1
|
Nguyen AA, Platt CD. Flow Cytometry-based Immune Phenotyping of T and B Lymphocytes in the Evaluation of Immunodeficiency and Immune Dysregulation. Immunol Allergy Clin North Am 2025; 45:189-203. [PMID: 40287168 DOI: 10.1016/j.iac.2025.01.008] [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: 04/29/2025]
Abstract
There are approximately 500 congenital disorders that impair immune cell development and/or function. Patients with these disorders may present with a wide range of symptoms, including increased susceptibility to infection, autoimmunity, autoinflammation, lymphoproliferation, and/or atopy. Flow cytometry-based immune phenotyping of T and B lymphocytes plays an essential role in the evaluation of patients with these presentations. In this review, we describe the clinical utility of flow cytometry as part of a comprehensive evaluation of immune function and how this testing may be used as a diagnostic tool to identify underlying aberrant immune pathways, monitor disease activity, and assess infection risk.
Collapse
Affiliation(s)
- Alan A Nguyen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Fegan Building 6th Floor, Boston, MA 02115, USA
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, 1 Blackfan Circle, Karp Building 10th Floor, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Day-Lewis ME, Berbert L, DeGrazia M, Yee C, Fried AJ, Nguyen AA, Hale JE, Counihan A, Comeau AM, Treffeisen ER, Reed MP, Platt CD, Chou J. Incidence and risk factors for CMV and EBV infection in infants with low T-cell receptor excision circles on newborn screen. Clin Immunol 2025; 277:110510. [PMID: 40316158 DOI: 10.1016/j.clim.2025.110510] [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: 02/13/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Newborn screening for severe combined immunodeficiency (SCID) using T cell receptor excision circles (TRECs) identifies patients with other causes of lymphopenia. The risk of opportunistic infection in patients with non-SCID lymphopenia is poorly understood. We aim to describe incidence and risk factors associated with cytomegalovirus (CMV) and Epstein-Barr virus (EBV) infection in patients with low TRECs. METHODS This retrospective study analyzed 289 patients with ≥1 abnormal TREC result. RESULTS Nineteen patients had CMV or EBV detected by PCR. Most had resolution of infection (n = 13). Two have chronic viremia, and four expired due to disseminated CMV. Risk factors included undetectable TRECs, consanguinity, family history, low NK, naïve CD4, naïve CD8 cells, and phytohemagglutinin. CONCLUSION Infection with CMV and EBV in patients with low TRECs is rare, however some may benefit from preventative measures. Consideration of risk factors may aid in decision-making and improve outcomes.
Collapse
Affiliation(s)
- Megan E Day-Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA.
| | - Laura Berbert
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, USA
| | - Michele DeGrazia
- Cardiovascular and Critical Care Services, Boston Children's Hospital, Boston, MA, USA; Division of Neonatology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Christina Yee
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ari J Fried
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Alan A Nguyen
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jaime E Hale
- New England Newborn Screening Program, ForHealth Consulting, UMass Chan Medical School, Worcester, MA, USA
| | - Anne Counihan
- New England Newborn Screening Program, ForHealth Consulting, UMass Chan Medical School, Worcester, MA, USA
| | - Anne Marie Comeau
- New England Newborn Screening Program, ForHealth Consulting, UMass Chan Medical School, Worcester, MA, USA; Department of Pediatrics, T.H. Chan School of Medicine, Worcester, MA, USA
| | - Elsa R Treffeisen
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Mary Poyner Reed
- Medicine Patient Services, Boston Children's Hospital, Boston, MA, USA
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Haskologlu S, Kocak S, Tufan LS, Aksoy FE, Bastug D, Oner DA, Islamoglu C, Baskin K, Esenboga S, Acican D, Ceylaner S, Guner SN, Keles S, Cagdas D, Reisli I, Tezel B, Dogu F, Tezcan I, Ikinciogullari A. Newborn screening for SCID: the very first prospective pilot study from Türkiye. Front Immunol 2024; 15:1384195. [PMID: 39483481 PMCID: PMC11526446 DOI: 10.3389/fimmu.2024.1384195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 09/02/2024] [Indexed: 11/03/2024] Open
Abstract
Purpose The measurement of T-cell receptor excision circle (TREC) is used for newborn screening (NBS) in dried blood spot (DBS) samples from Guthrie card for severe combined immunodeficiency (SCID). Here, we report the results of first newborn screening pilot program for SCID conducted in Türkiye. Methods The study was carried out together with Ankara University School of Medicine and The Ministry of Health, Public Health General Directorate, Pediatric and Adolescent Health Department. TREC measurements were performed in randomly selected Guthrie card samples obtained from 20253 babies born between October 2018 and October 2020. The TREC analyses were performed together with beta Actin (β-Actin) via RT-PCR (Real Time Polymerase Chain Reaction). Results TRECs found to be normal (≥15 copies/µl) in 98,6% of the newborns (n: 19975) but low (<15 copies/µl) in 1.4% (n:278) at the initial analyses. TRECs were retested in 278 suspected infants and found to be normal in 160 (0.8%) while low in 118 (0.58%). New DBS were obtained from the babies with low TRECs (new sample test). TRECs were normal in 108 (0.53%) of the new sample tests and low in 10 (0.049%). Two among 10 babies who had abnormal (undetectable) TRECs were diagnosed as SCID; ADA (P1) and RAG1 (P2) defects were confirmed respectively. They both received curative treatments [gene therapy (P1) and HSCT (P2)]. The remaining 6 of 8 newborns with abnormal TRECs were found normal after clinical and laboratory immune work-up, while medical records of other two revealed early postnatal death due to extreme prematurity. Conclusion In the light of this study the incidence of SCID was detected at least 1/10000 live births in Türkiye. This study shows the feasibility and usefulness of initiating SCID screening in Türkiye.
Collapse
Affiliation(s)
- Sule Haskologlu
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| | - Senem Kocak
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| | - Lale Satiroglu Tufan
- Department of Forensic Medicine, Forensic Genetics Laboratory, School of Medicine, Ankara University, Ankara, Türkiye
| | - Fethiye Eken Aksoy
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| | - Dilan Bastug
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| | - Deniz Aslar Oner
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| | - Candan Islamoglu
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| | - Kubra Baskin
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| | - Saliha Esenboga
- Department of Pediatrics, Division of Immunology, School of Medicine, Hacettepe University, Ankara, Türkiye
| | - Deniz Acican
- Pediatric and Adolescent Health Department, The Ministry of Health, Public Health General Directorate, Ankara, Türkiye
| | - Serdar Ceylaner
- Intergen Genetic and Rare Diseases Diagnosis Centre, Ankara, Türkiye
| | - Sukru Nail Guner
- Department of Pediatrics Division of Immunology and Allergy, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Türkiye
| | - Sevgi Keles
- Department of Pediatrics Division of Immunology and Allergy, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Türkiye
| | - Deniz Cagdas
- Department of Pediatrics, Division of Immunology, School of Medicine, Hacettepe University, Ankara, Türkiye
| | - Ismail Reisli
- Department of Pediatrics Division of Immunology and Allergy, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Türkiye
| | - Basak Tezel
- Pediatric and Adolescent Health Department, The Ministry of Health, Public Health General Directorate, Ankara, Türkiye
| | - Figen Dogu
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| | - Ilhan Tezcan
- Department of Pediatrics, Division of Immunology, School of Medicine, Hacettepe University, Ankara, Türkiye
| | - Aydan Ikinciogullari
- Department of Pediatrics, Division of Immunology and Allergy, School of Medicine, Ankara University, Ankara, Türkiye
| |
Collapse
|
4
|
Nguyen AA, Platt CD. Flow Cytometry-based Immune Phenotyping of T and B Lymphocytes in the Evaluation of Immunodeficiency and Immune Dysregulation. Clin Lab Med 2024; 44:479-493. [PMID: 39089753 DOI: 10.1016/j.cll.2024.04.009] [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: 08/04/2024]
Abstract
There are approximately 500 congenital disorders that impair immune cell development and/or function. Patients with these disorders may present with a wide range of symptoms, including increased susceptibility to infection, autoimmunity, autoinflammation, lymphoproliferation, and/or atopy. Flow cytometry-based immune phenotyping of T and B lymphocytes plays an essential role in the evaluation of patients with these presentations. In this review, we describe the clinical utility of flow cytometry as part of a comprehensive evaluation of immune function and how this testing may be used as a diagnostic tool to identify underlying aberrant immune pathways, monitor disease activity, and assess infection risk.
Collapse
Affiliation(s)
- Alan A Nguyen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Fegan Building 6th Floor, Boston, MA 02115, USA
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, 1 Blackfan Circle, Karp Building 10th Floor, Boston, MA 02115, USA.
| |
Collapse
|
5
|
Xiao N, Huang X, Yang D, Zang W, Kiselev S, Bolkov MA, Shinwari K, Tuzankina I, Chereshnev V. Health-related quality of life in patients with inborn errors of immunity: A systematic review and meta-analysis. Prev Med 2024; 186:108079. [PMID: 39053518 DOI: 10.1016/j.ypmed.2024.108079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Inborn Errors of Immunity (IEI) significantly affect patients' health-related quality of life (HRQOL), presenting greater challenges than those faced by the healthy population and other chronic disease sufferers. Current research lacks comprehensive integration of this critical issue. OBJECTIVE This study explores HRQOL in IEI patients, identifies impacting factors, and advocates for increased research focus on their quality of life. METHODS Following systematic review and meta-analysis guidelines, a search of Scopus and PubMed until November 15, 2023, yielded 1633 publications. We evaluated the literature, assessed study quality, and compared the HRQOL of IEI patients to that of healthy individuals and other chronic disease patients. RESULTS Of 90 articles and 10,971 IEI patients analyzed, study quality varied (nine good, 63 moderate, and 18 poor). The Short Form-36 (SF-36) and Pediatric Quality of Life Inventory generic core scales (PedsQL) were the primary generic instruments used among adults and children, respectively, with 12 studies each using the disease-specific instruments. Meta-analysis showed IEI patients have significantly lower scores in general health, physical and mental health, and social and emotional roles compared to healthy populations. We noted significant differences between self and proxy reports, indicating caregiver anxiety and perception disparities. CONCLUSION Despite limitations like small sample sizes and reliance on generic instruments, this research underscores the substantially lower HRQOL among IEI patients, emphasizing the need for a patient-centered, multidisciplinary approach to improve their life quality and calling for more focused attention on IEI patients and their caregivers' HRQOL.
Collapse
Affiliation(s)
- Ningkun Xiao
- Department of Immunochemistry, Institution of Chemical Engineering, Ural Federal University, Yekaterinburg, Russia; Laboratory for Brain and Neurocognitive Development, Department of Psychology, Institution of Humanities, Ural Federal University, Yekaterinburg, Russia.
| | - Xinlin Huang
- Laboratory for Brain and Neurocognitive Development, Department of Psychology, Institution of Humanities, Ural Federal University, Yekaterinburg, Russia.
| | - Dandan Yang
- Guang'an District Women and Children's Hospital, Guang'an, China
| | - Wanli Zang
- Postgraduate School, University of Harbin Sport, Harbin, China.
| | - Sergey Kiselev
- Laboratory for Brain and Neurocognitive Development, Department of Psychology, Institution of Humanities, Ural Federal University, Yekaterinburg, Russia.
| | - Mikhail A Bolkov
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Khyber Shinwari
- Department Biology, Nangrahar University, Nangrahar, Afghanistan
| | - Irina Tuzankina
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Valery Chereshnev
- Department of Immunochemistry, Institution of Chemical Engineering, Ural Federal University, Yekaterinburg, Russia; Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia.
| |
Collapse
|
6
|
Therrell BL, Padilla CD, Borrajo GJC, Khneisser I, Schielen PCJI, Knight-Madden J, Malherbe HL, Kase M. Current Status of Newborn Bloodspot Screening Worldwide 2024: A Comprehensive Review of Recent Activities (2020-2023). Int J Neonatal Screen 2024; 10:38. [PMID: 38920845 PMCID: PMC11203842 DOI: 10.3390/ijns10020038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 06/27/2024] Open
Abstract
Newborn bloodspot screening (NBS) began in the early 1960s based on the work of Dr. Robert "Bob" Guthrie in Buffalo, NY, USA. His development of a screening test for phenylketonuria on blood absorbed onto a special filter paper and transported to a remote testing laboratory began it all. Expansion of NBS to large numbers of asymptomatic congenital conditions flourishes in many settings while it has not yet been realized in others. The need for NBS as an efficient and effective public health prevention strategy that contributes to lowered morbidity and mortality wherever it is sustained is well known in the medical field but not necessarily by political policy makers. Acknowledging the value of national NBS reports published in 2007, the authors collaborated to create a worldwide NBS update in 2015. In a continuing attempt to review the progress of NBS globally, and to move towards a more harmonized and equitable screening system, we have updated our 2015 report with information available at the beginning of 2024. Reports on sub-Saharan Africa and the Caribbean, missing in 2015, have been included. Tables popular in the previous report have been updated with an eye towards harmonized comparisons. To emphasize areas needing attention globally, we have used regional tables containing similar listings of conditions screened, numbers of screening laboratories, and time at which specimen collection is recommended. Discussions are limited to bloodspot screening.
Collapse
Affiliation(s)
- Bradford L. Therrell
- Department of Pediatrics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
- National Newborn Screening and Global Resource Center, Austin, TX 78759, USA
| | - Carmencita D. Padilla
- Department of Pediatrics, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines;
| | - Gustavo J. C. Borrajo
- Detección de Errores Congénitos—Fundación Bioquímica Argentina, La Plata 1908, Argentina;
| | - Issam Khneisser
- Jacques LOISELET Genetic and Genomic Medical Center, Faculty of Medicine, Saint Joseph University, Beirut 1104 2020, Lebanon;
| | - Peter C. J. I. Schielen
- Office of the International Society for Neonatal Screening, Reigerskamp 273, 3607 HP Maarssen, The Netherlands;
| | - Jennifer Knight-Madden
- Caribbean Institute for Health Research—Sickle Cell Unit, The University of the West Indies, Mona, Kingston 7, Jamaica;
| | - Helen L. Malherbe
- Centre for Human Metabolomics, North-West University, Potchefstroom 2531, South Africa;
- Rare Diseases South Africa NPC, The Station Office, Bryanston, Sandton 2021, South Africa
| | - Marika Kase
- Strategic Initiatives Reproductive Health, Revvity, PL10, 10101 Turku, Finland;
| |
Collapse
|
7
|
Marakhonov AV, Efimova IY, Mukhina AA, Zinchenko RA, Balinova NV, Rodina Y, Pershin D, Ryzhkova OP, Orlova AA, Zabnenkova VV, Cherevatova TB, Beskorovainaya TS, Shchagina OA, Polyakov AV, Markova ZG, Minzhenkova ME, Shilova NV, Larin SS, Khadzhieva MB, Dudina ES, Kalinina EV, Mudaeva DA, Saydaeva DH, Matulevich SA, Belyashova EY, Yakubovskiy GI, Tebieva IS, Gabisova YV, Irinina NA, Nurgalieva LR, Saifullina EV, Belyaeva TI, Romanova OS, Voronin SV, Shcherbina A, Kutsev SI. Newborn Screening for Severe T and B Cell Lymphopenia Using TREC/KREC Detection: A Large-Scale Pilot Study of 202,908 Newborns. J Clin Immunol 2024; 44:93. [PMID: 38578360 DOI: 10.1007/s10875-024-01691-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 03/14/2024] [Indexed: 04/06/2024]
Abstract
Newborn screening (NBS) for severe inborn errors of immunity (IEI), affecting T lymphocytes, and implementing measurements of T cell receptor excision circles (TREC) has been shown to be effective in early diagnosis and improved prognosis of patients with these genetic disorders. Few studies conducted on smaller groups of newborns report results of NBS that also include measurement of kappa-deleting recombination excision circles (KREC) for IEI affecting B lymphocytes. A pilot NBS study utilizing TREC/KREC detection was conducted on 202,908 infants born in 8 regions of Russia over a 14-month period. One hundred thirty-four newborns (0.66‰) were NBS positive after the first test and subsequent retest, 41% of whom were born preterm. After lymphocyte subsets were assessed via flow cytometry, samples of 18 infants (0.09‰) were sent for whole exome sequencing. Confirmed genetic defects were consistent with autosomal recessive agammaglobulinemia in 1/18, severe combined immunodeficiency - in 7/18, 22q11.2DS syndrome - in 4/18, combined immunodeficiency - in 1/18 and trisomy 21 syndrome - in 1/18. Two patients in whom no genetic defect was found met criteria of (severe) combined immunodeficiency with syndromic features. Three patients appeared to have transient lymphopenia. Our findings demonstrate the value of implementing combined TREC/KREC NBS screening and inform the development of policies and guidelines for its integration into routine newborn screening programs.
Collapse
Affiliation(s)
| | | | - Anna A Mukhina
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | | | | | - Yulia Rodina
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Dmitry Pershin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | | | - Anna A Orlova
- Research Centre for Medical Genetics, Moscow, Russia
| | | | | | | | | | | | | | | | | | - Sergey S Larin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Maryam B Khadzhieva
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Ekaterina S Dudina
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Ekaterina V Kalinina
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | | | - Djamila H Saydaeva
- State Budgetary Institution "Maternity Hospital" of the Ministry of Healthcare of the Chechen Republic, Grozny, Russia
| | | | | | | | - Inna S Tebieva
- North-Ossetian State Medical Academy, Vladikavkaz, Russia
- Republican Childrens Clinical Hospital of the Republic of North Ossetia-Alania, Vladikavkaz, Russia
| | - Yulia V Gabisova
- Republican Childrens Clinical Hospital of the Republic of North Ossetia-Alania, Vladikavkaz, Russia
| | - Nataliya A Irinina
- State Budgetary Healthcare Institution of the Vladimir Region "Regional Clinical Hospital", Vladimir, Russia
| | | | | | - Tatiana I Belyaeva
- Clinical Diagnostic Center "Maternal and Child Health", Yekaterinburg, Russia
| | - Olga S Romanova
- Clinical Diagnostic Center "Maternal and Child Health", Yekaterinburg, Russia
| | | | - Anna Shcherbina
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | | |
Collapse
|
8
|
Xiao N, Huang X, Zang W, Kiselev S, Bolkov MA, Tuzankina IA, Chereshnev VA. Health-related quality of life in patients with inborn errors of immunity: a bibliometric analysis. Front Immunol 2024; 15:1371124. [PMID: 38515759 PMCID: PMC10954858 DOI: 10.3389/fimmu.2024.1371124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Background Inborn Errors of Immunity (IEI) are characterized by a heightened susceptibility to infections, allergies, and various other health complications. Health-Related Quality of Life (HRQOL) in patients with IEI is a critical area of research that demands attention due to the impact of IEI on patients' lives. This study utilized bibliometric methods, aiming to comprehensively explore the research content and hotspots in the field of HRQOL in patients with IEI. Methods This bibliometric analysis utilized data from the Science Citation Index Expanded (SCIE) and Social Sciences Citation Index (SSCI) within the Web of Science core datasets up to January 1, 2024. The study focused on literature that addressed HRQOL in IEI patients, involving a total of 1,807 authors and 309 articles published across 112 journals. The analysis included publication volume and growth trends, country and institutional contributions, authorship, and journal analysis. Results The research found that despite the importance of HRQOL in IEI, the volume of publications in this field remains consistently low, with no significant increase in trend. The USA leads in publication and citation volumes, reflecting a geographical imbalance in research contributions. Key journals in this field include the Journal of Clinical Immunology, Frontiers in Immunology, and the Journal of Allergy and Clinical Immunology. The study highlights that while treatments like hematopoietic stem cell transplants and gene therapy have improved patient IEI survival rates, they still often come with significant side effects impacting HRQOL. The analysis underlines the need for comprehensive HRQOL assessments in IEI, considering the physical and psychological impacts of treatments. Conclusions This study represents a bibliometric analysis focusing on HRQOL in patients with. It underscores the need for more extensive and systematic research in this area, emphasizing the importance of a multidisciplinary approach. Despite advancements in medical treatments for IEI, there is a crucial need to focus on HRQOL to enhance patient satisfaction and overall well-being. The findings advocate for more personalized treatment plans and a better understanding of the psychosocial needs of patients with IEI to improve their quality of life.
Collapse
Affiliation(s)
- Ningkun Xiao
- Department of Immunochemistry, Institution of Chemical Engineering, Ural Federal University, Yekaterinburg, Russia
- Laboratory for Brain and Neurocognitive Development, Department of Psychology, Institution of Humanities, Ural Federal University, Yekaterinburg, Russia
| | - Xinlin Huang
- Laboratory for Brain and Neurocognitive Development, Department of Psychology, Institution of Humanities, Ural Federal University, Yekaterinburg, Russia
| | - Wanli Zang
- Postgraduate School, University of Harbin Sport, Harbin, China
| | - Sergey Kiselev
- Laboratory for Brain and Neurocognitive Development, Department of Psychology, Institution of Humanities, Ural Federal University, Yekaterinburg, Russia
| | - Mikhail A. Bolkov
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Irina A. Tuzankina
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Valery A. Chereshnev
- Department of Immunochemistry, Institution of Chemical Engineering, Ural Federal University, Yekaterinburg, Russia
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| |
Collapse
|
9
|
Sonoda M, Ishimura M, Inoue H, Eguchi K, Ochiai M, Sakai Y, Doi T, Suzuki K, Inoue T, Mizukami T, Nakamura K, Takada H, Ohga S. Non-conditioned cord blood transplantation for infection control in athymic CHARGE syndrome. Pediatr Blood Cancer 2024; 71:e30809. [PMID: 38078568 DOI: 10.1002/pbc.30809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/04/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024]
Abstract
OBJECTIVE CHARGE syndrome is a congenital malformation syndrome caused by heterozygous mutations in the CHD7 gene. Severe combined immunodeficiency (SCID) arises from congenital athymia called CHARGE/complete DiGeorge syndrome. While cultured thymus tissue implantation (CTTI) provides an immunological cure, hematopoietic cell transplantation (HCT) is an alternative option for immuno-reconstitution of affected infants. We aimed to clarify the clinical outcomes of patients with athymic CHARGE syndrome after HCT. METHODS We studied the immunological reconstitution and outcomes of four patients who received non-conditioned unrelated donor cord blood transplantation (CBT) at Kyushu University Hospital from 2007 to 2022. The posttransplant outcomes were compared with the outcomes of eight reported patients. RESULTS Four index cases received CBT 70-144 days after birth and had no higher than grade II acute graft-versus-host disease. One infant was the first newborn-screened athymic case in Japan. They achieved more than 500/μL naïve T cells with balanced repertoire 1 month post transplant, and survived more than 12 months with home care. Twelve patients including the index cases received HCT at a median 106 days after birth (range: 70-195 days). One-year overall survival rate was significantly higher in patients who underwent non-conditioned HCT than in those who received conditioned HCT (100% vs. 37.5%, p = .02). Nine patients died, and the major cause of death was cardiopulmonary failure. CONCLUSIONS Athymic infants achieved a prompt reconstitution of non-skewing naïve T cells after non-conditioned CBT that led to home care in infancy without significant infections. Non-conditioned CBT is a useful bridging therapy for newborn-screened cases toward an immunological cure by CTTI.
Collapse
Affiliation(s)
- Motoshi Sonoda
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ishimura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirosuke Inoue
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuhide Eguchi
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masayuki Ochiai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takehiko Doi
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kyoko Suzuki
- Department of Pediatrics, Juntendo University, Urayasu Hospital, Chiba, Japan
| | - Takeshi Inoue
- Division of Neonatology, Perinatal Center, Kumamoto City Hospital, Kumamoto, Japan
| | - Tomoyuki Mizukami
- Department of Pediatrics, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Hidetoshi Takada
- Department of Child Health, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
10
|
Soomann M, Prader S, Pinto Monteiro A, Zeilhofer U, Hauri-Hohl M, Güngör T, Pachlopnik Schmid J, Trück J, Felber M. Reducing Mortality and Morbidity in Children with Severe Combined Immunodeficiency in Switzerland: the Role of Newborn Screening. J Clin Immunol 2024; 44:39. [PMID: 38165471 PMCID: PMC10761526 DOI: 10.1007/s10875-023-01640-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Newborn screening (NBS) for severe combined immunodeficiency (SCID) has been introduced in various countries with the aim of reducing morbidity and mortality. However, studies analyzing outcomes before and after the implementation of NBS programs remain limited. This study sought to compare the outcomes of SCID patients identified through Switzerland's national SCID NBS program, introduced in January 2019, with those of a historical cohort diagnosed between 2007 and 2019. The study included seven patients (32%) identified through NBS, and 15 (68%) born before NBS implementation and diagnosed based on clinical signs. Children in the NBS group were younger at diagnosis (median age 9 days vs 9 months, P = .002) and at hematopoietic stem cell transplantation (HSCT, median age 5 months vs 11 months, P = .003) compared to the clinical group. The NBS group had a lower incidence of infections before HSCT (29% vs 93%, P = .004). Although not statistically significant, the overall survival rate on last follow-up was higher in the NBS group (86% vs 67%, P = .62). Importantly, patients with active infections undergoing HSCT had a significantly lower overall survival probability compared to those without (P = .01). In conclusion, the introduction of NBS in Switzerland has led to earlier and often asymptomatic diagnosis of affected children, enabling timely intervention, infection prevention, and prompt treatment. These factors have contributed to higher survival rates in the NBS group. These findings underscore the critical importance of NBS for SCID, offering potential life-saving benefits through early detection and intervention.
Collapse
Affiliation(s)
- Maarja Soomann
- Division of Immunology and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Steinwiesstrasse 75, 8032, Zurich, Switzerland.
| | - Seraina Prader
- Division of Immunology and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Steinwiesstrasse 75, 8032, Zurich, Switzerland
| | - Aline Pinto Monteiro
- Division of Stem Cell Transplantation and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ulrike Zeilhofer
- Division of Stem Cell Transplantation and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mathias Hauri-Hohl
- Division of Stem Cell Transplantation and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Tayfun Güngör
- Division of Stem Cell Transplantation and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jana Pachlopnik Schmid
- Division of Immunology and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Steinwiesstrasse 75, 8032, Zurich, Switzerland
| | - Johannes Trück
- Division of Immunology and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Steinwiesstrasse 75, 8032, Zurich, Switzerland
| | - Matthias Felber
- Division of Stem Cell Transplantation and the Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Gaviglio A, Lasarev M, Sheller R, Singh S, Baker M. Newborn Screening for Severe Combined Immunodeficiency: Lessons Learned from Screening and Follow-Up of the Preterm Newborn Population. Int J Neonatal Screen 2023; 9:68. [PMID: 38132827 PMCID: PMC10744167 DOI: 10.3390/ijns9040068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Newborn screening (NBS) for Severe Combined Immunodeficiency (SCID) by measurement of T-cell receptor excision circles (TRECs) successfully identifies newborns with SCID and severe T-cell lymphopenia, as intended. At the same time, NBS programs face the challenge of false positive results, with a disproportionately high number in the premature newborn population. This study evaluates TREC values and SCID screening outcomes in premature newborns and elucidates evidence-based SCID screening practices that reduce unnecessary follow-up activities in this population. De-identified individual SCID newborn screening data and aggregate SCID screening data were obtained from seven states across the US for babies born between 2018 and 2020. Relevant statistics were performed on data pooled from these states to quantify screening performance metrics and clinical impact on various birth and gestational age categories of newborns. The data were normalized using multiples-of-the-median (MoM) values to allow for the aggregation of data across states. The aggregation of NBS data across a range of NBS programs highlighted the trajectory of TREC values over time, both between and within newborns, and provides evidence for improved SCID screening recommendations in the premature and low birth weight population.
Collapse
Affiliation(s)
- Amy Gaviglio
- Association of Public Health Laboratories, Silver Spring, MD 20910, USA; (A.G.)
| | - Michael Lasarev
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA;
| | - Ruthanne Sheller
- Association of Public Health Laboratories, Silver Spring, MD 20910, USA; (A.G.)
| | - Sikha Singh
- Association of Public Health Laboratories, Silver Spring, MD 20910, USA; (A.G.)
| | - Mei Baker
- Wisconsin State Laboratory of Hygiene, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA;
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
| |
Collapse
|
12
|
Lev A, Somech R, Somekh I. Newborn screening for severe combined immunodeficiency and inborn errors of immunity. Curr Opin Pediatr 2023; 35:692-702. [PMID: 37707504 DOI: 10.1097/mop.0000000000001291] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
PURPOSE OF REVIEW Severe combined immune deficiency (SCID) is the most devastating genetic disease of the immune system with an unfavorable outcome unless diagnosed early in life. Newborn screening (NBS) programs play a crucial role in facilitating early diagnoses and timely interventions for affected infants. RECENT FINDINGS SCID marked the pioneering inborn error of immunity (IEI) to undergo NBS, a milestone achieved 15 years ago through the enumeration of T-cell receptor excision circles (TRECs) extracted from Guthrie cards. This breakthrough has revolutionized our approach to SCID, enabling not only presymptomatic identification and prompt treatments (including hematopoietic stem cell transplantation), but also enhancing our comprehension of the global epidemiology of SCID. SUMMARY NBS is continuing to evolve with the advent of novel diagnostic technologies and treatments. Following the successful implementation of SCID-NBS programs, a call for the early identification of additional IEIs is the next step, encompassing a broader spectrum of IEIs, facilitating early diagnoses, and preventing morbidity and mortality.
Collapse
Affiliation(s)
- Atar Lev
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | | | | |
Collapse
|
13
|
Staudacher O, Klein J, Thee S, Ullrich J, Wahn V, Unterwalder N, Kölsch U, Lankes E, Stittrich A, Dedieu C, Dinges S, Völler M, Schuetz C, Schulte J, Boztug K, Meisel C, Kuehl JS, Krüger R, Blankenstein O, von Bernuth H. Screening Newborns for Low T Cell Receptor Excision Circles (TRECs) Fails to Detect Immunodeficiency, Centromeric Instability, and Facial Anomalies Syndrome. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2872-2883. [PMID: 37302792 DOI: 10.1016/j.jaip.2023.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Assessment of T-cell receptor excision circles (TRECs) in dried blood spots of newborns allows the detection of severe combined immunodeficiency (SCID) (T cells <300/μL at birth) with a presumed sensitivity of 100%. TREC screening also identifies patients with selected combined immunodeficiency (CID) (T cells >300/μL, yet <1500/μL at birth). Nevertheless, relevant CIDs that would benefit from early recognition and curative treatment pass undetected. OBJECTIVE We hypothesized that TREC screening at birth cannot identify CIDs that develop with age. METHODS We analyzed the number of TRECs in dried blood spots in archived Guthrie cards of 22 children who had been born in the Berlin-Brandenburg area between January 2006 and November 2018 and who had undergone hematopoietic stem-cell transplantation (HSCT) for inborn errors of immunity. RESULTS All patients with SCID would have been identified by TREC screening, but only 4 of 6 with CID. One of these patients had immunodeficiency, centromeric instability, and facial anomalies syndrome type 2 (ICF2). Two of 3 patients with ICF whom we have been following up at our institution had TREC numbers above the cutoff value suggestive of SCID at birth. Yet all patients with ICF had a severe clinical course that would have justified earlier HSCT. CONCLUSIONS In ICF, naïve T cells may be present at birth, yet they decline with age. Therefore, TREC screening cannot identify these patients. Early recognition is nevertheless crucial, as patients with ICF benefit from HSCT early in life.
Collapse
Affiliation(s)
- Olga Staudacher
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Jeanette Klein
- Newborn Screening Laboratory, Charité Universitätsmedizin, Berlin, Germany
| | - Stephanie Thee
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan Ullrich
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Volker Wahn
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nadine Unterwalder
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Uwe Kölsch
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Erwin Lankes
- Newborn Screening Laboratory, Charité Universitätsmedizin, Berlin, Germany; Department of Pediatric Endocrinology, Charité-Uninrsitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anna Stittrich
- Department of Human Genetics, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Cinzia Dedieu
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sarah Dinges
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mirjam Völler
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Catharina Schuetz
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Johannes Schulte
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; St. Anna Children's Cancer Research Institute, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Christian Meisel
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany; Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörn-Sven Kuehl
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Pediatric Oncology, Hematology and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Renate Krüger
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Horst von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany; Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.
| |
Collapse
|
14
|
Korkmaz SB, Karaselek MA, Aytekin SE, Tokgoz H, Reisli I, Guner S, Keles S. Retrospective analysis of patients with severe combined immunodeficiency and alternative diagnostic criteria: A 20-year single centre experience. Int J Immunogenet 2023; 50:177-184. [PMID: 37308802 DOI: 10.1111/iji.12624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/03/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023]
Abstract
Severe combined immunodeficiency (SCID) is an inborn errors of immunity (IEI) disorder characterized by impairment in the development and function of lymphocytes and could be fatal if not treated with hematopoietic stem cell transplant in the first 2 years of life. There are various diagnostic criteria for SCID among different primary immunodeficiency societies. We retrospectively evaluated clinical and laboratory findings of 59 patients followed up with the diagnosis of SCID at our clinic over the past 20 years in order to develop an algorithm that would help diagnosis of SCID for the countries where a high ratio of consanguineous marriage is present because these countries have not launched TREC assay in their newborn screening programs. The mean age at diagnosis was 5.80 ± 4.90 months, and the delay was 3.29 ± 3.99 months. The most common complaint and physical examination findings were cough (29.05%), eczematous rash (63%) and organomegaly (61%). ADA (17%), Artemis (14%), RAG1/2 (15%), MHC Class II (12%) and IL-2R (12%) deficiencies were the most common genetic defects. Lymphopenia (87.5%) was the most frequent abnormal laboratory finding and below 3000/mm3 in 95% of the patients. The CD3+ T cell count was 300/mm3 and below in 83% of the patients. As a result, a combination of low lymphocyte count and CD3 lymphopenia for SCID diagnosis would be more reliable for countries with high rate of consanguineous marriage. Physicians should consider diagnosis of SCID in a patient presenting with severe infections and lymphocyte counts below 3000/mm3 under 2 years of age.
Collapse
Affiliation(s)
- Sevim Busra Korkmaz
- Meram Medicine Faculty, Department of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Mehmet Ali Karaselek
- Meram Medicine Faculty, Department of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Selma Erol Aytekin
- Meram Medicine Faculty, Department of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Huseyin Tokgoz
- Meram Medicine Faculty, Department of Pediatric Hematology, Necmettin Erbakan University, Konya, Turkey
| | - Ismail Reisli
- Meram Medicine Faculty, Department of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Sukru Guner
- Meram Medicine Faculty, Department of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| | - Sevgi Keles
- Meram Medicine Faculty, Department of Pediatric Immunology and Allergy, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
15
|
Bosticardo M, Notarangelo LD. Human thymus in health and disease: Recent advances in diagnosis and biology. Semin Immunol 2023; 66:101732. [PMID: 36863139 PMCID: PMC10134747 DOI: 10.1016/j.smim.2023.101732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/30/2023] [Accepted: 02/14/2023] [Indexed: 03/04/2023]
Abstract
The thymus is the crucial tissue where thymocytes develop from hematopoietic precursors that originate from the bone marrow and differentiate to generate a repertoire of mature T cells able to respond to foreign antigens while remaining tolerant to self-antigens. Until recently, most of the knowledge on thymus biology and its cellular and molecular complexity have been obtained through studies in animal models, because of the difficulty to gain access to thymic tissue in humans and the lack of in vitro models able to faithfully recapitulate the thymic microenvironment. This review focuses on recent advances in the understanding of human thymus biology in health and disease obtained through the use of innovative experimental techniques (eg. single cell RNA sequencing, scRNAseq), diagnostic tools (eg. next generation sequencing), and in vitro models of T-cell differentiation (artificial thymic organoids) and thymus development (eg. thymic epithelial cell differentiation from embryonic stem cells or induced pluripotent stem cells).
Collapse
Affiliation(s)
- Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Al-Tamemi S, Al-Zadjali S, Bruwer Z, Naseem SUR, Al-Siyabi N, ALRawahi M, Alkharusi K, Al-Thihli K, Al-Murshedi F, AlSayegh A, Al-Maawali A, Dennison D. Genetic Causes, Clinical Features, and Survival of Underlying Inborn Errors of Immunity in Omani Patients: a Single-Center Study. J Clin Immunol 2023; 43:452-465. [PMID: 36324046 DOI: 10.1007/s10875-022-01394-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Early identification of inborn errors of immunity (IEIs) is crucial due to the significant risk of morbidity and mortality. This study aimed to describe the genetic causes, clinical features, and survival rate of IEIs in Omani patients. METHODS A prospective study of all Omani patients evaluated for immunodeficiency was conducted over a 17-year period. Clinical features and diagnostic immunological findings were recorded. Targeted gene testing was performed in cases of obvious immunodeficiency. For cases with less conclusive phenotypes, a gene panel was performed, followed by whole-exome sequencing if necessary. RESULTS A total of 185 patients were diagnosed with IEIs during the study period; of these, 60.5% were male. Mean ages at symptom onset and diagnosis were 30.0 and 50.5 months, respectively. Consanguinity and a family history of IEIs were present in 86.9% and 50.8%, respectively. Most patients presented with lower respiratory infections (65.9%), followed by growth and development manifestations (43.2%). Phagocytic defects were the most common cause of IEIs (31.9%), followed by combined immunodeficiency (21.1%). Overall, 109 of 132 patients (82.6%) who underwent genetic testing received a genetic diagnosis, while testing was inconclusive for the remaining 23 patients (17.4%). Among patients with established diagnoses, 37 genes and 44 variants were identified. Autosomal recessive inheritance was present in 81.7% of patients with gene defects. Several variants were novel. Intravenous immunoglobulin therapy was administered to 39.4% of patients and 21.6% received hematopoietic stem cell transplantation. The overall survival rate was 75.1%. CONCLUSION This study highlights the genetic causes of IEIs in Omani patients. This information may help in the early identification and management of the disease, thereby improving survival and quality of life.
Collapse
Affiliation(s)
- Salem Al-Tamemi
- Clinical Immunology & Allergy Unit, Department of Child Health, Sultan Qaboos University Hospital, Muscat, Oman.
| | - Shoaib Al-Zadjali
- Molecular Hematology Unit, Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Zandre Bruwer
- Department of Clinical Genetics, Sultan Qaboos University Hospital, Muscat, Oman
| | - Shafiq-Ur-Rehman Naseem
- Clinical Immunology & Allergy Unit, Department of Child Health, Sultan Qaboos University Hospital, Muscat, Oman
| | - Nabila Al-Siyabi
- Clinical Immunology & Allergy Unit, Directorate of Nursing, Sultan Qaboos University Hospital, Muscat, Oman
| | - Mohammed ALRawahi
- Molecular Hematology Unit, Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Khalsa Alkharusi
- Department of Clinical Genetics, Sultan Qaboos University Hospital, Muscat, Oman
| | - Khalid Al-Thihli
- Department of Clinical Genetics, Sultan Qaboos University Hospital, Muscat, Oman
| | - Fathiya Al-Murshedi
- Department of Clinical Genetics, Sultan Qaboos University Hospital, Muscat, Oman
| | - Abeer AlSayegh
- Department of Clinical Genetics, Sultan Qaboos University Hospital, Muscat, Oman
| | - Almundher Al-Maawali
- Department of Clinical Genetics, Sultan Qaboos University Hospital, Muscat, Oman
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - David Dennison
- Molecular Hematology Unit, Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
17
|
Walter JE, Ziegler JB, Ballow M, Cunningham-Rundles C. Advances and Challenges of the Decade: The Ever-Changing Clinical and Genetic Landscape of Immunodeficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:107-115. [PMID: 36610755 DOI: 10.1016/j.jaip.2022.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 01/06/2023]
Abstract
In the past 10 years, we have witnessed major advances in clinical immunology. Newborn screening for severe combined immunodeficiency has become universal in the United States and screening programs are being extended to severe combined immunodeficiency and other inborn errors of immunity globally. Early genetic testing is becoming the norm for many of our patients and allows for informed selection of targeted therapies including biologics repurposed from other specialties. During the COVID-19 pandemic, our understanding of essential immune responses expanded and the discovery of immune gene defects continued. Immunoglobulin products, the backbone of protection for antibody deficiency syndromes, came into use to minimize side effects. New polyclonal and monoclonal antibody products emerged with increasing options to manage respiratory viral agents such as SARS-CoV-2 and respiratory syncytial virus. Against these advances, we still face major challenges. Atypical is becoming typical as phenotypes of distinct genetic disease overlap whereas the clinical spectrum of the same genetic defect widens. Therefore, clinical judgment needs to be paired with repeated deep immune phenotyping and upfront genetic testing, as technologies rapidly evolve, and clinical disease often progresses with age. Managing patients with organ damage resulting from immune dysregulation poses a special major clinical challenge and management often lacks standardization, from autoimmune cytopenias, granulomatous interstitial lung disease, enteropathy, and liver disease to endocrine, rheumatologic, and neurologic complications. Clinical, translational, and basic science networks will continue to advance the field; however, cross-talk and education with practicing allergists/immunologists are essential to keep up with the ever-changing clinical and genetic landscape of inborn errors of immunity.
Collapse
Affiliation(s)
- Jolan E Walter
- Division of Pediatric Allergy and Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St Petersburg, Fla; Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston, Mass.
| | - John B Ziegler
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia; Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - Mark Ballow
- Department of Pediatrics, Division of Allergy and Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St Petersburg, Fla
| | | |
Collapse
|
18
|
Puck JM. A Spot of Good News: Israeli Experience With SCID Newborn Screening. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:2732-2733. [PMID: 36216463 PMCID: PMC9831241 DOI: 10.1016/j.jaip.2022.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022]
Affiliation(s)
- Jennifer M Puck
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics, San Francisco School of Medicine, University of California, San Francisco, Calif.
| |
Collapse
|
19
|
Lev A, Sharir I, Simon AJ, Levy S, Lee YN, Frizinsky S, Daas S, Saraf-Levy T, Broides A, Nahum A, Hanna S, Stepensky P, Toker O, Dalal I, Etzioni A, Stein J, Adam E, Hendel A, Marcus N, Almashanu S, Somech R. Lessons Learned From Five Years of Newborn Screening for Severe Combined Immunodeficiency in Israel. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:2722-2731.e9. [PMID: 35487367 DOI: 10.1016/j.jaip.2022.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/03/2022] [Accepted: 04/05/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Implementation of newborn screening (NBS) programs for severe combined immunodeficiency (SCID) have advanced the diagnosis and management of affected infants and undoubtedly improved their outcomes. Reporting long-term follow-up of such programs is of great importance. OBJECTIVE We report a 5-year summary of the NBS program for SCID in Israel. METHODS Immunologic and genetic assessments, clinical analyses, and outcome data from all infants who screened positive were evaluated and summarized. RESULTS A total of 937,953 Guthrie cards were screened for SCID. A second Guthrie card was requested on 1,169 occasions (0.12%), which resulted in 142 referrals (0.015%) for further validation tests. Flow cytometry immune-phenotyping, T cell receptor excision circle measurement in peripheral blood, and expression of TCRVβ repertoire for the validation of positive cases revealed a specificity and sensitivity of 93.7% and 75.9%, respectively, in detecting true cases of SCID. Altogether, 32 SCID and 110 non-SCID newborns were diagnosed, making the incidence of SCID in Israel as high as 1:29,000 births. The most common genetic defects in this group were associated with mutations in DNA cross-link repair protein 1C and IL-7 receptor α (IL-7Rα) genes. No infant with SCID was missed during the study time. Twenty-two SCID patients underwent hematopoietic stem cell transplantation, which resulted in a 91% survival rate. CONCLUSIONS Newborn screening for SCID should ultimately be applied globally, specifically to areas with high rates of consanguineous marriages. Accumulating data from follow-up studies on NBS for SCID will improve diagnosis and treatment and enrich our understanding of immune development in health and disease.
Collapse
Affiliation(s)
- Atar Lev
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Mina and Everard Goodman Faculty of Life Sciences, Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan, Israel
| | - Idan Sharir
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Amos J Simon
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Hemato-Immunology Unit, Hematology Lab, Sheba Medical Center, Tel HaShomer, Israel
| | - Shiran Levy
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yu Nee Lee
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shirly Frizinsky
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Suha Daas
- National Newborn Screening Program, Ministry of Health, Tel-HaShomer, Israel
| | - Talia Saraf-Levy
- National Newborn Screening Program, Ministry of Health, Tel-HaShomer, Israel
| | - Arnon Broides
- Pediatric Immunology, Soroka University Medical Center, Beer-Sheva, Israel; Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY
| | - Amit Nahum
- Pediatric Immunology, Soroka University Medical Center, Beer-Sheva, Israel; Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Primary Immunodeficiency Research Laboratory, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Suhair Hanna
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Ruth Children Hospital, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Polina Stepensky
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ori Toker
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Faculty of Medicine, Hebrew University of Jerusalem, Israel; Allergy and Immunology Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Ilan Dalal
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Department of Pediatrics, Pediatric Allergy Unit, E. Wolfson Medical Center, Holon, Israel, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Amos Etzioni
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Ruth Children Hospital, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Jerry Stein
- Department for Hemato-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Etai Adam
- Division of Pediatric Hematology and Oncology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Ayal Hendel
- Mina and Everard Goodman Faculty of Life Sciences, Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan, Israel
| | - Nufar Marcus
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Allergy and Immunology Unit, Schneider Children's Medical Center of Israel, Felsenstein Medical Research Center, Kipper Institute of Immunology, Petach Tikva, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Shlomo Almashanu
- National Newborn Screening Program, Ministry of Health, Tel-HaShomer, Israel.
| | - Raz Somech
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; National Lab for Confirming Primary Immunodeficiency in Newborn Screening Center for Newborn Screening, Ministry of Health, Tel HaShomer, Israel.
| |
Collapse
|
20
|
Yilmaz M, Potts DE, Geier C, Walter JE. Can we identify WHIM in infancy? Opportunities with the public newborn screening process. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:215-221. [PMID: 36210583 DOI: 10.1002/ajmg.c.32002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 06/16/2023]
Abstract
Newborn screening (NBS) for severe combined immunodeficiency (SCID) utilizing T-cell receptor excision circles (TRECs) has been implemented in all 50 states as of December 2018 and has been transformative for the clinical care of SCID patients. Though having high sensitivity for SCID, NBS-SCID has low specificity, therefore is able to detect other causes of lymphopenia in newborns including many inborn errors of immunity (IEIs). In a recent study, three of six newborns later diagnosed with Warts, Hypogammaglobulinemia, Infections, and Myelokathexis (WHIM) syndrome were found to have a low TRECs and lymphopenia at birth. This presents an opportunity to increase the detection and diagnosis of WHIM syndrome by NBS-SCID with immunological follow-up along with a combination of flow cytometry for immune cell subsets, absolute neutrophil count, and genetic testing, extending beyond the conventional bone marrow studies. Coupled with emerging technologies such as next-generation sequencing, transcriptomics and proteomics, dried blood spots used in NBS-SCID will promote earlier detection, diagnosis, and therefore treatment of IEIs such as WHIM syndrome.
Collapse
Affiliation(s)
- Melis Yilmaz
- Division of Allergy and Immunology, Department of Pediatrics and Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| | - David Evan Potts
- Division of Allergy and Immunology, Department of Pediatrics and Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| | - Christoph Geier
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
| | - Jolan E Walter
- Division of Allergy and Immunology, Department of Pediatrics and Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
- Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Habiballah SB, Whangbo JS, Cardona ID, Platt CD. Spontaneous resolution of severe idiopathic T cell lymphopenia. Clin Immunol 2022; 238:109014. [PMID: 35447312 DOI: 10.1016/j.clim.2022.109014] [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: 03/12/2022] [Revised: 04/10/2022] [Accepted: 04/10/2022] [Indexed: 11/03/2022]
Abstract
Potential etiologies of T-B + NK+ SCID include both hematopoietic defects and thymic aplasia. The management of patients with this phenotype, identified by newborn screen, may be unclear in the absence of a genetic diagnosis. We report an infant with lymphocyte flow cytometry consistent with T-B + NK+ SCID and reduced proliferative response to phytohemagglutinin. The patient had no genetic diagnosis after targeted panel and exome sequencing. The decision to trend laboratory values rather than move immediately to hematopoietic cell transplant was made given the absence of a genetic defect and the finding of a normal thymus on ultrasound. During the course of evaluation for transplant, the patient unexpectedly had normalization of T cell number and function. This case demonstrates a role for mediastinal ultrasound and the utility of trending laboratory values in patients with severe T cell lymphopenia but no genetic diagnosis, given the small but important possibility of spontaneous resolution.
Collapse
Affiliation(s)
- Saddiq B Habiballah
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States of America.
| | - Jennifer S Whangbo
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, MA, United States of America; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Ivan D Cardona
- Department of Pediatrics, Maine Medical Center Research Institute, Portland, ME, United States of America
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
22
|
Abstract
Over the past 20 years, the rapid evolution in the diagnosis and treatment of primary immunodeficiencies (PI) and the recognition of immune dysregulation as a feature in some have prompted the use of "inborn errors of immunity" (IEI) as a more encompassing term used to describe these disorders [1, 2] . This article aims to review the future of therapy of PI/IEI (referred to IEI throughout this paper). Historically, immune deficiencies have been characterized as monogenic disorders resulting in immune deficiencies affecting T cells, B cells, combination of T and B cells, or innate immune disorders. More recently, immunologists are also recognizing a variety of phenotypes associated with one genotype or similar phenotypes across genotypes and a role for incomplete penetrance or variable expressivity of some genes causing inborn errors of immunity [3]. The IUIS classification of immune deficiencies (IEIs) has evolved over time to include 10 categories, with disorders of immune dysregulation accounting for a new subset, some treatable with small molecule inhibitors or biologics. [1] Until recently, management options were limited to prompt treatment of infections, gammaglobulin replacement, and possibly bone marrow transplant depending on the defect. Available therapies have expanded to include small molecule inhibitors, biologics, gene therapy, and the use of adoptive transfer of virus-specific T cells to fight viral infections in immunocompromised patients. Several significant contributions to the field of clinical immunology have fueled the rapid advancement of therapies over the past two decades. Among these are educational efforts to recruit young immunologists to the field resulting in the growth of a world-wide community of clinicians and investigators interested in rare diseases, efforts to increase awareness of IEI globally contributing to international collaborations, along with advancements in diagnostic genetic testing, newborn screening, molecular biology techniques, gene correction, use of immune modulators, and ex vivo expansion of engineered T cells for therapeutic use. The development and widespread use of newborn screening have helped to identify severe combined immune deficiency (SCID) earlier resulting in better outcomes [4]. Continual improvements and accessibility of genetic sequencing have helped to identify new IEI diseases at an accelerated pace [5]. Advances in gene therapy and bone marrow transplant have made treatments possible in otherwise fatal diseases. Furthermore, the increased awareness of IEI across the world has driven networks of immunologists working together to improve the diagnosis and treatment of these rare diseases. These improvements in the diagnosis and treatment of IEI noted over the past 20 years bring hope for a better future for the IEI community. This paper will review future directions in a few of the newer therapies emerging for IEI. For easy reference, most of the diseases discussed in this paper are briefly described in a summary table, in the order mentioned within the paper (Appendix).
Collapse
Affiliation(s)
- Elena Perez
- Allergy Associates of the Palm Beaches, North Palm Beach, FL, USA.
| |
Collapse
|
23
|
Blom M, Pico-Knijnenburg I, Imholz S, Vissers L, Schulze J, Werner J, Bredius R, van der Burg M. Second Tier Testing to Reduce the Number of Non-actionable Secondary Findings and False-Positive Referrals in Newborn Screening for Severe Combined Immunodeficiency. J Clin Immunol 2021; 41:1762-1773. [PMID: 34370170 PMCID: PMC8604867 DOI: 10.1007/s10875-021-01107-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/20/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Newborn screening (NBS) for severe combined immunodeficiency (SCID) is based on the detection of T-cell receptor excision circles (TRECs). TRECs are a sensitive biomarker for T-cell lymphopenia, but not specific for SCID. This creates a palette of secondary findings associated with low T-cells that require follow-up and treatment or are non-actionable. The high rate of (non-actionable) secondary findings and false-positive referrals raises questions about the harm-benefit-ratio of SCID screening, as referrals are associated with high emotional impact and anxiety for parents. METHODS An alternative quantitative TREC PCR with different primers was performed on NBS cards of referred newborns (N = 56) and epigenetic immune cell counting was used as for relative quantification of CD3 + T-cells (N = 59). Retrospective data was used to determine the reduction in referrals with a lower TREC cutoff value or an adjusted screening algorithm. RESULTS When analyzed with a second PCR with different primers, 45% of the referrals (25/56) had TREC levels above cutoff, including four false-positive cases in which two SNPs were identified. With epigenetic qPCR, 41% (24/59) of the referrals were within the range of the relative CD3 + T-cell counts of the healthy controls. Lowering the TREC cutoff value or adjusting the screening algorithm led to lower referral rates but did not prevent all false-positive referrals. CONCLUSIONS Second tier tests and adjustments of cutoff values or screening algorithms all have the potential to reduce the number of non-actionable secondary findings in NBS for SCID, although second tier tests are more effective in preventing false-positive referrals.
Collapse
Affiliation(s)
- Maartje Blom
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Ingrid Pico-Knijnenburg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Sandra Imholz
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Lotte Vissers
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Janika Schulze
- Department of Research and Development, Epimune GmbH, Belin, Germany
| | - Jeannette Werner
- Department of Research and Development, Epimune GmbH, Belin, Germany
| | - Robbert Bredius
- Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Mirjam van der Burg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands.
| |
Collapse
|
24
|
Mantravadi V, Bednarski JJ, Ritter MA, Gu H, Kolicheski AL, Horner C, Cooper MA, Kitcharoensakkul M. Immunological Findings and Clinical Outcomes of Infants With Positive Newborn Screening for Severe Combined Immunodeficiency From a Tertiary Care Center in the U.S. Front Immunol 2021; 12:734096. [PMID: 34539671 PMCID: PMC8446381 DOI: 10.3389/fimmu.2021.734096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/19/2021] [Indexed: 02/03/2023] Open
Abstract
The implementation of severe combined immunodeficiency (SCID) newborn screening has played a pivotal role in identifying these patients early in life as well as detecting various milder forms of T cell lymphopenia (TCL). In this study we reviewed the diagnostic and clinical outcomes, and interesting immunology findings of term infants referred to a tertiary care center with abnormal newborn SCID screens over a 6-year period. Key findings included a 33% incidence of non-SCID TCL including infants with novel variants in FOXN1, TBX1, MYSM1, POLD1, and CD3E; 57% positivity rate of newborn SCID screening among infants with DiGeorge syndrome; and earlier diagnosis and improved transplant outcomes for SCID in infants diagnosed after compared to before implementation of routine screening. Our study is unique in terms of the extensive laboratory workup of abnormal SCID screens including lymphocyte subsets, measurement of thymic output (TREC and CD4TE), and lymphocyte proliferation to mitogens in nearly all infants. These data allowed us to observe a stronger positive correlation of the absolute CD3 count with CD4RTE than with TREC copies, and a weak positive correlation between CD4RTE and TREC copies. Finally, we did not observe a correlation between risk of TCL and history of prenatal or perinatal complications or low birth weight. Our study demonstrated SCID newborn screening improves disease outcomes, particularly in typical SCID, and allows early detection and discovery of novel variants of certain TCL-associated genetic conditions.
Collapse
Affiliation(s)
- Vasudha Mantravadi
- The Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Jeffrey J Bednarski
- The Division of Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Michelle A Ritter
- The Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Hongjie Gu
- The Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Ana L Kolicheski
- The Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Caroline Horner
- The Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Megan A Cooper
- The Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Maleewan Kitcharoensakkul
- The Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States.,The Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
25
|
Göngrich C, Ekwall O, Sundin M, Brodszki N, Fasth A, Marits P, Dysting S, Jonsson S, Barbaro M, Wedell A, von Döbeln U, Zetterström RH. First Year of TREC-Based National SCID Screening in Sweden. Int J Neonatal Screen 2021; 7:ijns7030059. [PMID: 34449549 PMCID: PMC8395826 DOI: 10.3390/ijns7030059] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
Screening for severe combined immunodeficiency (SCID) was introduced into the Swedish newborn screening program in August 2019 and here we report the results of the first year. T cell receptor excision circles (TRECs), kappa-deleting element excision circles (KRECs), and actin beta (ACTB) levels were quantitated by multiplex qPCR from dried blood spots (DBS) of 115,786 newborns and children up to two years of age, as an approximation of the number of recently formed T and B cells and sample quality, respectively. Based on low TREC levels, 73 children were referred for clinical assessment which led to the diagnosis of T cell lymphopenia in 21 children. Of these, three were diagnosed with SCID. The screening performance for SCID as the outcome was sensitivity 100%, specificity 99.94%, positive predictive value (PPV) 4.11%, and negative predictive value (NPV) 100%. For the outcome T cell lymphopenia, PPV was 28.77%, and specificity was 99.95%. Based on the first year of screening, the incidence of SCID in the Swedish population was estimated to be 1:38,500 newborns.
Collapse
Affiliation(s)
- Christina Göngrich
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden; (S.D.); (S.J.); (M.B.); (A.W.); (U.v.D.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17176 Stockholm, Sweden
- Correspondence: (C.G.); (R.H.Z.)
| | - Olov Ekwall
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden; (O.E.); (A.F.)
- Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
| | - Mikael Sundin
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17177 Stockholm, Sweden; (M.S.); (P.M.)
- Section of Pediatric Hematology, Immunology and HCT, Astrid Lindgren Children’s Hospital, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Nicholas Brodszki
- Department of Pediatric Immunology, Children’s Hospital, Lund University Hospital, 22242 Lund, Sweden;
| | - Anders Fasth
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden; (O.E.); (A.F.)
| | - Per Marits
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17177 Stockholm, Sweden; (M.S.); (P.M.)
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Sam Dysting
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden; (S.D.); (S.J.); (M.B.); (A.W.); (U.v.D.)
| | - Susanne Jonsson
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden; (S.D.); (S.J.); (M.B.); (A.W.); (U.v.D.)
| | - Michela Barbaro
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden; (S.D.); (S.J.); (M.B.); (A.W.); (U.v.D.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Anna Wedell
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden; (S.D.); (S.J.); (M.B.); (A.W.); (U.v.D.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Ulrika von Döbeln
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden; (S.D.); (S.J.); (M.B.); (A.W.); (U.v.D.)
- Department of Medical Biochemistry and Biophysics, Division of Molecular Metabolism, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Rolf H. Zetterström
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden; (S.D.); (S.J.); (M.B.); (A.W.); (U.v.D.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17176 Stockholm, Sweden
- Correspondence: (C.G.); (R.H.Z.)
| |
Collapse
|
26
|
Newborn Screening for Severe Combined Immunodeficiency: Do Preterm Infants Require Special Consideration? Int J Neonatal Screen 2021; 7:ijns7030040. [PMID: 34287233 PMCID: PMC8293075 DOI: 10.3390/ijns7030040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 11/17/2022] Open
Abstract
The Wisconsin Newborn Screening (NBS) Program began screening for severe combined immunodeficiency (SCID) in 2008, using real-time PCR to quantitate T-cell receptor excision circles (TRECs) in DNA isolated from dried blood NBS specimens. Prompted by the observation that there were disproportionately more screening-positive cases in premature infants, we performed a study to assess whether there is a difference in TRECs between full-term and preterm newborns. Based on de-identified SCID data from 1 January to 30 June 2008, we evaluated the TRECs from 2510 preterm newborns (gestational age, 23-36 weeks) whose specimens were collected ≤72 h after birth. The TRECs from 5020 full-term newborns were included as controls. The relationship between TRECs and gestational age in weeks was estimated using linear regression analysis. The estimated increase in TRECs for every additional week of gestation is 9.60%. The 95% confidence interval is 8.95% to 10.25% (p ≤ 0.0001). Our data suggest that TRECs increase at a steady rate as gestational age increases. These results provide rationale for Wisconsin's existing premature infant screening procedure of recommending repeat NBS following an SCID screening positive in a premature infant instead of the flow cytometry confirmatory testing for SCID screening positives in full-term infants.
Collapse
|