Novel heterozygous PIK3CD mutation presenting with only laboratory markers of combined immunodeficiency

Publication: LymphoSign Journal
6 May 2020

Abstract

Introduction: Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Delta (PIK3CD) is one part of a heterodimer forming the enzyme phosphoinositide 3-kinase (PI3K), found primarily in leukocytes. PIK3CD generates phosphatidyl-inositol 3,4,5-trisphosphate (PIP3), and is involved in cell growth, survival, proliferation, motility, and morphology. An increasing number of patients have been described with heterozygous PIK3CD gain-of-function (GOF) mutations, leading to combined immunodeficiency with both B- and T-cell dysfunction. Patients suffer recurrent respiratory infections, often associated with bronchiectasis and ear and sinus damage, as well as severe recurrent or persistent infections by herpesviruses, including EBV-induced lymphoproliferation.
Aim: To present the clinical phenotypic variability of a novel PI3KCD mutation within a family.
Methods: Patient information was collected prospectively and retrospectively from medical records. Comprehensive immune work up, genetic, and signaling evaluation was performed.
Results: We describe here 2 patients, daughter and mother, with heterozygous PIK3CD mutation identified by whole exome sequencing and Sanger confirmation. The child was screen-positive by newborn screening for severe combined immunodeficiency (SCID). Cellular assays revealed an increase in the baseline phosphorylation of T cells in the patient. Furthermore, both patients had hyper-activation of the catalytic domain, resulting in increased phosphorylation of AKT upon activation.
Discussion: GOF mutations affecting the PIK3CD gene are associated with an increased risk for lymphoproliferation leading to Activated PIK3-delta syndrome (APDS). The clinical course of APDS is highly variable, ranging from combined immunodeficiency with recurrent infections, autoimmune complications, and requiring stem cell transplantation, through isolated antibody deficiency, to asymptomatic adults. Our patient is the first to be identified by newborn screening for SCID. Surprisingly, the clinical course has so far been unremarkable, as well, the mother appears to be completely asymptomatic. Nevertheless, the persistent lymphopenia indicates PIK3CD dysfunction. Because of the wide gap between laboratory findings and clinical manifestations, this kindred poses both a diagnostic as well treatment challenge.
Statement of novelty: We report here a novel PIK3CD mutation diagnosed due to abnormal newborn screen for SCID.

Formats available

You can view the full content in the following formats:

REFERENCES

Coulter T.I., Chandra A., Bacon C.M., Babar J., Curtis J., Screaton N., Goodlad J.R., Farmer G., Steele C.L., Leahy T.R., Doffinger R., Baxendale H., Bernatoniene J., Edgar J.D., Longhurst H.J., Ehl S., Speckmann C., Grimbacher B., Sediva A., Milota T., Faust S.N., Williams A.P., Hayman G., Kucuk Z.Y., Hague R., French P., Brooker R., Forsyth P., Herriot R., Cancrini C., Palma P., Ariganello P., Conlon N., Feighery C., Gavin P.J., Jones A., Imai K., Ibrahim M.A., Markelj G., Abinun M., Rieux-Laucat F., Latour S., Pellier I., Fischer A., Touzot F., Casanova J.L., Durandy A., Burns S.O., Savic S., Kumararatne D.S., Moshous D., Kracker S., Vanhaesebroeck B., Okkenhaug K., Picard C., Nejentsev S., Condliffe A.M., and Cant A.J. 2017. Clinical spectrum and features of activated phosphoinositide 3-kinase δ syndrome: A large patient cohort study. J. Allergy Clin. Immunol. 139: 597–606.e4.
Crank M.C., Grossman J.K., Moir S., Pittaluga S., Buckner C.M., Kardava L., Agharahimi A., Meuwissen H., Stoddard J., Niemela J., Kuehn H., and Rosenzweig S.D. 2014. Mutations in PIK3CD can cause hyper IgM syndrome (HIGM) associated with increased cancer susceptibility. J. Clin. Immunol. 34: 272–276.
Dornan G.L., Siempelkamp B.D., Jenkins M.L., Vadas O., Lucas C.L., and Burke J.E. 2017. Conformational disruption of PI3Kδ regulation by immunodeficiency mutations in PIK3CD and PIK3R1. Proc. Natl. Acad. Sci. U.S.A. 114: 1982–1987.
Elgizouli M., Lowe D.M., Speckmann C., Schubert D., Hulsdunker J., Eskandarian Z., Dudek A., Schmitt-Graeff A., Wanders J., Jorgensen S.F., Fevang B., Salzer U., Nieters A., Burns S., and Grimbacher B. 2016. Activating PI3Kδ mutations in a cohort of 669 patients with primary immunodeficiency. Clin. Exp. Immunol. 183: 221–229.
Greenwell I.B., Ip A., and Cohen J.B. 2017. PI3K inhibitors: Understanding toxicity mechanisms and management. Oncology, 31: 821–828.
Hartman H.N., Niemela J., Hintermeyer M.K., Garofalo M., Stoddard J., Verbsky J.W., Rosenzweig S.D., and Routes J.M. 2015. Gain of function mutations of PIK3CD as a cause of primary sclerosing cholangitis. J. Clin. Immunol. 35: 11–14.
Lucas C.L., Kuehn H.S., Zhao F., Niemela J.E., Deenick E.K., Palendira U., Avery D.T., Moens L., Cannons J.L., Biancalana M., Stoddard J., Ouyang W., Frucht D.M., Rao V.K., Atkinson T.P., Agharahimi A., Hussey A.A., Folio L.R., Olivier K.N., Fleisher T.A., Pittaluga S., Holland S.M., Cohen J.I., Oliveira J.B., Tangye S.G., Schwartzberg P.L., Lenardo M.J., and Uzel G. 2014. Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110δ result in T cell senescence and human immunodeficiency. Nat. Immunol. 15: 88–97.
Rao V.K., Webster S., Dalm V.A.S.H., Šedivá A., van Hagen P.M., Holland S., Rosenzweig S.D., Christ A.D., Sloth B., Cabanski M., Joshi A.D., de Buck S., Doucet J., Guerini D., Kalis C., Pylvaenaeinen I., Soldermann N., Kashyap A., Uzel G., Lenardo M.J., Patel D.D., Lucas C.L., and Burkhart C. 2017. Effective “activated PI3Kδ syndrome”-targeted therapy with the PI3Kδ inhibitor leniolisib. Blood, 130: 2307–2316.
Roifman C.M., Somech R., Kavadas F., Pires L., Nahum A., Dalal I., and Grunebaum E. 2012. Defining combined immunodeficiency. J. Allergy Clin. Immunol. 130: 177–183.
Sharfe N., Nahum A., Newell A., Dadi H., Ngan B., Pereira S.L., Herbrick J.A., and Roifman C.M. 2014. Fatal combined immunodeficiency associated with heterozygous mutation in STAT1. J. Allergy Clin. Immunol. 133: 807–817.
Stavropoulos D.J., Merico D., Jobling R., Bowdin S., Monfared N., Thiruvahindrapuram B., Nalpathamkalam T., Pellecchia G., Yuen R.K.C., Szego M.J., Hayeems R.Z., Shaul R.Z., Brudno M., Girdea M., Frey B., Alipanahi B., Ahmed S., Babul-Hirji R., Porras R.B., Carter M.T., Chad L., Chaudhry A., Chitayat D., Doust S.J., Cytrynbaum C., Dupuis L., Ejaz R., Fishman L., Guerin A., Hashemi B., Helal M., Hewson S., Inbar-Feigenberg M., Kannu P., Karp N., Kim R., Kronick J., Liston E., MacDonald H., Mercimek-Mahmutoglu S., Mendoza-Londono R., Nasr E., Nimmo G., Parkinson N., Quercia N., Raiman J., Roifman M., Schulze A., Shugar A., Shuman C., Sinajon P., Siriwardena K., Weksberg R., Yoon G., Carew C., Erickson R., Leach R.A., Klein R., Ray P.N., Meyn M.S., Scherer S.W., Cohn R.D., and Marshall C.R. 2016. Whole genome sequencing expands diagnostic utility and improves clinical management in pediatric medicine. NPJ Genom Med. 1: 15012.
Wang K., Li M., and Hakonarson H. 2010. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38: e164.

Information & Authors

Information

Published In

cover image LymphoSign Journal
LymphoSign Journal
Volume 7Number 2June 2020
Pages: 49 - 55

History

Received: 10 February 2020
Accepted: 13 April 2020
Accepted manuscript online: 6 May 2020

Authors

Affiliations

Amarilla B. Mandola
Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON
The Canadian Centre for Primary Immunodeficiency and The Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, The Hospital for Sick Children, Toronto, ON
Harjit Dadi
Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON
The Canadian Centre for Primary Immunodeficiency and The Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, The Hospital for Sick Children, Toronto, ON
Brenda Reid
Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON
The Canadian Centre for Primary Immunodeficiency and The Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, The Hospital for Sick Children, Toronto, ON
Chaim M. Roifman [email protected]
Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON
The Canadian Centre for Primary Immunodeficiency and The Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, The Hospital for Sick Children, Toronto, ON

Metrics & Citations

Metrics

Other Metrics

Citations

Cite As

Export Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

View Options

View options

PDF

View PDF

Full Text

View Full Text

Get Access

Login options

Check if you access through your login credentials or your institution to get full access on this article.

Subscribe

Click on the button below to subscribe to LymphoSign Journal

Purchase options

Purchase this article to get full access to it.

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Media

Media

Other

Tables

Share Options

Share

Share the article link

Share on social media