Primary Combined Antibody and Cellular Immunodeficiencies

Published on 27/03/2015 by admin

Filed under Pediatrics

Last modified 27/03/2015

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 1174 times

Chapter 120 Primary Combined Antibody and Cellular Immunodeficiencies

Patients with combined antibody and cellular defects have severe, frequently opportunistic infections that lead to death in infancy or childhood unless they are provided hematopoietic stem cell transplantation early in life. These are thought to be rare defects, although the true incidences are unknown because there has been no newborn screening for any of these defects. It is possible that many affected children die of infection during infancy without being diagnosed. The defective gene products for many combined immunodeficiencies are identified (Table 120-1). Because life threatening infection may occur in infancy, screening for SCID has been recommended by the U.S. Secretary of Health and Human Services to be included in the state newborn screening programs. Live, vaccine-derived infections have occurred during this time of life and knowledge of SCID status could prevent these infections. In addition, early identification and subsequent bone marrow transplantation before life-threatening infections and end organ injury is the best approach to therapy.

120.1 Severe Combined Immunodeficiency (SCID)

Rebecca H. Buckley

The syndromes of SCID are caused by diverse genetic mutations that lead to absence of all adaptive immune function and, in some, a lack of natural killer (NK) cells. Patients with this group of disorders have the most severe immunodeficiency.

Pathogenesis

SCID results from mutations in any 1 of 13 known genes that encode components of the immune system crucial for lymphoid cell development (Table 120-2). All patients with SCID have very small thymuses (<1 g) that usually fail to descend from the neck, contain no thymocytes, and lack corticomedullary distinction or Hassall corpuscles. The thymic epithelium appears histologically normal. Both the follicular and paracortical areas of the spleen are depleted of lymphocytes. Lymph nodes, tonsils, adenoids, and Peyer patches are absent or extremely underdeveloped.

Table 120-2 PATHOPHYSIOLOGY MECHANISMS THAT ACCOUNT FOR SEVERE COMBINED IMMUNE DEFICIENCY (SCID)

DISEASE MECHANISM GENE DEFECTS
Increased apoptosis
• Due to mitochondrial energy failure AK2
• Due to accumulation of toxic metabolites ADA
• Due to abnormal actin polymerization CORO1A
Impaired cytokine-mediated signaling
• Due to defects of the common γ chain IG2RG (X-linked SCID)
• Due to defects of the IL-7R α chain IL7R
• Due to defects of Jak3 JAK3
Impaired signaling through the pre–T cell receptor
• Due to defective V(D)J recombination RAG1, RAG2, DCLRE1C, LIG4,* PRKDC
• Due to impaired expression of CD3 subunits CD3D, CD3E, CD3Z
Impaired signaling in the periphery ORA1
Unknown mechanism RMRP*

* These gene defects are most often associated with a milder clinical phenotype than SCID.

From Pessach I, Walter J, Notarangelo LD: Recent advances in primary immunodeficiencies: identification of novel genetic defects and unanticipated phenotypes, Pediatr Res 65:3R–12R, 2009.

Clinical Manifestations

Affected infants present within the 1st few months of life with recurrent or persistent diarrhea, pneumonia, otitis media, sepsis, and cutaneous infections. Growth may appear normal initially, but extreme wasting usually ensues after diarrhea and infections begin. Persistent infections with opportunistic organisms including Candida albicans, Pneumocystis jiroveci, parainfluenza 3 virus, adenovirus, respiratory syncytial virus, rotavirus vaccine virus, cytomegalovirus (CMV), Epstein-Barr virus (EBV), varicella-zoster virus, measles virus, MMR-V vaccine virus, or bacillus Calmette-Guérin (BCG) lead to death. Affected infants also lack the ability to reject foreign tissue and are therefore at risk for severe or fatal graft versus host disease (GVHD) from T lymphocytes in nonirradiated blood products or in allogeneic stem cell transplants or less severe GVHD from maternal immunocompetent T cells that crossed the placenta while the infant was in utero.

Because all molecular types of SCID lack T cells, infants with SCID have lymphopenia (<2,500/mm3) that is present at birth, indicating that the condition could be diagnosed in all affected infants if white blood cell counts with manual differential counts were routinely performed on all cord bloods and the absolute lymphocyte count calculated. These infants also have an absence of lymphocyte proliferative responses to mitogens, antigens, and allogeneic cells in vitro. Patients with adenosine deaminase (ADA) deficiency have the lowest absolute lymphocyte counts, usually <500/mm3. Serum immunoglobulin concentrations are low or absent, and no antibodies are formed after immunizations. Analyses of lymphocyte populations and subpopulations demonstrate distinctive phenotypes for the various genetic forms of SCID (see Table 120-2). T cells are extremely low or absent in all types; when detected, in most cases they are transplacentally derived maternal T cells.

X-Linked Severe Combined Immunodeficiency (SCIDX1) Due To Mutations in the Gene Encoding the Common Cytokine Receptor γ Chain (γC)

X-linked SCID (X-SCD) is the most common form of SCID in the USA, accounting for 47% of cases (Fig. 120-1). Clinically, immunologically, and histopathologically, affected individuals appear similar to those with other forms of SCID except for having uniformly low percentages of T and NK cells and an elevated percentage of B cells (T−, B+, NK−), a characteristic feature shared only with Janus kinase 3 (Jak3)–deficient SCID. The abnormal gene in X-SCD was mapped to Xq13, cloned, and found to encode the common γ chain (γc) for several cytokine receptors, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. The shared γc functions both to increase the affinity of the receptor for the respective cytokine and to enable the receptors to mediate intracellular signaling. Incapacitation of the receptors for all of these developmentally crucial cytokines by genetic mutations in γc provides an explanation for the severity of the immunodeficiency in SCIDX1. In the 1st 136 patients studied, 95 distinct mutations spanning all 8 IL2RG exons were identified, most of them consisting of small changes at the level of 1 to a few nucleotides. These mutations resulted in abnormal γc chains in two thirds of the cases and absent γc protein in the remainder. Carriers can be detected by demonstrating nonrandom X-chromosome inactivation or the deleterious mutation in their T, B, or NK lymphocytes. Unless donor B or NK cells develop, patients with X-SCID lack B- and NK-cell function after bone marrow transplantation because the abnormal γc persists in those host cells, despite excellent reconstitution of T-cell function by donor-derived T cells.

Autosomal Recessive Severe Combined Immunodeficiency

This pattern of inheritance of SCID is less common in the USA than in Europe. Mutated genes on autosomal chromosomes have been identified in 12 forms of SCID: ADA deficiency; Jak3 deficiency; IL-7 receptor α chain (IL-7Rα) deficiency; RAG1 or RAG2 deficiency; Artemis deficiency; ligase 4 deficiency; DNA–protein kinase catalytic subunit (DNA-PKcs) deficiency; CD3δ, CD3ε, CD3ζ deficiency; and CD45 deficiency (see Fig. 120-1).

ADA Deficiency

An absence of the enzyme adenosine deaminase (ADA) is observed in approximately 15% of patients, the second most common form of SCID, resulting from various point and deletional mutations in the ADA gene on chromosome 20q13-ter. Marked accumulations of adenosine, 2′-deoxyadenosine, and 2′-O-methyladenosine lead directly or indirectly to T-cell apoptosis, which causes the immunodeficiency. ADA-deficient patients usually have a much more profound lymphopenia than do infants with other types of SCID, with mean absolute lymphocyte counts of <500/mm3; the absolute numbers of T, B, and NK cells are very low. NK function is normal. After T-cell function is conferred by hematopoietic stem cell transplantation without pretransplant chemotherapy, there is generally excellent B-cell function despite the fact that the B cells are of host origin. This is because ADA deficiency affects primarily T-cell function. Milder forms of ADA deficiency have led to delayed diagnosis of immunodeficiency, even to adulthood. Other distinguishing features of ADA-deficient SCID include the presence of rib cage abnormalities similar to a rachitic rosary and numerous skeletal abnormalities of chondro-osseous dysplasia, which occur predominantly at the costochondral junctions, at the apophyses of the iliac bones, and in the vertebral bodies where a “bone-in-bone” effect is observed.

As with other types of SCID, ADA deficiency can be cured by HLA-identical or haploidentical T-cell–depleted stem cell transplantation without the need for pre- or post-transplant chemotherapy; this remains the treatment of choice. Enzyme replacement therapy should not be initiated if stem cell transplantation is possible because it confers graft-rejection capability. Enzyme replacement provides protective immunity but over time there is a decline of lymphocyte counts and mitogenic proliferative responses. Fifteen infants with ADA deficiency have become immune reconstituted by gene therapy; in all cases, PEG-ADA was withheld. Spontaneous reversion to normal of a mutation in the ADA gene has also been reported.

IL-7Rα Deficiency

Patients with IL-7Rα-deficient SCID have a distinctive lymphocyte phenotype in that, though lacking T cells, they have normal or elevated numbers of both B and NK cells (T−, B+, NK+). This is the third most common form of SCID, accounting for 12% of cases in the USA (see Fig. 120-1). In contrast to patients with γc– and Jak3-deficient SCID, the immunologic defect in these patients is completely correctable by bone marrow stem cell transplantation, as the host B and NK cells appear to be normal.