profile, and outcomes of patients in India. Methods Request to share data was sent to multiple centers in India that are involved in care and management of patients with PID. Six centers provided requisite data that were compiled and analyzed. Results In this multi-institutional cohort, clinical details of 108 patients who had a provisional diagnosis of WAS were received. Of these, 95 patients with 'definite WAS' were included Fourteen patients were classified as XLT and 81 patients as WAS. Median age at onset of symptoms of patients was 3 months (IQR 1.6, 6.0 months) and median age at diagnosis was 12 months (IQR 6,48 months). Clinical profile included bleeding episodes (92.6%), infections (84.2%), eczema (78.9%), various autoimmune manifestations (40%), and malignancy (2.1%). DNA analysis revealed 47 variants in 67 cases. Nonsense and missense variants were the most common (28.4% each), followed by small deletions (19.4%), and splice site defects (16.4%). We also report 24 novel variants, most of these being frameshift and nonsense mutations resulting in premature termination of protein synthesis. Prophylactic intravenous immunoglobulin (IVIg) was initiated in 52 patients (54.7%). Hematopoietic stem cell transplantation (HSCT) was carried out in 25 patients (26.3%). Of those transplanted, disease-free survival was seen in 15 patients (60%). Transplant related mortality was 36%. Outcome details were available for 89 patients. Of these, 37% had died till the time of this analysis. Median duration of follow-up was 36 months (range 2 weeks- 12 years; IQR 16.2 months- 70 months). Conclusions We report the first nationwide cohort of patients with WAS from India. Bleeding episodes and infections are common manifestations. Mortality continues to be high as curative therapy is not accessible to most of our patients.
Collapse
Affiliation(s)
- Deepti Suri
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Rashmi Rikhi
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ankur K. Jindal
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amit Rawat
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Murugan Sudhakar
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Pandiarajan Vignesh
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Anju Gupta
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Anit Kaur
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Jyoti Sharma
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Jasmina Ahluwalia
- Department of Haematology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Prateek Bhatia
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Alka Khadwal
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Revathi Raj
- Department of Paediatric Haematology and Oncology, Apollo Speciality Hospitals, Chennai, India
| | - Ramya Uppuluri
- Department of Paediatric Haematology and Oncology, Apollo Speciality Hospitals, Chennai, India
| | - Mukesh Desai
- Division of Immunology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Prasad Taur
- Division of Immunology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | | | - Vijaya Gowri
- Division of Immunology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Manisha R. Madkaikar
- Department of Paediatric Immunology and Leukocyte Biology, National Institute of Immunohematology, Mumbai, India
| | - Harsha Prasada Lashkari
- Department of Pediatrics, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Sagar Bhattad
- Pediatric Immunology and Rheumatology, Aster CMI Hospital, Bengaluru, India
| | - Harish Kumar
- Pediatric Immunology and Rheumatology, Aster CMI Hospital, Bengaluru, India
| | - Sanjeev Verma
- Department of King George Medical University, Lucknow, India
| | - Kohsuke Imai
- Department of Pediatrics, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shigeaki Nonoyama
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Chiba, Japan
| | - Koon W. Chan
- Department of Pediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong
| | - Pamela P. Lee
- Department of Pediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong
| | - Yu Lung Lau
- Department of Pediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong
| | - Surjit Singh
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
43
|
Tiri A, Masetti R, Conti F, Tignanelli A, Turrini E, Bertolini P, Esposito S, Pession A. Inborn Errors of Immunity and Cancer. BIOLOGY 2021; 10:biology10040313. [PMID: 33918597 PMCID: PMC8069273 DOI: 10.3390/biology10040313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/30/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022]
Abstract
Simple Summary Inborn Errors of Immunity (IEI) are a heterogeneous group of disorders characterized by a defect in the function of at least one, and often more, components of the immune system. The overall risk for cancer in children with IEI ranges from 4 to 25%. Several factors, namely, age of the patient, viral infection status and IEI type can influence the development of different cancer types. Immunologists and oncologists should interact to monitor and promptly diagnose the potential development of cancer in known IEI patients, as well as an underlying IEI in newly diagnosed cancers with suggestive medical history or high rate of therapy-related toxicity. The creation of an international registry of IEI cases with detailed information on the occurrence of cancer is fundamental to optimizing the diagnostic process and to evaluating the outcomes of new therapeutic options, with the aim of improving prognosis and reducing comorbidities. Abstract Inborn Errors of Immunity (IEI) are a heterogeneous group of disorders characterized by a defect in the function of at least one, and often more, components of the immune system. The aim of this narrative review is to discuss the epidemiology, the pathogenesis and the correct management of tumours in patients with IEI. PubMed was used to search for all of the studies published over the last 20 years using the keywords: “inborn errors of immunity” or “primary immunodeficiency” and “cancer” or “tumour” or “malignancy”. Literature analysis showed that the overall risk for cancer in children with IEI ranges from 4 to 25%. Several factors, namely, age of the patient, viral infection status and IEI type can influence the development of different cancer types. The knowledge of a specific tumour risk in the presence of IEI highlights the importance of a synergistic effort by immunologists and oncologists in tracking down the potential development of cancer in known IEI patients, as well as an underlying IEI in patients with newly diagnosed cancers. In the current genomic era, the creation of an international registry of IEI cases integrated with malignancies occurrence information is fundamental to optimizing the diagnostic process and to evaluating the outcomes of new therapeutic options, with the hope to obtain a better prognosis for these patients.
Collapse
Affiliation(s)
- Alessandra Tiri
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, 43126 Parma, Italy; (A.T.); (A.T.); (E.T.)
| | - Riccardo Masetti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, 40138 Bologna, Italy; (R.M.); (F.C.); (A.P.)
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, 40138 Bologna, Italy; (R.M.); (F.C.); (A.P.)
| | - Anna Tignanelli
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, 43126 Parma, Italy; (A.T.); (A.T.); (E.T.)
| | - Elena Turrini
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, 43126 Parma, Italy; (A.T.); (A.T.); (E.T.)
| | - Patrizia Bertolini
- Pediatric Oncohematology Unit, Pietro Barilla Children’s Hospital, 43126 Parma, Italy;
| | - Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, 43126 Parma, Italy; (A.T.); (A.T.); (E.T.)
- Correspondence: ; Tel.: +39-0521-903-524
| | - Andrea Pession
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, 40138 Bologna, Italy; (R.M.); (F.C.); (A.P.)
| |
Collapse
|
44
|
Rodriguez JA, Bang TJ, Restrepo CS, Green DB, Browne LP, Vargas D. Imaging Features of Primary Immunodeficiency Disorders. Radiol Cardiothorac Imaging 2021; 3:e200418. [PMID: 33969305 PMCID: PMC8098094 DOI: 10.1148/ryct.2021200418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 11/10/2020] [Accepted: 12/01/2020] [Indexed: 06/12/2023]
Abstract
Primary immunodeficiency disorders (PIDs), which are humoral, combined, and innate defects of the immune system, are relatively uncommon and may go undiagnosed in patients experiencing recurrent infections, resulting in increased morbidity and mortality. PIDs are clinically characterized by a broad spectrum of disorders, including repeated infections, autoimmune disorders, lymphoproliferative diseases, congenital anomalies, and increased risk of malignancy. Cardiothoracic imaging plays a crucial role in the diagnosis of PIDs owing to the high rates of repeated respiratory infections leading to bronchiectasis and other forms of chronic lung disease. Although PIDs as a group may seem similar in terms of radiologic features and clinical manifestations, there are specific entities that are pertinent to each PID on an individual level. For example, patients with common variable immunodeficiency may develop a unique granulomatous lymphocytic interstitial lung disease, and Good syndrome is associated with thymoma. Familiarity with the imaging characteristics of these disorders may expedite diagnosis and prognostication, and better direct therapy. Reviewing the thoracic manifestations of all PIDs is beyond the scope of this article; thus, the focus herein is on discussing the thoracic manifestations of the most common PIDs and their imaging features. © RSNA, 2021An earlier incorrect version appeared online. This article was corrected on March 25, 2021.
Collapse
|
45
|
A Novel Mutation in WAS Gene Causing a Phenotypic Presentation of Wiskott-Aldrich Syndrome: A Case Report. J Pediatr Hematol Oncol 2021; 43:e234-e236. [PMID: 32287099 DOI: 10.1097/mph.0000000000001790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 02/29/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Wiskott-Aldrich syndrome (WAS) is an X-linked disorder characterized by immunodeficiency, thrombocytopenia, and atopic dermatitis. OBSERVATIONS This infant presented at birth with petechiae and bruising, with severe neonatal thrombocytopenia. Genetic testing for WAS revealed a variant of unknown significance hemizygous missense mutation in the WAS gene. This variant has not previously been reported. On the basis of the patient's clinical course including bleeding, infection, abnormal immune evaluation, and dermatologic sequelae, he was diagnosed with WAS and underwent allogeneic hematopoietic stem cell transplantation. CONCLUSIONS We report a novel mutation in the WAS gene that causes a phenotypic presentation of Wiskott-Aldrich Syndrome.
Collapse
|
46
|
Wang C, Sample KM, Gajendran B, Kapranov P, Liu W, Hu A, Zacksenhaus E, Li Y, Hao X, Ben-David Y. FLI1 Induces Megakaryopoiesis Gene Expression Through WAS/WIP-Dependent and Independent Mechanisms; Implications for Wiskott-Aldrich Syndrome. Front Immunol 2021; 12:607836. [PMID: 33717090 PMCID: PMC7953068 DOI: 10.3389/fimmu.2021.607836] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Wiskott–Aldrich Syndrome, WAS/WAVE, is a rare, X-linked immune-deficiency disease caused by mutations in the WAS gene, which together with its homolog, N-WASP, regulates actin cytoskeleton remodeling and cell motility. WAS patients suffer from microthrombocytopenia, characterized by a diminished number and size of platelets, though the underlying mechanism is not fully understood. Here, we identified FLI1 as a direct transcriptional regulator of WAS and its binding partner WIP. Depletion of either WAS or WIP in human erythroleukemic cells accelerated cell proliferation, suggesting tumor suppressor function of both genes in leukemia. Depletion of WAS/WIP also led to a significant reduction in the percentage of CD41 and CD61 positive cells, which mark committed megakaryocytes. RNAseq analysis revealed common changes in megakaryocytic gene expression following FLI1 or WASP knockdown. However, in contrast to FLI1, WASP depletion did not alter expression of late-stage platelet-inducing genes. N-WASP was not regulated by FLI1, yet its silencing also reduced the percentage of CD41+ and CD61+ megakaryocytes. Moreover, combined knockdown of WASP and N-WASP further suppressed megakaryocyte differentiation, indicating a major cooperation of these related genes in controlling megakaryocytic cell fate. However, unlike WASP/WIP, N-WASP loss suppressed leukemic cell proliferation. WASP, WIP and N-WASP depletion led to induction of FLI1 expression, mediated by GATA1, and this may mitigate the severity of platelet deficiency in WAS patients. Together, these results uncover a crucial role for FLI1 in megakaryocyte differentiation, implicating this transcription factor in regulating microthrombocytopenia associated with Wiskott–Aldrich syndrome.
Collapse
Affiliation(s)
- Chunlin Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Klarke M Sample
- The National Health Commission's Key Laboratory of Immunological Pulmonary Disease, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou University, Guiyang, China
| | - Babu Gajendran
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Philipp Kapranov
- School of Biomedical Sciences, Institute of Genomics, Huaqiao University, Xiamen, China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Anling Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Eldad Zacksenhaus
- Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Advanced Diagnostics, Toronto General Research Institute-University Health Network, Toronto, ON, Canada
| | - Yanmei Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Xiaojiang Hao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.,The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| |
Collapse
|
47
|
Agarwal N, Citla Sridhar D, Malay S, Patil N, Shekar A, Ahuja S, Dalal J. Wiskott Aldrich syndrome: healthcare utilizations and disparities in transplant care. Sci Rep 2021; 11:4654. [PMID: 33633315 PMCID: PMC7907136 DOI: 10.1038/s41598-021-84328-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/15/2021] [Indexed: 11/09/2022] Open
Abstract
Wiskott Aldrich syndrome (WAS) is a rare disease and hematopoietic stem cell transplant (HCT) is considered the treatment modality of choice for WAS. We conducted a cross-sectional analysis on the KIDS' pediatric inpatient database and compared hospitalization rates, complications and healthcare utilizations in the transplant and non-transplant arms. Of the 383 pediatric admissions with diagnosis of WAS between 2006-2012, 114 underwent transplant and 269 did not. The non-transplant arm included older children, female patients and more African Americans. Death rates, income and payer source were similar in both arms, however the total charge for each admission was higher in the transplant arm. Emergency room visits were similar but non-elective admissions were more in the non-transplant arm. Length of stay was prolonged in the transplant arm. When comparing morbidities, lymphomas, ulcerative colitis and autoimmune complications of WAS were seen only in the non-transplant arm. Our study shows that transplant is the largest contributor to healthcare utilization in WAS patients. We identified healthcare disparities based on race and socioeconomic status and found that this rare disease is being appropriately directed to centers with HCT expertise. We noted a change in practice moving away from splenectomy in WAS patients.
Collapse
Affiliation(s)
- Nikki Agarwal
- Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| | - Divyaswathi Citla Sridhar
- Rainbow Babies and Children Hospital, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Sindhoosha Malay
- School of Medicine, Case Western Reserve University, 1200 Wolstein Research Building, Cleveland, OH, 44106, USA
| | - Nirav Patil
- University Hospitals Cleveland Medical Center, 1200 Wolstein Research Building, Cleveland, OH, 44106, USA
| | - Anjali Shekar
- Rainbow Babies and Children Hospital, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Sanjay Ahuja
- Rainbow Babies and Children Hospital, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Jignesh Dalal
- Rainbow Babies and Children Hospital, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH, 44106, USA
| |
Collapse
|
48
|
Ben-Shmuel A, Sabag B, Biber G, Barda-Saad M. The Role of the Cytoskeleton in Regulating the Natural Killer Cell Immune Response in Health and Disease: From Signaling Dynamics to Function. Front Cell Dev Biol 2021; 9:609532. [PMID: 33598461 PMCID: PMC7882700 DOI: 10.3389/fcell.2021.609532] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/11/2021] [Indexed: 01/13/2023] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells, which play key roles in elimination of virally infected and malignant cells. The balance between activating and inhibitory signals derived from NK surface receptors govern the NK cell immune response. The cytoskeleton facilitates most NK cell effector functions, such as motility, infiltration, conjugation with target cells, immunological synapse assembly, and cytotoxicity. Though many studies have characterized signaling pathways that promote actin reorganization in immune cells, it is not completely clear how particular cytoskeletal architectures at the immunological synapse promote effector functions, and how cytoskeletal dynamics impact downstream signaling pathways and activation. Moreover, pioneering studies employing advanced imaging techniques have only begun to uncover the architectural complexity dictating the NK cell activation threshold; it is becoming clear that a distinct organization of the cytoskeleton and signaling receptors at the NK immunological synapse plays a decisive role in activation and tolerance. Here, we review the roles of the actin cytoskeleton in NK cells. We focus on how actin dynamics impact cytolytic granule secretion, NK cell motility, and NK cell infiltration through tissues into inflammatory sites. We will also describe the additional cytoskeletal components, non-muscle Myosin II and microtubules that play pivotal roles in NK cell activity. Furthermore, special emphasis will be placed on the role of the cytoskeleton in assembly of immunological synapses, and how mutations or downregulation of cytoskeletal accessory proteins impact NK cell function in health and disease.
Collapse
Affiliation(s)
- Aviad Ben-Shmuel
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Batel Sabag
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Guy Biber
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Mira Barda-Saad
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
49
|
Wang Y, Hao CL. [Gene analysis of a family with Wiskott-Aldrich syndrome]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 41:593-594. [PMID: 32810968 PMCID: PMC7449772 DOI: 10.3760/cma.j.issn.0253-2727.2020.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Y Wang
- Department of Hematology, Affiliated Hospital of Chengde Medical university, Chengde
| | - C L Hao
- Department of Hematology, Affiliated Hospital of Chengde Medical university, Chengde
| |
Collapse
|
50
|
Mastio J, Saeed MB, Wurzer H, Krecke M, Westerberg LS, Thomas C. Higher Incidence of B Cell Malignancies in Primary Immunodeficiencies: A Combination of Intrinsic Genomic Instability and Exocytosis Defects at the Immunological Synapse. Front Immunol 2020; 11:581119. [PMID: 33240268 PMCID: PMC7680899 DOI: 10.3389/fimmu.2020.581119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Congenital defects of the immune system called primary immunodeficiency disorders (PID) describe a group of diseases characterized by a decrease, an absence, or a malfunction of at least one part of the immune system. As a result, PID patients are more prone to develop life-threatening complications, including cancer. PID currently include over 400 different disorders, however, the variety of PID-related cancers is narrow. We discuss here reasons for this clinical phenotype. Namely, PID can lead to cell intrinsic failure to control cell transformation, failure to activate tumor surveillance by cytotoxic cells or both. As the most frequent tumors seen among PID patients stem from faulty lymphocyte development leading to leukemia and lymphoma, we focus on the extensive genomic alterations needed to create the vast diversity of B and T lymphocytes with potential to recognize any pathogen and why defects in these processes lead to malignancies in the immunodeficient environment of PID patients. In the second part of the review, we discuss PID affecting tumor surveillance and especially membrane trafficking defects caused by altered exocytosis and regulation of the actin cytoskeleton. As an impairment of these membrane trafficking pathways often results in dysfunctional effector immune cells, tumor cell immune evasion is elevated in PID. By considering new anti-cancer treatment concepts, such as transfer of genetically engineered immune cells, restoration of anti-tumor immunity in PID patients could be an approach to complement standard therapies.
Collapse
Affiliation(s)
- Jérôme Mastio
- Department of Oncology, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Mezida B Saeed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hannah Wurzer
- Department of Oncology, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Max Krecke
- Department of Oncology, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Clément Thomas
- Department of Oncology, Cytoskeleton and Cancer Progression, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| |
Collapse
|