Fact-checked by Grok 2 weeks ago

Chronic granulomatous disease

Chronic granulomatous disease (CGD) is a rare inherited disorder in which , a type of , fail to effectively kill certain and fungi due to defects in the enzyme complex, resulting in recurrent severe infections and the formation of granulomas—clusters of immune cells that can cause tissue damage and organ dysfunction. This condition typically manifests in , with an incidence of approximately 1 in 200,000 to 250,000 live births worldwide, and affects the by impairing the production of necessary for microbial destruction. Without treatment, CGD can lead to life-threatening complications, including , abscesses, and chronic inflammatory conditions. CGD arises from mutations in one of six genes that encode the subunits of the NADPH oxidase complex, with the most common form being X-linked recessive due to mutations in the CYBB gene on the , accounting for about 70% of cases and predominantly affecting males. The remaining cases are autosomal recessive, involving genes such as NCF1, NCF2, or CYBA, and can affect both sexes equally. These genetic defects prevent like neutrophils, monocytes, and macrophages from generating superoxide radicals, leaving patients susceptible to catalase-positive pathogens such as , , and species. The disease's not only promotes infections but also triggers excessive , leading to formation in organs like the lungs, liver, , and urinary system. Clinically, individuals with CGD often present with repeated bacterial or fungal infections starting in infancy or early childhood, including , skin abscesses, , and gastrointestinal issues resembling , along with symptoms such as fever, swollen lymph nodes, and organ-specific pain. Granulomas may obstruct airways, bowels, or urinary tracts, potentially causing additional complications like , , or autoimmune disorders. Diagnosis is confirmed through functional assays of oxidative burst, such as the dihydrorhodamine (DHR) test or nitroblue tetrazolium (NBT) reduction , supplemented by genetic sequencing to identify specific mutations. Management of CGD focuses on preventing infections and controlling inflammation through lifelong prophylactic antibiotics like trimethoprim-sulfamethoxazole and antifungals such as , often combined with subcutaneous interferon-gamma injections to enhance immune function. For severe or refractory cases, (HSCT) offers a potential cure by replacing defective immune cells, with success rates exceeding 90% in matched sibling donor transplants. Emerging therapies, including targeting the CYBB mutation, are under investigation, showing promise in ongoing clinical trials as of 2025. Early and multidisciplinary care are essential to improve outcomes and for affected individuals.

Clinical Presentation

Symptoms and Signs

Chronic granulomatous disease (CGD) is characterized by recurrent and severe bacterial and fungal infections, which typically begin in infancy or early childhood, often within the first year of life and with a median age of onset around 1 to 3 years. These infections are the hallmark of the disorder and can affect multiple organ systems, leading to significant morbidity if untreated. The most common sites of infection include the lungs, where frequently occurs, often caused by pathogens such as Aspergillus species or Burkholderia cepacia, presenting with symptoms like persistent , , fever, and . Skin infections manifest as abscesses, boils, or ulcers, commonly due to , while lymphadenitis involves swollen and painful lymph nodes, particularly in the cervical or axillary regions. Liver abscesses, (bone infections), and perirectal abscesses are also prevalent, with the latter often leading to painful swelling and drainage in the anal area. Gastrointestinal involvement may present with , , or bloody stools from resembling , frequently triggered by enteric pathogens. A distinctive feature of CGD is susceptibility to infections by catalase-positive organisms, such as Nocardia species, Serratia marcescens, and Aspergillus, which evade phagocyte killing due to the absence of reactive oxygen species production. These atypical infections can disseminate hematogenously, resulting in complications like brain abscesses or widespread sepsis, and are more aggressive than typical community-acquired infections. For instance, nocardiosis may initially appear as pulmonary nodules mimicking pneumonia, progressing to skin lesions or central nervous system involvement if unchecked. Non-infectious signs in CGD include persistent fever, fatigue, and , often accompanying inflammatory responses to unresolved . Swollen lymph nodes without overt and can cause obstructive symptoms, such as urinary tract blockage or gastrointestinal strictures, leading to colicky pain or altered bowel habits. These manifestations underscore the chronic inflammatory state in CGD, where formation contributes to tissue damage beyond direct microbial invasion.

Complications and Associated Conditions

Chronic granulomatous disease (CGD) is associated with significant noninfectious complications arising from dysregulated inflammatory responses, particularly granulomatous inflammation that can lead to tissue damage in multiple organs. In the gastrointestinal (GI) tract, obstructive granulomas commonly form in the , , , cecum, rectum, and perirectal areas, mimicking and resulting in strictures, fistulas, and that cause . These granulomatous lesions contribute to chronic morbidity, with GI involvement reported in 32.8% to 46% of patients across large cohorts. In the lungs, granulomas can cause obstructive lesions and interstitial changes, exacerbating respiratory compromise. A prominent auto-inflammatory condition in CGD is CGD-associated (CGD-IBD), which affects up to 50% to 61% of patients and presents with symptoms such as , (including bloody stools), , , fever, and . CGD-IBD is particularly severe in X-linked forms, often leading to , nutritional deficits, and a higher risk of misdiagnosis as conventional ; fistulas and fissures are common, with median onset around age 5 years. Organ-specific complications further compound the disease burden. Pulmonary fibrosis develops in a subset of patients, particularly those with X-linked CGD, often linked to repeated inflammatory insults and affecting up to 34% with non-infectious lung issues like . Liver dysfunction arises from chronic abscesses and granulomas, occurring in up to 35% of cases, leading to portal venopathy, nodular regenerative hyperplasia, , and . Growth delays and are frequent in children, primarily due to severe GI complications causing and obstruction. Patients with CGD face an elevated, though rare, risk of malignancy attributable to chronic inflammation, including gastric cancer and , with reports suggesting a potentially higher incidence in autosomal recessive forms despite overall low rates in large series. Other associations include autoimmune phenomena, such as (affecting approximately 2.7% of patients, characterized by photosensitive rashes and oral ulcers) and (often linked to or as idiopathic thrombocytopenic ). These conditions highlight the broad spectrum of inflammatory dysregulation in CGD beyond infectious risks.

Genetics and Inheritance

Genetic Mutations

Chronic granulomatous disease (CGD) results from pathogenic variants in one of six genes that encode subunits of the phagocyte NADPH oxidase complex, which is essential for generating reactive oxygen species (ROS) to combat microbial infections. The most frequently affected gene is CYBB, located on the X chromosome and encoding the gp91phox subunit (also known as NOX2), accounting for approximately 65-70% of all CGD cases. This X-linked form predominates due to the severity of mutations disrupting the catalytic core of the oxidase. The autosomal recessive forms involve NCF1 (encoding p47phox, about 25% of cases), NCF2 (encoding p67phox, around 5%), CYBA (encoding p22phox, approximately 5%), NCF4 (encoding p40phox, rare with fewer than 1% of cases), and CYBC1 (encoding cytochrome b-245 chaperone 1, also known as EROS, exceptionally rare with <1% of cases). Mutations across these genes are diverse, including missense, , frameshift, splice-site alterations, and large deletions or insertions that impair subunit expression, stability, or assembly. In CYBB, and frameshift mutations often lead to absent protein and complete loss of activity, resulting in severe phenotypes, while hypomorphic missense variants may allow partial function and milder manifestations. For NCF1, a common founder is a 2-base pair (GT) deletion in 2, which disrupts the and is prevalent in certain populations, often correlating with less severe compared to CYBB defects. NCF2 and CYBA typically involve point changes or small indels that prevent proper translocation of cytosolic components to the , and NCF4 and CYBC1 variants, such as homozygous missense changes, similarly abolish but are exceptionally rare. These genetic alterations functionally compromise by preventing the proper assembly of its flavocytochrome b558 (membrane-bound) and cytosolic (p47phox, p67phox, p40phox) subunits upon activation, thereby abolishing or severely reducing production and downstream ROS generation. In CYBB-deficient cases, the absence of the catalytic subunit halts from NADPH to oxygen, leading to no detectable ROS; analogous disruptions in autosomal genes yield similar outcomes, though residual activity (1-10% of normal) can occur with certain hypomorphic alleles, influencing disease severity. Carrier detection is particularly relevant for X-linked CYBB mutations, where heterozygous females exhibit random X-chromosome inactivation (Lyonization), resulting in populations of normal and defective neutrophils that can be quantified via showing intermediate ROS production levels. For autosomal recessive forms, carriers are typically but identifiable through targeted sequencing. via next-generation sequencing of a multigene panel encompassing CYBB, NCF1, NCF2, CYBA, NCF4, and CYBC1 confirms the in over 95% of cases by detecting pathogenic variants, with deletion/duplication analysis required for genes like NCF1 where copy number variations are common.

Inheritance Patterns

Chronic granulomatous disease (CGD) is primarily inherited in an X-linked recessive manner, accounting for approximately 70% of cases due to pathogenic variants in the CYBB gene on the . This pattern predominantly affects males, as they inherit a single from their mother; carrier mothers have a 50% chance of transmitting the variant to each son, who would then be affected, while daughters have a 50% chance of becoming carriers but are typically unaffected. Affected males transmit the variant to all of their daughters, who become obligate carriers, but no male-to-male transmission occurs. The remaining 30% of CGD cases follow an autosomal recessive inheritance pattern, resulting from biallelic pathogenic variants in genes such as NCF1, NCF2, CYBA, NCF4, or CYBC1. This form affects males and females equally, with each of two parents carrying a 25% risk of an affected child, a 50% risk of an unaffected , and a 25% risk of an unaffected non-carrier. in parents increases the likelihood of autosomal recessive CGD. De novo mutations, which arise spontaneously without prior family history, are rare and account for less than 5% of all CGD cases, though they are more commonly observed in sporadic presentations of the X-linked form. A notable proportion of CGD cases have a positive family history of affected relatives, underscoring the importance of genetic counseling for at-risk families to assess carrier status and reproductive risks. Genetic counseling is recommended for all identified carriers to discuss inheritance risks, prenatal testing options, and family planning. Due to the predominance of the X-linked form, males are affected 5 to 10 times more frequently than females overall.

Pathophysiology

Defect in Phagocyte Function

Chronic granulomatous disease (CGD) arises from a primary defect in the complex, a multi-subunit essential for the respiratory burst in . This complex consists of membrane-bound components gp91phox (also known as ) and p22phox, encoded by the CYBB and CYBA genes, respectively, along with cytosolic subunits p47phox, p67phox, p40phox, and the chaperone EROS encoded by CYBC1, as well as p47phox, p67phox, and p40phox, encoded by NCF1, NCF2, NCF4, and CYBC1. Additionally, the Rac2 facilitates activation. Upon pathogen recognition, the cytosolic subunits translocate to the phagosomal membrane, where they assemble with the membrane-bound flavocytochrome b558 (gp91phox/p22phox) to form the active oxidase, transferring electrons from NADPH to oxygen to initiate the oxidative burst. Mutations in any of these genes disrupt assembly or function, resulting in a profound inability to generate anion (O2) and subsequent downstream (ROS), including (H2O2) and (HOCl). These ROS are critical for direct microbial killing within the , as they damage bacterial proteins, DNA, and . Without functional oxidase, cannot effectively eliminate ingested pathogens, leading to recurrent infections. The defect primarily impairs neutrophils, monocytes, and eosinophils, which rely heavily on NADPH oxidase for ROS production during phagocytosis. Macrophages exhibit less severe dysfunction but still show reduced oxidative capacity, contributing to overall immune compromise. This selective impairment explains the vulnerability to specific microbes: catalase-negative bacteria, such as certain streptococci, can be partially controlled via accumulation of their own H2O2 within the phagosome. In contrast, catalase-positive organisms like Staphylococcus aureus and Aspergillus species degrade peroxides using their own catalase enzyme, evading ROS-dependent killing mechanisms. Severity correlates with residual NADPH oxidase activity: X-linked forms (most commonly CYBB mutations) typically exhibit less than 5% activity, resulting in classic severe disease, while milder autosomal recessive variants, such as those in NCF1 (p47phox), often exhibit no detectable or minimal residual function (typically <10%), contributing to a relatively attenuated compared to X-linked forms.

Granuloma Formation and Inflammation

In chronic granulomatous disease (CGD), the failure of to produce (ROS) impairs microbial clearance, leading to persistent antigenic stimulation and hyperinflammation. This triggers prolonged release of proinflammatory cytokines such as TNF-α, IL-1, and IL-6 from activated and neutrophils, which recruit additional immune cells and perpetuate tissue damage. The dysregulated response results in excessive activation and fusion into multinucleated giant cells, independent of ongoing in many cases. Granulomas in CGD are characteristically non-caseating collections of epithelioid histiocytes, surrounded by lymphocytes and fibroblasts, often forming in fibrotic tissues of hollow organs. These structures commonly obstruct the , intestines, and urinary tract, causing symptoms like , , and . Unlike infectious granulomas, many in CGD are sterile, arising from intrinsic immune dysregulation rather than viable pathogens. The auto-inflammatory component of CGD manifests as macrophage hyperactivity, driven by defective autophagy and efferocytosis, which sustains without external triggers and contributes to conditions like CGD-associated (CGD-IBD). This leads to sterile pyogranulomatous reactions in affected tissues. In CGD patients evaluated by , granulomas can lead to significant morbidity, including colonic strictures observed in approximately 24% of such cases, often requiring surgical . Histologically, CGD granulomas resemble those in due to their non-caseating nature, but they stem specifically from deficiency rather than idiopathic T-cell dysregulation.

Diagnosis

Clinical Evaluation

Clinical evaluation of chronic granulomatous disease (CGD) begins with a thorough and to identify patterns suggestive of this , particularly in patients presenting with recurrent or severe infections. The process aims to raise suspicion for CGD based on clinical features before proceeding to confirmatory testing, focusing on the characteristic of infections and associated findings. During history taking, clinicians assess for recurrent infections, defined as at least three infections in a lifetime or at least two serious infections such as or abscesses in sites like the lungs, lymph nodes, liver, bones, or . A family history of early deaths, similar illnesses, or known immunodeficiencies is crucial, especially for X-linked forms affecting males, while raises suspicion for autosomal recessive variants. The age of onset is typically before 5 years, with a median around 2.5–3 years, though autosomal recessive cases may present later in childhood or even adulthood due to milder phenotypes. On , hepatosplenomegaly is a common finding, often resulting from chronic infections or inflammatory responses in the liver and spleen. , particularly suppurative, , skin lesions such as abscesses or , and signs of respiratory distress from pulmonary involvement are frequently observed. Red flags include infections with opportunistic pathogens like or , which are unusual in otherwise healthy individuals, and a poor response to standard antibiotic therapy despite appropriate initial management. These features highlight the need for urgent evaluation in patients with unexplained persistent or recurrent infections. Differential diagnosis encompasses other immunodeficiencies, such as leukocyte adhesion deficiency (LAD) or hyper-IgE syndrome, but CGD is distinguished by its unique pattern of formation leading to obstructive complications in sites like the or .

Laboratory Tests

The diagnosis of chronic granulomatous disease (CGD) relies on laboratory tests that confirm defective (ROS) production by and identify underlying genetic mutations. Functional assays are the cornerstone for initial confirmation, while provides definitive subtype classification. Supportive laboratory findings help assess and infection but are nonspecific. Functional assays evaluate activity in neutrophils and monocytes. The dihydrorhodamine 123 (DHR) test is the gold standard, measuring ROS through intensity after stimulation with phorbol myristate ; it detects absent or reduced ROS in over 90% of CGD cases, including complete, partial (hypomorphic), and forms, with high sensitivity for all inheritance patterns except rare NCF4 variants. The nitroblue tetrazolium (NBT) slide test, an older qualitative method, assesses generation by observing precipitate formation under ; normal neutrophils reduce NBT in >95% of cells, but CGD patients show failure or markedly reduced reduction (<10%), making it useful for rapid screening though less sensitive and specific than DHR. Genetic testing involves targeted sequencing and deletion/duplication analysis of the six genes encoding NADPH oxidase components—CYBB (X-linked, ~65% of cases), NCF1 (autosomal recessive, ~20%), NCF2, NCF4, CYBA, and CYBC1—identifying pathogenic variants in approximately 95% of patients when combined methods are used. This testing not only confirms the diagnosis but also distinguishes X-linked from autosomal recessive forms and enables prenatal diagnosis via amniocentesis or chorionic villus sampling, as well as carrier testing for at-risk relatives. In female carriers of X-linked CGD, DHR flow cytometry variants detect mosaicism due to skewed X-chromosome inactivation, revealing two neutrophil populations with differing ROS production. Supportive laboratory tests include complete blood count (CBC), which often shows neutrophilia during infections; elevated erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) as markers of ongoing inflammation; and microbial cultures from blood, tissue, or abscesses, which frequently isolate catalase-positive organisms such as or . Diagnostic criteria for CGD require a positive functional assay (e.g., DHR or NBT) in the context of compatible clinical history, with genetic testing to classify the inheritance pattern and guide family counseling; isolated supportive labs are insufficient for diagnosis.

Management and Treatment

Prophylactic Therapies

Prophylactic therapies for chronic granulomatous disease (CGD) primarily focus on preventing bacterial, fungal, and inflammatory complications through targeted antimicrobial and immunomodulatory agents. Antimicrobial prophylaxis is a cornerstone, with trimethoprim-sulfamethoxazole (TMP-SMX) administered at 5 mg/kg/day (trimethoprim component, divided into two doses) to provide broad bacterial coverage, including against Staphylococcus aureus, Burkholderia cepacia, and Nocardia species. This regimen has been shown to reduce the incidence of bacterial infections by approximately 56-66%, extending the time between infection episodes from every 10 months to every 40 months in treated patients. For fungal prevention, itraconazole is recommended at 5 mg/kg/day (up to 200 mg daily, preferably as oral solution) to target Aspergillus species, significantly lowering the rate of invasive fungal infections from 0.053 to 0.027 per patient-year. Posaconazole serves as an alternative azole, dosed at 120-200 mg every 12 hours for children under 40 kg or 300 mg daily for adults and larger children, particularly in cases of itraconazole intolerance or emerging resistance. Anti-inflammatory prophylaxis with interferon-gamma (IFN-γ) enhances phagocyte function and is particularly beneficial in X-linked CGD. Administered subcutaneously at 50 μg/m² three times per week, IFN-γ reduces the frequency of serious infections by 67-70% and decreases hospitalization days, with 79% of X-linked patients remaining infection-free at 12 months compared to 33% on placebo. This therapy improves overall survival by modulating macrophage activity and is routinely used in combination with antimicrobials in many centers. Guidelines from expert consensus, including those from the National Institutes of Health and European Society for Immunodeficiencies, recommend initiating lifelong prophylaxis with TMP-SMX and an azole antifungal at diagnosis for all CGD patients to minimize infection risk. IFN-γ is advised particularly for X-linked cases, though its use varies by institution. Monitoring for antimicrobial resistance is essential, with routine drug level assessments for azoles and switches to alternatives such as atovaquone (for sulfa-allergic patients unable to tolerate TMP-SMX) or other antibiotics like dicloxacillin if needed. Vaccination plays a supportive role in prophylaxis, with all standard inactivated childhood vaccines recommended per national schedules to protect against common pathogens. Live viral vaccines, such as measles-mumps-rubella and varicella, are generally safe in CGD patients not undergoing imminent hematopoietic stem cell transplantation (HSCT), but live bacterial vaccines like BCG or oral Salmonella typhi should be avoided due to heightened risk. Lifestyle modifications further reduce exposure to environmental pathogens, particularly molds like . Patients should avoid activities involving decayed organic matter, such as gardening, mulching, handling potting soil, or exposure to rotting plants and animal manure, which harbor high fungal loads. Good hygiene practices, including prompt wound care and avoiding dusty environments, complement these measures to prevent opportunistic infections.

Treatment of Acute Infections

The treatment of acute infections in patients with chronic granulomatous disease (CGD) emphasizes prompt, aggressive, and pathogen-directed interventions to compensate for defective phagocyte killing and prevent life-threatening complications. Identifying the causative organism through cultures, imaging, and biopsy is essential to guide therapy, with empiric broad-spectrum antimicrobials initiated immediately upon suspicion of infection. For bacterial pathogens like Staphylococcus aureus, a common culprit in CGD, intravenous combinations such as vancomycin plus piperacillin-tazobactam are standard to cover methicillin-resistant strains and gram-negative organisms. Fungal infections, particularly those involving Aspergillus species, require targeted antifungals like voriconazole (administered intravenously with therapeutic drug monitoring) or liposomal amphotericin B, often started empirically in patients with compatible symptoms such as persistent fever or pulmonary infiltrates despite antibacterial therapy. Surgical management is frequently necessary for localized suppurative infections, including drainage of abscesses in the liver or lungs, which affect 20-30% of CGD patients with acute presentations, and debridement for osteomyelitis to remove necrotic tissue and necrotic debris. These interventions, performed under imaging guidance when possible, are combined with ongoing antimicrobial therapy to eradicate residual infection. Treatment courses are extended due to impaired host immunity, generally spanning 4-6 weeks or longer for bacterial infections and 3-6 months for fungal ones, with clinical response assessed through repeated cultures, inflammatory markers, and serial imaging to guide de-escalation or prolongation. Breakthrough infections despite prophylactic regimens necessitate intensified acute management to restore control. Supportive measures enhance antimicrobial efficacy and address secondary effects of infection, including granulocyte colony-stimulating factor (G-CSF) administration to boost neutrophil counts and function in cases of neutropenia or severe leukopenia. Nutritional support, such as enteral or parenteral feeding, is provided for patients with gastrointestinal involvement, like inflammatory bowel disease exacerbations triggered by infection, to maintain immune competence and promote healing. Care is coordinated by a multidisciplinary team, including infectious disease specialists, surgeons, immunologists, and pharmacists, ensuring rapid diagnostics, optimized dosing, and holistic monitoring. This approach has dramatically lowered mortality from acute infections, from historical rates near 50% to under 10% with timely intervention.

Curative Interventions

Hematopoietic stem cell transplantation (HSCT) represents the primary curative intervention for (CGD), replacing the patient's defective phagocytes with functional donor cells to restore NADPH oxidase activity. When performed early in the disease course, particularly before age 5 years, HSCT achieves cure rates approaching 90-100% overall survival in pediatric cohorts using matched sibling donors and reduced-intensity conditioning regimens. Matched sibling donors are preferred due to lower rates of (GVHD) and higher engraftment success compared to unrelated or haploidentical donors. Reduced-intensity conditioning, often involving fludarabine and treosulfan or melphalan, has improved outcomes by minimizing toxicity, with reported survival exceeding 95% in low-risk patients and stable myeloid chimerism in over 90% of survivors. However, risks include acute and chronic (affecting 20-30% of patients, with severe cases contributing to 33% of post-transplant mortality), infections during the early post-transplant period (accounting for 42% of deaths), and organ toxicity from conditioning. Gene therapy offers an alternative curative approach by genetically correcting patient-derived hematopoietic stem cells ex vivo, primarily targeting the in X-linked CGD. Lentiviral vectors have shown promise in clinical trials, with transduction of autologous CD34+ cells leading to restored NADPH oxidase function in 6 of 9 patients at 12 months follow-up, enabling resolution of infections without myeloablative conditioning. Ongoing trials are evaluating similar lentiviral approaches for autosomal recessive forms, such as p47phox-deficient CGD, with initial data indicating safe engraftment and improved phagocyte function. Autologous HSCT following gene correction is emerging as a strategy to avoid donor-related complications, though long-term efficacy and risks like insertional mutagenesis remain under investigation. Indications for curative interventions prioritize patients with severe or refractory , including those with recurrent life-threatening infections, inflammatory complications unresponsive to prophylaxis, or progressive organ damage despite conventional management. Availability of a suitable family donor is crucial for , while gene therapy suits cases lacking matched donors or with specific genetic subtypes amenable to correction. No other interventions fully cure , underscoring the importance of early referral before irreversible damage occurs; prophylactic therapies may serve as a bridge to transplantation in eligible patients.

Prognosis and Epidemiology

Prognosis

With advances in medical care, the prognosis for individuals with (CGD) has improved substantially, with survival rates exceeding 90% at age 10 years and a median survival of over 40 years in modern cohorts. In contrast, prior to the 1980s, median survival was less than 10 years due to limited treatment options. Fungal infections remain a leading cause of mortality, accounting for approximately 55% of deaths in large patient series. Key prognostic factors include early diagnosis and adherence to prophylactic therapies, which significantly reduce infection risk and improve long-term outcomes. Hematopoietic stem cell transplantation (HSCT) offers a curative potential, with overall survival rates of 85-96% and disease-free survival of 81-93% in pediatric and young adult recipients, depending on donor matching and pre-transplant status. Patients with X-linked CGD generally face a worse prognosis than those with autosomal recessive forms, with median survival estimates of 37.8 years versus 49.6 years, respectively, reflecting differences in disease severity and residual NADPH oxidase activity. Quality of life in CGD is often impacted by chronic complications, including lung disease in about 50% of patients and gastrointestinal involvement in 33-60%, leading to issues such as recurrent pneumonia, bronchiectasis, colitis, and strictures. These manifestations contribute to reduced functional status and frequent hospitalizations, though prophylactic and can mitigate severity and enhance daily living. In adulthood, approximately 20% of patients develop severe complications, such as progressive pulmonary fibrosis or inflammatory bowel disease-like conditions, necessitating ongoing multidisciplinary monitoring. Additionally, CGD confers an elevated risk of malignancies, particularly gastrointestinal cancers, underscoring the importance of routine screening in long-term surveillance protocols. As of 2025, emerging gene therapy trials using lentiviral and CRISPR-based approaches have shown promising early results in individual pediatric patients, including restoration of phagocyte function in initial cases.

Epidemiology

Chronic granulomatous disease (CGD) has an estimated worldwide incidence of 1 in 200,000 to 250,000 live births. This rate is higher in populations with high rates of consanguinity, such as certain Middle Eastern communities, where autosomal recessive forms predominate and incidence can reach approximately 1 in 100,000 live births in affected regions. The overall prevalence of CGD is approximately 1 in 150,000 to 250,000 individuals, though it remains underdiagnosed in low-resource settings due to limited access to diagnostic testing. Demographically, about 70% of cases occur in males, reflecting the predominance of the X-linked form, while the condition affects all ethnic groups equally in terms of genetic susceptibility but shows higher detection rates in developed countries with advanced healthcare infrastructure. Geographic variations in incidence and inheritance patterns are notable; for example, the United States reports an incidence of 1 in 200,000 live births, while Sweden has a prevalence of around 1 in 450,000. Autosomal recessive forms are more prevalent in Asia and Africa, contributing to elevated rates in those regions compared to X-linked dominance in Western populations. Recent trends indicate improved early diagnosis through pilot newborn screening programs in select areas, enhancing detection without major shifts in overall incidence as of 2025. Increased awareness has also followed advancements in gene therapy trials, potentially leading to better case identification in the coming years.

History

Discovery and Early Descriptions

The initial recognition of what would later be known as chronic granulomatous disease (CGD) occurred in the early 1950s through scattered reports of children experiencing severe, recurrent infections accompanied by granuloma formation, often leading to fatal outcomes. These cases were frequently misattributed to tuberculosis, sarcoidosis, or malignancies due to the presence of granulomatous lesions and persistent suppuration. A pivotal description came in 1957 when Berendes et al. detailed the cases of three siblings—two boys and one girl—who suffered recurrent bacterial and fungal infections, hepatosplenomegaly, lymphadenopathy, and granulomatous inflammation, dubbing the entity "fatal granulomatous disease of childhood" based on its lethal progression and characteristic pathology. In parallel, Landing and Shirkey reported two boys with a similar syndrome of repeated infections starting in infancy, marked by visceral infiltration from lipid-laden histiocytes and noncaseating granulomas, underscoring the reticuloendothelial system's role in the disease process. These accounts, led by key figures like Robert A. Good (co-author on the Berendes report) and Brian H. Landing, first highlighted the underlying immunodeficiency rather than isolated infectious or neoplastic processes. The syndrome was described in detail in 1959 by Bridges et al., who termed it "a fatal granulomatous disease of childhood" based on autopsy findings in affected children that revealed widespread, noncaseating granulomas without evidence of typical infectious agents like acid-fast bacilli, distinguishing it from conventional granulomatous disorders. The term "chronic granulomatous disease" emerged in subsequent literature, such as in studies by Holmes et al. in the 1960s. Early clinical observations noted a striking male predominance, consistent with an X-linked pattern of inheritance, and emphasized the dismal prognosis, with most untreated patients succumbing to overwhelming infections within the first few years of life despite supportive antibiotic and surgical interventions.

Key Milestones

In 1967, Robert L. Baehner and David G. Nathan developed the nitroblue tetrazolium (NBT) reduction test, the first functional assay to detect the defective oxidative burst in neutrophils from patients with (CGD), enabling reliable diagnosis of the oxidase deficiency. This breakthrough shifted CGD from a poorly understood clinical syndrome to a defined phagocyte disorder, facilitating carrier detection and family screening. During the 1970s, researchers identified the core defect in CGD as a failure of the respiratory burst in phagocytes, where neutrophils and macrophages could not generate superoxide via the , leading to impaired microbial killing. Key studies, including those by Klebanoff and colleagues, linked this to reduced hydrogen peroxide production, explaining the susceptibility to catalase-positive bacteria and fungi. This mechanistic insight paved the way for targeted therapies beyond empirical antibiotics. In the 1980s, the cloning of the on the X chromosome, encoding the gp91phox subunit of , marked a pivotal advance in understanding X-linked CGD, which accounts for about 70% of cases. Dinauer et al.'s work in 1987 confirmed as the cause, allowing genetic diagnosis and prenatal testing. Concurrently, the identification of cytochrome b-558 components further elucidated the oxidase assembly. The 1990s brought clinical breakthroughs in prophylaxis. Prophylactic use of trimethoprim-sulfamethoxazole (TMP-SMX) was introduced in the late 1980s and confirmed effective in a 1990 randomized trial showing a significant drop in non-fungal infections without increasing fungal risks. In 1991, a placebo-controlled trial demonstrated that subcutaneous reduced serious infections by 70% in CGD patients, likely by enhancing non-oxidative killing mechanisms, establishing it as standard adjunctive therapy. Additionally, mapping of autosomal recessive genes progressed, with cloned in 1989 and in 1990, enabling comprehensive genetic subtyping. Entering the 2000s, hematopoietic stem cell transplantation (HSCT) emerged as curative, with matched sibling donor outcomes exceeding 80% overall survival by the mid-decade, particularly when performed early in childhood to prevent organ damage. A 2003 European series reported disease-free survival in over 80% of cases using reduced-intensity conditioning. Lentiviral vector-based gene therapy for CGD, offering a safer alternative to early retroviral approaches by reducing insertional mutagenesis risk, advanced through preclinical models in the 2000s and entered clinical trials in the 2010s, with promising results in restoring oxidase function reported in the 2020s. In the 2020s, lentiviral gene therapy trials have shown durable clinical benefits, including restored NADPH oxidase function in X-CGD patients (as of 2023). As of 2025, prime editing approaches like PM359 demonstrated early efficacy in Phase 1/2 trials, with one patient achieving 66% dihydrorhodamine (DHR) positivity by Day 30 post-treatment. These milestones transformed CGD from a typically fatal childhood illness, with median survival under 10 years in the 1950s, to a manageable chronic condition, extending life expectancy into adulthood through combined prophylaxis, transplantation, and emerging gene correction strategies.

Research Directions

Current Research

Ongoing research into chronic granulomatous disease (CGD) emphasizes advancements in diagnostics, mechanistic insights into inflammation, preclinical infection models, biomarker development, and longitudinal data from patient registries to improve early detection and management. Efforts to enhance diagnostics include studies on functional assays for CGD identification. Investigations into inflammatory pathways have revealed key roles for interleukin-1 (IL-1) and signal transducer and activator of transcription 3 (STAT3) in CGD-associated inflammatory bowel disease (IBD), prompting studies on targeted inhibitors. Anakinra, an IL-1 receptor antagonist, has demonstrated efficacy in reducing inflammation in CGD-IBD patients, with case series reporting symptom resolution and mucosal healing without increased infection risk. Additionally, metabolomics analyses indicate that interferon-gamma (IFN-γ) reprograms defective monocyte metabolism in CGD, restoring glycolysis, mitochondrial function, and amino acid levels (such as glutamine and serine) while suppressing excessive IL-1β and IL-6 production to mitigate hyperinflammation. This IFN-γ-mediated rewiring, detailed in a 2025 study, underscores its prophylactic benefits by enhancing fungal clearance and trained immunity in CGD cells. Preclinical models utilizing gp91phox knockout mice, which recapitulate human X-linked CGD, are being employed to test vaccines against opportunistic pathogens like Burkholderia species. These mice exhibit heightened susceptibility to Burkholderia cepacia complex infections, allowing evaluation of vaccine candidates that elicit protective immunity without relying on NADPH oxidase activity; recent studies have shown intranasal immunization with Burkholderia outer membrane vesicles improving survival in this model by 50-70%. Biomarker research focuses on circulating cytokines for predicting inflammatory flares in CGD. Elevated serum IL-8 levels have been associated with inflammation in CGD. Emerging applications of artificial intelligence in imaging analysis aim to detect granulomas more accurately; machine learning models trained on CT scans have achieved 85-90% accuracy in identifying granulomatous nodules in related pulmonary diseases, with potential adaptation for CGD-specific lung manifestations. Patient registries such as the United States Immunodeficiency Network (USIDNET) and dedicated CGD databases continue to track over 1,000 individuals globally, providing real-world data on outcomes like infection rates and inflammatory complications. Analyses from USIDNET, encompassing more than 300 CGD cases, highlight improved survival with early prophylaxis but persistent challenges from non-infectious inflammation, informing personalized care strategies.

Emerging Therapies

Emerging therapies for chronic granulomatous disease (CGD) focus on gene-based approaches to correct the underlying defects in the , offering potential curative options beyond traditional hematopoietic stem cell transplantation (HSCT). These investigational treatments target specific genetic mutations in genes such as CYBB, CYBA, NCF1, and NCF2, aiming to restore phagocyte function in (HSCs). As of 2025, clinical trials and preclinical studies have demonstrated promising safety and efficacy profiles, though challenges like off-target effects and long-term durability persist. CRISPR/Cas9-based gene editing has advanced for correcting CYBB mutations, the most common cause of X-linked CGD, by enabling precise targeted editing and cDNA insertion to restore gp91phox expression. A 2025 study developed strategies for variants in both CYBA (autosomal recessive CGD) and CYBB, achieving 50-70% correction efficiency in patient-derived HSCs through and cDNA knock-in at endogenous loci. This approach preserved genomic integrity and restored activity in edited myeloid cells, outperforming viral integration methods in preclinical models. Prime editing, a more precise CRISPR derivative, is being evaluated in the PM359 trial by Prime Medicine for autosomal recessive CGD due to NCF1 (p47phox) deficiency. In the phase 1/2 study initiated in 2025, a single infusion of prime-edited autologous CD34+ HSCs restored dihydrorhodamine (DHR) positivity in the first patient, indicating functional neutrophil recovery, with no serious adverse events reported and up to 80% restoration of NADPH oxidase function by day 30 post-infusion. Initial data from the first patient showed rapid engraftment and 58-66% DHR positivity within the first month, marking the first clinical proof-of-concept for prime editing in a genetic disease (as of May 2025). Lentiviral gene therapy trials target p47phox-deficient CGD through autologous HSCT with corrected cells. The phase 1/2 trial at (GOSH) and (UCL), launched in 2023, uses a lentiviral vector to deliver a functional NCF1 copy into patient HSCs, with infection-free follow-up periods exceeding one year in the first treated adolescent patient. This short ex vivo culture protocol minimizes risks associated with older retroviral methods. Other approaches include nucleases (ZFNs) for NCF1 correction, which target sequences to resurrect functional p47phox expression in iPSC-derived cells, restoring production in preclinical studies. Reduced-intensity HSCT protocols, often combined with these gene corrections, have improved overall survival to 95% in recent cohorts by reducing while maintaining engraftment. Despite these advances, challenges include potential off-target edits leading to unintended mutations and uncertainties in long-term engraftment and immune reconstitution. An August 2025 FDA patient listening session underscored the need for therapies addressing unmet needs in CGD, such as durable cures without HSCT-related complications, informing ongoing designs.

References

  1. [1]
    Chronic Granulomatous Disease (CGD) | NIAID
    May 22, 2020 · Chronic granulomatous disease (CGD) is a genetic disorder in which white blood cells called phagocytes are unable to kill certain types of bacteria and fungi.
  2. [2]
    Chronic granulomatous disease - Symptoms and causes - Mayo Clinic
    Mar 28, 2025 · Chronic granulomatous (gran-u-LOM-uh-tus) disease (CGD) is a genetic condition in which infection-fighting white blood cells don't work properly.Overview · Symptoms · Risk Factors
  3. [3]
    Chronic Granulomatous Disease - StatPearls - NCBI Bookshelf
    Chronic granulomatous diseases (CGD) are a group of recurrent life-threatening infections caused by a hereditary defect in forming reactive oxygen species.Continuing Education Activity · Introduction · Etiology · Treatment / Management
  4. [4]
    Chronic Granulomatous Disease - GeneReviews® - NCBI Bookshelf
    Aug 9, 2012 · Chronic granulomatous disease (CGD) is characterized by severe recurrent bacterial and fungal infections and dysregulated inflammatory responses ...
  5. [5]
    Inflammatory Complications in Chronic Granulomatous Disease - PMC
    Feb 15, 2024 · Inflammatory complications are also common, often affecting the gastrointestinal, respiratory, urinary tracts and other tissues.
  6. [6]
    Noninfectious Manifestations and Complications of Chronic ... - NIH
    May 9, 2018 · These complications can include, for example, a wide range of inflammatory diseases that affect the gastrointestinal tract, lung, skin, and genitourinary tract ...
  7. [7]
    A Review of Chronic Granulomatous Disease - PMC - NIH
    Nov 22, 2017 · Infections are primarily with a subset of catalase-positive microorganisms, and the most common sites of infection are the lungs, skin, lymph ...
  8. [8]
    X-Linked CGD and Carrier Inheritance - Actimmune HCP
    About 10%-15% of X-linked mutations are the result of new random mutations. X-linked CGD often presents early in life with most cases diagnosed by age 5.Missing: percentage | Show results with:percentage
  9. [9]
    Clinical and demographic pattern of chronic granulomatous disease ...
    May 17, 2021 · Twenty patients were diagnosed as CGD. Males (N = 13, 65%) outnumber females (N = 7, 35%). CGD is commonest amongst the Malays (65%) followed by ...
  10. [10]
    Chronic Granulomatous Disease - Medscape Reference
    Jul 29, 2019 · Patients with CGD are at increased risk of life-threatening infections with catalase-positive bacteria and fungi and inflammatory complications ...
  11. [11]
    NADPH Oxidase Deficiency: A Multisystem Approach - PMC
    NADPH oxidase (NOX) plays a pivotal role in the production of ROS, and the defect of its different subunits leads to the development of chronic granulomatous ...
  12. [12]
    Recent advances in chronic granulomatous disease - PMC - NIH
    Chronic granulomatous disease (CGD) is an inherited defect of phagocyte function due to defective NADPH oxidase. Patients with CGD are not able to effectively ...
  13. [13]
    Inflammation and repeated infections in CGD: two sides of a coin
    Nov 15, 2011 · IL-17 also synergizes with other cytokines, such as IL-1, IL-6, and TNF-α that promote activation of tissue infiltrating neutrophils to ...
  14. [14]
    Chronic Granulomatous Disease: Epidemiology, Pathophysiology ...
    May 9, 2018 · In this review, we briefly discuss the epidemiology, genetics, and pathophysiology of CGD; we focus first on the genetic mutations responsible for the disease.
  15. [15]
    Gastrointestinal Features of Chronic Granulomatous Disease Found ...
    Bowel strictures and fistulae are present in a significant number of patients. Upper gastrointestinal tract inflammatory disease is common, though typically not ...
  16. [16]
  17. [17]
    Chronic granulomatous disease - Diagnosis and treatment
    Mar 28, 2025 · To diagnose CGD, a healthcare professional will review a family and medical history and do a physical exam. There are several tests used to diagnose CGD.
  18. [18]
    Considerations in the Diagnosis of Chronic Granulomatous Disease
    May 9, 2018 · The purpose of this review is to highlight considerations for and challenges in the diagnosis of CGD. Keywords: chronic granulomatous disease, ...Missing: physical | Show results with:physical
  19. [19]
    Prevention of Infectious Complications in Patients With Chronic ...
    May 9, 2018 · An ideal antibacterial prophylactic regimen for patients with CGD, therefore, would include bactericidal activity against all 5 of the pathogens ...
  20. [20]
    A Controlled Trial of Interferon Gamma to Prevent Infection in ...
    Feb 21, 1991 · This study shows that interferon gamma is an effective and safe therapy for patients with chronic granulomatous disease, since the drug ...
  21. [21]
    2013 IDSA Clinical Practice Guideline for Vaccination of the ...
    Inactivated vaccines should be administered ≥2 weeks prior to immunosuppression (strong, moderate). RECOMMENDATIONS FOR VACCINES FOR HOUSEHOLD MEMBERS OF ...
  22. [22]
    Lessons About the Pathogenesis and Management of Aspergillosis ...
    Management of CGD patients requires minimizing exposure to Aspergillus by avoiding environmental risk factors such as mulch, rotting plants and animal manure.
  23. [23]
    Chronic Granulomatous Disease Treatment & Management
    Jul 29, 2019 · Modern therapy for chronic granulomatous disease (CGD) includes aggressive and prolonged administration of antibiotics and prednisone.Medical Care · Consultations · Prevention
  24. [24]
    Chronic granulomatous disease: Treatment and prognosis - UpToDate
    Jan 30, 2023 · The average patient now survives at least 40 years. Overall, survival rates are better for females (autosomal recessive) than males (most often ...<|separator|>
  25. [25]
    Chronic Granulomatous Disease - PMC - NIH
    Similarly, Muoy (28) found an 8-year survival rate of 70.5% for children born before 1978 but a 92.9% survival rate for those born later. Before the ...
  26. [26]
    Common Severe Infections in Chronic Granulomatous Disease - PMC
    Fungal infection accounted for 21 deaths (21/38 [55%]). Overall Aspergillus-specific mortality was 9%. Death due to fungal infection was confirmed in 37% of the ...
  27. [27]
    Bone marrow transplant shows promise for chronic granulomatous ...
    Oct 22, 2022 · The results of the study are promising. The trials show a 96 percent survival rate – one person died from underlying lung disease – and 93 percent of people in ...
  28. [28]
    Chronic Granulomatous Disease: The European Experience
    Of the 429 patients, 351 were male (82%) and 78 female (18%), including 6 heterozygous female carriers with an extremely unfavorable lyonization (Table 2) [19].Missing: ratio | Show results with:ratio<|separator|>
  29. [29]
    Chronic Granulomatous Disease in Patients Reaching Adulthood
    Concerning the digestive tract, 8.8% of patients showed chronic complications at age 16 chronic diarrhea (9%), bowel stenosis (7%), or chronic fistulae (7%), ...
  30. [30]
    New gene therapy is life changing for teenage patient | UCL News
    Aug 18, 2025 · Remi was the first patient to receive this gene therapy in 2024 and the trial will treat five patients in total. p47 CGD is an inherited genetic ...<|separator|>
  31. [31]
    CRISPR Clinical Trials: A 2025 Update - Innovative Genomics Institute
    Jul 9, 2025 · Chronic Granulomatous Disease (CGD). Two clinical trials have been initiated that address chronic granulomatous disease (CGD), each targeting ...
  32. [32]
    Frontiers | Chronic granulomatous disease in the United Arab Emirates
    Nov 1, 2023 · Consanguinity was noted in all patients with AR CGD, whereas positive family history was identified in 50% of cases. The median age of onset of ...Missing: Sweden | Show results with:Sweden
  33. [33]
    Diagnosis of Chronic Granulomatous Disease: Strengths and ... - MDPI
    NCF1 (neutrophil cytosolic factor 1) encodes p47phox, and mutation of this gene accounts for about 20% of all CGD [67,68,69]. NCF1-related CGD is generally mild ...3. Molecular And Cellular... · 6. Testing Of Nadph Oxidase... · 9. Mutations In Cgd
  34. [34]
    Chronic granulomatous disease - Orphanet
    CGD follows an X-linked pattern of inheritance in those with a CYBB mutation. It can also be inherited autosomal recessively (due to CYBA, NCF1, NCF2, NCF4 and ...
  35. [35]
    Geographic Variability and Pathogen-Specific Considerations in the ...
    Chronic granulomatous disease (CGD) is a rare but serious primary immunodeficiency with varying prevalence and rates of X-linked and autosomal recessive disease ...Missing: 2023-2025 | Show results with:2023-2025
  36. [36]
    Emerging diagnostics & therapies for chronic granulomatous ...
    Aug 5, 2024 · Emerging diagnostics & therapies for chronic granulomatous disease (CGD): 2024 PI Conference ... Learn the potential clinical implications of recent studies and ...
  37. [37]
    Great Ormond Street Hospital Tackling Chronic Granulomatous ...
    Aug 18, 2025 · The nonrandomized, open-label clinical trial for pCHIM-p47, which launched on March 20, 2023, is taking place at Great Ormond Street Hospital in ...
  38. [38]
    A fatal granulomatous disease of childhood; the clinical ... - PubMed
    A fatal granulomatous disease of childhood; the clinical, pathological, and laboratory features of a new syndrome. AMA J Dis Child. 1959 Apr;97(4):387-408.Missing: et al
  39. [39]
    A fatal granulomatosus of childhood: the clinical study of a ... - PubMed
    A fatal granulomatosus of childhood: the clinical study of a new syndrome. Minn Med. 1957 May;40(5):309-12. Authors. H BERENDES, R A BRIDGES, R A GOOD.
  40. [40]
    A syndrome of recurrent infection and infiltration of viscera ... - PubMed
    1957 Sep;20(3):431-8. Authors. B H LANDING, H S SHIRKEY. PMID: 13465232. No abstract available. MeSH terms. Child; Communicable Diseases*; Disease*; Histiocytes ...
  41. [41]
    Leukocyte Oxidase: Defective Activity in Chronic Granulomatous ...
    The intact leukocytes of two children with chronic granulomatous disease fail to reduce nitroblue tetrazolium during phagocytosis.<|separator|>
  42. [42]
    Chronic Granulomatous Disease; fundamental stages in our ...
    Holmes et al. [5] suggested that the cellular defect was 'a deficiency in one enzyme, either located within polymorph lysosomes or among those responsible for ...
  43. [43]
    The Respiratory Burst and Its Physiological Significance
    The Respiratory burst and its physiological significance I edited by Anthony J. Sbarra and Robert R. Strauss. p. cm. Includes bibliographies and index.<|separator|>
  44. [44]
    Trimethoprim-Sulfamethoxazole Prophylaxis in the Management of ...
    TMP-SMX prophylaxis is indicated for the management of patients with CGD and decreases the incidence of nonfungal infections without increasing the incidence ...
  45. [45]
    Genes for Two Autosomal Recessive Forms of Chronic ... - PubMed
    Two autosomal recessive forms of CGD result from defects in cytosolic components of the phagocyte NADPH oxidase system, p47phox (NCF1) and p67phox (NCF2).
  46. [46]
    Significant Improvement in Survival After Allogeneic Hematopoietic ...
    The study reported herein describes the changes in the alloHCT patient population and their overall survival (OS) rates over a 12-year period. PATIENTS AND ...
  47. [47]
    Lentiviral Vector Gene Therapy Protects XCGD Mice From Acute ...
    Sep 6, 2016 · Our results support the clinical development of gene therapy protocols using lentiviral vectors for the protection against infections and inflammation.
  48. [48]
    The value of DHR-enzyme-linked immunosorbent assay ... - PubMed
    Jun 5, 2025 · The DHR-ELISA method demonstrated a specificity of 90% and a sensitivity ranging from 90.5% to 100% in detecting CGD. Conclusion: The DHR-ELISA ...Missing: newborn dried blood spots pilots US Europe 2023-2025
  49. [49]
    Chronic granulomatous disease with inflammatory bowel ... - NIH
    May 1, 2023 · Anakinra, an IL-1 receptor antagonist, has also been used in CGD-IBD patients and shown to be effective, however one study noted that the early ...Missing: STAT3 2023-2025
  50. [50]
    Interferon gamma rebalances immunopathological signatures in ...
    CGD monocytes exhibit profound glycolytic and mitochondrial metabolic defects, and reduced intracellular amino acid levels. IFN-γ rebalances metabolic and ...
  51. [51]
    Virulence of Burkholderia cepacia complex strains in gp91phox-/- mice
    We conclude that the Bcc infection model in CGD mice is well suited for the assessment of Bcc virulence. Publication types. Research Support, Non-U.S. Gov't ...Missing: vaccine 2020-2025
  52. [52]
    Intranasal immunization with a Bucl8-based vaccine ameliorates ...
    Jul 19, 2023 · To evaluate vaccine efficacy, we developed a surrogate Burkholderia infection model that employs outbred CD-1 mice which imitates the ...Missing: gp91phox | Show results with:gp91phox
  53. [53]
    Inflammatory Complications in Chronic Granulomatous Disease
    Feb 15, 2024 · Chronic granulomatous disease (CGD) is a rare inborn error of immunity that typically manifests with infectious complications.1. Introduction · 2. Prevalence And Clinical... · 5. Aetiopathogenesis
  54. [54]
    CRDet: A circle representation detector for lung granulomas based ...
    In this paper, we propose an automatic method CRDet to localize granulomas in histopathological images and deal with these challenges.Missing: machine chronic
  55. [55]
    Original Article Chronic Granulomatous Disease With Inflammatory ...
    We analyzed patients with CGD from the US Immunodeficiency Network (USIDNET) registry to determine whether IBD changes the presentation, treatment, and outcomes ...
  56. [56]
    Registry Data and Queries - USIDNET
    Within the USIDNET registry, thousands of clinical, molecular, and laboratory data points are collected from individuals with primary immunodeficiencies ...Missing: real- world outcomes 2023-2025
  57. [57]
    Targeted gene editing and near-universal cDNA insertion of CYBA ...
    Aug 12, 2025 · Consequently, transplantation of autologous ex vivo gene-edited HSCs has emerged as a promising treatment option for IEIs, including CGD.<|separator|>
  58. [58]
    Targeted gene editing and near-universal cDNA insertion of CYBA ...
    Aug 12, 2025 · 2025 Aug ... Here, we develop CRISPR/Cas9-based gene editing strategies for correction of variants in the CYBA and CYBB genes causing CGD.Missing: study | Show results with:study
  59. [59]
    Gene Editing Enables cDNA Therapy for Chronic Granulomatous ...
    Sep 6, 2025 · The targeted nature of CRISPR-Cas9 mediated cDNA insertion into endogenous loci represents an elegant solution to preserve genomic context and ...
  60. [60]
    Prime Medicine: Rapid DHR Positivity After PM359 Infusion for CGD
    May 19, 2025 · Prime Medicine Announces Breakthrough Clinical Data Showing Rapid Restoration of DHR Positivity After Single Infusion of PM359, an ...
  61. [61]
    First-Ever Prime-Editing Therapy Shows Safety and Efficacy in ...
    May 21, 2025 · In May 2024, Prime Medicine announced that the FDA had cleared its IND application for PM359 for the treatment of CGD, making PM359 the first ...
  62. [62]
    Prime Medicine Announces Breakthrough Clinical Data Showing
    May 19, 2025 · Initial data from first patient dosed in Phase 1/2 trial finds single dose of PM359 led to 58% DHR positivity by Day 15 and 66% by Day 30.
  63. [63]
    UCL and GOSH gene therapy for rare immunological condition ...
    Aug 18, 2025 · The new gene therapy is the first clinical trial in the world for patients with the p47 type of CGD. Remi was the first patient to receive ...Missing: Lentiviral p47phox
  64. [64]
    CRISPR-Directed Therapeutic Correction at the NCF1 Locus Is ... - NIH
    Here we demonstrate that concurrent targeting and correction of mutated NCF1 and its pseudogenes results in therapeutic CGD phenotype correction, but also ...
  65. [65]
    Reliable genetic diagnosis of NCF1 (p47 phox )-deficient chronic ...
    Aug 17, 2025 · Each patient with 1 intact GTGT copy had a single mutation, while P1, with 2 GTGT alleles, carried 2 separate mutations; each variant had a ...
  66. [66]
    Gene-edited pseudogene resurrection corrects p47phox-deficient ...
    Jan 10, 2017 · Here, we examined strategies for p47-CGD gene correction using engineered zinc-finger nucleases (ZFNs) targeting the exon 2 ΔGT in induced ...
  67. [67]
    Fludarabine Based Reduced-Intensity Conditioning Allogenic Stem ...
    The overall survival (OS) rate was improved to 70% to 90% using Reduce Intensity Conditioning (RIC) regimen However, it can lead to establishment mixed ...
  68. [68]
    [PDF] FDA Patient Listening Session - Chronic Granulomatous Disease
    Sep 15, 2025 · ○ Disease status: cured. ○ Treatments: CGD prophylaxis prior to unrelated umbilical cord blood transplant and not no treatments other than ...
  69. [69]
    Coffee & Clinicians on gene therapy; News from Ensoma, Prime
    Sep 25, 2025 · Summary of FDA Patient Listening Session for Chronic Granulomatous Disease is now available! On August 11, 2025, the CGDAA had the honor of ...