Abstract

Adenosine deaminase (ADA) deficiency is a form of severe combined immunodeficiency. Aberrant mutations in the ADA gene result in loss of ADA activity and the toxic accumulation of metabolites that damage both immune and non-immune organs. While patients with complete ADA deficiency present during infancy with failure to thrive, recurrent bacterial, viral and fungal infections, those with incomplete (partial) deficiency may present at a later age with milder symptoms associated with reduced T, B, and NK cell subpopulations. Based on experience in Canadian centres, we provide management guidelines for patients with ADA deficiency, including a treatment algorithm for use of hematopoietic stem cell transplantation, gene therapy, and enzyme replacement therapy.
Statement of novelty: Herein, we define guidelines for the management and treatment of patients with ADA deficiency.

Introduction

Adenosine deaminase (ADA) is a ubiquitously expressed enzyme of the purine salvage pathway which catalyzes the irreversible deamination of adenosine and deoxyadenosine. It is essential for normal lymphoid development, with particularly high levels found in the thymus (Adams and Harkness 1976; Poliani et al. 2009). Deficiency of ADA, caused by mutations in the ADA gene and subsequent impairment of ADA activity, leads to an autosomal recessive form of severe combined immunodeficiency (SCID) (Sauer et al. 2012; Bradford et al. 2017). The toxic accumulation of ADA substrates and metabolites interferes with downstream metabolic pathways, including inhibition of ribonucleotide reductase and subsequent blockade of DNA synthesis, as well as S-adenosylhomocysteine hydrolase-dependent transmethylation (Flinn and Gennery 2018). The detrimental effects are most pronounced in pathways regulating lymphocyte maturation and function, although non-immunological organ systems, including the hepatic, renal, pulmonary, skeletal, peripheral and central nervous systems, can also be affected (reviewed by(Whitmore and Gaspar (2016)).
ADA deficiency affects 1:200 000 live births (Blackburn and Kellems 2005), with a higher frequency reported in Canadian Inuit and Mennonite populations (Grunebaum et al. 2013). Without treatment it is usually fatal within the first year of life. Genotype-phenotype correlations have revealed greater metabolic disturbance in those with more severe biallelic defects in ADA (Arredondo-Vega et al. 1998; Cagdas et al. 2018). By contrast, hypomorphic mutations result in somewhat reduced ADA activity and are associated with less severe or late-onset phenotypes. Prior to the advent of newborn screening, patients with complete ADA deficiency traditionally presented during infancy with recurrent bacterial, viral and fungal infections and failure to thrive, while laboratory evaluations revealed severe lymphopenia, hypogammaglobulinemia, and neutropenia. Skeletal abnormalities, cognitive impairment, and hearing loss are common (Albuquerque and Gaspar 2004; Titman et al. 2008; Sauer et al. 2009; Manson et al. 2013). Those with incomplete (partial) deficiency may present with milder but gradually worsening manifestations, associated with reduced populations of T, B, and NK cells (Santisteban et al. 1993; Shovlin et al. 1993). In either case, measurement of ADA enzyme activity and associated levels of ADA substrates/metabolites (adenosine, 2′deoxyadenosine (dAXP), deoxyadenosine triphosphate) is often the first step in identifying ADA deficiency.
The implementation of newborn screening for SCID in Canada (first introduced in Ontario in 2013 and now routine across Nova Scotia, New Brunswick, Prince Edward Island, and Alberta (Biggs et al. 2017; Reid et al. 2017)) has resulted in the early detection of ADA deficient patients as well as improved survival of this cohort (Scott et al. 2019). Management currently relies on immediate enzyme replacement therapy (ERT), immunoglobulin replacement, protective isolation procedures, and antibiotic prophylaxis until definitive treatment is initiated (Figure 1). Patients require close monitoring to reduce the risk of infectious and non-infectious complications (Table 1).
Figure 1:
Figure 1: Algorithm for management of patients with ADA deficiency. ADA, adenosine deaminase; BM, bone marrow; CMV, cytomegalovirus; ERT, enzyme replacement therapy; HSC-GT, hematopoietic stem cell gene therapy; HSCT, hematopoietic stem cell transplantation; MRD, matched related donor; MUD, matched unrelated donor; RIC, reduced intensity conditioning; SCID, severe combined immunodeficiency.
Table 1:
Table 1: Recommended immune evaluation while on ERT.a
a
For elapegademase (Revcovi) naïve patients, guidelines in the product monograph may be followed.
b
Testing for antibodies to elapegademase (Revcovi) should be performed if a persistent fall in plasma ADA activity trough levels below 15 mmol/h/L occurs.
ERT using polyethylene glycol-modified bovine ADA has proved to be effective in correcting the immune and some non-immune abnormalities conferred by ADA deficiency, however, in some patients immune function declined over time (Booth et al. 2007). The bovine-derived ADA product was replaced in 2019 by a recombinant form of the enzyme (elapegademase) which has shown similar in-vitro and possibly better in-vivo activity compared to its predecessor (Murguia-Favela et al. 2020). ERT remains a costly treatment. Thus, it is considered a bridging modality before curative treatment is initiated. To date, hematopoietic stem cell transplantation (HSCT) is the treatment of choice when a human leukocyte antigen (HLA)-matched related donor (MRD) is available (Griffith et al. 2008). Another option that has been shown to correct for the absence of ADA is ex vivo-modified autologous hematopoietic stem cell gene therapy (HSC-GT) (Kohn et al. 1995; Aiuti et al. 2002; Ferrua et al. 2010; Cicalese et al. 2016). Experiences with HSCT with matched unrelated donors (MUD) or haploidentical donors have shown lower success rates (Hassan et al. 2012), although these options can still be considered if MRD HSCT or HSC-GT are not available.
Herein we, the Canadian Expert Committee, define guidelines on the management of ADA deficiency based on our collective experience and available literature in managing this group of patients.

Diagnosis of ADA deficiency

Complete deficiency

Mandatory findings1
(1) <1% of normal ADA activity in red blood cells or peripheral blood lymphocytes2
(2) Severe lymphopenia
(3) Low to absent responses to mitogens
(4) Biallelic pathogenic mutations in the ADA gene
Additional but not mandatory findings
(5) Increased deoxyadenosine in urine, plasma, or dried blood spots
(6) <1% of normal ADA activity in T cells
Additional clues to aid in diagnosis
(7) Recent/persistent infections consistent with SCID
(8) Neurological, skeletal, lung and liver anomalies, and
(9) Hematopoietic anomalies (most commonly neutropenia)

Incomplete (partial) deficiency

Mandatory findings
(1) Low ADA activity in red blood cells
(2) 20%–75% of normal ADA activity in lymphocytes
(3) T cell lymphopenia
(4) Low responses to mitogens
Additional but not mandatory findings
(5) Increased deoxyadenosine in urine, plasma, or dried blood spots
Additional clues to aid in diagnosis
(6) Can have delayed and (or) milder onset of clinical features listed under complete deficiency

Clinical and laboratory features

Non-immune features of ADA deficiency

Skeletal
- Rib and small bone anomalies
Neurological
- Developmental delay, hearing anomalies, seizures
Lungs
- Bronchiectasis, alveolar proteinosis
Liver
- Increased liver enzymes, autoimmune hepatitis, hepatic failure
Renal
- Hemolytic uremic syndrome

Hematopoietic and immune anomalies

Lymphoid cells
- Decreased number and function of T, B cells, NK cells
Lymphoid tissues
- Dysplastic thymus and lymph nodes
Hematopoietic
- Neutropenia, myeloid dysplasia

Management of patients with ADA deficiency

General

- Protective isolation at home or in hospital until reconstituted immune function3
- Immunoglobulin replacement
- Prophylaxis against Pneumocystis jirovecii pneumonia (trimethoprim-sulfamethoxazole) at 1 month of age
- Avoid breast feeding if CMV IgG is positive in the mother
- Use CMV negative and irradiated or leuko-reduced blood products, when needed, discuss with local blood bank as per policy for immunosuppressed patients
- Avoid live vaccines (MMRV, rotavirus, BCG)

Specific treatment

Enzyme replacement (ERT)
- For newly diagnosed patients it is recommended to start recombinant PEG-ADA, elapegademase (Revcovi) at a total weekly dose of 0.4 mg/kg, divided and administered twice a week (0.2 mg/kg per injection) intramuscularly, as per product insert.
- Reassessment of dosing is recommended 6 months from treatment initiation and could be gradually reduced and (or) consolidated to 1 injection per week, based on immune reconstitution and (or) ADA activity of more than 30 mmol/h/L and dAXP level under 0.02 mmol/L.
- For patients already on pegademase (Adagen) treatment, we recommend changing to elapegademase (Revcovi) with a starting dose of 0.2 mg/kg once a week while periodically increasing or decreasing doses by increments of 0.033 mg/kg weekly to monitor immune reconstitution; ADA activity above 30 mmol/h/L and (or) dAXP below 0.02 mmol/L.
Treatment duration
- For most patients ERT should be a temporary option until definitive treatment such as HSCT or HSC-GT are available.
- ERT should be stopped just prior or shortly after HSCT4.
- Before HSC-GT, ERT should be administered according to the approved GT protocol used.
- Long term ERT can be used when HSCT or HSC-GT cannot be performed, and can be continued in patients who were treated for long durations (>10 years) and have no other option or where the risk of HSCT is deemed to be too high.
- Long term ERT can be effective in patients with partial ADA-deficiency
Dosing adjustment
- Doses should be adjusted over time according to ADA activity trough levels and dAXP levels.
- Antibodies to elapegademase (Revcovi) should be tested in cases where a persistent decline in plasma ADA activity is recorded.

Footnotes

1
It is acknowledged that mandatory findings are ideal criteria to follow. However, in some circumstances where a sibling has been diagnosed with ADA deficiency or when all mandatory findings cannot be met but administration of treatment is urgent, criteria No. 4 plus one of the other findings could be sufficient.
2
In patients who received blood transfusion, ADA level measurement in peripheral blood lymphocytes should be performed.
3
The decision to isolate at home or in the hospital usually depends on the patient’s medical condition and home compliance.
4
Some suggest continuing ERT for 2 weeks.

REFERENCES

Adams A. and Harkness R.A. 1976. Adenosine deaminase activity in thymus and other human tissues. Clin. Exp. Immunol. 26: 647–649.
Aiuti A., Slavin S., Aker M., Ficara F., Deola S., Mortellaro A., Morecki S., Andolfi G., Tabucchi A., Carlucci F., Marinello E., Cattaneo F., Vai S., Servida P., Miniero R., Roncarolo M.G., and Bordignon C. 2002. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science, 296: 2410–2413.
Albuquerque W. and Gaspar H.B. 2004. Bilateral sensorineural deafness in adenosine deaminase-deficient severe combined immunodeficiency. J. Pediatr. 144: 278–280.
Arredondo-Vega F.X., Santisteban I., Daniels S., Toutain S., and Hershfield M.S. 1998. Adenosine deaminase deficiency: Genotype-phenotype correlations based on expressed activity of 29 mutant alleles. Am. J. Hum. Genet. 63: 1049–1059.
Biggs C.M., Haddad E., Issekutz T.B., Roifman C.M., and Turvey S.E. 2017. Newborn screening for severe combined immunodeficiency: A primer for clinicians. CMAJ, 189: E1551–E1557.
Blackburn M.R. and Kellems R.E. 2005. Adenosine deaminase deficiency: Metabolic basis of immune deficiency and pulmonary inflammation. Adv. Immunol. 86: 1–41.
Booth C., Hershfield M., Notarangelo L., Buckley R., Hoenig M., Mahlaoui N., Cavazzana-Calvo M., Aiuti A., and Gaspar H.B. 2007. Management options for adenosine deaminase deficiency; proceedings of the EBMT satellite workshop (Hamburg, March 2006). Clin. Immunol. 123: 139–147.
Bradford K.L., Moretti F.A., Carbonaro-Sarracino D.A., Gaspar H.B., and Kohn D.B. 2017. Adenosine deaminase (ADA)-deficient severe combined immune deficiency (SCID): Molecular pathogenesis and clinical manifestations. J. Clin. Immunol. 37: 626–637.
Cagdas D., Gur Cetinkaya P., Karaatmaca B., Esenboga S., Tan C., Yilmaz T., Gumus E., Baris S., Kuskonmaz B., Ozgur T.T., Bali P., Santisteban I., Orhan D., Yuce A., Cetinkaya D., Boztug K., Hershfield M., Sanal O., and Tezcan I. 2018. ADA deficiency: Evaluation of the clinical and laboratory features and the outcome. J. Clin. Immunol. 38: 484–493.
Cicalese M.P., Ferrua F., Castagnaro L., Pajno R., Barzaghi F., Giannelli S., Dionisio F., Brigida I., Bonopane M., Casiraghi M., Tabucchi A., Carlucci F., Grunebaum E., Adeli M., Bredius R.G., Puck J.M., Stepensky P., Tezcan I., Rolfe K., De Boever E., Reinhardt R.R., Appleby J., Ciceri F., Roncarolo M.G., and Aiuti A. 2016. Update on the safety and efficacy of retroviral gene therapy for immunodeficiency due to adenosine deaminase deficiency. Blood, 128: 45–54.
Ferrua F., Brigida I., and Aiuti A. 2010. Update on gene therapy for adenosine deaminase-deficient severe combined immunodeficiency. Curr. Opin. Allergy Clin. Immunol. 10: 551–556.
Flinn A.M. and Gennery A.R. 2018. Adenosine deaminase deficiency: A review. Orphanet J. Rare Dis. 13: 65.
Griffith L.M., Cowan M.J., Kohn D.B., Notarangelo L.D., Puck J.M., Schultz K.R., Buckley R.H., Eapen M., Kamani N.R., O’Reilly R.J., Parkman R., Roifman C.M., Sullivan K.E., Filipovich A.H., Fleisher T.A., and Shearer W.T. 2008. Allogeneic hematopoietic cell transplantation for primary immune deficiency diseases: Current status and critical needs. J. Allergy Clin. Immunol. 122: 1087–1096.
Grunebaum E., Cohen A., and Roifman C.M. 2013. Recent advances in understanding and managing adenosine deaminase and purine nucleoside phosphorylase deficiencies. Curr. Opin. Allergy Clin. Immunol. 13: 630–638.
Hassan A., Booth C., Brightwell A., Allwood Z., Veys P., Rao K., Honig M., Friedrich W., Gennery A., Slatter M., Bredius R., Finocchi A., Cancrini C., Aiuti A., Porta F., Lanfranchi A., Ridella M., Steward C., Filipovich A., Marsh R., Bordon V., Al-Muhsen S., Al-Mousa H., Alsum Z., Al-Dhekri H., Al Ghonaium A., Speckmann C., Fischer A., Mahlaoui N., Nichols K.E., Grunebaum E., Al Zahrani D., Roifman C.M., Boelens J., Davies E.G., Cavazzana-Calvo M., Notarangelo L., Gaspar H.B., and Inborn Errors Working Party of the European Group for Blood and Marrow Transplantation and European Society for Immunodeficiency. 2012. Outcome of hematopoietic stem cell transplantation for adenosine deaminase-deficient severe combined immunodeficiency. Blood, 120: 3615–3624; quiz 3626.
Kohn D.B., Weinberg K.I., Nolta J.A., Heiss L.N., Lenarsky C., Crooks G.M., Hanley M.E., Annett G., Brooks J.S., El-Khoureiy A., Lawrence K., Wells S., Moen R.C., Bastian J., Williams-Herman D.E., Elder M., Wara D., Bowen T., Hershfield M.S., Mullen C.A., Blaese R.M., and Parkman R. 1995. Engraftment of gene-modified umbilical cord blood cells in neonates with adenosine deaminase deficiency. Nat. Med. 1: 1017–1023.
Manson D., Diamond L., Oudjhane K., Hussain F.B., Roifman C., and Grunebaum E. 2013. Characteristic scapular and rib changes on chest radiographs of children with ADA-deficiency SCIDS in the first year of life. Pediatr. Radiol. 43: 589–592.
Murguia-Favela L., Min W., Loves R., Leon-Ponte M., and Grunebaum E. 2020. Comparison of elapegademase and pegademase in ADA-deficient patients and mice. Clin. Exp. Immunol. 200: 176–184.
Poliani P.L., Facchetti F., Ravanini M., Gennery A.R., Villa A., Roifman C.M., and Notarangelo L.D. 2009. Early defects in human T-cell development severely affect distribution and maturation of thymic stromal cells: Possible implications for the pathophysiology of Omenn syndrome. Blood, 114: 105–108.
Reid B., Ovadia A., and Dinur Schejter Y. 2017. Managing newborn screening for SCID in a referral centre. LymphoSign J. 4: 77–79.
Santisteban I., Arredondo-Vega F.X., Kelly S., Mary A., Fischer A., Hummell D.S., Lawton A., Sorensen R.U., Stiehm E.R., Uribe L., Weinberg K., and Hershfield M.S. 1993. Novel splicing, missense, and deletion mutations in seven adenosine deaminase-deficient patients with late/delayed onset of combined immunodeficiency disease. Contribution of genotype to phenotype. J. Clin. Invest. 92: 2291–2302.
Sauer A.V., Mrak E., Hernandez R.J., Zacchi E., Cavani F., Casiraghi M., Grunebaum E., Roifman C.M., Cervi M.C., Ambrosi A., Carlucci F., Roncarolo M.G., Villa A., Rubinacci A., and Aiuti A. 2009. ADA-deficient SCID is associated with a specific microenvironment and bone phenotype characterized by RANKL/OPG imbalance and osteoblast insufficiency. Blood, 114: 3216–3226.
Sauer A.V., Brigida I., Carriglio N., and Aiuti A. 2012. Autoimmune dysregulation and purine metabolism in adenosine deaminase deficiency. Front. Immunol. 3: 265.
Scott O., Brager R., Dent P., Siu V., Reid B., Kim V.H.-D., and Roifman C.M. 2019. Survival of patients with severe combined immunodeficiency in the newborn screening era: A single-centre experience 2013–2019 (in Abstracts from the Immunodeficiency Canada—7th SCID Symposium, Montreal, QC, 24 October 2019). LymphoSign J. 6: 148–163.
Shovlin C.L., Hughes J.M., Simmonds H.A., Fairbanks L., Deacock S., Lechler R., Roberts I., and Webster A.D. 1993. Adult presentation of adenosine deaminase deficiency. Lancet, 341: 1471.
Titman P., Pink E., Skucek E., O’Hanlon K., Cole T.J., Gaspar J., Xu-Bayford J., Jones A., Thrasher A.J., Davies E.G., Veys P.A., and Gaspar H.B. 2008. Cognitive and behavioral abnormalities in children after hematopoietic stem cell transplantation for severe congenital immunodeficiencies. Blood, 112: 3907–3913.
Whitmore K.V. and Gaspar H.B. 2016. Adenosine deaminase deficiency—More than just an immunodeficiency. Front. Immunol. 7: 314.

Information & Authors

Information

Published In

cover image LymphoSign Journal
LymphoSign Journal
Volume 7Number 3September 2020
Pages: 109 - 115

History

Received: 21 August 2020
Accepted: 24 August 2020
Accepted manuscript online: 30 August 2020

Authors

Affiliations

Rae Brager
Division of Rheumatology, Immunology, and Allergy, Department of Paediatrics, McMaster Children’s Hospital, McMaster University, Hamilton, ON
Parwinder Gill
Division of Clinical Immunology, St. Joseph’s Hospital, Western University, London, ON
Alison Haynes
Division of Pediatrics, Faculty of Medicine, Memorial University, St John’s, NL
Kyla Hildebrand
Division of Allergy & Immunology, Department of Pediatrics, University of British Columbia, Vancouver, BC
Child & Family Research Institute, Vancouver, BC
Vy H.D. Kim
Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON
University of Toronto, Toronto, ON
Thomas B. Issekutz
Division of Immunology, Department of Pediatrics, IWK Health Centre, Dalhousie University, Halifax, NS
Christine McCusker
Division of Pediatric Allergy Immunology and Dermatology, Department of Pediatrics, McGill University Health Center, Montreal, QC
Luis Murguia-Favela
Section of Hematology/Immunology, Department of Pediatrics, University of Calgary, Calgary, AB
Brenda Reid
Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON
University of Toronto, Toronto, ON
Tamar Rubin
Winnipeg Children’s Hospital, Division of Pediatric Allergy and Immunology, University of Manitoba, Winnipeg, MB
Julia Upton
Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON
University of Toronto, Toronto, ON
Chaim M. Roifman [email protected]
Division of Immunology & Allergy, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON
University of Toronto, Toronto, ON
Canadian Centre for Primary Immunodeficiency, 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.

There are no citations for this item

View Options

View options

PDF

View PDF

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