1
|
Freeman AF, Gonzalez CE, Yates B, Cole K, Little L, Flannelly E, Steinberg SM, Mo G, Piette N, Hughes TE, Cuellar-Rodriguez J, Gea-Banacloche J, Heller T, Hammoud DA, Holland SM, Kong HH, Young FD, Jing H, Kayaoglu B, Su HC, Pai SY, Hickstein DD, Shah NN. Hematopoietic cell transplantation for DOCK8 deficiency: Results from a prospective clinical trial. J Allergy Clin Immunol 2024:S0091-6749(24)00904-7. [PMID: 39233015 DOI: 10.1016/j.jaci.2024.08.021] [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: 02/22/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND DOCK8 deficiency is a primary immunodeficiency in which allogeneic hematopoietic cell transplantation (HCT) represents the only known cure. We tested the ability of a busulfan-based regimen to achieve reliable engraftment and high levels of donor chimerism with acceptable toxicity in a prospective clinical trial in DOCK8 deficiency. OBJECTIVES To both evaluate the ability of HCT to reverse the clinical phenotype and to correct the immunologic abnormalities by 1 year post HCT. METHODS We conducted a prospective HCT trial for recipients with DOCK8 deficiency. Subjects were recruited from October 5, 2010, to December 30, 2022. Donor sources included fully matched related and unrelated donors and haploidentical donors. The reduced toxicity, myeloablative conditioning regimen contained no serotherapy. Graft-versus-host disease (GVHD) prophylaxis included either a calcineurin inhibitor with methotrexate or post-HCT cyclophosphamide (PT/Cy) followed by tacrolimus and mycophenolate mofetil. The trial was later amended to study PT/Cy in all patients. (Pilot Study of Reduced-Intensity Hematopoietic Stem Cell Transplant of DOCK8 [NCT01176006].) RESULTS: Thirty-six subjects, both children and adults (median age 16.4 years), underwent HCT for DOCK8 deficiency. Most patients, 33 of 36 (92%), achieved full (≥98%) donor chimerism in whole blood as early as day +30. With a median potential follow-up of 7.4 years, 29 (80.6%) were alive with no evidence of new DOCK8 deficiency-related complications. PT/Cy was effective in reducing the risk of acute GVHD in patients who had received matched unrelated donor and haploidentical transplants, but it was associated with transient delays in immune-reconstitution and hemorrhagic cystitis. CONCLUSIONS A busulfan-based HCT regimen using PT/Cy for GVHD prophylaxis and a broad range of donor types and hematopoietic cell sources were well tolerated, leading to the reversal of the clinical immunophenotype.
Collapse
Affiliation(s)
- Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Corina E Gonzalez
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md.
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | - Kristen Cole
- Transplant and Cell Therapy Program, Clinical Center, Bethesda, Md
| | - Lauren Little
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | - Erin Flannelly
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Seth M Steinberg
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, Md
| | - George Mo
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | | | | | - Jennifer Cuellar-Rodriguez
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Juan Gea-Banacloche
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Theo Heller
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Md
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, Bethesda, Md
| | - Steve M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Fernanda D Young
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Huie Jing
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Basak Kayaoglu
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Sung-Yun Pai
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Dennis D Hickstein
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| |
Collapse
|
2
|
Biglari S, Moghaddam AS, Tabatabaiefar MA, Sherkat R, Youssefian L, Saeidian AH, Vahidnezhad F, Tsoi LC, Gudjonsson JE, Hakonarson H, Casanova JL, Béziat V, Jouanguy E, Vahidnezhad H. Monogenic etiologies of persistent human papillomavirus infections: A comprehensive systematic review. Genet Med 2024; 26:101028. [PMID: 37978863 PMCID: PMC10922824 DOI: 10.1016/j.gim.2023.101028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
PURPOSE Persistent human papillomavirus infection (PHPVI) causes cutaneous, anogenital, and mucosal warts. Cutaneous warts include common warts, Treeman syndrome, and epidermodysplasia verruciformis, among others. Although more reports of monogenic predisposition to PHPVI have been published with the development of genomic technologies, genetic testing is rarely incorporated into clinical assessments. To encourage broader molecular testing, we compiled a list of the various monogenic etiologies of PHPVI. METHODS We conducted a systematic literature review to determine the genetic, immunological, and clinical characteristics of patients with PHPVI. RESULTS The inclusion criteria were met by 261 of 40,687 articles. In 842 patients, 83 PHPVI-associated genes were identified, including 42, 6, and 35 genes with strong, moderate, and weak evidence for causality, respectively. Autosomal recessive inheritance predominated (69%). PHPVI onset age was 10.8 ± 8.6 years, with an interquartile range of 5 to 14 years. GATA2,IL2RG,DOCK8, CXCR4, TMC6, TMC8, and CIB1 are the most frequently reported PHPVI-associated genes with strong causality. Most genes (74 out of 83) belong to a catalog of 485 inborn errors of immunity-related genes, and 40 genes (54%) are represented in the nonsyndromic and syndromic combined immunodeficiency categories. CONCLUSION PHPVI has at least 83 monogenic etiologies and a genetic diagnosis is essential for effective management.
Collapse
Affiliation(s)
- Sajjad Biglari
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Sherkat
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Youssefian
- Department of Pathology and Laboratory Medicine, UCLA Clinical Genomics Center, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Amir Hossein Saeidian
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI
| | | | - Hakon Hakonarson
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France; Imagine Institute, Paris Cité University, France; Department of Pediatrics, Necker Hospital for Sick Children, Paris, France, EU; Howard Hughes Medical Institute, Chevy Chase, MD
| | - Vivien Béziat
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France; Imagine Institute, Paris Cité University, France
| | - Emmanuelle Jouanguy
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France; Imagine Institute, Paris Cité University, France
| | - Hassan Vahidnezhad
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA.
| |
Collapse
|
3
|
Limnander A, Kaur N, Asrat S, Tasker C, Boyapati A, Ben LH, Janczy J, Pedraza P, Abreu P, Chen WC, Godin S, Daniel BJ, Chin H, DeVeaux M, Rodriguez Lorenc K, Sirulnik A, Harari O, Stahl N, Sleeman MA, Murphy AJ, Yancopoulos GD, Orengo JM. A therapeutic strategy to target distinct sources of IgE and durably reverse allergy. Sci Transl Med 2023; 15:eadf9561. [PMID: 38091405 DOI: 10.1126/scitranslmed.adf9561] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Immunoglobulin E (IgE) is a key driver of type 1 hypersensitivity reactions and allergic disorders, which are globally increasing in number and severity. Although eliminating pathogenic IgE may be a powerful way to treat allergy, no therapeutic strategy reported to date can fully ablate IgE production. Interleukin-4 receptor α (IL-4Rα) signaling is required for IgE class switching, and IL-4Rα blockade gradually reduces, but does not eliminate, IgE. The persistence of IgE after IL-4Rα blockade may be due to long-lived IgE+ plasma cells that maintain serological memory to allergens and thus may be susceptible to plasma cell-targeted therapeutics. We demonstrate that transient administration of a B cell maturation antigen x CD3 (BCMAxCD3) bispecific antibody markedly depletes IgE, as well as other immunoglobulins, by ablating long-lived plasma cells, although IgE and other immunoglobulins rapidly rebound after treatment. Concomitant IL-4Rα blockade specifically and durably prevents the reemergence of IgE by blocking IgE class switching while allowing the restoration of other immunoglobulins. Moreover, this combination treatment prevented anaphylaxis in mice. Together with additional cynomolgus monkey and human data, our studies demonstrate that allergic memory is primarily maintained by both non-IgE+ memory B cells that require class switching and long-lived IgE+ plasma cells. Our combination approach to durably eliminate pathogenic IgE has potential to benefit allergy in humans while preserving antibody-mediated immunity.
Collapse
Affiliation(s)
| | - Navneet Kaur
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Carley Tasker
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Anita Boyapati
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Li-Hong Ben
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - John Janczy
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Pablo Abreu
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Wen-Chi Chen
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Stephen Godin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | - Harvey Chin
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Olivier Harari
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | - Neil Stahl
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| | | | | | | | - Jamie M Orengo
- Regeneron Pharmaceuticals, Tarrytown, New York, 10591, USA
| |
Collapse
|
4
|
Sams L, Wijetilleka S, Ponsford M, Gennery A, Jolles S. Atopic manifestations of inborn errors of immunity. Curr Opin Allergy Clin Immunol 2023; 23:478-490. [PMID: 37755421 PMCID: PMC10621644 DOI: 10.1097/aci.0000000000000943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
PURPOSE OF REVIEW Allergy and atopic features are now well recognized manifestations of many inborn errors of immunity (IEI), and indeed may be the hallmark in some, such as DOCK8 deficiency. In this review, we describe the current IEI associated with atopy, using a comprehensive literature search and updates from the IUIS highlighting clinical clues for underlying IEI such as very early onset of atopic disease or treatment resistance to enable early and accurate genetic diagnosis. RECENT FINDINGS We focus on recently described genes, their categories of pathogenic mechanisms and the expanding range of potential therapies. SUMMARY We highlight in this review that patients with very early onset or treatment resistant atopic disorders should be investigated for an IEI, as targeted and effective therapies exist. Early and accurate genetic diagnosis is crucial in this cohort to reduce the burden of disease and mortality.
Collapse
Affiliation(s)
- Laura Sams
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Royal Victoria Infirmary, Queen Victoria Road
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne
| | - Sonali Wijetilleka
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - Mark Ponsford
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - Andrew Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Royal Victoria Infirmary, Queen Victoria Road
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| |
Collapse
|
5
|
Kono A, Wakamatsu M, Umezawa Y, Muramatsu H, Fujiwara H, Tomomasa D, Inoue K, Hattori K, Mitsui T, Takada H, Minegishi Y, Takahashi Y, Yamamoto M, Mori T, Kanegane H. Successful treatment of DOCK8 deficiency by allogeneic hematopoietic cell transplantation from alternative donors. Int J Hematol 2023; 118:519-525. [PMID: 37131080 DOI: 10.1007/s12185-023-03613-y] [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: 02/07/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
Dedicator of cytokinesis 8 (DOCK8) deficiency is a rare autosomal recessive inborn error of immunity (IEI) characterized by eczematous dermatitis, elevated serum IgE, and recurrent infections, comprising a seemingly hyper-IgE syndrome (HIES). DOCK8 deficiency is only curable with allogeneic hematopoietic cell transplantation (HCT), but the outcome of HCT from alternative donors is not fully understood. Here, we describe the cases of two Japanese patients with DOCK8 deficiency who were successfully treated by allogeneic HCT from alternative donors. Patient 1 underwent cord blood transplantation at the age of 16 years, and Patient 2 underwent haploidentical peripheral blood stem cell transplantation with post-transplant cyclophosphamide at the age of 22 years. Each patient received a fludarabine-based conditioning regimen. Their clinical manifestations, including refractory molluscum contagiosum, promptly improved post-HCT. They achieved successful engraftment and immune reconstitution without serious complications. Alternative donor sources such as cord blood and haploidentical donors can be options for allogeneic HCT for DOCK8 deficiency.
Collapse
Affiliation(s)
- Asuka Kono
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Manabu Wakamatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Umezawa
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Fujiwara
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Dan Tomomasa
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kento Inoue
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Keiichiro Hattori
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tetsuo Mitsui
- Department of Pediatrics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hidetoshi Takada
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshiyuki Minegishi
- Division of Molecular Medicine, Institute of Advanced Enzyme Research, Tokushima University, Tokushima, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahide Yamamoto
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Takehiko Mori
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
6
|
Sharma D, Ben Yakov G, Kapuria D, Viana Rodriguez G, Gewirtz M, Haddad J, Kleiner DE, Koh C, Bergerson JRE, Freeman AF, Heller T. Tip of the iceberg: A comprehensive review of liver disease in Inborn errors of immunity. Hepatology 2022; 76:1845-1861. [PMID: 35466407 DOI: 10.1002/hep.32539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/30/2022] [Accepted: 04/17/2022] [Indexed: 12/08/2022]
Abstract
Inborn errors of immunity (IEIs) consist of numerous rare, inherited defects of the immune system that affect about 500,000 people in the United States. As advancements in diagnosis through genetic testing and treatment with targeted immunotherapy and bone marrow transplant emerge, increasing numbers of patients survive into adulthood posing fresh clinical challenges. A large spectrum of hepatobiliary diseases now present in those with immunodeficiency diseases, leading to morbidity and mortality in this population. Awareness of these hepatobiliary diseases has lagged the improved management of the underlying disorders, leading to missed opportunities to improve clinical outcomes. This review article provides a detailed description of specific liver diseases occurring in various inborn errors of immunity. A generalized approach to diagnosis and management of hepatic complications is provided, and collaboration with hepatologists, immunologists, and pathologists is emphasized as a requirement for optimizing management and outcomes.
Collapse
Affiliation(s)
- Disha Sharma
- Department of Internal MedicineMedStar Washington Hospital Center & Georgetown UniversityWashingtonDCUSA.,Liver Diseases Branch, Translational Hepatology SectionNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA
| | - Gil Ben Yakov
- Liver Diseases Branch, Translational Hepatology SectionNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA.,26744Center for Liver DiseaseSheba Medical CenterTel HaShomerIsrael
| | - Devika Kapuria
- Liver Diseases Branch, Translational Hepatology SectionNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA.,Department of GastroenterologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Gracia Viana Rodriguez
- Liver Diseases Branch, Translational Hepatology SectionNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA
| | - Meital Gewirtz
- Liver Diseases Branch, Translational Hepatology SectionNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA
| | - James Haddad
- Liver Diseases Branch, Translational Hepatology SectionNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA
| | - David E Kleiner
- 3421Laboratory of PathologyNational Cancer InstituteBethesdaMarylandUSA
| | - Christopher Koh
- Liver Diseases Branch, Translational Hepatology SectionNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA
| | - Jenna R E Bergerson
- Laboratory of Clinical Immunology and MicrobiologyNIAID, NIHBethesdaMarylandUSA
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and MicrobiologyNIAID, NIHBethesdaMarylandUSA
| | - Theo Heller
- Liver Diseases Branch, Translational Hepatology SectionNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA
| |
Collapse
|
7
|
Liquidano-Pérez E, Maza-Ramos G, Yamazaki-Nakashimada MA, Barragán-Arévalo T, Lugo-Reyes SO, Scheffler-Mendoza S, Espinosa-Padilla SE, González-Serrano ME. [Combined immunodeficiency due to DOCK8 deficiency. State of the art]. REVISTA ALERGIA MÉXICO 2022; 69:31-47. [PMID: 36927749 DOI: 10.29262/ram.v69i1.1104] [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: 03/20/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
Combinedimmunodeficiency (CID) due to DOCK8 deficiency is an inborn error of immunity (IBD) characterized by dysfunctional T and B lymphocytes; The spectrum of manifestations includes allergy, autoimmunity, inflammation, predisposition to cancer, and recurrent infections. DOCK8 deficiency can be distinguished from other CIDs or within the spectrum of hyper-IgE syndromes by exhibiting profound susceptibility to viral skin infections, associated skin cancers, and severe food allergies. The 9p24.3 subtelomeric locus where DOCK8 is located includes numerous repetitive sequence elements that predispose to the generation of large germline deletions and recombination-mediated somatic DNA repair. Residual production DOCK8 protein contributes to the variable phenotype of the disease. Severe viral skin infections and varicella-zoster virus (VZV)-associated vasculopathy, reflect an essential role of the DOCK8 protein, which is required to maintain lymphocyte integrity as cells migrate through the tissues. Loss of DOCK8 causes immune deficiencies through other mechanisms, including a cell survival defect. In addition, there are alterations in the response of dendritic cells, which explains susceptibility to virus infection and regulatory T lymphocytes that could help explain autoimmunity in patients. Hematopoietic stem cell transplantation (HSCT) is the only curative treatment; it improves eczema, allergies, and susceptibility to infections.
Collapse
Affiliation(s)
- Eduardo Liquidano-Pérez
- Instituto Nacional de Pediatría, Unidad de Investigación en Inmunodeficiencias, Ciudad de México, México
| | | | | | - Tania Barragán-Arévalo
- Fundación de Asistencia Privada, Instituto de Oftalmología Conde de Valenciana, Departamento de Genética, Ciudad de México, México
| | - Saúl Oswaldo Lugo-Reyes
- Instituto Nacional de Pediatría, Unidad de Investigación en Inmunodeficiencias, Ciudad de México, México
| | | | - Sara Elva Espinosa-Padilla
- Instituto Nacional de Pediatría, Unidad de Investigación en Inmunodeficiencias, Ciudad de México, México
| | | |
Collapse
|
8
|
Vaseghi-Shanjani M, Snow AL, Margolis DJ, Latrous M, Milner JD, Turvey SE, Biggs CM. Atopy as Immune Dysregulation: Offender Genes and Targets. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1737-1756. [PMID: 35680527 DOI: 10.1016/j.jaip.2022.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 06/15/2023]
Abstract
Allergic diseases are a heterogeneous group of disorders resulting from exaggerated type 2 inflammation. Although typically viewed as polygenic multifactorial disorders caused by the interaction of several genes with the environment, we have come to appreciate that allergic diseases can also be caused by monogenic variants affecting the immune system and the skin epithelial barrier. Through a myriad of genetic association studies and high-throughput sequencing tools, many monogenic and polygenic culprits of allergic diseases have been described. Identifying the genetic causes of atopy has shaped our understanding of how these conditions occur and how they may be treated and even prevented. Precision diagnostic tools and therapies that address the specific molecular pathways implicated in allergic inflammation provide exciting opportunities to improve our care for patients across the field of allergy and immunology. Here, we highlight offender genes implicated in polygenic and monogenic allergic diseases and list targeted therapeutic approaches that address these disrupted pathways.
Collapse
Affiliation(s)
- Maryam Vaseghi-Shanjani
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - David J Margolis
- Department of Dermatology and Dermatologic Surgery, University of Pennsylvania Medical Center, Philadelphia, Pa; Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Medical Center, Philadelphia, Pa
| | - Meriem Latrous
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Catherine M Biggs
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; St Paul's Hospital, Vancouver, British Columbia, Canada.
| |
Collapse
|
9
|
El-Sayed ZA, El-Ghoneimy DH, Ortega-Martell JA, Radwan N, Aldave JC, Al-Herz W, Al-Nesf MA, Condino-Neto A, Cole T, Eley B, Erwa NH, Espinosa-Padilla S, Faria E, Rosario Filho NA, Fuleihan R, Galal N, Garabedian E, Hintermeyer M, Imai K, Irani C, Kamal E, Kechout N, Klocperk A, Levin M, Milota T, Ouederni M, Paganelli R, Pignata C, Qamar FN, Quinti I, Qureshi S, Radhakrishnan N, Rezaei N, Routes J, Singh S, Siniah S, Abdel-Hakam Taha I, Tanno LK, Van Dort B, Volokha A, Sullivan K. Allergic manifestations of inborn errors of immunity and their impact on the diagnosis: A worldwide study. World Allergy Organ J 2022; 15:100657. [PMID: 35783543 PMCID: PMC9218584 DOI: 10.1016/j.waojou.2022.100657] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background Allergies have long been observed in Inborn Errors of Immunity (IEI) and might even be the first presentation resulting in delayed diagnosis or misdiagnosis in some cases. However, data on the prevalence of allergic diseases among IEI patients are limited and contradictory. Objective To provide a worldwide view of allergic diseases, across a broad spectrum of IEI, and their impact on the timely diagnosis of IEI. Methods This is a worldwide study, conceived by the World Allergy Organization (WAO) Inborn Errors of Immunity Committee. A questionnaire was developed and pilot-tested and was sent via email to collect data from 61 immunology centers known to treat pediatric and/or adult IEI patients in 41 countries. In addition, a query was submitted to The United States Immunodeficiency Network (USIDNET) at its website. Results Thirty centers in 23 countries caring for a total 8450 IEI patients responded. The USIDNET dataset included 2332 patients. Data from responders showed that a median (IQR) of 16.3% (10-28.8%) of patients experienced allergic diseases during the course of their IEI as follows: 3.6% (1.3-11.3%) had bronchial asthma, 3.6% (1.9-9.1%) atopic dermatitis, 3.0% (1.0-7.8%) allergic rhinitis, and 1.3% (0.5-3.3%) food allergy. As per the USIDNET data, the frequency of allergy among IEI patients was 68.8% (bronchial asthma in 46.9%). The percentage of IEI patients who presented initially with allergic disorders was 8% (5-25%) and diagnosis delay was reported in 7.5% (0.9-20.6%). Predominantly antibody deficiencies had the highest frequency of allergic disease followed by combined immunodeficiency with a frequency of 40.3% (19.2-62.5%) and 20.0% (10-32%) respectively. As per the data of centers, anaphylaxis occurred in 25/8450 patients (0.3%) whereas per USIDNET dataset, it occurred in 249/2332 (10.6%); drugs and food allergy were the main causes in both datasets. Conclusions This multinational study brings to focus the relation between allergic diseases and IEI. Major allergies do occur in IEI patients but were less frequent than the general population. Initial presentation with allergy could adversely affect the timely diagnosis of IEI. There is a need for policies to raise awareness and educate primary care and other referring specialties on the association of allergic diseases with IEI. This study provides a network among centers for future prospective studies in the field.
Collapse
Affiliation(s)
- Zeinab A. El-Sayed
- Pediatric Allergy, Immunology and Rheumatology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | - Dalia H. El-Ghoneimy
- Pediatric Allergy, Immunology and Rheumatology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | | | - Nesrine Radwan
- Pediatric Allergy, Immunology and Rheumatology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | - Juan C. Aldave
- Allergy and Clinical Immunology, Hospital Nacional Edgardo Rebagliati Martins, Peru
| | - Waleed Al-Herz
- Allergy and Clinical Immunology Unit, Pediatric Department, AlSabah Hospital, Faculty of Medicine, Kuwait University, Kuwait
| | - Maryam A. Al-Nesf
- Department of Pediatrics, Pulmonary Division- Allergy and Immunology Section, Qatar
| | - Antonio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Theresa Cole
- Department of Allergy and Immunology- Royal Children's Hospital, Australia
| | - Brian Eley
- Paediatric Infectious Diseases Unit, Red Cross War Memorial Children's Hospital and the Department of Paediatrics and Child Health, University of Cape Town, South Africa
| | - Nahla H.H. Erwa
- Allergy and Immunology Unit- Soba University Hospital and Faculty of Medicine, University of Khartoum, Sudan
| | | | - Emilia Faria
- Immunoallergy Department, Coimbra Hospital and University Centre (CHUC), Portugal
| | | | - Ramsay Fuleihan
- Pediatric Allergy, Immunology and Rheumatology, Columbia University Medical Center, New York, USA
| | - Nermeen Galal
- Department of Pediatrics- Division of Immunology, Cairo University, Egypt
| | | | - Mary Hintermeyer
- Department of Allergy and Clinical Immunology- Children's Hospital of Wisconsin, Medical College of Wisconsin, USA
| | - Kohsuke Imai
- Department of Pediatrics-Tokyo Medical and Dental University, Japan
| | - Carla Irani
- Department of Internal Medicine and Clinical Immunology, Hotel Dieu de France Hospital, Beirut, Lebanon
| | - Ebtihal Kamal
- Allergy and Immunology Unit- Soba University Hospital and Faculty of Medicine, University of Khartoum, Sudan
| | - Nadia Kechout
- Department of Immunology, Pasteur Institute of Algeria, Algeria
| | - Adam Klocperk
- Department of Immunology, University Hospital Motol and the 2nd Faculty of Medicine, Charles University, Czech Republic
| | - Michael Levin
- Division of Pediatric Allergy, University of Cape Town, South Africa
| | - Tomas Milota
- Department of Immunology, University Hospital Motol and the 2nd Faculty of Medicine, Charles University, Czech Republic
| | - Monia Ouederni
- Department of Pediatrics, Immuno-hematology and Stem Cell Transplantation- Bone Marrow Transplantation Center Tunisia
| | - Roberto Paganelli
- Department of Medicine and Sciences of Aging, University “G. D’Annunzio”, Chieti-Pescara – and YDA, Institute of Clinical Immunotherapy and Advanced Biological Treatments, Pescara, Italy
| | | | - Farah N. Qamar
- Department of Pediatrics and Child Health- Aga Khan University Hospital, Karachi, Pakistan
| | - Isabella Quinti
- Department of Molecular Medicine, PID Reference Centre, Sapienza University of Rome, Italy
| | - Sonia Qureshi
- Department of Pediatrics and Child Health- Aga Khan University Hospital, Karachi, Pakistan
| | - Nita Radhakrishnan
- Department of Pediatric Hematology Oncology, Super Specialty Pediatric Hospital and Post Graduate Teaching Institute Noida, Delhi NCR 201303, India
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - John Routes
- Department of Allergy and Clinical Immunology- Children's Hospital of Wisconsin, Medical College of Wisconsin, USA
| | - Surjit Singh
- Department of Pediatrics and Chief, Allergy Immunology Unit, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sangeetha Siniah
- Hospital Tunku Azizah, Women and Children Hospital Kuala Lumpur, Malaysia
| | | | - Luciana K. Tanno
- Department of Pulmonology, Division of Allergy, University Hospital of Montpellier, Montpellier, France
- Desbrest Institute of Epidemiology and Public Health, UMR UA11, INSERM, University of Montpellier, Montpellier, France
- WHO Collaborating Centre on Classification Scientific Support, Montpellier, France
| | - Ben Van Dort
- Department of Allergy and Immunology- Royal Children's Hospital, Australia
| | - Alla Volokha
- Department of Pediatric Infectious Diseases and Immunology- Shupyk National Medical Academy of Postgraduate Education, Ukraine
| | | |
Collapse
|
10
|
Ravendran S, Hernández SS, König S, Bak RO. CRISPR/Cas-Based Gene Editing Strategies for DOCK8 Immunodeficiency Syndrome. Front Genome Ed 2022; 4:793010. [PMID: 35373187 PMCID: PMC8969908 DOI: 10.3389/fgeed.2022.793010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/14/2022] [Indexed: 12/17/2022] Open
Abstract
Defects in the DOCK8 gene causes combined immunodeficiency termed DOCK8 immunodeficiency syndrome (DIDS). DIDS previously belonged to the disease category of autosomal recessive hyper IgE syndrome (AR-HIES) but is now classified as a combined immunodeficiency (CID). This genetic disorder induces early onset of susceptibility to severe recurrent viral and bacterial infections, atopic diseases and malignancy resulting in high morbidity and mortality. This pathological state arises from impairment of actin polymerization and cytoskeletal rearrangement, which induces improper immune cell migration-, survival-, and effector functions. Owing to the severity of the disease, early allogenic hematopoietic stem cell transplantation is recommended even though it is associated with risk of unintended adverse effects, the need for compatible donors, and high expenses. So far, no alternative therapies have been developed, but the monogenic recessive nature of the disease suggests that gene therapy may be applied. The advent of the CRISPR/Cas gene editing system heralds a new era of possibilities in precision gene therapy, and positive results from clinical trials have already suggested that the tool may provide definitive cures for several genetic disorders. Here, we discuss the potential application of different CRISPR/Cas-mediated genetic therapies to correct the DOCK8 gene. Our findings encourage the pursuit of CRISPR/Cas-based gene editing approaches, which may constitute more precise, affordable, and low-risk definitive treatment options for DOCK8 deficiency.
Collapse
Affiliation(s)
| | | | | | - Rasmus O. Bak
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
11
|
Wang Z, Zhang Y, Li G, Huang L, Chen J. Dedicator of cytokinesis 8 deficiency and hyperimmunoglobulin E syndrome: A case report. Medicine (Baltimore) 2022; 101:e28807. [PMID: 35119052 PMCID: PMC8812656 DOI: 10.1097/md.0000000000028807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Hyperimmunoglobulin E syndrome (HIES) is a rare and complex immunoregulatory multisystem disorder characterized by recurrent eczema, skin and sinopulmonary infections, elevated serum immunoglobulin E levels, and eosinophilia. Onset is most likely in childhood, although infrequent adult cases have been reported. Early diagnosis is important. The use of the National Institutes of Health scoring system and the HIES signal transducer and activation of transcription 3 score can standardize the diagnosis of HIES. PATIENT CONCERNS A 19-year-old woman presented with complaints of dry cough, pyrexia, dyspnea, and recurrent pneumonia. She had a history of milk allergy, recurrent eczema, suppurative otitis media, chalazia, and aphthous ulcers. Her parents had a consanguineous marriage. DIAGNOSIS HIES; severe pneumonia. INTERVENTIONS Voriconazole (200 mg iv 2 times/d) and flucytosine (1 g orally 4 times/d) for 3 weeks were administered, followed by oral administration of fluconazole for 3 weeks. OUTCOMES The patient experienced near-complete remission of her respiratory symptoms. The patient was followed-up for one and a half years. During the follow-up, the patient presented again with cough and dyspnea and was again admitted to hospital. After being hospitalized for 3 weeks of antibiotic treatment, the patient experienced near-complete relief of her respiratory symptoms. LESSONS Regardless of patient age, it is important to consider the possibility of HIES when a patient has recurrent eczema, skin and sinopulmonary infections, elevated serum immunoglobulin E levels, and eosinophilia. Early diagnosis and intervention are essential to improve prognosis.
Collapse
Affiliation(s)
- Zhaojun Wang
- Department of Pulmonary and Critical care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yanan Zhang
- Department of Pulmonary and Critical care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Gang Li
- Central of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lingyan Huang
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Juan Chen
- Department of Pulmonary and Critical care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
12
|
Tangye SG, Gray PE, Pillay BA, Yap JY, Figgett WA, Reeves J, Kummerfeld SK, Stoddard J, Uzel G, Jing H, Su HC, Campbell DE, Sullivan A, Burnett L, Peake J, Ma CS. Hyper-IgE Syndrome due to an Elusive Novel Intronic Homozygous Variant in DOCK8. J Clin Immunol 2022; 42:119-129. [PMID: 34657245 PMCID: PMC10461790 DOI: 10.1007/s10875-021-01152-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022]
Abstract
Rare, biallelic loss-of-function mutations in DOCK8 result in a combined immune deficiency characterized by severe and recurrent cutaneous infections, eczema, allergies, and susceptibility to malignancy, as well as impaired humoral and cellular immunity and hyper-IgE. The advent of next-generation sequencing technologies has enabled the rapid molecular diagnosis of rare monogenic diseases, including inborn errors of immunity. These advances have resulted in the implementation of gene-guided treatments, such as hematopoietic stem cell transplant for DOCK8 deficiency. However, putative disease-causing variants revealed by next-generation sequencing need rigorous validation to demonstrate pathogenicity. Here, we report the eventual diagnosis of DOCK8 deficiency in a consanguineous family due to a novel homozygous intronic deletion variant that caused aberrant exon splicing and subsequent loss of expression of DOCK8 protein. Remarkably, the causative variant was not initially detected by clinical whole-genome sequencing but was subsequently identified and validated by combining advanced genomic analysis, RNA-seq, and flow cytometry. This case highlights the need to adopt multipronged confirmatory approaches to definitively solve complex genetic cases that result from variants outside protein-coding exons and conventional splice sites.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
| | - Paul E Gray
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Sydney, New South Wales, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Bethany A Pillay
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jin Yan Yap
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
| | - William A Figgett
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
| | - John Reeves
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Sarah K Kummerfeld
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
| | - Jennifer Stoddard
- Immunology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Huie Jing
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dianne E Campbell
- Department of Allergy and Immunology, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Anna Sullivan
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
- Queensland Children's Hospital and University of Queensland, South Brisbane, Queensland, Australia
| | - Leslie Burnett
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Jane Peake
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
- Queensland Children's Hospital and University of Queensland, South Brisbane, Queensland, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia.
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia.
| |
Collapse
|
13
|
Holland EM, Gonzalez C, Levy E, Valera VA, Chalfin H, Klicka-Skeels J, Yates B, Kleiner DE, Hadigan C, Dave H, Shalabi H, Hickstein DD, Su HC, Grimley M, Freeman AF, Shah NN. Case Report: Fatal Complications of BK Virus-Hemorrhagic Cystitis and Severe Cytokine Release Syndrome Following BK Virus-Specific T-Cells. Front Immunol 2021; 12:801281. [PMID: 34975916 PMCID: PMC8718506 DOI: 10.3389/fimmu.2021.801281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/01/2021] [Indexed: 11/13/2022] Open
Abstract
BK virus (BKV)-hemorrhagic cystitis (HC) is a well-known and rarely fatal complication of hematopoietic stem cell transplantation (HSCT). Treatment for BKV-HC is limited, but virus-specific T-cells (VST) represent a promising therapeutic option feasible for use posttransplant. We report on the case of a 16-year-old male with dedicator of cytokinesis 8 (DOCK8) deficiency who underwent haploidentical HSCT complicated by severe BKV-HC, catastrophic renal hemorrhage, and VST-associated cytokine release syndrome (CRS). Gross hematuria refractory to multiple interventions began with initiation of posttransplant cyclophosphamide (PT/Cy). Complete left renal arterial embolization (day +43) was ultimately indicated to control intractable renal hemorrhage. Subsequent infusion of anti-BK VSTs was complicated by CRS and progressive multiorgan failure, with postmortem analysis confirming diagnosis of hepatic sinusoidal obstruction syndrome (SOS). This case illustrates opportunities for improvement in the management of severe BKV-HC posttransplant while highlighting rare and potentially life-threatening complications of BKV-HC and VST therapy.
Collapse
Affiliation(s)
- Elizabeth M. Holland
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Corina Gonzalez
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
- Immune Deficiency- Cellular Therapy Program, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Elliot Levy
- Radiology and Imaging Sciences, NIH Clinical Center (CC), Bethesda, MD, United States
| | - Vladimir A. Valera
- Urologic Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Heather Chalfin
- Urologic Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | | | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - David E. Kleiner
- Laboratory of Pathology, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Colleen Hadigan
- Pediatric Gastroenterology, NIH Clinical Center (CC), Bethesda, MD, United States
| | - Hema Dave
- Pediatric Oncology, Children’s National Medical Center, Washington, DC, United States
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Dennis D. Hickstein
- Immune Deficiency- Cellular Therapy Program, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Helen C. Su
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Disease, NIH Clinical Center (CC), Bethesda, MD, United States
| | - Michael Grimley
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Disease, NIH Clinical Center (CC), Bethesda, MD, United States
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
14
|
Raedler J, Magg T, Rohlfs M, Klein C, Vallée T, Hauck F, Albert MH. Lineage-Specific Chimerism and Outcome After Hematopoietic Stem Cell Transplantation for DOCK8 Deficiency. J Clin Immunol 2021; 41:1536-1548. [PMID: 34080085 PMCID: PMC8452590 DOI: 10.1007/s10875-021-01069-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/18/2021] [Indexed: 11/30/2022]
Abstract
Bi-allelic variants in the dedicator of cytokinesis 8 (DOCK8) gene cause a combined immunodeficiency, characterized by recurrent sinopulmonary and skin infections, food allergies, eczema, eosinophilia, and elevated IgE. Long-term outcome is poor given susceptibility to infections, malignancy, and vascular complications. Allogeneic hematopoietic stem cell transplantation is currently the only curative treatment option and has shown promising outcome. The impact of mixed chimerism on long-term outcome is unclear. We reasoned that reversal of disease phenotype would depend on cell lineage-specific chimerism. DOCK8 variants were confirmed by Sanger and/or exome sequencing and immunoblot and/or intracellular flow cytometry. Donor chimerism was analyzed by XY-fluorescence in situ hybridization or quantitative short tandem repeat PCR. Outcome was assessed by laboratory tests, lymphocyte subsets, intracellular DOCK8 protein flow cytometry, T-cell proliferation analysis, and multiparameter immunoblot allergy screening. We report on nine patients, four of whom with mixed chimerism, with a median follow-up of 78 months after transplantation. Overall, we report successful transplantation with improvement of susceptibility to infections and allergies, and resolution of eczema in all patients. Immunological outcome in patients with mixed chimerism suggests a selective advantage for wild-type donor T-cells but lower donor B-cell chimerism possibly results in a tendency to hypogammaglobulinemia. No increased infectious and allergic complications were associated with mixed chimerism. Aware of the relatively small cohort size, we could not demonstrate a consistent detrimental effect of mixed chimerism on clinical outcomes. We nevertheless advocate aiming for complete donor chimerism in treating DOCK8 deficiency, but recommend reduced toxicity conditioning.
Collapse
Affiliation(s)
- Johannes Raedler
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Magg
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Meino Rohlfs
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christoph Klein
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Centre for Rare Diseases (M-ZSELMU), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tanja Vallée
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fabian Hauck
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Centre for Rare Diseases (M-ZSELMU), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael H Albert
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
15
|
Ollech A, Mashiah J, Lev A, Simon AJ, Somech R, Adam E, Barzilai A, Hagin D, Greenberger S. Treatment options for DOCK8 deficiency-related severe dermatitis. J Dermatol 2021; 48:1386-1393. [PMID: 34043252 DOI: 10.1111/1346-8138.15955] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cutaneous manifestations of dedicator of cytokinesis 8 gene (DOCK8) deficiency, a combined type of T and B cell immunodeficiency, previously designated as autosomal recessive hyper IgE syndrome, includes dermatitis and skin infections. There are limited treatment options for dermatitis related to the syndrome. OBJECTIVE To describe a cohort of patients with DOCK8 deficiency with a focus on the treatment of their cutaneous manifestations. METHODS A retrospective study on all children with the genetic diagnosis of DOCK8 deficiency treated at the Sheba Medical Center between 1/1/2003 and 1/1/2021 was preformed. Collected data included: demographic features, family history, laboratory, genetic testing, skin manifestations, treatment, and disease course. Description of two cases of severe recalcitrant dermatitis treated with dupilumab is detailed. RESULTS Nine children with a genetic diagnosis of DOCK8 deficiency were included, of whom six were girls (66%) with a median age of 8.5 (±2.2 SD) years. The median age at diagnosis was 2.8 (±2.6 SD) years. Six patients were born to consanguineous parents. Five out of six patients who received hematopoietic stem cell transplantation (HSCT) had a complete response, and one was recently transplanted. Of note, two patients, while awaiting HSCT, were treated with dupilumab for their severe dermatitis resulting in a marked improvement of the cutaneous manifestations and pruritus. CONCLUSIONS Hematopoietic stem cell transplantation is the gold standard and most effective therapy for patients with DOCK8 deficiency. Dupilumab, a biological therapy indicated for atopic dermatitis and other Th2 derived dermatoses, is an excellent option for dermatitis in patients with DOCK8 deficiency and can be used as a bridge before HSCT. Larger studies are needed to confirm this observation.
Collapse
Affiliation(s)
- Ayelet Ollech
- Department of Dermatology, Pediatric Dermatology Service, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jacob Mashiah
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Dermatology and Venereology, Pediatric Dermatology Unit, Dana Children's Hospital, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Atar Lev
- Department of Pediatrics, Immunodeficiency Unit, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Amos J Simon
- Department of Pediatrics, Immunodeficiency Unit, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Pediatrics, Immunodeficiency Unit, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Etai Adam
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Aviv Barzilai
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Dermatology and Venereology, Pediatric Dermatology Unit, Dana Children's Hospital, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - David Hagin
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Dermatology and Venereology, Pediatric Dermatology Unit, Dana Children's Hospital, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shoshana Greenberger
- Department of Dermatology, Pediatric Dermatology Service, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
16
|
Seo E, Lee BH, Lee JH, Park YS, Im HJ, Lee J. Hematopoietic stem cell transplantation in an infant with dedicator of cytokinesis 8 (DOCK8) deficiency associated with systemic lupus erythematosus: A case report. Medicine (Baltimore) 2021; 100:e20866. [PMID: 33787566 PMCID: PMC8021304 DOI: 10.1097/md.0000000000020866] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 05/21/2020] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION DOCK8 deficiency is a primary immunodeficiency characterized by recurrent infections, severe allergic disease, and autoimmunity. Here, we report a patient with DOCK8 deficiency that was initially presented as systemic lupus erythematosus (SLE) without recurrent infections and treated with hematopoietic stem cell transplantation (HSCT). PATIENT CONCERNS A 16-month-old boy with a previous history of eczema developed high fever and hand and foot swelling. Over time, multiple purpura, oral ulcers, and oliguria developed with a persistent fever. His laboratory findings showed anemia, thrombocytopenia, and coagulopathy with a high level of C-reactive protein (CRP). No definite pathogens were identified. The complement fractions C3, C4, and CH50 were low. Autoantibodies including antinuclear antibody (ANA) and anti-ds DNA antibody were positive. He definitively satisfied the 2015 ACR/SLICC revised criteria for the diagnosis of SLE (7 points out of 16); therefore, he was treated with a steroid. Lupus nephritis was confirmed by renal biopsy later. Considering the early-onset SLE, partial exome sequencing was performed. DIAGNOSIS One heterozygous missense variant, c.5536A>G (p.Lys1846Glu), which was inherited from his father, and heterozygous deletion of exon 1 to 8 inherited from his mother were found. Through the results of the genetic testing, the patient was confirmed to have DOCK8 deficiency. INTERVENTIONS At the age of 28 months, he received haploidentical HSCT from his mother as a donor. OUTCOMES Laboratory findings including complement fractions C3, C4, CH50, anti-ds DNA antibody, and the ANA became normal after HSCT. Currently, at 12 months post-HSCT, he is doing well, without any autoimmune features or infections. CONCLUSIONS DOCK8 deficiency can be presented as autoimmune disease such as SLE. Encountering a child diagnosed with SLE at a very young age, pediatricians should consider immunodeficiency syndrome including DOCK8 deficiency.
Collapse
Affiliation(s)
- Euri Seo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| | - Joo Hoon Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| | - Young Seo Park
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| | - Ho Joon Im
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| | - Jina Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| |
Collapse
|
17
|
Haskologlu S, Kostel Bal S, Islamoglu C, Aytekin C, Guner S, Sevinc S, Keles S, Kendirli T, Ceylaner S, Dogu F, Ikinciogullari A. Clinical, immunological features and follow up of 20 patients with dedicator of cytokinesis 8 (DOCK8) deficiency. Pediatr Allergy Immunol 2020; 31:515-527. [PMID: 32108967 PMCID: PMC7228270 DOI: 10.1111/pai.13236] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 01/27/2020] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
Biallelic mutations in the dedicator of cytokinesis 8 gene (DOCK8) cause a progressive combined immunodeficiency (CID) characterized by susceptibility to severe viral skin infections, atopic diseases, recurrent respiratory infections, and malignancy. Hematopoietic stem cell transplantation (HSCT) is only curative treatment for the disease. However, there is limited information about long-term outcome of HSCT and its effect to protect against cancer development in DOCK8-deficient patients. In this study, we retrospectively evaluated clinical and immunologic characteristics of 20 DOCK8-deficient patients and outcome of 11 patients who underwent HSCT. We aimed to report the experience of our center and the result of the largest transplantation series of DOCK8 deficiency in our country. Median follow-up time is 71 months (min-max: 16-172) in all patients and 48 months (min-max: 5-84) in transplanted patients. Atopic dermatitis (18/20), recurrent respiratory tract infections (17/20), and food allergy (14/20) were the most frequent clinical manifestations. Failure to thrive (13/20), liver problems (12/20), bronchiectasis (11/20), chronic diarrhea (10/21), and autism spectrum disorders (3/20) were remarkable findings in our series. Elevated IgE level (20/20) and eosinophilia (17/20), low IgM level (15/20), and decreased CD3+ T (10/20) and CD4+ T (11/20) cell count were prominent laboratory findings. HSCT was performed in 11 patients. All patients achieved adequate engraftment and showed improvement in their clinical and immunologic findings. Atopic dermatitis and food allergies improved in all patients, and their dietary restriction was stopped except one patient who was transplanted recently. The frequency of infections was decreased. The overall survival is 91% in HSCT-received patients and 80% in all. HSCT at the earliest possible period with most suitable donor- and patient-specific appropriate conditioning regimen and GvHD prophylaxis is lifesaving for DOCK8 deficiency cases.
Collapse
Affiliation(s)
- Sule Haskologlu
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sevgi Kostel Bal
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Candan Islamoglu
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Caner Aytekin
- Department of Pediatric Immunology, Dr.Sami Ulus Maternity and Children's Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Sukru Guner
- Department of Pediatrics, Division of Immunology and Allergy, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Selin Sevinc
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sevgi Keles
- Department of Pediatrics, Division of Immunology and Allergy, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Tanil Kendirli
- Pediatric Intensive Care Unit, Ankara University School of Medicine, Ankara, Turkey
| | | | - Figen Dogu
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Aydan Ikinciogullari
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
18
|
Mitchell R. Hematopoietic Stem Cell Transplantation Beyond Severe Combined Immunodeficiency: Seeking a Cure for Primary Immunodeficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 7:776-785. [PMID: 30832892 DOI: 10.1016/j.jaip.2018.12.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/27/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) can provide definitive therapy for patients with primary immunodeficiency disease (PIDD). Modern HSCT techniques and supportive care have significantly improved outcomes for patients with PIDD. This review examines current HSCT practice for PIDD other than severe combined immunodeficiency, and explores indications, risks, and long-term outcomes for this group of challenging diseases.
Collapse
Affiliation(s)
- Richard Mitchell
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia; School of Women and Children's Health, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
19
|
Freeman AF, Yazigi N, Shah NN, Kleiner DE, Parta M, Atkinson P, Heller T, Holland SM, Kaufman SS, Khan KM, Hickstein DD. Tandem Orthotopic Living Donor Liver Transplantation Followed by Same Donor Haploidentical Hematopoietic Stem Cell Transplantation for DOCK8 Deficiency. Transplantation 2019; 103:2144-2149. [PMID: 30720689 PMCID: PMC6667308 DOI: 10.1097/tp.0000000000002649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND An 11-year-old girl with dedicator of cytokinesis 8 (DOCK8) deficiency was proposed for potentially curative hematopoietic stem cell transplantation (HSCT), the donor being her haploidentical mother. However, end-stage liver disease caused by chronic Cryptosporidium infection required liver transplantation before HSCT. METHODS Consequently, a staged approach of a sequential liver transplant followed by a HSCT was planned with her mother as the donor for both liver and HSCT. RESULTS The patient successfully underwent a left-lobe orthotopic liver transplant; however, she developed a biliary leak delaying the HSCT. Notably, the recipient demonstrated 3% donor lymphocyte chimerism in her peripheral blood immediately before HSCT. Haploidentical-related donor HSCT performed 2 months after liver transplantation was complicated by the development of acyclovir-resistant herpes simplex virus viremia, primary graft failure, and sinusoidal obstruction syndrome. The patient died from sinusoidal obstruction syndrome-associated multiorgan failure with Candida sepsis on day +40 following HSCT. CONCLUSIONS We discuss the many considerations inherent to planning for HSCT preceded by liver transplant in patients with primary immunodeficiencies, including the role of prolonged immunosuppression and the risk of infection before immune reconstitution. We also discuss the implications of potential recipient sensitization against donor stem cells precipitated by exposure of the recipient to the donor lymphocytes from the transplanted organ.
Collapse
Affiliation(s)
- Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology,
National Institute of Allergy and Infectious Diseases, National Institutes of
Health, Bethesda, MD
| | - Nada Yazigi
- Pediatric Liver Transplantation, Department of Pediatrics,
MedStar Georgetown University Hospital, Washington DC
| | - Nirali N. Shah
- Pediatric Oncology Branch, National Cancer Institute,
National Institutes of Health, Bethesda, MD
| | - David E. Kleiner
- Laboratory of Pathology, National Cancer Institute,
National Institutes of Health, Bethesda, MD
| | - Mark Parta
- Clinical Monitoring Research Program Directorate, Frederick
National Laboratory for Cancer Research sponsored by the National Cancer
Institute
| | - Prescott Atkinson
- Division of Pediatric Allergy, Asthma and Immunology,
University of Alabama at Birmingham, Birmingham, AL
| | - Theo Heller
- Liver Diseases Branch, National Institute of Digestive,
Diabetes, and Kidney Disease Institute, National Institutes of Health, Bethesda,
MD
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology,
National Institute of Allergy and Infectious Diseases, National Institutes of
Health, Bethesda, MD
| | - Stuart S. Kaufman
- Pediatric Liver Transplantation, Department of Pediatrics,
MedStar Georgetown University Hospital, Washington DC
| | - Khalid M. Khan
- Pediatric Liver Transplantation, Department of Pediatrics,
MedStar Georgetown University Hospital, Washington DC
| | - Dennis D. Hickstein
- Experimental Transplantation and Immunology Branch,
National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
20
|
Lopes J, Teixeira D, Sousa C, Baptista A, Moreira D, Ferreira EO. Autosomal recessive hyper-IgE syndrome successfully treated with hematopoietic stem cell transplantation. Pediatr Dermatol 2019; 36:693-696. [PMID: 31338855 DOI: 10.1111/pde.13919] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Autosomal recessive hyper-IgE syndrome is a primary immunodeficiency that results from a mutation in the DOCK8 gene. We report a case of a patient presenting with severe eczema, atopy, and recurrent skin infections since the first months of life. The diagnosis of autosomal recessive hyper-IgE syndrome was made at the age of 7 by a positive DOCK8 genetic test. The patient underwent hematopoietic stem cell transplantation, with complete remission of the various manifestations.
Collapse
Affiliation(s)
- Jorge Lopes
- Department of Dermatology, Vila Nova de Gaia/Espinho Hospital Center, Vila Nova de Gaia, Portugal
| | - Diogo Teixeira
- Department of Dermatology, Vila Nova de Gaia/Espinho Hospital Center, Vila Nova de Gaia, Portugal
| | - Cristina Sousa
- Department of Dermatology, Vila Nova de Gaia/Espinho Hospital Center, Vila Nova de Gaia, Portugal
| | - Armando Baptista
- Department of Dermatology, Vila Nova de Gaia/Espinho Hospital Center, Vila Nova de Gaia, Portugal
| | - Diana Moreira
- Department of Pediatrics, Vila Nova de Gaia/Espinho Hospital Center, Vila Nova de Gaia, Portugal
| | - Eduarda O Ferreira
- Department of Dermatology, Vila Nova de Gaia/Espinho Hospital Center, Vila Nova de Gaia, Portugal
| |
Collapse
|
21
|
Pillay BA, Avery DT, Smart JM, Cole T, Choo S, Chan D, Gray PE, Frith K, Mitchell R, Phan TG, Wong M, Campbell DE, Hsu P, Ziegler JB, Peake J, Alvaro F, Picard C, Bustamante J, Neven B, Cant AJ, Uzel G, Arkwright PD, Casanova JL, Su HC, Freeman AF, Shah N, Hickstein DD, Tangye SG, Ma CS. Hematopoietic stem cell transplant effectively rescues lymphocyte differentiation and function in DOCK8-deficient patients. JCI Insight 2019; 5:127527. [PMID: 31021819 DOI: 10.1172/jci.insight.127527] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bi-allelic inactivating mutations in DOCK8 cause a combined immunodeficiency characterised by severe pathogen infections, eczema, allergies, malignancy and impaired humoral responses. These clinical features result from functional defects in most lymphocyte lineages. Thus, DOCK8 plays a key role in immune cell function. Hematopoietic stem cell transplantation (HSCT) is curative for DOCK8 deficiency. While previous reports have described clinical outcomes for DOCK8 deficiency following HSCT, the effect on lymphocyte reconstitution and function has not been investigated. Our study determined whether defects in lymphocyte differentiation and function in DOCK8-deficient patients were restored following HSCT. DOCK8-deficient T and B lymphocytes exhibited aberrant activation and effector function in vivo and in vitro. Frequencies of αβ T and MAIT cells were reduced while γδT cells were increased in DOCK8-deficient patients. HSCT improved, abnormal lymphocyte function in DOCK8-deficient patients. Elevated total and allergen-specific IgE in DOCK8-deficient patients decreased over time following HSCT. Our results document the extensive catalogue of cellular defects in DOCK8-deficient patients, and the efficacy of HSCT to correct these defects, concurrent with improvements in clinical phenotypes. Overall, our findings provide mechanisms at a functional cellular level for improvements in clinical features of DOCK8 deficiency post-HSCT, identify biomarkers that correlate with improved clinical outcomes, and inform the general dynamics of immune reconstitution in patients with monogenic immune disorders following HSCT.
Collapse
Affiliation(s)
- Bethany A Pillay
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Danielle T Avery
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Joanne M Smart
- Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Theresa Cole
- Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Sharon Choo
- Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Damien Chan
- Women and Children's Hosp==ital, Adelaide, South Australia, Australia
| | - Paul E Gray
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Sydney, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia.,Clinical Immunogenomics Research Consortium of Australia (CIRCA), Sydney, New South Wales, Australia
| | - Katie Frith
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Sydney, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Richard Mitchell
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.,Clinical Immunogenomics Research Consortium of Australia (CIRCA), Sydney, New South Wales, Australia
| | - Melanie Wong
- Clinical Immunogenomics Research Consortium of Australia (CIRCA), Sydney, New South Wales, Australia.,Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Dianne E Campbell
- Clinical Immunogenomics Research Consortium of Australia (CIRCA), Sydney, New South Wales, Australia.,Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Peter Hsu
- Clinical Immunogenomics Research Consortium of Australia (CIRCA), Sydney, New South Wales, Australia.,Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - John B Ziegler
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Sydney, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia.,Clinical Immunogenomics Research Consortium of Australia (CIRCA), Sydney, New South Wales, Australia
| | - Jane Peake
- Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Frank Alvaro
- Pediatric Hematology, John Hunter Hospital, New Lambton, New South Wales, Australia
| | - Capucine Picard
- Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, INSERM UMR 1163, Imagine institut, Paris, France.,Study Center for Primary Immunodeficiencies, Assistance Publique-Hôpitaux de Paris (AP-HP), Necker Hospital for Sick Children, Paris, France.,Pediatric Hematology and Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Jacinta Bustamante
- Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, INSERM UMR 1163, Imagine institut, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Institut IMAGINE, Necker Medical School, University Paris Descartes Paris, France.,St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Benedicte Neven
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Andrew J Cant
- Great North Children's Hospital, Newcastle upon Tyne Hospitals, NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Primary Immunodeficiency Group, Institute of Cellular Medicine, Newcastle upon Tyne University, Newcastle upon Tyne, United Kingdom
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Peter D Arkwright
- Lydia Becker Institute of Immunology & Inflammation, University of Manchester, Manchester, United Kingdom
| | - Jean-Laurent Casanova
- Pediatric Hematology and Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Institut IMAGINE, Necker Medical School, University Paris Descartes Paris, France.,St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA.,Howard Hughes Medical Institute, New York, New York, USA
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | - Dennis D Hickstein
- Experimental Transplantation and Immunology Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.,Clinical Immunogenomics Research Consortium of Australia (CIRCA), Sydney, New South Wales, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.,Clinical Immunogenomics Research Consortium of Australia (CIRCA), Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Albert MH, Freeman AF. Wiskott-Aldrich Syndrome (WAS) and Dedicator of Cytokinesis 8- (DOCK8) Deficiency. Front Pediatr 2019; 7:451. [PMID: 31750279 PMCID: PMC6848221 DOI: 10.3389/fped.2019.00451] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/17/2019] [Indexed: 01/04/2023] Open
Abstract
Both Wiskott-Aldrich syndrome (WAS) and dedicator of cytokinesis 8 (DOCK8) deficiency are primary immunodeficiency diseases caused by mutations in genes that result in defective organization of the cytoskeleton in hematopoietic tissues. They share some overlapping features such as a combined immunodeficiency, eczema and a predisposition to autoimmunity and malignancy, but also have some unique features that make them relatively easy to diagnose by clinical means. Both diseases can be cured by HSCT in a large proportion of patients. In WAS it is sometimes difficult to establish an indication for HSCT due to the large variability of disease severity, while HSCT is probably indicated in all patients affected by DOCK8 deficiency. There is considerably more published HSCT experience for WAS than for DOCK8 deficiency, but many open questions remain, which will be discussed in this review.
Collapse
Affiliation(s)
- Michael H Albert
- Dr. von Hauner University Children's Hospital, Ludwig-Maximilians Universität, Munich, Germany
| | - Alexandra F Freeman
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| |
Collapse
|
23
|
Castagnoli R, Delmonte OM, Calzoni E, Notarangelo LD. Hematopoietic Stem Cell Transplantation in Primary Immunodeficiency Diseases: Current Status and Future Perspectives. Front Pediatr 2019; 7:295. [PMID: 31440487 PMCID: PMC6694735 DOI: 10.3389/fped.2019.00295] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/03/2019] [Indexed: 12/29/2022] Open
Abstract
Primary immunodeficiencies (PID) are disorders that for the most part result from mutations in genes involved in immune host defense and immunoregulation. These conditions are characterized by various combinations of recurrent infections, autoimmunity, lymphoproliferation, inflammatory manifestations, atopy, and malignancy. Most PID are due to genetic defects that are intrinsic to hematopoietic cells. Therefore, replacement of mutant cells by healthy donor hematopoietic stem cells (HSC) represents a rational therapeutic approach. Full or partial ablation of the recipient's marrow with chemotherapy is often used to allow stable engraftment of donor-derived HSCs, and serotherapy may be added to the conditioning regimen to reduce the risks of graft rejection and graft versus host disease (GVHD). Initially, hematopoietic stem cell transplantation (HSCT) was attempted in patients with severe combined immunodeficiency (SCID) as the only available curative treatment. It was a challenging procedure, associated with elevated rates of morbidity and mortality. Overtime, outcome of HSCT for PID has significantly improved due to availability of high-resolution HLA typing, increased use of alternative donors and new stem cell sources, development of less toxic, reduced-intensity conditioning (RIC) regimens, and cellular engineering techniques for graft manipulation. Early identification of infants affected by SCID, prior to infectious complication, through newborn screening (NBS) programs and prompt genetic diagnosis with Next Generation Sequencing (NGS) techniques, have also ameliorated the outcome of HSCT. In addition, HSCT has been applied to treat a broader range of PID, including disorders of immune dysregulation. Yet, the broad spectrum of clinical and immunological phenotypes associated with PID makes it difficult to define a universal transplant regimen. As such, integration of knowledge between immunologists and transplant specialists is necessary for the development of innovative transplant protocols and to monitor their results during follow-up. Despite the improved outcome observed after HSCT, patients with severe forms of PID still face significant challenges of short and long-term transplant-related complications. To address this issue, novel HSCT strategies are being implemented aiming to improve both survival and long-term quality of life. This article will discuss the current status and latest developments in HSCT for PID, and present data regarding approach and outcome of HSCT in recently described PID, including disorders associated with immune dysregulation.
Collapse
Affiliation(s)
- Riccardo Castagnoli
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Ottavia Maria Delmonte
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Enrica Calzoni
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Molecular and Translational Medicine, A. Nocivelli Institute for Molecular Medicine, University of Brescia, Brescia, Italy
| | - Luigi Daniele Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
24
|
Immune Reconstitution Therapy for Immunodeficiency. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
25
|
Su HC, Jing H, Angelus P, Freeman AF. Insights into immunity from clinical and basic science studies of DOCK8 immunodeficiency syndrome. Immunol Rev 2019; 287:9-19. [PMID: 30565250 PMCID: PMC6350515 DOI: 10.1111/imr.12723] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 12/29/2022]
Abstract
DOCK8 immunodeficiency syndrome (DIDS) is a progressive combined immunodeficiency that can be distinguished from other combined immunodeficiencies or hyperimmunoglobulinemia E syndromes in featuring (a) profound susceptibility to virus infections of the skin, with associated skin cancers, and (b) severe food allergies. The DOCK8 locus has many repetitive sequence elements that predispose to the generation of large germline deletions as well as recombination-mediated somatic DNA repair. Residual DOCK8 protein contributes to the variable disease phenotype. The severe virus infections of the skin, and probably also VZV-associated vasculopathy, reflect an important function of DOCK8, which is normally required to maintain lymphocyte shape integrity as the cells migrate through dense tissues. Loss of DOCK8 also causes immune deficits through other mechanisms including a milder generalized cell survival defect and skewing of T helper cell subsets. Recent work has uncovered the roles for DOCK8 in dendritic cell responses that can also help explain the virus susceptibility, as well as in regulatory T cells that might help explain autoimmunity in a minority of patients. Fortunately, hematopoietic stem cell transplantation cures the eczema and infection susceptibility of DIDS, but not necessarily the other disease manifestations including food allergies.
Collapse
Affiliation(s)
- Helen C. Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| | - Huie Jing
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| | - Pam Angelus
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| |
Collapse
|
26
|
Aydin SE, Freeman AF, Al-Herz W, Al-Mousa HA, Arnaout RK, Aydin RC, Barlogis V, Belohradsky BH, Bonfim C, Bredius RG, Chu JI, Ciocarlie OC, Doğu F, Gaspar HB, Geha RS, Gennery AR, Hauck F, Hawwari A, Hickstein DD, Hoenig M, Ikinciogullari A, Klein C, Kumar A, Ifversen MRS, Matthes S, Metin A, Neven B, Pai SY, Parikh SH, Picard C, Renner ED, Sanal Ö, Schulz AS, Schuster F, Shah NN, Shereck EB, Slatter MA, Su HC, van Montfrans J, Woessmann W, Ziegler JB, Albert MH. Hematopoietic Stem Cell Transplantation as Treatment for Patients with DOCK8 Deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 7:848-855. [PMID: 30391550 DOI: 10.1016/j.jaip.2018.10.035] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/21/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Biallelic variations in the dedicator of cytokinesis 8 (DOCK8) gene cause a combined immunodeficiency with eczema, recurrent bacterial and viral infections, and malignancy. Natural disease outcome is dismal, but allogeneic hematopoietic stem cell transplantation (HSCT) can cure the disease. OBJECTIVE To determine outcome of HSCT for DOCK8 deficiency and define possible outcome variables. METHODS We performed a retrospective study of the results of HSCT in a large international cohort of DOCK8-deficient patients. RESULTS We identified 81 patients from 22 centers transplanted at a median age of 9.7 years (range, 0.7-27.2 years) between 1995 and 2015. After median follow-up of 26 months (range, 3-135 months), 68 (84%) patients are alive. Severe acute (III-IV) or chronic graft versus host disease occurred in 11% and 10%, respectively. Causes of death were infections (n = 5), graft versus host disease (5), multiorgan failure (2), and preexistent lymphoma (1). Survival after matched related (n = 40) or unrelated (35) HSCT was 89% and 81%, respectively. Reduced-toxicity conditioning based on either treosulfan or reduced-dose busulfan resulted in superior survival compared with fully myeloablative busulfan-based regimens (97% vs 78%; P = .049). Ninety-six percent of patients younger than 8 years at HSCT survived, compared with 78% of those 8 years and older (P = .06). Of the 73 patients with chimerism data available, 65 (89%) had more than 90% donor T-cell chimerism at last follow-up. Not all disease manifestations responded equally well to HSCT: eczema, infections, and mollusca resolved quicker than food allergies or failure to thrive. CONCLUSIONS HSCT is curative in most DOCK8-deficient patients, confirming this approach as the treatment of choice. HSCT using a reduced-toxicity regimen may offer the best chance for survival.
Collapse
Affiliation(s)
- Susanne E Aydin
- Dr von Hauner University Children's Hospital, Ludwig Maximilians Universität, Munich, Germany
| | - Alexandra F Freeman
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Waleed Al-Herz
- Department of Pediatrics, Al-Sabah Hospital, Kuwait, Kuwait
| | - Hamoud A Al-Mousa
- Department of Pediatrics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Rand K Arnaout
- Department of Medicine, Allergy & Immunology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Roland C Aydin
- Dr von Hauner University Children's Hospital, Ludwig Maximilians Universität, Munich, Germany
| | - Vincent Barlogis
- Pediatric Hematology, Assistance publique des Hopitaux de Marseille, Marseille, France
| | | | - Carmem Bonfim
- Pediatric Blood and Marrow Transplantation Program, Hospital de Clinicas, Federal University of Parana, Curitiba, Brazil
| | | | - Julia I Chu
- Department of Pediatrics, Stanford University School of Medicine, Stanford, Calif
| | - Oana C Ciocarlie
- Department of Bone Marrow Transplantation, Great Ormond Street Hospital NHS Trust, London, United Kingdom
| | - Figen Doğu
- Department of Pediatric Immunology & Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Hubert B Gaspar
- Molecular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Raif S Geha
- Department of Immunology, Boston Children's Hospital, Boston, Mass
| | - Andrew R Gennery
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Fabian Hauck
- Dr von Hauner University Children's Hospital, Ludwig Maximilians Universität, Munich, Germany
| | - Abbas Hawwari
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | | | - Manfred Hoenig
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Aydan Ikinciogullari
- Department of Pediatric Immunology & Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Christoph Klein
- Dr von Hauner University Children's Hospital, Ludwig Maximilians Universität, Munich, Germany
| | - Ashish Kumar
- BMT/Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Marianne R S Ifversen
- Department for Children and Adolescents, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Matthes
- Stem Cell Transplantation, St Anna Children's Hospital, Vienna, Austria
| | - Ayse Metin
- Pediatric Immunology, Ankara Children's Hematology Oncology Training and Research Hospital, Ankara, Turkey
| | - Benedicte Neven
- Department for Pediatric Immuno-Hematology and Rheumatology, Necker Hospital, Paris, France
| | - Sung-Yun Pai
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Mass
| | - Suhag H Parikh
- Pediatric Blood and Marrow Transplant Program, Duke University Medical Center, Durham, NC
| | - Capucine Picard
- Study Center of Primary Immunodeficiency, Necker Children's Hospital, Paris, France
| | | | - Özden Sanal
- Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Ansgar S Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Friedhelm Schuster
- Department of Pediatrics, Düsseldorf University Hospital, Düsseldorf, Germany
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Md
| | - Evan B Shereck
- Pediatric Hematology/Oncology, Oregon & Health Science University, Portland, Ore
| | - Mary A Slatter
- Paediatric BMT, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, Md
| | - Joris van Montfrans
- Pediatric Immunology and Infectious Diseases, UMC Utrecht, Utrecht, The Netherlands
| | - Wilhelm Woessmann
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - John B Ziegler
- Immunology & Infectious Diseases, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Michael H Albert
- Dr von Hauner University Children's Hospital, Ludwig Maximilians Universität, Munich, Germany.
| | | |
Collapse
|
27
|
Rae W, Ward D, Mattocks C, Pengelly RJ, Eren E, Patel SV, Faust SN, Hunt D, Williams AP. Clinical efficacy of a next-generation sequencing gene panel for primary immunodeficiency diagnostics. Clin Genet 2018; 93:647-655. [PMID: 29077208 DOI: 10.1111/cge.13163] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/18/2017] [Accepted: 10/23/2017] [Indexed: 01/04/2023]
Abstract
Primary immunodeficiencies (PIDs) are rare monogenic inborn errors of immunity that result in impairment of functions of the human immune system. PIDs have a broad phenotype with increased morbidity and mortality, and treatment choices are often complex. With increased accessibility of next-generation sequencing (NGS), the rate of discovery of genetic causes for PID has increased exponentially. Identification of an underlying monogenic diagnosis provides important clinical benefits for patients with the potential to alter treatments, facilitate genetic counselling, and pre-implantation diagnostics. We investigated a NGS PID panel of 242 genes within clinical care across a range of PID phenotypes. We also evaluated Phenomizer to predict causal genes from human phenotype ontology (HPO) terms. Twenty-seven participants were recruited, and a total of 15 reportable variants were identified in 48% (13/27) of the participants. The panel results had implications for treatment in 37% (10/27) of participants. Phenomizer identified the genes harbouring variants from HPO terms in 33% (9/27) of participants. This study shows the clinical efficacy that genetic testing has in the care of PID. However, it also highlights some of the disadvantages of gene panels in the rapidly moving field of PID genomics and current challenges in HPO term assignment for PID.
Collapse
Affiliation(s)
- W Rae
- Department of Immunology, University Hospital Southampton NHSFT, Southampton, UK.,Southampton National Institute for Health Research Clinical Research Facility, University Hospital Southampton NHSFT, Southampton, UK
| | - D Ward
- Wessex Investigational Sciences Hub Laboratory, University Hospital Southampton NHSFT, Southampton, UK
| | - C Mattocks
- Wessex Investigational Sciences Hub Laboratory, University Hospital Southampton NHSFT, Southampton, UK
| | - R J Pengelly
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - E Eren
- Department of Immunology, University Hospital Southampton NHSFT, Southampton, UK
| | - S V Patel
- Paediatric Immunology and Infectious Disease, Children's Hospital Southampton, Southampton, UK
| | - S N Faust
- Southampton National Institute for Health Research Clinical Research Facility, University Hospital Southampton NHSFT, Southampton, UK.,Faculty of Medicine, University of Southampton, Southampton, UK.,Paediatric Immunology and Infectious Disease, Children's Hospital Southampton, Southampton, UK
| | - D Hunt
- Wessex Clinical Genetics Service, University Hospital Southampton NHSFT, Southampton, UK
| | - A P Williams
- Department of Immunology, University Hospital Southampton NHSFT, Southampton, UK.,Wessex Investigational Sciences Hub Laboratory, University Hospital Southampton NHSFT, Southampton, UK.,Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
28
|
Slatter MA, Gennery AR. Hematopoietic cell transplantation in primary immunodeficiency - conventional and emerging indications. Expert Rev Clin Immunol 2018; 14:103-114. [PMID: 29300535 DOI: 10.1080/1744666x.2018.1424627] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Hematopoietic stem cell transplantation (HSCT) is an established curative treatment for many primary immunodeficiencies. Advances in donor selection, graft manipulation, conditioning and treatment of complications, mean that survival for many conditions is now around 90%. Next generation sequencing is identifying new immunodeficiencies, many of which are treatable with HSCT. Challenges remain however with short and long-term sequalae. This article reviews latest developments in HSCT for conventional primary immunodeficiencies and presents data on outcome for emerging diseases, Areas covered: This article reviews recently published literature detailing advances, particularly in conditioning regimens and new methods of T-lymphocyte depletion, as well as new information regarding approach and out come of transplanting patients with conventional primary immunodeficiencies. The article reviews data regarding transplant outcomes for newly described primary immunodeficiencies, particularly those associated with gain-of-function mutations. Expert commentary: New methods of graft manipulation have had significant impact on HSCT outcomes, with the range of PIDs treated using T-lymphocyte depletion significantly expanded. Outcomes for newly described diseases with variable phenotypes and clinical features, transplanted when the diagnosis was unknown are beginning to be described, and will improve as patients are identified earlier, and targeted therapies such as JAK inhibitors are used as a bridge to transplantation.
Collapse
Affiliation(s)
- Mary A Slatter
- a Institute of Cellular Medicine , Newcastle University , Newcastle Upon Tyne , UK.,b Paediatric Immunology and HSCT , Great North Children's Hospital , Newcastle Upon Tyne , UK
| | - Andrew R Gennery
- a Institute of Cellular Medicine , Newcastle University , Newcastle Upon Tyne , UK.,b Paediatric Immunology and HSCT , Great North Children's Hospital , Newcastle Upon Tyne , UK
| |
Collapse
|
29
|
Successful hematopoietic stem cell transplantation after myeloablative conditioning in three patients with dedicator of cytokinesis 8 deficiency (DOCK8) related Hyper IgE syndrome. Bone Marrow Transplant 2017; 53:339-343. [PMID: 29269803 DOI: 10.1038/s41409-017-0040-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 11/11/2017] [Accepted: 11/19/2017] [Indexed: 11/08/2022]
|
30
|
Brunet BA, Rodriguez R. Unusual presentation of combined immunodeficiency in a child with homozygous DOCK8 mutation. Ann Allergy Asthma Immunol 2017; 119:294-295. [PMID: 28890024 DOI: 10.1016/j.anai.2017.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Barbara A Brunet
- Divisions of Clinical Immunology and Allergy, Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi.
| | - Ray Rodriguez
- Divisions of Clinical Immunology and Allergy, Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
31
|
Kuşkonmaz B, Ayvaz D, Barış S, Ünal Ş, Tezcan İ, Uçkan D. Acute myeloid leukemia in a child with dedicator of cytokinesis 8 (DOCK8) deficiency. Pediatr Blood Cancer 2017. [PMID: 28627091 DOI: 10.1002/pbc.26695] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Barış Kuşkonmaz
- Division of Bone Marrow Transplantation Unit, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Deniz Ayvaz
- Division of Immunology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Safa Barış
- Division of Pediatric Immunology/Allergy, Marmara University, Istanbul, Turkey
| | - Şule Ünal
- Division of Hematology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - İlhan Tezcan
- Division of Immunology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Duygu Uçkan
- Division of Bone Marrow Transplantation Unit, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
32
|
Uygun DFK, Uygun V, Reisli İ, Keleş S, Özen A, Yılmaz M, Sayar EH, Daloğlu H, Öztürkmen SI, Çakı S, Karasu GT, Yeşilipek A. Hematopoietic stem cell transplantation from unrelated donors in children with DOCK8 deficiency. Pediatr Transplant 2017; 21. [PMID: 28664550 DOI: 10.1111/petr.13015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/02/2017] [Indexed: 01/01/2023]
Abstract
DIDS is a unique form of combined immune deficiency characterized by an unusual susceptibility to cutaneous viral infections, severe allergies with eosinophilia and elevated immunoglobulin E titers, autoimmunity, and cancer. HSCT is considered the standard of care for this deadly disease. We have retrospectively analyzed the outcome of allogeneic HSCT from unrelated donors in patients with DIDS. Data from four patients, with five transplants, are presented. All patients received transplants from unrelated donors' BM, except for one patient who received a cord blood transplant. The conditioning regimens were based on myeloablative protocols for BM derived transplants; a NM regimen was pursued for the patient who received a cord blood transplant, which resulted in graft rejection. Although recurrent pneumonia and skin infections resolved immediately after transplantation, all patients subsequently developed human herpesvirus infection, including cutaneous herpetic lesions, cytomegalovirus reactivation, and zona zoster, which could be attributed to the use of ATG. Despite the presence of serious morbidities prior to transplantation, all patients recovered successfully. DIDS can be successfully treated with allogeneic HSCT from unrelated donors following a myeloablative conditioning regimen, with a reasonable safety profile.
Collapse
Affiliation(s)
- Dilara Fatma K Uygun
- Department of Pediatric Immunology, Antalya Training and Research Hospital, Antalya, Turkey
| | - Vedat Uygun
- Faculty of Medicine, Department of Pediatric Bone Marrow Transplantation Unit, MedicalPark Antalya Hospital, Bahçeşehir University, Antalya, Turkey
| | - İsmail Reisli
- Meram Medical Faculty, Division of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Sevgi Keleş
- Meram Medical Faculty, Division of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Ahmet Özen
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, İstanbul, Turkey
| | - Mustafa Yılmaz
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Cukurova University, Adana, Turkey
| | - Esra H Sayar
- Meram Medical Faculty, Division of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Hayriye Daloğlu
- Faculty of Medicine, Department of Pediatric Bone Marrow Transplantation Unit, MedicalPark Antalya Hospital, Bahçeşehir University, Antalya, Turkey
| | - Seda I Öztürkmen
- Faculty of Medicine, Department of Pediatric Bone Marrow Transplantation Unit, MedicalPark Antalya Hospital, Bahçeşehir University, Antalya, Turkey
| | - Suar Çakı
- Faculty of Medicine, Department of Pediatric Bone Marrow Transplantation Unit, MedicalPark Göztepe Hospital, Bahçeşehir University, İstanbul, Turkey
| | - Gülsün T Karasu
- Faculty of Medicine, Department of Pediatric Bone Marrow Transplantation Unit, MedicalPark Göztepe Hospital, Bahçeşehir University, İstanbul, Turkey
| | - Akif Yeşilipek
- Faculty of Medicine, Department of Pediatric Bone Marrow Transplantation Unit, MedicalPark Antalya Hospital, Bahçeşehir University, Antalya, Turkey
| |
Collapse
|
33
|
Adams SP, Kricke S, Ralph E, Gilmour N, Gilmour KC. A comparison of TRECs and flow cytometry for naive T cell quantification. Clin Exp Immunol 2017; 191:198-202. [PMID: 28976005 DOI: 10.1111/cei.13062] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2017] [Indexed: 12/24/2022] Open
Abstract
Assessment of thymic output by measurement of naive T cells is carried out routinely in clinical diagnostic laboratories, predominantly using flow cytometry with a suitable panel of antibodies. Naive T cell measurements can also be made using molecular analyses to quantify T cell receptor excision circle (TRECs) levels in sorted cells from peripheral blood. In this study we have compared TRECs levels retrospectively with CD45RA+ CD27+ T cells and also with CD45RA+ CD31+ T cells in 134 patient samples at diagnosis or during follow-up. Both panels provide naive T cell measurements that have a strongly positive correlation with TRECs numbers and are suitable for use with enumerating naive T cell levels in a clinical laboratory.
Collapse
Affiliation(s)
- S P Adams
- SIHMDS Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - S Kricke
- SIHMDS Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - E Ralph
- Immunology, Camelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - N Gilmour
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - K C Gilmour
- Immunology, Camelia Botnar Laboratories, Great Ormond Street Hospital for Children NHS Trust, London, UK
| |
Collapse
|
34
|
Alroqi FJ, Charbonnier LM, Keles S, Ghandour F, Mouawad P, Sabouneh R, Mohammed R, Almutairi A, Chou J, Massaad MJ, Geha RS, Baz Z, Chatila TA. DOCK8 Deficiency Presenting as an IPEX-Like Disorder. J Clin Immunol 2017; 37:811-819. [PMID: 29058101 DOI: 10.1007/s10875-017-0451-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 10/09/2017] [Indexed: 12/13/2022]
Abstract
PURPOSE The dedicator of cytokinesis 8 (DOCK8) deficiency is an autosomal recessive-combined immunodeficiency whose clinical spectra include recurrent infections, autoimmunity, malignancies, elevated serum IgE, eczema, and food allergies. Here, we report on patients with loss of function DOCK8 mutations with profound immune dysregulation suggestive of an immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX)-like disorder. METHODS Immunophenotyping of lymphocyte subpopulations and analysis of DOCK8 protein expression were evaluated by flow cytometry. T regulatory (Treg) cells were isolated by cell sorting, and their suppressive activity was analyzed by flow cytometry. Gene mutational analysis was performed by whole-exome and Sanger sequencing. RESULTS Patient 1 (P1) presented at 10 months of age with chronic severe diarrhea and active colitis in the absence of an infectious trigger, severe eczema with elevated serum IgE, and autoimmune hemolytic anemia, suggestive of an IPEX-related disorder. Whole-exome sequencing revealed a homozygous nonsense mutation in DOCK8 at the DOCK-homology region (DHR)-1 (c.1498C>T; p. R500X). Patient P2, a cousin of P1 who carries the same DOCK8 nonsense mutation, presented with eczema and recurrent ear infections in early infancy, and she developed persistent diarrhea by 3 years of age. Patient P3 presented with lymphoproliferation, severe eczema with allergic dysregulation, and chronic diarrhea with colitis. She harbored a homozygous loss of function DOCK8 mutation (c.2402 -1G→A). Treg cell function was severely compromised by both DOCK8 mutations. CONCLUSION DOCK8 deficiency may present severe immune dysregulation with features that may overlap with those of IPEX and other IPEX-like disorders.
Collapse
Affiliation(s)
- Fayhan J Alroqi
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Karp Family Building, Room 10-214. 1 Blackfan Street, Boston, MA, 02115, USA
- Department of Pediatrics, King Abdulaziz Medical City, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Louis-Marie Charbonnier
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Karp Family Building, Room 10-214. 1 Blackfan Street, Boston, MA, 02115, USA
| | - Sevgi Keles
- Division of Pediatric Allergy and Immunology, Necmettin Erbakan University, Konya, Turkey
| | - Fatima Ghandour
- Department of Pathology, St George Hospital University Medical Center, Beirut, Lebanon
| | - Pierre Mouawad
- Department of Pediatrics, St George Hospital University Medical Center, Beirut, Lebanon
| | - Rami Sabouneh
- Department of Pediatrics, St George Hospital University Medical Center, Beirut, Lebanon
| | - Reem Mohammed
- Department of Pediatrics, King Abdulaziz Medical City, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abduarahman Almutairi
- Department of Pediatrics, King Abdulaziz Medical City, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Karp Family Building, Room 10-214. 1 Blackfan Street, Boston, MA, 02115, USA
| | - Michel J Massaad
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Karp Family Building, Room 10-214. 1 Blackfan Street, Boston, MA, 02115, USA
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Karp Family Building, Room 10-214. 1 Blackfan Street, Boston, MA, 02115, USA
| | - Zeina Baz
- Department of Pediatrics, St George Hospital University Medical Center, Beirut, Lebanon
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Karp Family Building, Room 10-214. 1 Blackfan Street, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Dedicator of cytokinesis 8 mutation related combined immune deficiency: A single centre experience from India. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2017. [DOI: 10.1016/j.phoj.2017.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
36
|
Chinen J, Badran YR, Geha RS, Chou JS, Fried AJ. Advances in basic and clinical immunology in 2016. J Allergy Clin Immunol 2017; 140:959-973. [DOI: 10.1016/j.jaci.2017.07.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/12/2017] [Accepted: 07/22/2017] [Indexed: 10/19/2022]
|
37
|
Ruffner MA, Sullivan KE, Henrickson SE. Recurrent and Sustained Viral Infections in Primary Immunodeficiencies. Front Immunol 2017; 8:665. [PMID: 28674531 PMCID: PMC5474473 DOI: 10.3389/fimmu.2017.00665] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/22/2017] [Indexed: 01/25/2023] Open
Abstract
Viral infections are commonplace and often innocuous. Nevertheless, within the population of patients with primary immunodeficiencies (PIDDs), viral infections can be the feature that drives a diagnostic evaluation or can be the most significant morbidity for the patient. This review is focused on the viral complications of PIDDs. It will focus on respiratory viruses, the most common type of viral infection in the general population. Children and adults with an increased frequency or severity of respiratory viral infections are often referred for an immunologic evaluation. The classic teaching is to investigate humoral function in people with recurrent sinopulmonary infections, but this is often interpreted to mean recurrent bacterial infections. Recurrent or very severe viral infections may also be a harbinger of a primary immunodeficiency as well. This review will also cover persistent cutaneous viral infections, systemic infections, central nervous system infections, and gastrointestinal infections. In each case, the specific viral infections may drive a diagnostic evaluation that is specific for that type of virus. This review also discusses the management of these infections, which can become problematic in patients with PIDDs.
Collapse
Affiliation(s)
- Melanie A Ruffner
- The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | | | - Sarah E Henrickson
- The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
38
|
Biggs CM, Keles S, Chatila TA. DOCK8 deficiency: Insights into pathophysiology, clinical features and management. Clin Immunol 2017. [PMID: 28625885 DOI: 10.1016/j.clim.2017.06.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Dedicator of cytokinesis 8 (DOCK8) deficiency is a combined immunodeficiency that exemplifies the broad clinical features of primary immunodeficiencies (PIDs), extending beyond recurrent infections to include atopy, autoimmunity and cancer. It is caused by loss of function mutations in DOCK8, encoding a guanine nucleotide exchange factor highly expressed in lymphocytes that regulates the actin cytoskeleton. Additional roles of DOCK8 have also emerged, including regulating MyD88-dependent Toll-like receptor signaling and the activation of the transcription factor STAT3. DOCK8 deficiency impairs immune cell migration, function and survival, and it impacts both innate and adaptive immune responses. Clinically, DOCK8 deficiency is characterized by allergic inflammation as well as susceptibility towards infections, autoimmunity and malignancy. This review details the pathophysiology, clinical features and management of DOCK8 deficiency. It also surveys the recently discovered combined immunodeficiency due to DOCK2 deficiency, highlighting in the process the emerging spectrum of PIDs resulting from DOCK protein family abnormalities.
Collapse
Affiliation(s)
- Catherine M Biggs
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Sevgi Keles
- Division of Pediatric Allergy and Immunology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Shah NN, Freeman AF, Su H, Cole K, Parta M, Moutsopoulos NM, Baris S, Karakoc-Aydiner E, Hughes TE, Kong HH, Holland SM, Hickstein DD. Haploidentical Related Donor Hematopoietic Stem Cell Transplantation for Dedicator-of-Cytokinesis 8 Deficiency Using Post-Transplantation Cyclophosphamide. Biol Blood Marrow Transplant 2017; 23:980-990. [PMID: 28288951 PMCID: PMC5757872 DOI: 10.1016/j.bbmt.2017.03.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 03/09/2017] [Indexed: 01/02/2023]
Abstract
Dedicator-of-cytokinesis 8 (DOCK8) deficiency, a primary immunodeficiency disease, can be reversed by allogeneic hematopoietic stem cell transplantation (HSCT); however, there are few reports describing the use of alternative donor sources for HSCT in DOCK8 deficiency. We describe HSCT for patients with DOCK8 deficiency who lack a matched related or unrelated donor using bone marrow from haploidentical related donors and post-transplantation cyclophosphamide (PT/Cy) for graft-versus-host disease (GVHD) prophylaxis. Seven patients with DOCK8 deficiency (median age, 20 years; range, 7 to 25 years) received a haploidentical related donor HSCT. The conditioning regimen included 2 days of low-dose cyclophosphamide, 5 days of fludarabine, 3 days of busulfan, and 200 cGy total body irradiation. GVHD prophylaxis consisted of PT/Cy 50 mg/kg/day on days +3 and +4 and tacrolimus and mycophenolate mofetil starting at day +5. The median times to neutrophil and platelet engraftment were 15 and 19 days, respectively. All patients attained >90% donor engraftment by day +30. Four subjects developed acute GVHD (1 with maximum grade 3). No patient developed chronic GVHD. With a median follow-up time of 20.6 months (range, 9.5 to 31.7 months), 6 of 7 patients are alive and disease free. Haploidentical related donor HSCT with PT/Cy represents an effective therapeutic approach for patients with DOCK8 deficiency who lack a matched related or unrelated donor.
Collapse
Affiliation(s)
- Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Alexandra F Freeman
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Helen Su
- Laboratory of Host Defense, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Kristen Cole
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark Parta
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Niki M Moutsopoulos
- Oral Immunity and Inflammation Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Safa Baris
- Division of Pediatric Allergy and Immunology, Ministry of Health, Marmara University, Training and Research Hospital, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Division of Pediatric Allergy and Immunology, Ministry of Health, Marmara University, Training and Research Hospital, Istanbul, Turkey
| | - Thomas E Hughes
- Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, Maryland
| | - Heidi H Kong
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Steve M Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Dennis D Hickstein
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
40
|
Connelly JA. Hematopoietic Stem Cell Transplant for a New Primary Immunodeficiency Disorder: A Voyage Where No Transplant Physician Has Gone Before. Biol Blood Marrow Transplant 2017; 23:863-864. [PMID: 28411176 DOI: 10.1016/j.bbmt.2017.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 04/07/2017] [Indexed: 01/10/2023]
Affiliation(s)
- James A Connelly
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
41
|
Dimitriades VR, Devlin V, Pittaluga S, Su HC, Holland SM, Wilson W, Dunleavy K, Shah NN, Freeman AF. DOCK 8 Deficiency, EBV+ Lymphomatoid Granulomatosis, and Intrafamilial Variation in Presentation. Front Pediatr 2017; 5:38. [PMID: 28293550 PMCID: PMC5328973 DOI: 10.3389/fped.2017.00038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/13/2017] [Indexed: 01/09/2023] Open
Abstract
Dedicator of cytokinesis 8 (DOCK8) deficiency is an autosomal recessive, combined immunodeficiency within the spectrum of hyper-IgE syndromes. Epstein-Barr virus-positive lymphomatoid granulomatosis (LYG) (EBV + LYG) is a rare diagnosis and a previously unreported presentation of DOCK8 deficiency. A 10-year-old girl was initially evaluated for mild eczema and recurrent sinopulmonary infections. She had normal immunoglobulins with elevated IgE, poor polysaccharide response with low switched memory B cells, low CD4 count, and normal mitogen and antigen responses. Despite clinical improvement following immunoglobulin replacement, a prolonged cough prompted a CT scan, which showed nodules. Biopsy identified a Grade 2 EBV + LYG. Due to an inadequate response with chemotherapy, further workup for primary immunodeficiency was performed. With her symptoms of eczema and IgE elevation, along with her brother's history of recurrent sinopulmonary infections and warts, targeted sequencing of DOCK8 was performed revealing compound heterozygous mutations for the two siblings. Both patients were successfully transplanted with resolution of the LYG and warts, respectively. This is the first reported case of LYG in DOCK8 deficiency. The EBV-driven lymphoproliferative disease along with the infection history in the brother led to the diagnosis of DOCK8 deficiency and curative hematopoietic stem cell transplants.
Collapse
Affiliation(s)
- Victoria R Dimitriades
- Department of Pediatrics, Division of Infectious Diseases, Immunology & Allergy, University of California Davis Medical Center , Sacramento, CA , USA
| | - Vincent Devlin
- Department of Pediatrics, Louisiana State University Health Sciences Center , New Orleans, LA , USA
| | | | | | | | | | | | | | | |
Collapse
|