Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Mar 5.
Published in final edited form as: Curr Allergy Asthma Rep. 2021 Mar 5;21(3):14. doi: 10.1007/s11882-020-00984-8

Disease presentation, treatment options and outcomes for myeloid immunodeficiencies

Elizabeth Kang 1
PMCID: PMC8162003  NIHMSID: NIHMS1697869  PMID: 33666780

Abstract

Purpose:

Up to date review on various types of immunodeficiencies with a significant myeloid component including some more recently described congenital disorders.

Summary:

Neutrophils (or granulocytes) are a major contributor to infection surveillance and clearance and defective neutrophils characteristically lead to pyogenic infections. Deficiency in numbers, either iatrogenic or congenital, functional defects, and/or inability to target to the sites of infection can all lead to serious morbidity and mortality; however, myeloid based immunodeficiencies are not all the same. Having absent neutrophils, that is neutropenia, has implications different to those of having dysfunctional neutrophils as will become evident as the various disorders are reviewed.

Recent Findings:

While a number of disorders have been described in the past, genetic sequencing has led to the identification of the specific disorders and clarified their pathophysiology. Advances in genetic therapies including genetic editing should provide future treatments beyond hematopoieitic stem cell transplant for patients with these rare disorders

Introduction

A simplified way to classify neutrophilic disorders is by categorizing them based on their primary defect. Although various pathways may be affected, there is often a predominant phenotype that characterizes these disorders based either on 1) compromised mobility, either due to an inability to respond to chemotactants or adhere properly, 2) inadequate ability to phagocytose and/or create the respiratory burst needed to kill various microbes or 3) inadequate numbers (neutropenias). A few genetic mutations though appear to interfere significantly in multiple pathways and therefore do not necessarily fit a specific pattern (See Table 1). However, patients with primarily neutrophilic deficiencies typically present at an early age with a pyogenic infection, or even earlier with signs such as delayed cord healing/separation and omphalitis. Other immunodeficiencies such as Wiskott-Aldrich Syndrome or Hyper IgM’s may also involve the myeloid component either through aberrant cytokine or ligand signaling, defective phagocytosis or even due to decreased granulocytic numbers, so those will also be briefly reviewed here.

Table 1.

Myeloid Immunodeficiencies by phenotype/underlying deficiency. A few genetic mutations appear to interfere significantly in multiple pathways

Type Disease Inheritance Genes Incidence Defect Current Management
Mobility/Signaling Leukocyte adhesion deficiency I (LAD I) AR ITGB2 1 in 1000000 B-2 integrin (CD18) deficiency, HSCT- gene therapy studies ongoing
LAD 2 (Congenital disorder of glycosylation CDG) AR SLC35Ca <1 in 1000000 Aberrant GDP fucose transporter leading to lack of selectins needed for rolling. HSCT- gene therapy studies ongoing
LAD3 (LAD V1) AR FERMT3 (Kindlin 3) <1 in 1000000 Defective Rap-1 activation leading to loss of tight adherence and Glanzmann’s thrombocytopenia HSCT- gene therapy studies ongoing
CARD9 deficiency AR CARD9 Rare CARD9 deficiency, poor cytokine production, impaired phagocytosis Antifungals, surgical resection, GMCSF, HSCT in selected cases?
WDR1 deficiency AR WDR1 Rare Abnormal actininteracting protein 1 (Aip1) resulting in abnormal actin depolymerization Treatment of infections, HSCT
MyD88/IRAK4 deficiency AR MyD88/IRAK4 Rare Altered signalling through TLRs and IL-1 Prophylactic antibiotics until adulthood
Impaired killing
Respiratory burst CGD X-linked, AR CYBB, CYBA, NCF1, NCF2, NCF4 1 in 250000 Defective respiratory burst- due to NADPH deficiency Prophylactic Antibiotics, HSCT, Gene therapy
MPO AR MPO 1 in 2--4000 Europeans/NA and 1 in 55000 Japanese Inability to produce HOCL Treatment of Infections
Severe G6PD deficiency X-linked G6PD most common enzyme deficiency Severe G6PD can cause reduction in the NADPH oxidation Avoidance of triggers
Granule Specific granule deficiency AR CEBP epsilon RARE Lack of myeloid differentiation, altered secondary granule formation, abnormal chemotaxis HSCT
Miscellaneous RAC2 deficiency AR RAC2 gene one case Multiple abnormalities including chemotaxis defects, respiratory burst decrease, adhesion defects HSCT
Neutropenias Severe congenital neutropenia (SCN)and Cyclical neutropenias AR/AD HAX1, ELANE, CSFR3 1 in 200000 Decreased total or oscillations in numbers of neutrophils- pathways of development or survival affected GCSF, monitoring for leukemia, occasional HSCT
Autoimmune neutropenia of infancy unknown 1 in 100000 Autoimmune mediated clearance of neutrophils Usually resolves spontaneously
Glutathione synthetase deficiency (5- oxoprolinuria) AR GSS Accumulation of oxidative damage to neutrophils Symptomatic management
Myeloid component Wiskott Aldrich/X-linked Neutropenia X-linked WAS/Exon 9 mutations 1 in 1000000 WAS protein mutations affecting actin polymerization HSCT/gene therapy
Chediak-Higashi AR LYST mutations- <1 in 1000000 Abnormal granule formation leading to pigment problems, granule malformation, platelet dysfunction HSCT
X-linked HyperIgM/CD40 deficiency X-linked/AR CD40 ligand, CD40 1 in 250000 Failure to B cell switch HSCT

1). Disorders affecting neutrophil migration or cytokine signaling

Myeloid differentiation primary response 88 (MYD88)/Interleukin 1 receptor associated kinase 4 (IRAK4) deficiencies.

Mutations in MYD88 or its downstream signaling molecule IRAK4 have been described in patients presenting with bacterial infections, the nature and sites of which seem to be well delineated: mostly invasive pneumococcal disease, cutaneous and invasive staphylococcal disease, and Pseudomonas infection of the upper respiratory tract. 1, 2

MYD88 and IRAK4 are required in TLR2, TLR4 and Il-1 signaling but not in TLR3 and TNF-alpha signaling. The phenotype may vary though depending on the type of infection as one patient with a homozygous truncation in MYD88 had decreased TNF-alpha as well as decreased Il-6, and presented with delayed umbilical cord separation as well as BCGitis in addition to a pseudomonal infection. Another set of siblings were also described with a unique homozygous mutation and presented with Salmonellosis along with other more typical infections3,4. The majority of patients appear to outgrow their infection susceptibility though, possibly due to the development of adaptive T and B cell responses and/or maturation of other innate immune responses.2 Thus management consists of antibacterial prophylaxis to cover those specific bacteria in childhood which can be then discontinued in adulthood.

Leukocyte Adhesion Deficiencies (LAD)

The first case series of 6 infants with delayed separation of their umbilical cords was described in 1979. Five of the 6 patients succumbed to severe bacterial infections and ascorbic acid was hypothesized to aid in the immunodeficiency, but was not used.5 Subsequently, it was recognized to be a disorder of adhesion attributed to a CR3 deficiency.69 Leukocyte adhesion deficiency (LAD) type I as the disorder is now known results from mutations of the B2 integrin (also known as CD18) gene ITGR2 on Chromosome 21 and is inherited in an autosomal recessive fashion with severity of the phenotype dependent on the level of expression of the protein.10 CD18 complexes with CD11a to form LFA-1, with CD11b to form MAC-1, and CD11c to form p150,95 which after activation are required for binding to ICAM-1, fibrinogen and other ligands to be able to migrate to and phagocytose bacteria properly.11, 12

LAD type II is a separate disorder due to mutations in the GDP fucose transporter gene resulting in a failure of glycosylation of a fucose leading to lack of a Sialyl-Lewis-X- the carbohydrate ligand for P and E selectins.13 However fucose is also required for the carbohydrate structures responsible for ABO typing of blood. Patients thus have a characteristic Bombay (H-) blood phenotype.14 Patients present with a myriad of problems including neurologic developmental delays, growth retardation, anemia, and bleeding as well as infections. This is a much rarer form of LAD and has been described in only a handful of families.15

LAD type III is often referred to as a variant of LAD type I as the phenotype is similar. Mutations in the Kindlin3 (or FERMT3) gene result in aberrant integrin binding to selectins on the endothelium.16 Neutrophils are unable to roll and again do not reach the sites of infection required for their function.17

The management for all patients with the LAD subtypes, particularly those with severe disease, is hematopoietic stem cell transplant (HSCT) from an unaffected sibling donor, or if not available, a matched unrelated donor. 18, 19 Given the variance in phenotype depending on the expression levels of the proteins, particularly in LAD type I, mixed chimerism may be sufficient for phenotypic resolution in these patients. Centres have been trying reduced toxicity or nonmyeloablative conditioning regimens with some success.20 As with all genetic disorders there are efforts to develop a gene therapy using in vitro gene correction of hematopoietic stem cells. There is a canine model of LAD type I and dogs have been successfully treated with both lentiviral vectors and more recently foamy virus vectors expressing the B2 integrin, providing proof of principal. 21, 22 Currently, a clinical study is ongoing for pediatric patients with severe LAD type I in the US with high gene marking, 47% restored CD18 expression (<1% pre-gene therapy), and clinical improvement in inflammatory skin lesions in one patient with at least 6 months followup as presented at the 2020 American Society for Gene and Cell Therapy Annual Meeting.

Caspase recruitment domain-containing protein 9 (CARD9) deficiency

CARD9 deficiency is an unusual myeloid immunodeficiency inherited in an autosomal recessive fashion, whereby patients have a specific susceptibility to fungal infections which can range from chronic mucocutaneous infections due to Candida, to deep invasive infections including extrapulmonary aspergillosis.23, 24 Mutations lead to impaired cytokine and chemokine production by macrophages, reduced neutrophil recruitment, impaired fungal killing and a decrease in Th17 cells. Treatment has been antifungals and surgical resection of infected masses as well as GCSF or GMCSF.25 HSCT has been used with varying results. 26, 27 Gene therapy using a lentiviral vector is being developed (personal communication).

WDR1 deficiency

This is a very rare disorder initially described in a patient who presented with various infections and an unusual morphology to her neutrophils, wherein her neutrophils migrated abnormally, and extruded their nuclei in cytoplasmic blebs.28 Other patients have also been described with a type of lazy leucocyte syndrome; however the pathophysiology was not understood at the time, although hypothesized to involve altered membrane proteins.29, 30 The defect was later characterized as a deficiency in the WDR1 gene which encodes for the Actin-interacting protein 1 (AIP1). HSCT was then used to treat one patient.31 Further description of mutations in other patients suggest more profound defects can result in immune dysregulation and inflammasome activation as well as the stomatitis and recurrent infections initially described.32 Overall the prognosis is mixed with some patients seeming to improve symptomatically over time and others succumbing to infection 31 (personal communication).

2). Disorders affecting mechanisms of microbial killing

Myeloperoxidase (MPO) deficiency

MPO deficiency is the most common enzymatic disorder affecting white blood cells with an estimated incidence of 1 in 2000 for moderate deficiency. Originally it was thought to be much rarer with a few patients presenting with infectious complications. However biochemical and phenotypic characterization have found the prevalence to be much higher with some variation in ethnic populations (1 in 55000 described in the Japanese population versus 1 in 4000 (severe deficiency) in the European and North American groups).33 The defect occurs due to mutations in the MPO gene and is inherited in an autosomal recessive manner. MPO is required for the production of hypochlorous acid (HOCL) by oxidation of chloride anions. 34 Although in vitro, this HOCL production appears necessary for the killing of Candida, the vast majority of patients have few to no symptoms, as this HOCL production appears to be redundant as part of the microbial killing pathway in vivo. 35

Treatment of infections as they occur appears adequate with no prophylaxis normally recommended. There are a few case reports of patients presenting with severe/life threatening infections, but these have been described prior to the implementation of whole genome sequencing and therefore there may be adjuvant factors contributing to these rarer presentations.

Chronic Granulomatous Disease (CGD)

CGD is a much rarer disorder of neutrophil function estimated to occur in 1 of 250000 births, but results in significantly more morbidity and mortality.36 CGD is characterized by a failure of neutrophils to produce NADPH oxidase which is crucial to the suppression and killing of certain types of bacteria (most commonly catalase positive) such as Burkholderia and Serratia as well as Staphylococcus aureus in addition to atypical bacteria, most significantly Nocardia. Patients are also very susceptible to fungal infections, particularly aspergillus but with more common use of azoles, there is now an increased incidence of other dematiaceous mold infections. CGD patients however are not particularly susceptible to Candida.37, 38

Possibly due to the ineffective killing, patients also develop granulomas, hence the name of the disease. These often involve tubular organs, such as the urinary tract, or esophagus and can become obstructive but respond quickly to a course of steroids. Patients also develop microabscesses that resemble small microtumours and the addition of steroids to treat these infections has proven helpful if somewhat paradoxically in the setting of an immunodeficiency.39 Patients can also develop hyperinflammatory response to exposure of microbial elements such as aspergillus or nocardia and require steroids in addition to aggressive antifungal treatment for resolution. 40

Distinct from the granulomas and the hyperinflammation, a significant proportion of patients also have autoinflammatory disorders, such as pneumonitis, arthritides and lupus, with estimates of 40 to 50% having inflammatory bowel disease (IBD) alone.41 Their innate immunity appears relatively intact with good responses to vaccines and normal immunoglobulin levels and the cause of the autoinflammation is not well understood although there may be some genetic factors.42

There are 5 subtypes of CGD with the most common form being the X-linked involving the CYBB gene, resulting in a failure to produce GP91phox. Autosomal recessive forms consist of P22, P47 and P67 deficiencies due to mutations in the CYBA, NCF1 and NCF2 genes respectively. 43 A 5th type has been more recently described, with mutations in the NCF4 gene affecting expression of the P40phox; however these patients presented with significant inflammatory disease and not infection. In fact, their respiratory burst appears mostly intact and laboratory diagnosis using Dihydrorhodamine (DHR) based flow cytometry is equivocal. 44, 45 DHR however is the preferred method of diagnosis as it can be quantitative, and even diagnostic for the various forms. Of course genetic sequencing is also helpful, but nitroblue tetrazolium staining should no longer be used. Patients who produce some oxidase (more commonly in the P47 form) appear to have better prognosis than those with little to no oxidase production.46 Significant skewing of the X-chromosome lyonization in female carriers, has resulted in CGD specific infections when levels of 10% to 20% or less of oxidase expression are reached. 47 Treatment for patients was traditionally prophylaxis with Bactrim (or ciprofloxacin in sulfa allergic patients) and itraconazole; however now more often third generation azoles such as posaconazole or voriconazole. 48 Interferon gamma was also shown to be useful in a randomized trial, but is not necessarily well tolerated.4951 It is usually held during an active infection. Pioglitazone has also been suggested to improve infection responses.52

Corticosteroids are very successful in the treatment of the granulomas but less so for the autoimmune type disorders seen, with many patients requiring high doses or the use of other agents such as monoclonal antibodies/biologics for steroid reduction. These alternate therapies can be effective but may increase the risk of infection in this immunocompromised population.53, 54

HSCT has become much more common and overall survival ranges from 80–90% in various studies. 55 Regimens vary but the majority now are of a reduced intensity/reduced toxicity. Graft rejection rates are higher than many of the other ‘classical immunodeficiencies’ with rates ranging from 12–20% often occurring late.5660 With improving outcomes using matched donors, various centres have been exploring the use of mismatched donors to expand the donor pool for appropriate patients with mixed results. 61, 62 Transplant has also been used successfully in a few rare cases for carriers with skewed lyonization (personal experience). As mentioned, data from X-linked carriers suggests that for phenotypic correction of the infection susceptibility, 10–20% oxidase positivity is sufficient. Thus mixed myeloid chimerism may be sufficient for cure; however the threshold required for reversal of autoimmunity is less clear. 63 The Primary Immune Deficiency Treatment Consortium (PIDTC) evaluated a number of patients treated within North America with HSCT and found that IBD resolves in all patients, but there were no patients who had myeloid engraftment of less than 85%. Also of note, patients with inflammatory bowel disease did not have a statistically higher rate of GvHD nor was there an impact on survival thus supporting the use of HSCT for management of patients with recalcitrant inflammatory disease. 64

Given the benefits of mixed chimerism, CGD is a natural candidate for gene modification of hematopoietic stem cells and was in fact one of the first immunodeficiencies to be studied clinically. Results were encouraging, but without conditioning, gene marking did not persist. 65, 66 The addition of busulfan improved results; however, the use of a retroviral vector was complicated by the development of myelodysplasia with activation of genes causing clonal outgrowth in one trial and eventual silencing and in other studies, inadequate marking for long term clinical benefit. 67, 68

More recently, lentiviral based gene therapy has had more success, with levels of greater than 10% oxidase positive cells detectable in many of the patients persisting for more than 2 years. However, again the level of correction necessary for autoimmune disease resolution is not clear, and patients in these trials did not have significant inflammation. Further a few patients have appeared to reject the grafts despite high dose busulfan. 69

Patients with large mutations of the CYBB gene may also present with McLeod’s syndrome due to the gene’s proximity which can complicate transplant but does not preclude it. Other genetic disorders affecting neighbouring genes have also been seen in conjunction with CGD such as the dystrophin gene, Ornithine transcarbamylase, and retinitis pigmentosa GTPase.70 Patients with severe Glucose 6 phosphate dehydrogenase (G6PD) deficiency can also have a CGD like phenotype as NADPH oxidase production is dependent upon the glycolytic pathway.

Specific granule deficiency (SGD)

First described in a young male presenting with infections from birth, the patient had bilobed nuclei with abnormal appearing specific granules and was found to have a lack of lactoferrin production. The disorder was ultimately found to be due to a mutation in the CCAAT/enhancer binding protein epsilon which is expressed exclusively in myeloid and T cell lineages. This class of proteins are key regulators of cellular differentiation and the myeloid cells appear to be blocked in their differentiation, resulting in decreased numbers with the few neutrophils having poor chemotaxis, impaired phagocytosis and microbial killing. 7173, 74, 75 A consideration for HDAC1 inhibitor use has been proposed for specific mutations based on invitro studies; however these have not been used clinically to date. 76 This is a rare autosomal recessive disorder but there is a report of one patient being successfully transplanted. 77

RAC2 deficiency

Probably the rarest disorder of neutrophils, this has been described in a patient who presented with a severe bacterial infection at 5 weeks of age. Genetic analysis revealed a point mutation in the RAC2 gene whose protein is a member of the Rho GTPAse family. The patient’s mutation resulted in an aspartic acid to asparagine change at the GTP binding site. The resulting neutrophils had defects in chemotaxis due to actin abnormalities, as well as a deficiency in respiratory burst formation as RAC 2 is necessary for NADPH formation. This patient successfully underwent allogeneic transplantation.78, 79 No other cases have been described to date, but this disorder points to other pathways and/or proteins that should be considered when evaluating for genetic defects involving myeloid based immunodeficiencies. 80, 81

3). Disorders affecting neutrophil numbers

Congenital Neutropenias

Severe congenital neutropenias (SCN) as well as cyclical neutropenias arise from the same genetic defects and are differentiated simply by the chronicity of the neutropenia and whether the counts oscillate. Mutations in ELANE, the gene responsible for neutrophil elastase production, accounts for the majority of patients with either SCN or cyclical neutropenia.82 HAX 1 mutations (also known as Kostmann’s) are the second most common cause of congenital neutropenias. Other genes that have been described include CSFR3,GF11, and G6PC3, which function in various pathways of myeloid development.83

Patients often present at birth or early age with low neutrophil counts, and a pyogenic infection or omphalitis similar to patients with LAD.

Management is typically with GCSF.84 A number of patients have also undergone allogeneic hematopoietic stem cell transplant with the Europeans reporting an overall survival of 82%.85 There is an increased predisposition to the development of myeloid leukemias in patients with these disorders, and there was concern that this might be related to the chronic GCSF use; however this may in fact be intrinsic to the diseases and not necessarily due to stimulation by the growth factor. 8688

Autoimmune neutropenia of infancy (AIN)

Another neutropenia diagnosed early in life is AIN. Due to the presence of autoimmune antibodies to neutrophils, patients can present with a congenital neutropenia type picture and are typically diagnosed within the first 7 to 9 months of life. In the vast majority of cases, the antibody titres wane and the neutropenia resolves by the 1st year of life. 89, 90 The development of these antibodies appears idiopathic and is not well understood.

Glutathione synthetase deficiency (GSD)

Finally, a very rare cause of congenital neutropenia has been attributed to GSD, although not a typical myeloid immunodeficiency per se as patients present with a multitude of symptoms. There has been a description of a patient being diagnosed with GSD after developing bacterial infections in the setting of neutropenia. The buildup of oxolopurine within the neutrophils leads to their destruction and thus reduced numbers and increased susceptibility to infections. 91 There is currently no treatment available.

4). Immunodeficiency Syndromes involving the myeloid lineage.

Wiskott-Aldrich Syndrome (WAS)

WAS is an X-linked disorder whose hallmark is that of thrombocytopenia with small platelets, but in which the defect affects multiple hematopoietic lineages and presents in a very heterogeneous manner. Patients are categorized as having mild disease (often referred to as X-linked thrombocytopenia) to severe, based on the development of infections, autoimmunity and/or malignancy.92 However patients can also progress from mild to severe disease and thus patients need lifelong monitoring particularly as patients with mild disease have a similar life expectancy to those with severe disease. 93

The WAS protein is part of a family of actin nucleation-promoting factors which is required for actin polymerization and cytoskeletal remodeling. Neutrophils are thus unable to respond to chemotactants, phagocytose adequately and also have reduced integrin-induced respiratory bursts. 9497 As all the white blood cell lineages are affected, patients can develop a wide variety of infections, including viral, bacterial, fungal, and opportunistic. They can also have significant problems from atopy, autoimmunity, and an increased risk of malignancy probably as a result of their disordered immune surveillance and regulation.

Notably there is a condition referred to as X-linked Neutropenia (XLN) that occurs from mutations in exon 9 of the WAS gene and these patients present with neutropenia and myelodysplasia without the other manifestations of Wiskott-Aldrich syndrome.98, 99

Allogeneic transplantation can be curative and is recommended for those with severe disease. It is less commonly considered in those with mild disease particularly as there appears to be a relatively high rate of autoimmune complications post-transplant, and mixed chimerism is not sufficient for full phenotypic reversion. 100, 101, 102, 103, 104, 105 Gene therapy has been hampered by the development of malignancies in a retroviral vector study; however the use of lentiviral vectors appears promising. 106110

Chediak-Higashi Syndrome

Chediak-Higashi patients have mutations in the LYST gene which is required for lysosomal granule formation. Patients have albinism, platelet dysfunction with a bleeding diathesis but also develop infections related to a neutrophilic granule defect where the neutrophils are characterized by abnormally large azurophilic granules. A number of patients will develop an accelerated phase of the disease which resembles hemophagocytic lymphohistiocytosis and may be related to macrophage or Natural Killer cell dysfunction. Patients have a shortened life expectancy and the majority are treated with HSCT including cord blood transplants. 111, 112

Hyper IgM Syndrome

HIGM syndromes are a group of disorders that are characterized by failure of B cells to class switch, leading to decreased IgG and IgA serum levels, but normal or elevated serum IgM. The X-linked form (XHIGM) is the most common subtype and arises from mutations in the CD40 ligand gene. These patients are particularly susceptible to Pneumocystis carinii pneumonia, diarrhea secondary to Cryptosporidium and 67% of patients will also present with neutropenia. 113,114 Despite intravenous immunoglobulin supplementation, use of GCSF and antibiotic prophylaxis, patients do not do well with an estimated 20% survival beyond the age of 25 in one European study.113 Cryptosporidial infections are particularly significant for development of chronic cholangitis and liver failure; thus patients are restricted to boiled water.114 Most patients are offered HSCT using a variety of type of donors; however outcomes are better when using a matched donor and patients are transplanted prior to infection.115, 116 CD40 deficiency which is a rare autosomal recessive form of the disease presents similarly in patients; however transplantation is less successful as the CD40 deficiency is not corrected in the nonhematopoietic cell lineages by HSCT.117,118 Although gene therapy has been considered, dominant negative effects have been shown in in vitro modeling and unregulated expression may be detrimental, thus therapies may depend on the success of gene editing as opposed to gene addition. 119, 120

Conclusions

As this review demonstrates, myeloid based immunodeficiencies are varied in their pathophysiology but present similarly with recurrent bacterial infections early in life and therefore should be considered in any patient presenting with a pyogenic infection within the first or second year of life. Genetic sequencing has significantly helped with the elucidation of many of the pathophysiologies; however, genetic mutations do not always explain the variety of phenotypes and presentations. As gene therapy and eventually gene editing improve and become more available clinically, the barriers of transplantation including availability of donors and the toxicities of conditioning regimens as well as the risks of graft versus host disease will hopefully be replaced by curative treatments with less adverse effects and better outcomes overall. Finally, as further research continues to elucidate the pathways of neutrophil function, novel therapies such as small molecule adjuncts or monoclonal antibodies will mostly likely be developed.

Footnotes

Compliance with Ethics Guidelines

Conflict of Interest

The authors declare no conflicts of interest relevant to this manuscript.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of a an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Picard C, von Bernuth H, Ghandil P, et al. Clinical features and outcome of patients with IRAK-4 and MyD88 deficiency. Medicine (Baltimore) 2010; 89: 403–425. 2010/November/09. DOI: 10.1097/MD.0b013e3181fd8ec3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ku CL, von Bernuth H, Picard C, et al. Selective predisposition to bacterial infections in IRAK-4-deficient children: IRAK-4-dependent TLRs are otherwise redundant in protective immunity. J Exp Med 2007; 204: 2407–2422. 2007/September/26. DOI: 10.1084/jem.20070628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Gokturk B, Casanova JL, Picard C, et al. A Novel Homozygous Mutation With Different Clinical Presentations in 2 IRAK-4-Deficient Siblings: First Case With Recurrent Salmonellosis and Non-Hodgkin Lymphoma. J Investig Allergol Clin Immunol 2018; 28: 271–273. 2018/August/04. DOI: 10.18176/jiaci.0261. [DOI] [PubMed] [Google Scholar]
  • 4.Platt CD, Zaman F, Wallace JG, et al. A novel truncating mutation in MYD88 in a patient with BCG adenitis, neutropenia and delayed umbilical cord separation. Clin Immunol 2019; 207: 40–42. 2019/July/14. DOI: 10.1016/j.clim.2019.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hayward AR, Harvey BA, Leonard J, et al. Delayed separation of the umbilical cord, widespread infections, and defective neutrophil mobility. Lancet 1979; 1: 1099–1101. 1979/May/26. DOI: 10.1016/s0140-6736(79)91786-0. [DOI] [PubMed] [Google Scholar]
  • 6.Bowen TJ, Ochs HD, Altman LC, et al. Severe recurrent bacterial infections associated with defective adherence and chemotaxis in two patients with neutrophils deficient in a cell-associated glycoprotein. J Pediatr 1982; 101: 932–940. 1982/December/01. DOI: 10.1016/s0022-3476(82)80013-9. [DOI] [PubMed] [Google Scholar]
  • 7.Springer TA, Thompson WS, Miller LJ, et al. Inherited deficiency of the Mac-1, LFA-1, p150,95 glycoprotein family and its molecular basis. J Exp Med 1984; 160: 1901–1918. 1984/December/01. DOI: 10.1084/jem.160.6.1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Malech HL and Gallin JI. Current concepts: immunology. Neutrophils in human diseases. N Engl J Med 1987; 317: 687–694. 1987/09/10. DOI: 10.1056/NEJM198709103171107. [DOI] [PubMed] [Google Scholar]
  • 9.Buchanan MR, Crowley CA, Rosin RE, et al. Studies on the interaction between GP-18–0-deficient neutrophils and vascular endothelium. Blood 1982; 60: 160–165. 1982/July/01. [PubMed] [Google Scholar]
  • 10.Anderson DC, Schmalsteig FC, Finegold MJ, et al. The severe and moderate phenotypes of heritable Mac-1, LFA-1 deficiency: their quantitative definition and relation to leukocyte dysfunction and clinical features. J Infect Dis 1985; 152: 668–689. 1985/October/01. DOI: 10.1093/infdis/152.4.668. [DOI] [PubMed] [Google Scholar]
  • 11.Stewart M and Hogg N. Regulation of leukocyte integrin function: affinity vs. avidity. J Cell Biochem 1996; 61: 554–561. 1996/June/15. DOI: . [DOI] [PubMed] [Google Scholar]
  • 12.McDowall A, Leitinger B, Stanley P, et al. The I domain of integrin leukocyte function-associated antigen-1 is involved in a conformational change leading to high affinity binding to ligand intercellular adhesion molecule 1 (ICAM-1). J Biol Chem 1998; 273: 27396–27403. 1998/October/09. DOI: 10.1074/jbc.273.42.27396. [DOI] [PubMed] [Google Scholar]
  • 13.Lubke T, Marquardt T, Etzioni A, et al. Complementation cloning identifies CDG-IIc, a new type of congenital disorders of glycosylation, as a GDP-fucose transporter deficiency. Nat Genet 2001; 28: 73–76. 2001/April/28. DOI: 10.1038/ng0501-73. [DOI] [PubMed] [Google Scholar]
  • 14.Bhende YM, Deshpande CK, Bhatia HM, et al. A “new” blood group character related to the ABO system. Lancet 1952; 1: 903–904. 1951/May/03. [PubMed] [Google Scholar]
  • 15.Gazit Y, Mory A, Etzioni A, et al. Leukocyte adhesion deficiency type II: long-term follow-up and review of the literature. J Clin Immunol 2010; 30: 308–313. 2010/January/26. DOI: 10.1007/s10875-009-9354-0. [DOI] [PubMed] [Google Scholar]
  • 16.DeLisser HM, Christofidou-Solomidou M, Sun J, et al. Loss of endothelial surface expression of E-selectin in a patient with recurrent infections. Blood 1999; 94: 884–894. 1999/July/27. [PubMed] [Google Scholar]
  • 17.Moser M, Bauer M, Schmid S, et al. Kindlin-3 is required for beta2 integrin-mediated leukocyte adhesion to endothelial cells. Nat Med 2009; 15: 300–305. 2009/February/24. DOI: 10.1038/nm.1921. [DOI] [PubMed] [Google Scholar]
  • 18.Fischer A, Trung PH, Descamps-Latscha B, et al. Bone-marrow transplantation for inborn error of phagocytic cells associated with defective adherence, chemotaxis, and oxidative response during opsonised particle phagocytosis. Lancet 1983; 2: 473–476. 1983/August/27. DOI: 10.1016/s0140-6736(83)90509-3. [DOI] [PubMed] [Google Scholar]
  • 19.Essa MF, Elbashir E, Alroqi F, et al. Successful hematopoietic stem cell transplant in leukocyte adhesion deficiency type III presenting primarily as malignant infantile osteopetrosis. Clin Immunol 2020; 213: 108365. 2020/February/25. DOI: 10.1016/j.clim.2020.108365. [DOI] [PubMed] [Google Scholar]
  • 20.Engel ME, Hickstein DD, Bauer TR Jr., et al. Matched unrelated bone marrow transplantation with reduced-intensity conditioning for leukocyte adhesion deficiency. Bone Marrow Transplant 2006; 37: 717–718. 2006/February/21. DOI: 10.1038/sj.bmt.1705301. [DOI] [PubMed] [Google Scholar]
  • *21.Bauer TR Jr., Allen JM, Hai M, et al. Successful treatment of canine leukocyte adhesion deficiency by foamy virus vectors. Nat Med 2008; 14: 93–97. 2007/December/25. DOI: 10.1038/nm1695.Description of the results using a novel (foamy) viral vector for the treatment of canine LAD.
  • 22.Bauer TR Jr., Tuschong LM, Calvo KR, et al. Long-term follow-up of foamy viral vector-mediated gene therapy for canine leukocyte adhesion deficiency. Mol Ther 2013; 21: 964–972. 2013/March/28. DOI: 10.1038/mt.2013.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *23.Corvilain E, Casanova JL and Puel A. Inherited CARD9 Deficiency: Invasive Disease Caused by Ascomycete Fungi in Previously Healthy Children and Adults. J Clin Immunol 2018; 38: 656–693. 2018/August/24. DOI: 10.1007/s10875-018-0539-2.Description of a novel disease and elucidation of a pertinent pathway for immunity.
  • 24.Drummond RA, Franco LM and Lionakis MS. Human CARD9: A Critical Molecule of Fungal Immune Surveillance. Front Immunol 2018; 9: 1836. 2018/August/22. DOI: 10.3389/fimmu.2018.01836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Celmeli F, Oztoprak N, Turkkahraman D, et al. Successful Granulocyte Colony-stimulating Factor Treatment of Relapsing Candida albicans Meningoencephalitis Caused by CARD9 Deficiency. Pediatr Infect Dis J 2016; 35: 428–431. 2015/December/15. DOI: 10.1097/INF.0000000000001028. [DOI] [PubMed] [Google Scholar]
  • 26.Rieber N, Gazendam RP, Freeman AF, et al. Extrapulmonary Aspergillus infection in patients with CARD9 deficiency. JCI Insight 2016; 1: e89890. 2016/October/26. DOI: 10.1172/jci.insight.89890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Queiroz-Telles F, Mercier T, Maertens J, et al. Successful Allogenic Stem Cell Transplantation in Patients with Inherited CARD9 Deficiency. J Clin Immunol 2019; 39: 462–469. 2019/June/22. DOI: 10.1007/s10875-019-00662-z. [DOI] [PubMed] [Google Scholar]
  • 28.Gallin JI, Malech HL, Wright DG, et al. Recurrent severe infections in a child with abnormal leukocyte function: possible relationship to increased microtubule assembly. Blood 1978; 51: 919–933. 1978/May/01. [PubMed] [Google Scholar]
  • 29.Pinkerton PH, Robinson JB and Senn JS. Lazy leucocyte syndrome--disorder of the granulocyte membrane? J Clin Pathol 1978; 31: 300–308. 1978/April/01. DOI: 10.1136/jcp.31.4.300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Constantopoulos A, Karpathios T, Nicolaidou P, et al. Lazy-leukocyte syndrome. A case report. J Pediatr 1975; 87: 945–946. 1975/December/01. DOI: 10.1016/s0022-3476(75)80913-9. [DOI] [PubMed] [Google Scholar]
  • 31.Kuhns DB, Fink DL, Choi U, et al. Cytoskeletal abnormalities and neutrophil dysfunction in WDR1 deficiency. Blood 2016; 128: 2135–2143. 2016/October/30. DOI: 10.1182/blood-2016-03-706028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Standing AS, Malinova D, Hong Y, et al. Autoinflammatory periodic fever, immunodeficiency, and thrombocytopenia (PFIT) caused by mutation in actin-regulatory gene WDR1. J Exp Med 2017; 214: 59–71. 2016/December/21. DOI: 10.1084/jem.20161228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kitahara M, Eyre HJ, Simonian Y, et al. Hereditary myeloperoxidase deficiency. Blood 1981; 57: 888–893. 1981/May/01. [PubMed] [Google Scholar]
  • 34.Nauseef WM, Root RK and Malech HL. Biochemical and immunologic analysis of hereditary myeloperoxidase deficiency. J Clin Invest 1983; 71: 1297–1307. 1983/May/01. DOI: 10.1172/jci110880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Klebanoff SJ, Kettle AJ, Rosen H, et al. Myeloperoxidase: a front-line defender against phagocytosed microorganisms. J Leukoc Biol 2013; 93: 185–198. 2012/October/16. DOI: 10.1189/jlb.0712349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Winkelstein JA, Marino MC, Johnston RB Jr., et al. Chronic granulomatous disease. Report on a national registry of 368 patients. Medicine (Baltimore) 2000; 79: 155–169. 2000/June/09. [DOI] [PubMed] [Google Scholar]
  • 37.Marciano BE, Spalding C, Fitzgerald A, et al. Common severe infections in chronic granulomatous disease. Clin Infect Dis 2015; 60: 1176–1183. 2014/December/30. DOI: 10.1093/cid/ciu1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.De Ravin SS, Challipalli M, Anderson V, et al. Geosmithia argillacea: an emerging cause of invasive mycosis in human chronic granulomatous disease. Clin Infect Dis 2011; 52: e136–143. 2011/March/04. DOI: 10.1093/cid/ciq250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Straughan DM, McLoughlin KC, Mullinax JE, et al. The Changing Paradigm of Management of Liver Abscesses in Chronic Granulomatous Disease. Clin Infect Dis 2018; 66: 1427–1434. 2017/November/18. DOI: 10.1093/cid/cix1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Freeman AF, Marciano BE, Anderson VL, et al. Corticosteroids in the treatment of severe nocardia pneumonia in chronic granulomatous disease. Pediatr Infect Dis J 2011; 30: 806–808. 2011/March/18. DOI: 10.1097/INF.0b013e318218181d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.De Ravin SS, Naumann N, Cowen EW, et al. Chronic granulomatous disease as a risk factor for autoimmune disease. J Allergy Clin Immunol 2008; 122: 1097–1103. 2008/October/01. DOI: 10.1016/j.jaci.2008.07.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Huang C, De Ravin SS, Paul AR, et al. Genetic Risk for Inflammatory Bowel Disease Is a Determinant of Crohn’s Disease Development in Chronic Granulomatous Disease. Inflamm Bowel Dis 2016; 22: 2794–2801. 2016/November/20. DOI: 10.1097/MIB.0000000000000966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Segal BH, Leto TL, Gallin JI, et al. Genetic, biochemical, and clinical features of chronic granulomatous disease. Medicine (Baltimore) 2000; 79: 170–200. Review 2000/June/09. [DOI] [PubMed] [Google Scholar]
  • 44.Matute JD, Arias AA, Dinauer MC, et al. p40phox: the last NADPH oxidase subunit. Blood Cells Mol Dis 2005; 35: 291–302. 2005/August/17. DOI: S1079–9796(05)00105–1 [pii] 10.1016/j.bcmd.2005.06.010. [DOI] [PubMed] [Google Scholar]
  • 45.Matute JD, Arias AA, Wright NA, et al. A new genetic subgroup of chronic granulomatous disease with autosomal recessive mutations in p40 phox and selective defects in neutrophil NADPH oxidase activity. Blood 2009; 114: 3309–3315. 2009/August/21. DOI: blood-2009–07-231498 [pii] 10.1182/blood-2009-07-231498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *46.Kuhns DB, Alvord WG, Heller T, et al. Residual NADPH oxidase and survival in chronic granulomatous disease. N Engl J Med 2010; 363: 2600–2610. 2010/December/31. DOI: 10.1056/NEJMoa1007097.First paper to suggest a clinically applicable prognostic factor for outcomes in CGD.
  • 47.Marciano BE, Zerbe CS, Falcone EL, et al. X-linked carriers of chronic granulomatous disease: Illness, lyonization, and stability. J Allergy Clin Immunol 2018; 141: 365–371. 2017/May/22. DOI: 10.1016/j.jaci.2017.04.035. [DOI] [PubMed] [Google Scholar]
  • 48.Gallin JI, Alling DW, Malech HL, et al. Itraconazole to prevent fungal infections in chronic granulomatous disease. N Engl J Med 2003; 348: 2416–2422. 2003/June/13. DOI: 10.1056/NEJMoa021931348/24/2416 [pii]. [DOI] [PubMed] [Google Scholar]
  • 49.A controlled trial of interferon gamma to prevent infection in chronic granulomatous disease. The International Chronic Granulomatous Disease Cooperative Study Group. N Engl J Med 1991; 324: 509–516. 1991/February/21. DOI: 10.1056/NEJM199102213240801. [DOI] [PubMed] [Google Scholar]
  • 50.Mouy R, Seger R, Bourquin JP, et al. Interferon gamma for chronic granulomatous disease. N Engl J Med 1991; 325: 1516–1517. 1991/November/21. DOI: 10.1056/NEJM199111213252115. [DOI] [PubMed] [Google Scholar]
  • 51.Marciano BE, Wesley R, De Carlo ES, et al. Long-term interferon-gamma therapy for patients with chronic granulomatous disease. Clin Infect Dis 2004; 39: 692–699. 2004/September/10. DOI: 10.1086/422993CID32208 [pii]. [DOI] [PubMed] [Google Scholar]
  • 52.Fernandez-Boyanapalli RF, Falcone EL, Zerbe CS, et al. Impaired efferocytosis in human chronic granulomatous disease is reversed by pioglitazone treatment. J Allergy Clin Immunol 2015; 136: 1399–1401 e1393. 2015/09/21. DOI: 10.1016/j.jaci.2015.07.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Uzel G, Orange JS, Poliak N, et al. Complications of tumor necrosis factor-alpha blockade in chronic granulomatous disease-related colitis. Clin Infect Dis 2010; 51: 1429–1434. 2010/November/10. DOI: 10.1086/657308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Butte MJ, Park KT and Lewis DB. Treatment of CGD-associated Colitis with the IL-23 Blocker Ustekinumab. J Clin Immunol 2016; 36: 619–620. 2016/July/29. DOI: 10.1007/s10875-016-0318-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Martinez CA, Shah S, Shearer WT, et al. Excellent survival after sibling or unrelated donor stem cell transplantation for chronic granulomatous disease. J Allergy Clin Immunol 2012; 129: 176–183. 2011/November/15. DOI: 10.1016/j.jaci.2011.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gungor T, Teira P, Slatter M, et al. Reduced-intensity conditioning and HLA-matched haemopoietic stem-cell transplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet 2014; 383: 436–448. DOI: 10.1016/S0140-6736(13)62069-3. [DOI] [PubMed] [Google Scholar]
  • 57.Mark Parta CK, Kwatemaa Nana, Theobald Narda, Hilligoss Dianne, Qin Jing, Kuhns Douglas B., Zerbe Christa, Holland Steven M, Malech Harry, Kang Elizabeth M.. Allogeneic Reduced-Intensity Hematopoietic Stem Cell Transplatation for Chronic Granulomatous Disease: a Single-Center Prospective Trial. Journal of Clinical Immunology 2017. Original Article 28 July 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Soncini E, Slatter M, Jones L, et al. Haematopoeitic stem cell transplantation for chronic granulomatous disease - a single-centre experience. Bone Marrow Transplantation 2008; 41: S28–S28. [Google Scholar]
  • 59.Horwitz ME, Barrett AJ, Brown MR, et al. Treatment of chronic granulomatous disease with nonmyeloablative conditioning and a T-cell-depleted hematopoietic allograft. N Engl J Med 2001; 344: 881–888. 2001/March/22. DOI: 10.1056/NEJM200103223441203. [DOI] [PubMed] [Google Scholar]
  • 60.Morillo-Gutierrez B, Beier R, Rao K, et al. Treosulfan-based conditioning for allogeneic HSCT in children with chronic granulomatous disease: a multicenter experience. Blood 2016; 128: 440–448. 2016/May/25. DOI: 10.1182/blood-2016-03-704015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Parta M, Hilligoss D, Kelly C, et al. Failure to Prevent Severe Graft-Versus-Host Disease in Haploidentical Hematopoietic Cell Transplantation with Post-Transplant Cyclophosphamide in Chronic Granulomatous Disease. J Clin Immunol 2020 2020/April/22. DOI: 10.1007/s10875-020-00772-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Parta M, Hilligoss D, Kelly C, et al. Haploidentical Hematopoietic Cell Transplantation with Post-Transplant Cyclophosphamide in a Patient with Chronic Granulomatous Disease and Active Infection: A First Report. J Clin Immunol 2015; 35: 675–680. 2015/October/11. DOI: 10.1007/s10875-015-0204-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Marciano BE, Zerbe CS, Falcone EL, et al. X-linked carriers of chronic granulomatous disease: Illness, lyonization, and stability. J Allergy Clin Immunol 2017. DOI: 10.1016/j.jaci.2017.04.035. [DOI] [PubMed] [Google Scholar]
  • 64.Marsh RA, Leiding JW, Logan BR, et al. Chronic Granulomatous Disease-Associated IBD Resolves and Does Not Adversely Impact Survival Following Allogeneic HCT. J Clin Immunol 2019; 39: 653–667. 2019/August/04. DOI: 10.1007/s10875-019-00659-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Malech HL, Maples PB, Whiting-Theobald N, et al. Prolonged production of NADPH oxidase-corrected granulocytes after gene therapy of chronic granulomatous disease. Proc Natl Acad Sci U S A 1997; 94: 12133–12138. 1997/October/29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Dinauer MC, Li LL, Bjorgvinsdottir H, et al. Long-term correction of phagocyte NADPH oxidase activity by retroviral-mediated gene transfer in murine X-linked chronic granulomatous disease. Blood 1999; 94: 914–922. [PubMed] [Google Scholar]
  • 67.Ott MG, Schmidt M, Schwarzwaelder K, et al. Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat Med 2006; 12: 401–409. 2006/April/04. DOI: nm1393 [pii] 10.1038/nm1393. [DOI] [PubMed] [Google Scholar]
  • 68.Kang EM, Choi U, Theobald N, et al. Retrovirus gene therapy for X-linked chronic granulomatous disease can achieve stable long-term correction of oxidase activity in peripheral blood neutrophils. Blood 2010; 115: 783–791. 2009/December/08. DOI: blood-2009–05-222760 [pii] 10.1182/blood-2009-05-222760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **69.Kohn DB, Booth C, Kang EM, et al. Lentiviral gene therapy for X-linked chronic granulomatous disease. Nat Med 2020; 26: 200–206. 2020/January/29. DOI: 10.1038/s41591-019-0735-5.First clinical gene therapy showing long term clinically relevant levels of marking in gene therapy for CGD
  • 70.Peng J, Redman CM, Wu X, et al. Insights into extensive deletions around the XK locus associated with McLeod phenotype and characterization of two novel cases. Gene 2007; 392: 142–150. 2007/February/16. DOI: 10.1016/j.gene.2006.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gallin JI. Neutrophil specific granule deficiency. Annu Rev Med 1985; 36: 263–274. 1985/January/01. DOI: 10.1146/annurev.me.36.020185.001403. [DOI] [PubMed] [Google Scholar]
  • 72.Gombart AF, Shiohara M, Kwok SH, et al. Neutrophil-specific granule deficiency: homozygous recessive inheritance of a frameshift mutation in the gene encoding transcription factor CCAAT/enhancer binding protein--epsilon. Blood 2001; 97: 2561–2567. 2001/April/21. DOI: 10.1182/blood.v97.9.2561. [DOI] [PubMed] [Google Scholar]
  • 73.Lekstrom-Himes JA, Dorman SE, Kopar P, et al. Neutrophil-specific granule deficiency results from a novel mutation with loss of function of the transcription factor CCAAT/enhancer binding protein epsilon. J Exp Med 1999; 189: 1847–1852. 1999/June/08. DOI: 10.1084/jem.189.11.1847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Gallin JI, Fletcher MP, Seligmann BE, et al. Human neutrophil-specific granule deficiency: a model to assess the role of neutrophil-specific granules in the evolution of the inflammatory response. Blood 1982; 59: 1317–1329. 1982/June/01. [PubMed] [Google Scholar]
  • 75.McIlwaine L, Parker A, Sandilands G, et al. Neutrophil-specific granule deficiency. Br J Haematol 2013; 160: 735. 2013/January/09. DOI: 10.1111/bjh.12207. [DOI] [PubMed] [Google Scholar]
  • 76.Muraoka M, Akagi T, Ueda A, et al. C/EBPepsilon DeltaRS derived from a neutrophil-specific granule deficiency patient interacts with HDAC1 and its dysfunction is restored by trichostatin A. Biochem Biophys Res Commun 2019; 516: 293–299. 2019/July/02. DOI: 10.1016/j.bbrc.2019.06.130. [DOI] [PubMed] [Google Scholar]
  • 77.Wynn RF, Sood M, Theilgaard-Monch K, et al. Intractable diarrhoea of infancy caused by neutrophil specific granule deficiency and cured by stem cell transplantation. Gut 2006; 55: 292–293. 2006/January/13. DOI: 10.1136/gut.2005.081927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kurkchubasche AG, Panepinto JA, Tracy TF Jr., et al. Clinical features of a human Rac2 mutation: a complex neutrophil dysfunction disease. J Pediatr 2001; 139: 141–147. 2001/July/11. DOI: 10.1067/mpd.2001.114718. [DOI] [PubMed] [Google Scholar]
  • *79.Williams DA, Tao W, Yang F, et al. Dominant negative mutation of the hematopoietic-specific Rho GTPase, Rac2, is associated with a human phagocyte immunodeficiency. Blood 2000; 96: 1646–1654. 2000/August/29.The use of genetic sequencing to elucidate a rare disorder.
  • 80.Abell AN, DeCathelineau AM, Weed SA, et al. Rac2D57N, a dominant inhibitory Rac2 mutant that inhibits p38 kinase signaling and prevents surface ruffling in bone-marrow-derived macrophages. J Cell Sci 2004; 117: 243–255. 2003/December/17. DOI: 10.1242/jcs.00853. [DOI] [PubMed] [Google Scholar]
  • 81.Brechard S, Salsmann A and Tschirhart EJ. OAG induces an additional PKC-, PI3K-, and Rac2-mediated signaling pathway up-regulating NOX2 activity, independently of Ca2+ entry. J Leukoc Biol 2009; 85: 638–647. 2009/January/02. DOI: jlb.0508330 [pii] 10.1189/jlb.0508330. [DOI] [PubMed] [Google Scholar]
  • 82.Makaryan V, Zeidler C, Bolyard AA, et al. The diversity of mutations and clinical outcomes for ELANE-associated neutropenia. Curr Opin Hematol 2015; 22: 3–11. 2014/November/27. DOI: 10.1097/MOH.0000000000000105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Makaryan V, Rosenthal EA, Bolyard AA, et al. TCIRG1-associated congenital neutropenia. Hum Mutat 2014; 35: 824–827. 2014/April/23. DOI: 10.1002/humu.22563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Dale DC and Bolyard AA. An update on the diagnosis and treatment of chronic idiopathic neutropenia. Curr Opin Hematol 2017; 24: 46–53. 2016/November/15. DOI: 10.1097/MOH.0000000000000305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Fioredda F, Iacobelli S, van Biezen A, et al. Stem cell transplantation in severe congenital neutropenia: an analysis from the European Society for Blood and Marrow Transplantation. Blood 2015; 126: 1885–1892; quiz 1970. 2015/July/18. DOI: 10.1182/blood-2015-02-628859. [DOI] [PubMed] [Google Scholar]
  • 86.Dale DC, Bolyard A, Marrero T, et al. Long-Term Effects of G-CSF Therapy in Cyclic Neutropenia. N Engl J Med 2017; 377: 2290–2292. 2017/December/07. DOI: 10.1056/NEJMc1709258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Rosenberg PS, Alter BP, Link DC, et al. Neutrophil elastase mutations and risk of leukaemia in severe congenital neutropenia. Br J Haematol 2008; 140: 210–213. 2007/November/22. DOI: 10.1111/j.1365-2141.2007.06897.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Rosenberg PS, Zeidler C, Bolyard AA, et al. Stable long-term risk of leukaemia in patients with severe congenital neutropenia maintained on G-CSF therapy. Br J Haematol 2010; 150: 196–199. 2010/May/12. DOI: 10.1111/j.1365-2141.2010.08216.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Farruggia P and Dufour C. Diagnosis and management of primary autoimmune neutropenia in children: insights for clinicians. Ther Adv Hematol 2015; 6: 15–24. 2015/February/03. DOI: 10.1177/2040620714556642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Fioredda F, Calvillo M, Burlando O, et al. Infectious complications in children with severe congenital, autoimmune or idiopathic neutropenia: a retrospective study from the Italian Neutropenia Registry. Pediatr Infect Dis J 2013; 32: 410–412. 2012/December/20. DOI: 10.1097/INF.0b013e3182814b5a. [DOI] [PubMed] [Google Scholar]
  • 91.Spielberg SP, Boxer LA, Oliver JM, et al. Oxidative damage to neutrophils in glutathione synthetase deficiency. Br J Haematol 1979; 42: 215–223. 1979/June/01. DOI: 10.1111/j.13652141.1979.tb01126.x. [DOI] [PubMed] [Google Scholar]
  • 92.Candotti F. Clinical Manifestations and Pathophysiological Mechanisms of the Wiskott-Aldrich Syndrome. J Clin Immunol 2018; 38: 13–27. 2017/November/01. DOI: 10.1007/s10875-0170453-z. [DOI] [PubMed] [Google Scholar]
  • 93.Albert MH, Bittner TC, Nonoyama S, et al. X-linked thrombocytopenia (XLT) due to WAS mutations: clinical characteristics, long-term outcome, and treatment options. Blood 2010; 115: 3231–3238. 2010/February/23. DOI: 10.1182/blood-2009-09-239087. [DOI] [PubMed] [Google Scholar]
  • 94.Zicha D, Allen WE, Brickell PM, et al. Chemotaxis of macrophages is abolished in the Wiskott-Aldrich syndrome. Br J Haematol 1998; 101: 659–665. 1998/July/23. DOI: 10.1046/j.1365-2141.1998.00767.x. [DOI] [PubMed] [Google Scholar]
  • 95.Badolato R, Sozzani S, Malacarne F, et al. Monocytes from Wiskott-Aldrich patients display reduced chemotaxis and lack of cell polarization in response to monocyte chemoattractant protein-1 and formyl-methionyl-leucyl-phenylalanine. J Immunol 1998; 161: 1026–1033. 1998/July/22. [PubMed] [Google Scholar]
  • 96.Zhang H, Schaff UY, Green CE, et al. Impaired integrin-dependent function in Wiskott-Aldrich syndrome protein-deficient murine and human neutrophils. Immunity 2006; 25: 285295. 2006/August/12. DOI: 10.1016/j.immuni.2006.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Lorenzi R, Brickell PM, Katz DR, et al. Wiskott-Aldrich syndrome protein is necessary for efficient IgG-mediated phagocytosis. Blood 2000; 95: 2943–2946. 2000/April/26. [PubMed] [Google Scholar]
  • 98.Westerberg LS, Meelu P, Baptista M, et al. Activating WASP mutations associated with X-linked neutropenia result in enhanced actin polymerization, altered cytoskeletal responses, and genomic instability in lymphocytes. J Exp Med 2010; 207: 1145–1152. 2010/June/02. DOI: 10.1084/jem.20091245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Thrasher AJ and Burns SO. WASP: a key immunological multitasker. Nat Rev Immunol 2010; 10: 182–192. 2010/February/26. DOI: 10.1038/nri2724. [DOI] [PubMed] [Google Scholar]
  • 100.Ozsahin H, Cavazzana-Calvo M, Notarangelo LD, et al. Long-term outcome following hematopoietic stem-cell transplantation in Wiskott-Aldrich syndrome: collaborative study of the European Society for Immunodeficiencies and European Group for Blood and Marrow Transplantation. Blood 2008; 111: 439–445. Multicenter Study 2007/September/29. DOI: 10.1182/blood-2007-03-076679. [DOI] [PubMed] [Google Scholar]
  • 101.Glasmacher Jannik S TCB, MD, Ochs Hans D, MD, Aiuti Alessandro, MD PhD, Arkwright Peter D, MD, Balashov Dmitry, Behrends Uta, Belohradsky Bernd H., MD, Bertoni Elisa, Buchbinder David K., MD MSHS, Browning Michael, Bondarenko Anastasiia, Candotti Fabio, MD, Cattoni Alessandro, Chernyshova Liudmyla, Chewning Joseph H., MD, Ciznar Peter, Cole Theresa, Costa-Carvalho Beatriz T, Czogala Wojciech, Dueckers Gregor, Edgar David M, Erbey Fatih, Fasth Anders, MD PhD, Formankova Renata, MD PhD, Freiberger Tomas, Gambineri Eleonora, Gennery Andrew, MD, Goldman Frederick D, MD, Gonzalez-Granado Luis I, Gulmaraes Tiago N, Hagin David, MD PhD, Hauck Fabian, MD PhD, Heiskanen Tarja, Hoenig Manfred, MD, Juntti Hanna, Kanegane Hirokazu, Kainulainen Leena, Karaca Neslihan E, Kilic Sara S, Klein Christoph, MD PhD, Koltan Sylwia, Kondratenko Irina, Liu Dawei, MD, Matthes Susanne, MD, Mazzucchelli Juliana T L, Meyts Isabelle, Misbah Siraj, Nademi Zohreh, Nasrullayeva Gulnara, Notarangelo Lucia D, Soler-Palacin Pere, Pashchenko Olga, Pasic Srdjan, Pellier Isabelle, MD, Pignata Claudio, Roepstorff Camilla, Schuetz Catharina, Schulz Ansgar S, MD PhD Prof., Segundo Gesmar R S, Shcherbina Anna, Smart Joanne, Sokolic Robert A., MD, Stepensky Polina, MD, Torgerson Troy, MD PhD, Vakhlyarskaya Svetlana, van Montfrans Joris, MD PhD, Vettenranta Kim, MD PD, Wolska-Kusnierz Beata, Zhao Xiaodong, Ziegler John B, Zhang Xuan, Albert Michael H., MD. Wiskott-Aldrich Syndrome: A Retrospective Study on 575 Patients Analyzing the Impact of Splenectomy, Stem Cell Transplantation, or No Definitive Treatment on Frequency of Disease-Related Complications and Physician-Perceived Quality of Life. American Society of Hematology. American Society of Hematology, 2016, p. 366. [Google Scholar]
  • 102.Kharya G, Nademi Z, Leahy TR, et al. Haploidentical T-cell alpha beta receptor and CD19-depleted stem cell transplant for Wiskott-Aldrich syndrome. J Allergy Clin Immunol 2014; 134: 1199–1201. 2014/July/06. DOI: 10.1016/j.jaci.2014.04.041. [DOI] [PubMed] [Google Scholar]
  • 103.Shin CR, Kim MO, Li D, et al. Outcomes following hematopoietic cell transplantation for Wiskott-Aldrich syndrome. Bone Marrow Transplant 2012; 47: 1428–1435. 2012/March/20. DOI: 10.1038/bmt.2012.31. [DOI] [PubMed] [Google Scholar]
  • 104.Shekhovtsova Z, Bonfim C, Ruggeri A, et al. A risk factor analysis of outcomes after unrelated cord blood transplantation for children with Wiskott-Aldrich syndrome. Haematologica 2017; 102: 1112–1119. 2017/March/04. DOI: 10.3324/haematol.2016.158808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Pai SY and Notarangelo LD. Hematopoietic cell transplantation for Wiskott-Aldrich syndrome: advances in biology and future directions for treatment. Immunol Allergy Clin North Am 2010; 30: 179–194. 2010/May/25. DOI: 10.1016/j.iac.2010.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ferrua F, Cicalese MP, Galimberti S, et al. Lentiviral haemopoietic stem/progenitor cell gene therapy for treatment of Wiskott-Aldrich syndrome: interim results of a non-randomised, open-label, phase 1/2 clinical study. Lancet Haematol 2019; 6: e239–e253. 2019/April/15. DOI: 10.1016/S2352-3026(19)30021-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sereni L, Castiello MC, Di Silvestre D, et al. Lentiviral gene therapy corrects platelet phenotype and function in patients with Wiskott-Aldrich syndrome. J Allergy Clin Immunol 2019; 144: 825–838. 2019/March/31. DOI: 10.1016/j.jaci.2019.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Boztug K, Schmidt M, Schwarzer A, et al. Stem-cell gene therapy for the Wiskott-Aldrich syndrome. N Engl J Med 2010; 363: 1918–1927. Clinical Trial Research Support, Non-U.S. Gov’t 2010/November/12. DOI: 10.1056/NEJMoa1003548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *109.Braun CJ, Boztug K, Paruzynski A, et al. Gene therapy for Wiskott-Aldrich syndrome--long-term efficacy and genotoxicity. Sci Transl Med 2014; 6: 227ra233. 2014/March/14. DOI: 10.1126/scitranslmed.3007280.Demonstration of insertational mutagenesis in a retroviral gene therapy study for WAS.
  • 110.Aiuti A, Biasco L, Scaramuzza S, et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science 2013; 341: 1233151. Clinical Trial, Phase I Clinical Trial, Phase II Research Support, Non-U.S. Gov’t 2013/July/13. DOI: 10.1126/science.1233151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Haddad E, Le Deist F, Blanche S, et al. Treatment of Chediak-Higashi syndrome by allogenic bone marrow transplantation: report of 10 cases. Blood 1995; 85: 3328–3333. 1995/June/01. [PubMed] [Google Scholar]
  • 112.Eapen M, DeLaat CA, Baker KS, et al. Hematopoietic cell transplantation for Chediak-Higashi syndrome. Bone Marrow Transplant 2007; 39: 411–415. 2007/February/13. DOI: 10.1038/sj.bmt.1705600. [DOI] [PubMed] [Google Scholar]
  • 113.Notarangelo LD, Peitsch MC, Abrahamsen TG, et al. CD40lbase: a database of CD40L gene mutations causing X-linked hyper-IgM syndrome. Immunol Today 1996; 17: 511–516. 1996/November/01. DOI: 10.1016/0167-5699(96)30059-5. [DOI] [PubMed] [Google Scholar]
  • 114.Hayward AR, Levy J, Facchetti F, et al. Cholangiopathy and tumors of the pancreas, liver, and biliary tree in boys with X-linked immunodeficiency with hyper-IgM. J Immunol 1997; 158: 977–983. 1997/January/15. [PubMed] [Google Scholar]
  • 115.Thomas C, de Saint Basile G, Le Deist F, et al. Brief report: correction of X-linked hyper-IgM syndrome by allogeneic bone marrow transplantation. N Engl J Med 1995; 333: 426–429. 1995/August/17. DOI: 10.1056/NEJM199508173330705. [DOI] [PubMed] [Google Scholar]
  • 116.Gennery AR, Khawaja K, Veys P, et al. Treatment of CD40 ligand deficiency by hematopoietic stem cell transplantation: a survey of the European experience, 1993–2002. Blood 2004; 103: 1152–1157. DOI: 10.1182/blood-2003-06-2014. [DOI] [PubMed] [Google Scholar]
  • 117.Ferrari S, Giliani S, Insalaco A, et al. Mutations of CD40 gene cause an autosomal recessive form of immunodeficiency with hyper IgM. Proc Natl Acad Sci U S A 2001; 98: 12614–12619. 2001/October/25. DOI: 10.1073/pnas.221456898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kutukculer N, Aksoylar S, Kansoy S, et al. Outcome of hematopoietic stem cell transplantation in hyper-IgM syndrome caused by CD40 deficiency. J Pediatr 2003; 143: 141–142. 2003/August/14. DOI: 10.1016/S0022-3476(03)00274-9. [DOI] [PubMed] [Google Scholar]
  • 119.Seyama K, Osborne WR and Ochs HD. CD40 ligand mutants responsible for X-linked hyper-IgM syndrome associate with wild type CD40 ligand. J Biol Chem 1999; 274: 11310–11320. 1999/April/10. DOI: 10.1074/jbc.274.16.11310. [DOI] [PubMed] [Google Scholar]
  • *120.Hubbard N, Hagin D, Sommer K, et al. Targeted gene editing restores regulated CD40L function in X-linked hyper-IgM syndrome. Blood 2016; 127: 2513–2522. 2016/February/24. DOI: 10.1182/blood-2015-11-683235.One of the first demonstrations of the use of gene editing in a myeloid immunodeficiency

RESOURCES