You are on page 1of 8

Primary Immunodeficiencies

89
Michael Albert, Arjan Lankester,
and Andrew Gennery

89.1 Introduction 89.2 Diseases

Primary immunodeficiency (PID) diseases arise For HSCT purposes and thus for this handbook,
from genetic defects that lead to abnormalities in PID may be broadly categorized into severe com-
immune cell development or function with a wide bined immunodeficiencies (SCID) and non-­
spectrum in severity and clinical manifestations. SCID.  To further subdivide non-SCID, the
A subgroup of patients with an immunodeficiency phenotypic classification as suggested by the
present as a medical emergency which is associ- International Union of Immunological Societies
ated with a chronic disabling and life-­threatening (IUIS) Inborn Errors of Immunity Committee
clinical course. In these cases, allo-­HSCT pro- can be used, which encompasses >300 genetic
vides a life-saving and curative treatment modal- causes of PID (Table 89.1).
ity. Replacement of the defective cell lineage by Overall guidelines for HSCT for SCID and
HSCT from healthy allogeneic donors remains non-SCID diseases together with detailed proto-
the curative approach for these patients. Other cols have been produced by the EBMT Inborn
management options including enzyme replace- Errors Working Party (EBMT IEWP) and can be
ment therapy, gene transfer into autologous hema- found online at https://www.ebmt.org/sites/
topoietic stem cells, and targeted therapies (see default/files/migration_legacy_files/document/
below) may provide an alternative approach to Inborn%20Errors%20Working%20Party%20
HSCT in specific immune deficiencies. ESID%20EBMT%20HSCT%20Guidelines%20

Table 89.1  Phenotypic classification of PID as sug-


gested by the International Union of Immunological
M. Albert
Societies (IUIS) Inborn Errors of Immunity Committee
Pediatric SCT Program, Dr. von Hauner University
(Picard et al. 2018)
Children’s Hospital, Ludwig-Maximilians
Universität, Munich, Germany 1. Combined immunodeficiency (CID)
2. CID with associated or syndromic features
A. Lankester
Department of Pediatrics, Stem Cell Transplantation 3. Predominantly antibody deficiencies
Program, Willem-Alexander Children’s Hospital, 4. Diseases of immune dysregulation
Leiden University Medical Center, Leiden, 5. Congenital defects of phagocyte number, function,
The Netherlands or both
6. Defects in intrinsic and innate immunity
A. Gennery (*)
7. Auto-inflammatory disorders
Paediatric Immunology + HSCT, Great North Children’s
Hospital, Newcastle University, Newcastle upon Tyne, UK 8. Complement deficiencies
e-mail: andrew.gennery@newcastle.ac.uk 9. Phenocopies of PID

© EBMT and the Author(s) 2019 663


E. Carreras et al. (eds.), The EBMT Handbook, https://doi.org/10.1007/978-3-030-02278-5_89
664 M. Albert et al.

2017.pdf. An update of these guidelines is 3–6 months with unusually severe and recurrent
planned for 2018 based on current IEWP infections or with opportunistic infections, the
studies. most common being Pneumocystis jiroveci pneu-
monia. Other common symptoms include diar-
rhea, dermatitis, and failure to thrive. Survival in
89.3 SCID SCID patients depends on expeditious T-cell
reconstitution, and in the absence of successful
The overall frequency of SCID was for a long HSCT, or in selected cases autologous stem cell
time estimated to be 1  in 50,000–100,000 live gene therapy, most children die usually during
births. However, in recent years newborn screen- the first year of life from overwhelming infec-
ing programs making use of the T-cell receptor tion. It is recognized that as many as 50% of
excision circles (TREC) technology have demon- SCID patients are engrafted with maternal T-cells
strated that the frequency may actually be two- or but in most instances these cells do not initiate
more-fold higher with clear geographical and GvHD.  Transfusion-associated GvHD, on the
ethnic differences (Kwan et  al. 2014; Rechavi other hand, is frequently lethal in SCID, and any
et al. 2017). patient with a possible diagnosis of SCID should
The immunological phenotypes of SCID are receive irradiated blood products. Bacille
shown in Table  89.2 representing monogenic Calmette-Guérin (BCG) vaccination can give rise
inherited defects in T-, B-, and NK-cell differen- to disseminated BCG-osis in SCID patients and
tiation leading to the absence or inactivity of cor- should be avoided at birth if there is any suspi-
responding mature cells. Over the past two cion or family history of immunodeficiency.
decades, the genetic basis of an increasing num-
ber of SCID variants has been identified
(Table  89.2) leading to modifications in trans- 89.3.1 General Principles in Allo-­
plant strategy dependent on the underlying defect HSCT for SCID
particularly in SCID variants caused by defects in
DNA repair genes. The Stem CEll Transplant for primary Immune
In the absence of newborn screening pro- Deficiencies in Europe (SCETIDE) registry has
grams, most patients present within the first now collected data on SCID transplants compris-
ing 50 years of HSCT experience, and a number
of important publications have documented the
Table 89.2  Gene defects typically associated with spe-
outcomes and important risk factors (Fischer
cific SCID phenotypes
et  al. 1990; Antoine et  al. 2003; Gennery et  al.
T-B+NK- T-B+NK+ T-B-NK- T-B-NK+
2010). Recently, studies from the North American
IL2RG IL7R ADA LIG4
(SCID-X1) group have reported similar findings (Pai et  al.
JAK3 CD3D AK2 RAG1 2014; Heimall et  al. 2017). The major factors
(reticular influencing outcome reported in these studies
dysgenesis include:
CD3E RAG2
CD247 DCLRE1C
(CD3ζ) (Artemis 1. Preceding comorbidity (particularly infec-

def.) tious complications at HSCT) adversely
CORO1A NHEJ1 affecting outcome
(Cernunnos 2. The type of donor with matched sibling donors
XLF)
having the best outcome1
PTPRC PRKDC
(CD45 (DNA-PKcs
def.) def.)
Recent data show that the type of donor and the immuno-
1 
FOXN1 logical SCID phenotype have an ever-diminishing influ-
Adapted from Picard et al. (2018) ence on outcome.
89  Primary Immunodeficiencies 665

3. The type of SCID, with T-B- forms of SCID 89.3.4 HLA-Mismatched Family
having an inferior outcome (see footnote 1) Donor for SCID
4. Age at transplant with patients <3.5  months
having a favorable outcome Virtually all children have a haploidentical paren-
tal donor, and this is an alternative option espe-
cially as the donor is readily available. HLA
89.3.2 Matched Sibling Donor HSCT disparity necessitates rigorous in vitro or in vivo
for SCID TCD in order to avoid GvHD. Using mobilized
PBSC as a preferred stem cell source, most cen-
During the last decade, the overall survival for ters employ either CD34-positive selection or
MSD HSCT in SCID has improved to over 90%. CD3-/CD19-negative depletion methods to
Somewhat remarkably, sibling donor BM may be achieve a 4–5 log TCD achieving a threshold of
infused into SCID recipients without the require- 1–5  ×  104/kg CD3+ cells, below which GvHD
ment for conditioning or GvHD prophylaxis. prophylaxis is not required.
Infusion of sibling BM leads to the rapid devel- More recently, alternative haploidentical pro-
opment of T- and B-cell function post-HSCT, cedures including TCR alpha/beta depletion
although usually only T-cells of donor origin (Balashov et  al. 2015; Shah et  al. 2018) and
develop and myeloid and erythroid cells remain PT-CY have emerged as HSCT options. Although
of recipient origin. In T-/B+ SCID, the majority promising survival rates have been reported, lon-
of patients achieve humoral reconstitution despite ger follow-up in a larger cohort of patients is
lack of donor B-cells, whereas following uncon- required to determine the position of these
ditioned HSCT in T-/B- SCID immunoglobulin approaches.
dependence often persists. Some centers advocate performing transplants
without the use of any conditioning, and survival
rates of over 80% have been reported (Dvorak
89.3.3 Other Matched Family et al. 2014). However, the best results are seen in
and URD HSCT for SCID those transplanted at <3.5 months of age and in
the absence of active infections. Despite general
Overall survival rate following phenotypically improvements in survival rate, the best results are
matched related as well as URD transplants has still seen in the T-B+ subgroup of SCID patients.
steadily improved and is approaching MSD results Even in these cases, B-cell function is only
(Gennery et al. 2010; Pai et al. 2014; Heimall et al. restored in the minority of patients. Conditioning
2017). It is generally considered that the risk of regimes can be used to improve outcome, but the
rejection and particularly GvHD is too high for use of MAC regimes in children often <1 year of
simple infusion of phenotypically matched mar- age is associated with significant comorbidity
row into SCID patients, so conditioning/GvHD and leads to survival figures of 50–60%.
prophylaxis is recommended. In a recent transat- Individualized approaches making use of thera-
lantic study, survival rate in unconditioned URD peutic drug monitoring or antibody-based condi-
HSCT was comparable with MSD HSCT, how- tioning strategies may provide novel and less
ever at the expense of increased acute GvHD and toxic options to improve HSCT outcome in these
inferior B-cellular immune reconstitution (Dvorak vulnerable young infants.
et  al. 2014). A variety of conditioning regimes
have been used, and current IEWP recommenda-
tions include the use of an IV BU/FLU- or TREO/ 89.3.5 Unrelated CBT for SCID
FLU-based protocol (details at http://www.ebmt.
org/5WorkingParties/IEWP/wparties-ie5.html). During the last decade, the availability of CBU
Comparison of survival rates and immune func- plus the increased level of HLA matching degree
tion with these regimens is part of ongoing studies. has made CB a suitable alternative source of stem
666 M. Albert et al.

cells (Fernandes et  al. 2012). There are some studies are available, but a low-dose FLU/CY
theoretical advantages for the use of cord blood regime has been suggested by the EBMT IEWP
stem cells for SCID, namely, rapid availability, as (http://www.ebmt.org/5WorkingParties/IEWP/
with haplotype-matched parental donors but with wparties-ie5.html).
no requirement for TCD; less risk of GvHD com-
pared to adult URD; no medical risk to the donor;
and a greater proliferative life span which might 89.4 Non-SCID Immunodeficiency
be particularly important in such young recipi-
ents. Moreover, the usual limitation of cell dose From a HSCT viewpoint, the major difference
in CBT is usually no issue in infants with with non-SCID patients in comparison with
SCID.  There are also some specific disadvan- SCID patients is the requirement for a condition-
tages including slower engraftment, lack of viral-­ ing regimen to achieve engraftment. It is the goal
specific cytotoxic T-cells, and lack of availability to establish sufficient long-term donor chimerism
of the donor for a boost HSCT. in the affected cell lineage. The required degree
of donor chimerism for full disease correction
varies depending on PID and has not been estab-
89.3.6 Omenn’s Syndrome lished for all entities.
Many children with non-SCID PID have
Omenn’s syndrome (OS) is characterized by ­significant comorbidities at the time of HSCT.
SCID typically associated with the triad of eryth- Conventional MAC preparation with BU-/
roderma, hepatosplenomegaly, and lymphade- CY-based regimes has historically been associ-
nopathy. There is a marked eosinophilia and a ated with significant treatment-related toxicity
variable number of autologous, activated, and and TRM. The Inborn Errors Working Party of
oligoclonal T lymphocytes (leaky SCID/CID), EBMT has therefore in 2005 published detailed
which infiltrate target organs and are generally recommendations for conditioning and PID as
poorly responsive to mitogens. Whereas out- discussed above. These recommendations
comes in HSCT for OS were traditionally more include:
difficult compared the classical SCID, results
have improved in recent years (Gennery et  al. 1. Replacement of CY with FLU, as the combi-
2010; Heimall et al. 2017). The overall mortality nation of BU/FLU appears to be better toler-
in these studies was lower than previously ated in these patients
reported and was due to early recognition of OS 2. Adding the option to replace BU with a struc-
and rapid initiation of treatment with topical/sys- tural analogue, TREO, which is similarly
temic immune suppression with steroids and/or immuno- and myelosuppressive but causes
cyclosporin A to control immune dysreactivity less hepatic SOS/VOD (Slatter et al. 2018)
before proceeding to HSCT. 3. Establishing RIC to achieve stable engraft-
ment of immunocompetent donor cells with
reduced procedure-related morbidity and
89.3.7 HSCT for Radiosensitive SCID mortality (Veys 2010)

Patients with T-B- SCID due to radiosensitive The latest outcome data for HSCT in non-­
disorders such as DNA ligase 4 deficiency, SCID patients come from Europe (Gennery et al.
Cernunnos deficiency, DNA-PKcs deficiency are 2010). In the 2000–2005 period, HSCT using an
increasingly being identified and being consid- URD (n = 124) gave a 3-year survival rate similar
ered for HSCT. As many of the conditioning regi- to a genoidentical donor (n = 73), 79% for both.
mens are particularly damaging to DNA, less Survival was 76% in phenoidentical transplants
toxic regimens are required to successfully treat (n = 23) and worse in mismatched related donor
these patients (Slack et  al. 2018). No definitive transplants (n = 47, 46%, p = 0.016), in contrast
89  Primary Immunodeficiencies 667

to SCID patients (see above). Ten-year survival in comparison to non-HSCT approaches


was significantly better for patients with WAS, (Speckmann et al. 2017; Barzaghi et al. 2018).
phagocytic, and hemophagocytic disorders than • Adolescents and young adults with PID are
for patients with T-lymphocyte immunodeficien- appreciated as candidates for HSCT, and out-
cies (71, 63, 58, 47%, respectively). comes are encouragingly good (Albert et  al.
Unrelated CB donors appear to also give 2018; Fox et al. 2018).
promising results in non-SCID immunodefi- • Patients with DNA double-strand repair
ciency with 29/32 (91%) patients surviving CBT disorders can safely undergo HSCT with
­
matched for 4–6/6 HLA antigens. irradiation-­
free, reduced-intensity regimens
The landscape of non-SCID PID and HSCT (Slack et al. 2018).
has dramatically changed over the last decade:
The consequence of all this has been that
• HSCT outcomes have further improved with many more patients with PID are today consid-
80–90% overall survival and low GVHD rates ered for, referred for, and counselled about
after MSD or MUD HSCT in almost every HSCT. On the other hand, it has made decision
non-SCID PID entity (Güngör et al. 2014; making with respect to HSCT much more diffi-
Moratto et al. 2011). cult. A genetic diagnosis may make the decision
• Haplo-HSCT has become a safe alternative, at to proceed to HSCT easier in patients with severe
least in the hands of experienced centers symptoms, but a genetic diagnosis alone should
(Balashov et al. 2015; Shah et al. 2018). never be sufficient to indicate HSCT.
• New genetic causes of PID are being described
in accelerating frequency thanks to next-­
generation sequencing techniques. 89.5 Alternative Therapies
• The concept of “pure” immunodeficiencies
with predisposition to infections has been Alternative treatments to HSCT have been devel-
abandoned with newly described autoim- oped for specific immunodeficiencies over the
mune, auto-inflammatory conditions or syn- last three decades.
dromal disorders with immunodeficiency.
Many of these diseases can be cured by HSCT,
while in syndromal disorders only the hema- 89.5.1 Enzyme Replacement Therapy
topoietic portion of the disease can be cor- (ERT) for Adenosine
rected, which may nevertheless be indicated Deaminase Deficiency
and result in not just increased survival but (ADA-SCID)
also quality of life in selected patients.
• More PID patients are discovered with very Enzyme replacement has been used in the treat-
mild or atypical phenotypes of well-known ment of ADA deficiency since 1987 (Chan et al.
PID, and these often hypomorphic genetic 2005). PEG-ADA is administered weekly or
variants are especially challenging with twice weekly by IM injection and leads to rapid
respect to timely recognition and management metabolic correction with normalization of meta-
(Notarangelo et al. 2016). bolic parameters which is then followed by cel-
• Especially in these “milder” cases, quality of lular and humoral immune reconstitution. The
life is increasingly a factor in HSCT decision extent of immune recovery is variable, and a sig-
making, especially compared to diseases with nificant number (~50%) remain on Ig replace-
an immediate indication for HSCT such as ment. Over a longer time period, patients show a
SCID (Cole et al. 2013). decline in T-cell numbers and remain lymphope-
• The importance of DFS as compared to OS is nic. Long-term follow-up shows that patients
increasingly appreciated and addressed in remain clinically well, but a number of cases of
medium- to long-term outcome studies, also EBV-related lymphoma have been reported, sug-
668 M. Albert et al.

gesting decreased immune surveillance with mature cells in diseases like CD40 ligand defi-
time. Given the improved outcomes of HSCT in ciency and IPEX Syndrome, as well as HSC.
recent times and the availability of gene therapy, In theory autologous stem cell gene therapy
ERT may predominantly be considered a bridge offers the appealing prospect of avoiding alloim-
to stem cell-based curative therapy. mune reactions such as GVHD or rejection and a
lower conditioning-related toxicity compared to
allo-HSCT. But its exact role in treatment algo-
89.5.2 Gene Therapy for Specific rithms still needs to be defined in the absence of
Immune Deficiencies comparative studies. Also, logistic, regulatory,
and economic hurdles still have to be overcome
Autologous stem cell gene therapy (GT) via before its widespread application in the treatment
vector-­mediated transfer of healthy copies of an of PID.  Nevertheless, it has widened the thera-
affected gene into autologous CD34+ cells has peutic repertoire for patients with some PID. The
progressed from a highly experimental therapy to rapid evolution of novel gene correction
the first licensed gene therapy for a PID (ADA-­ approaches promises to lead to even safer and
SCID) within the last two decades. One of the more effective treatment options.
major advantages of GT is the elimination of the
inherent risk of GVHD connected to any HSCT
procedures. 89.5.3 Targeted Therapies
Clinical trials performed with gamma retro-
viral vectors for ADA-SCID, X-linked SCID The unravelling of new genetic PID entities,
(SCID-X1), chronic granulomatous disease especially those caused by gain-of-function
(CGD), and Wiskott-Aldrich syndrome (WAS) (GOF) variants and their pathophysiology, has
demonstrated that gene therapy can be an effec- for the first time opened the possibility to treat
tive treatment option in patients lacking an these diseases with highly specific, often small
HLA-­identical donor (Hacein-Bey-Abina et al. molecule inhibitors, some of which are already
2002; Boztug et  al. 2010; Stein et  al. 2010; approved for other indications. These include but
Aiuti et al. 2009). However, a high rate of inser- are not limited to abatacept for CTLA4 haploin-
tional mutagenesis was observed in trials for sufficiency, ruxolitinib for STAT1 GOF, leni-
SCID-X1, WAS, and CGD but not for ADA- olisib for PIK3CD and PIK3R1, etanercept for
SCID (Ott et al. 2006; Hacein-Bey-Abina et al. ADA2 deficiency, and IL-1-targeted therapies
2003; Braun et al. 2014). This has prompted the (anakinra, rilonacept, and canakinumab) for
development of safer vectors based on self- auto-inflammatory recurrent fever syndromes
inactivating retroviral or lentiviral vectors. (Jhamnani and Rosenzweig 2017; Ochs and
Currently, a number of trials are ongoing or Petroni 2018). At this point in time, the exact role
concluded for the diseases named above. All of these agents in the treatment algorithm of PID
share the concept of submyeloablative or lym- is however unclear. Ideally, they could make
phodepleting conditioning followed by the HSCT unnecessary for some patients. On the
infusion of auto-HSCT with added copies of other hand, concerns about long-term infection
the gene of interest. Promising results were (and lymphoma) risk exist. In any case, in some
published, especially for ADA-SCID (Cicalese patients with excessive autoimmunity and/or
et  al. 2016), WAS (Aiuti et  al. 2013) and inflammation, these therapies can be viewed as
SCID-X1 (Hacein-Bey-Abina et al. 2014). It is an ideal bridge to HSCT and considered as a
expected that gene editing approaches as alter- remission induction strategy to control the under-
native for gene addition technologies as cur- lying PID, because they have the ability to bring
rently employed will be developed in the next the patient into the best possible clinical condi-
few years and may be employed to correct tion for HSCT.
89  Primary Immunodeficiencies 669

Cicalese MP, Ferrua F, Castagnaro L, et  al. Update on


Key Points the safety and efficacy of retroviral gene therapy for
immunodeficiency due to adenosine deaminase defi-
• PIDs require a tailored approach to ciency. Blood. 2016;128:45–54.
management, and transplant-specific Cole T, McKendrick F, Titman P, et  al. Health related
protocols have been developed for these quality of life and emotional health in children with
chronic granulomatous disease: a comparison of
diseases. those managed conservatively with those that have
• Preceding comorbidity, particularly undergone haematopoietic stem cell transplant. J Clin
infectious complications at HSCT and Immunol. 2013;33:8–13.
concurrent end-organ damage, adversely Dvorak CC, Hassan A, Slatter MA, et al. Comparison of
outcomes of hematopoietic stem cell transplantation
affects outcome; for many diseases, trans- without chemotherapy conditioning by using matched
plant at an early age is recommended. sibling and unrelated donors for treatment of severe
• Alternative therapies instead of, or as a combined immunodeficiency. J Allergy Clin Immunol.
bridge to, transplant are increasingly 2014;134:935–943.e15.
Fernandes JF, Rocha V, Labopin M, et al. Transplantation
available to improve patient outcomes. in patients with SCID: mismatched related stem cells
or unrelated cord blood? Blood. 2012;119:2949–55.
Fischer A, Landais P, Friedrich W, et al. European experi-
ence of bone-marrow transplantation for severe com-
bined immunodeficiency. Lancet. 1990;336:850–4.
References Fox TA, Chakraverty R, Burns S, et  al. Successful out-
come following allogeneic hematopoietic stem cell
Aiuti A, Cattaneo F, Galimberti S, et al. Gene therapy for transplantation in adults with primary immunodefi-
immunodeficiency due to adenosine deaminase defi- ciency. Blood. 2018;131:917–31.
ciency. N Engl J Med. 2009;360:447–58. Gennery AR, Slatter MA, Grandin L, Inborn Errors
Aiuti A, Biasco L, Scaramuzza S, et al. Lentiviral hemato- Working Party of the European Group for Blood
poietic stem cell gene therapy in patients with Wiskott-­ and Marrow Transplantation, European Society for
Aldrich syndrome. Science. 2013;341:1233151. Immunodeficiency, et  al. Transplantation of hema-
Albert MH, Hauck F, Wiebking V, et al. Allogeneic stem topoietic stem cells and long-term survival for pri-
cell transplantation in adolescents and young adults mary immunodeficiencies in Europe: entering a new
with primary immunodeficiencies. J Allergy Clin century, do we do better? J Allergy Clin Immunol.
Immunol Pract. 2018;6:298–301. 2010;126:602–10.
Antoine C, Müller S, Cant A, European Group for Blood Güngör T, Albert MH, Teira P, et  al. Reduced-intensity
and Marrow Transplantation; European Society for conditioning and HLA-matched haemopoietic stem-­
Immunodeficiency, et  al. Long-term survival and cell transplantation in patients with chronic granu-
transplantation of haemopoietic stem cells for immu- lomatous disease: a prospective multicentre study.
nodeficiencies: report of the European experience Lancet. 2014;838:436–48.
1968-99. Lancet. 2003;361:553–60. Hacein-Bey-Abina S, Le Deist F, Carlier F, et al. Sustained
Balashov D, Shcherbina A, Maschan M, et  al. Single-­ correction of X-linked severe combined immuno-
center experience of unrelated and haploidentical stem deficiency by ex  vivo gene therapy. N Engl J Med.
cell transplantation with TCRαβ and CD19 depletion in 2002;346:1185–93.
children with primary immunodeficiency syndromes. Hacein-Bey-Abina S, von Kalle C, Schmidt M, et  al.
Biol Blood Marrow Transplant. 2015;21:1955–62. LMO2-associated clonal T cell proliferation in two
Barzaghi F, Amaya Hernandez LC, Ambrosi A, et  al. patients after gene therapy for SCID-X1. Science.
IPEX syndrome survivors following different thera- 2003;302:415–9.
peutic strategies: an international multicenter retro- Hacein-Bey-Abina S, Pai SY, Gaspar HB, et al. A modi-
spective study. J Allergy Clin Immunol. 2018;141: fied γ-retrovirus vector for X-linked severe combined
1036–49. immunodeficiency. N Engl J Med. 2014;371:1407–17.
Boztug K, Schmidt M, Schwarzer A, et al. Stem-cell gene Heimall J, Logan BR, Cowan MJ, et al. Immune recon-
therapy for the Wiskott-Aldrich syndrome. N Engl J stitution and survival of 100 SCID patients post-­
Med. 2010;363:1918–27. hematopoietic cell transplant: a PIDTC natural history
Braun CJ, Boztug K, Paruzynski A, et al. Gene therapy for study. Blood. 2017;130:2718–27.
Wiskott-Aldrich syndrome—long-term efficacy and Jhamnani RD, Rosenzweig SD.  An update on gain-­
genotoxicity. Sci Transl Med. 2014;6:227ra33. of-­function mutations in primary immunodefi-
Chan B, Wara D, Bastian J, et al. Long-term efficacy of ciency diseases. Curr Opin Allergy Clin Immunol.
enzyme replacement therapy for adenosine deaminase 2017;17:391–7.
(ADA)-deficient Severe Combined Immunodeficiency Kwan A, Abraham RS, Currier R, et al. Newborn screen-
(SCID). Clin Immunol. 2005;117:133–43. ing for severe combined immunodeficiency in 11
670 M. Albert et al.

screening programs in the United States. JAMA. Shah RM, Elfeky R, Nademi Z, et al. T-cell receptor αβ+
2014;312:729–38. and CD19+ cell-depleted haploidentical and mis-
Moratto D, Giliani S, Bonfim C, et  al. Long-term out- matched hematopoietic stem cell transplantation in
come and lineage-specific chimerism in 194 patients primary immune deficiency. J Allergy Clin Immunol.
with Wiskott-Aldrich syndrome treated by hema- 2018;141:1417–26.
topoietic cell transplantation in the period 1980-­ Slack J, Albert MH, Balashov D, Inborn Errors Working
2009: an international collaborative study. Blood. Party of the European Society for Blood and
2011;118:1675–84. Marrow Transplantation and the European Society
Notarangelo LD, Kim MS, Walter JE, et al. Human RAG for Immunodeficiencies, Stem Cell Transplant for
mutations: biochemistry and clinical implications. Nat Immunodeficiencies in Europe (SCETIDE), Center for
Rev Immunol. 2016;16:234–46. International Blood and Marrow Transplant Research,
Ochs HD, Petroni D.  From clinical observations and Primary Immunodeficiency Treatment Consortium,
molecular dissection to novel therapeutic strategies et al. Outcome of haematopoietic stem cell transplan-
for primary immunodeficiency disorders. Am J Med tation for DNA-double strand breakage repair disor-
Genet A. 2018;176:784–803. ders. J Allergy Clin Immunol. 2018;141:322–8.
Ott MG, Schmidt M, Schwarzwaelder K, et al. Correction Slatter M, Rao K, Nademi Z, et al. Treosulfan and fludara-
of X-linked chronic granulomatous disease by bine conditioning for haematopoietic stem cell trans-
gene therapy, augmented by insertional activation plantation in children with Primary Immunodeficiency:
of MDS1-EVI1, PRDM16 or SETBP1. Nat Med. UK experience. Biol Blood Marrow Transplant.
2006;12:401–9. 2018;24:529–36.
Pai S-Y, Logan BR, Griffith LM, et  al. Transplantation Speckmann C, Doerken S, Aiuti A, PCID-study of the
outcomes for severe combined immunodefi- Inborn Errors Working Party of the EBMT, et  al. A
ciency, 2000–2009. N Engl J Med. 2014;371: prospective study on the natural history of patients
434–46. with profound combined immunodeficiency (P-CID):
Picard C, Bobby Gaspar H, Al-Herz W, et al. International a “first fifty” analysis. J Allergy Clin Immunol.
Union of Immunological Societies: 2017 pri- 2017;139:1302–10.
mary immunodeficiency diseases committee report Stein S, Ott MG, Schultze-Strasser S, et al. Genomic insta-
on inborn errors of immunity. J Clin Immunol. bility and myelodysplasia with monosomy 7 conse-
2018;38:96–128. quent to EVI1 activation after gene therapy for chronic
Rechavi E, Lev A, Simon AJ, et  al. First Year of Israeli granulomatous disease. Nat Med. 2010;16:198–204.
newborn screening for severe combined immunode- Veys P.  Reduced intensity transplantation for primary
ficiency—clinical achievements and insights. Front immunodeficiency disorders. Immunol Allergy Clin N
Immunol. 2017;8:1448. Am. 2010;30:103–24.

Open Access  This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license,
unless indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons
license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to
obtain permission directly from the copyright holder.

You might also like