Fact-checked by Grok 2 weeks ago

Fanconi anemia

Fanconi anemia (FA) is a inherited characterized by progressive failure, congenital physical abnormalities, and a markedly increased risk of developing cancers, particularly and solid tumors of the head and neck. It occurs worldwide with an estimated frequency of 1 in 100,000 to 160,000 individuals, though it is more prevalent in certain populations such as those of Ashkenazi Jewish, , Afrikaner, and descent. The arises from defects in a DNA repair pathway that handles interstrand crosslinks, leading to genomic instability, cell death in rapidly dividing tissues like the , and heightened susceptibility to DNA damage from environmental agents. Genetically, FA results from biallelic pathogenic variants in one of at least 23 genes that function in the FA pathway (including the recently identified FANCX complementation group), with the FANCA gene accounting for approximately 60-70% of cases. Inheritance is predominantly autosomal recessive, requiring two mutated copies of the gene (one from each parent), but rare instances involve autosomal dominant transmission (e.g., via RAD51) or X-linked recessive patterns (e.g., via FANCB). Carriers of a single mutated gene are typically asymptomatic but have a 25% chance of having an affected child if both parents are carriers. Clinically, FA presents with a wide spectrum of features, though not all individuals exhibit every manifestation. Common congenital anomalies include , skeletal malformations such as absent or underdeveloped thumbs and radii, café-au-lait spots or of the skin, and genitourinary or cardiac defects. Bone marrow failure typically develops between ages 5 and 10, resulting in —low levels of red blood cells (), white blood cells (), and platelets ()—which predisposes patients to fatigue, recurrent infections, and easy bruising or bleeding. Cancer risk is substantial, with about 10-30% of individuals developing by age 40 and a cumulative incidence of up to 86% for any by age 50, including gynecologic and gastrointestinal tumors. Diagnosis of FA relies on clinical suspicion prompted by family history, physical findings, or early-onset bone marrow issues, confirmed by specialized tests such as diepoxybutane (DEB)- or (MMC)-induced chromosomal breakage assays, which reveal hypersensitivity to DNA crosslinking agents, or targeted molecular to identify causative variants. Management is multidisciplinary and supportive, with (HSCT) from a matched donor serving as the only curative option for bone marrow failure, ideally performed before age 10 to minimize complications. Other interventions include androgen therapy (e.g., ) to stimulate blood cell production, (G-CSF) for , transfusions for or , and rigorous surveillance protocols for early cancer detection through regular blood counts, imaging, and endoscopies. Ongoing research into shows promise, with clinical trials demonstrating potential for correcting the underlying genetic defects.

Introduction

Definition and characteristics

Fanconi anemia (FA) is a rare inherited bone marrow failure syndrome characterized by progressive failure of the bone marrow to produce sufficient blood cells, leading to genomic instability due to defects in the DNA interstrand cross-link repair pathway. This pathway, known as the Fanconi anemia pathway, is essential for repairing DNA damage from interstrand cross-links, and its impairment results in chromosomal fragility and cellular hypersensitivity to DNA-damaging agents. FA is classified as one of the inherited bone marrow failure syndromes (IBMFS), distinguished by its involvement of all three hematopoietic lineages—erythroid, myeloid, and megakaryocytic—manifesting as pancytopenia that typically develops in the first decade of life. Key clinical hallmarks of FA include progressive , with reduced production of red blood cells (), white blood cells (), and platelets (), often progressing to severe aplasia. Congenital malformations are present in 60-75% of affected individuals, commonly affecting skeletal, renal, and cardiac systems, though these vary widely in severity. Additionally, FA confers a markedly elevated cancer risk, with (AML) occurring in approximately 10-30% of cases and solid tumors (such as those of the head and neck, , and gynecological organs) developing at rates significantly higher than in the general population. The disorder was first described in 1927 by pediatrician Guido Fanconi, who reported on three siblings exhibiting a combination of physical anomalies and progressive .

Historical background

Fanconi anemia was first described in 1927 by the pediatrician Guido Fanconi, who reported a sibship of three brothers exhibiting progressive , physical malformations including and skeletal anomalies, and eventual death from bone marrow failure. This initial report highlighted the familial nature of the condition and distinguished it from other forms of childhood through its association with congenital defects, though early understandings viewed it as a variant of inherited . The disorder later became known as "Fanconi anemia" to honor the describers' contributions, evolving from prior terms like "congenital hypoplastic anemia" used for similar bone marrow failure syndromes. In the early , researchers observed elevated rates of spontaneous chromosomal breakage in cultured cells from affected individuals, marking the recognition of Fanconi anemia as a chromosomal instability disorder. This finding, pioneered by studies such as those by Schroeder in , laid the groundwork for diagnostic advancements. Subsequent work in the early refined this by demonstrating hypersensitivity to DNA cross-linking agents; for instance, exposure to diepoxybutane (DEB) or induced significantly higher chromosome aberrations in Fanconi anemia cells compared to controls, establishing these agents as hallmarks for cytogenetic . The 1980s and 1990s saw major progress in unraveling the genetic basis, with the identification of multiple complementation groups indicating . The first Fanconi anemia gene, FANCC, was cloned in 1992 through functional complementation studies, revealing mutations responsible for a subset of cases. This was followed by the cloning of FANCA in 1996, the most common genetic subtype accounting for about 60-70% of patients, and the delineation of at least eight complementation groups by the late 1990s, confirming the disorder's multisubunit nuclear complex involvement. From the 2000s onward, discoveries linked Fanconi anemia to broader pathways, notably the identification of as FANCD1 in 2002 via studies of biallelic mutations causing severe early-onset disease. This connection illuminated the Fanconi anemia/BRCA pathway's role in interstrand cross-link repair and its overlap with susceptibility, expanding understanding of the disorder's predisposition to malignancies. Subsequent research through 2025 has identified additional genes, bringing the total to 23 complementation groups, and advanced approaches that show promise in clinical trials for correcting the genetic defects.

Epidemiology

Incidence and prevalence

Fanconi anemia (FA) is a rare with a global incidence estimated at 1 in 100,000 to 160,000 live births. In Western populations, the incidence is similarly reported as approximately 1 in 130,000 to 160,000 live births. These figures reflect the autosomal recessive inheritance pattern of the disease, where both parents must be carriers for a child to be affected. The prevalence of FA is estimated to be around 1 to 9 cases per million individuals worldwide, though this may be an underestimate due to underdiagnosis, particularly in low-resource settings where access to and specialized diagnostics is limited. In the United States, approximately 20 to 30 new cases are diagnosed annually, based on patient registries and birth statistics. Certain populations exhibit higher prevalence due to founder mutations. Among individuals of Ashkenazi Jewish descent, the incidence is about 1 in 32,000, primarily linked to mutations in the . The (Gypsy) population in has the world's highest reported prevalence, with a carrier frequency of 1 in 64 to 70 attributable to a specific . Similarly, in Afrikaner populations of , the birth incidence is at least 1 in 22,000, driven by a in the . Elevated prevalence is also observed in the population due to specific founder mutations.

Demographic patterns

Certain ethnic groups exhibit elevated carrier frequencies due to mutations, notably in Ashkenazi Jewish populations. For instance, the FANCC c.456+4A>T contributes to carrier rates of approximately 1-2% in this group. Geographic disparities in Fanconi anemia prevalence are pronounced in regions with high rates of consanguineous marriages, such as the , where autosomal recessive disorders are amplified. For example, the condition is more common in Saudi Arabian cohorts due to cultural practices favoring relatedness. The implementation of expanded carrier screening programs, particularly in high-risk populations like since the 2010s, aims to reduce incidence through informed reproductive choices, including prenatal diagnosis and preimplantation . These efforts have mirrored successes in other recessive disorders, such as Tay-Sachs disease.

Clinical manifestations

Congenital anomalies

Congenital anomalies are present in approximately 60-75% of individuals with Fanconi anemia, manifesting as a wide range of structural malformations that primarily affect the skeletal, renal, cardiac, and craniofacial systems. These birth defects often occur in combination and contribute to the variable observed across patients, with some exhibiting multiple anomalies while others have none. Skeletal anomalies are among the most characteristic, particularly those involving the radial ray, such as absent or hypoplastic thumbs (affecting 30-50% of cases) and radial aplasia. Other skeletal features include hip dysplasia and syndactyly of the lower limbs. Renal malformations occur in 30-40% of patients and commonly include horseshoe kidney and renal ectopia, which can predispose to functional impairments. Cardiac defects, seen in 10-15% of cases, frequently involve ventricular septal defects or patent ductus arteriosus. Craniofacial abnormalities, such as microcephaly and low-set ears, are reported in about 20-30% of individuals. Skin pigmentation irregularities, including café-au-lait spots or hyperpigmentation, affect 50-60% of patients and are often evident from birth. Growth disturbances, including short stature and low birth weight, are intrinsic features observed in the majority of cases, reflecting early developmental disruptions. These anomalies arise from defects in the Fanconi anemia pathway, which leads to accumulated DNA damage during critical periods of embryonic development, such as limb bud formation and . This genomic instability disrupts normal cellular and in the , resulting in the observed malformations without direct involvement of later-onset hematologic issues.

Hematologic abnormalities

Hematologic abnormalities represent a hallmark of Fanconi anemia, characterized by progressive failure that leads to in more than 90% of patients during their lifetime. This failure typically manifests as a reduction in all three major lineages: blood cells, , and platelets. in Fanconi anemia is often macrocytic, with elevated , though it can appear normocytic in some cases; thrombocytopenia and follow, contributing to the full pancytopenic state. The onset of bone marrow failure occurs at a median age of 7 years, though progression varies by complementation group, with earlier development observed in some, such as FANCA. By , nearly all affected individuals exhibit significant cytopenias, with the condition advancing to severe in the majority. Clinical manifestations include fatigue and pallor due to , recurrent infections from , and easy bruising or bleeding from ; reticulocytopenia often serves as an early laboratory indicator of impending marrow failure. Bone marrow examination in these patients reveals hypocellularity, typically with cellularity less than 25%, alongside variable dysplastic changes in hematopoietic precursors that may precede or accompany progression to (MDS). The cumulative incidence of MDS reaches approximately 40% by age 50, frequently evolving into (AML), for which patients face a 600-fold increased compared to the general population. This hematopoietic dysfunction stems from underlying defects that hasten exhaustion, as detailed in the of the disorder.

Other systemic features

Fanconi anemia is associated with a range of endocrine dysfunctions, affecting 50-75% of affected individuals. Hypogonadism manifests as delayed puberty in approximately 60% of females, often leading to premature ovarian failure, and as small testes or azoospermia in up to 64% of males. Growth hormone deficiency occurs in 12-25% of patients, contributing to short stature, while hypothyroidism affects 30-60% and requires lifelong monitoring. Glucose intolerance is prevalent, with 8-10% developing diabetes and 25-70% showing impaired glucose tolerance or insulin resistance. Neurologic features beyond structural anomalies include in approximately 21% of patients, in 24%, and mixed hearing loss in 10%, often necessitating audiologic evaluation. Mild cognitive impairments, such as learning disabilities and attention deficits, along with behavioral problems like anxiety or , are observed in 20-30% of individuals, potentially impacting . Gastrointestinal involvement includes or in 5-10% of cases, frequently overlapping with features like vertebral or renal anomalies. Other anomalies, such as or , occur in up to 14% of patients with gastrointestinal malformations. Beyond neutropenia-related risks, patients exhibit immune dysregulation, including impaired function and reduced T-lymphocyte responses, leading to increased susceptibility to infections in a significant proportion even without severe bone marrow failure. Later-onset features encompass liver fibrosis or persistent liver injury in 10-42% of patients, particularly following or androgen therapy, with monitoring essential to detect progression to .

Genetics

Inheritance and penetrance

Fanconi anemia is primarily inherited in an autosomal recessive manner, accounting for approximately 95% of cases, which requires biallelic pathogenic variants in one of the associated genes for the disorder to manifest. In this pattern, both parents of an affected individual are typically carriers, each harboring one pathogenic variant. Rarely, autosomal dominant occurs in approximately 2% of cases, specifically associated with pathogenic variants in RAD51 (also known as FANCR), where a single heterozygous variant is sufficient to cause the condition. Even more infrequently, X-linked is observed in approximately 2% of cases due to pathogenic variants in FANCB, affecting males more severely while females may be carriers. Penetrance for bone marrow failure in classic Fanconi anemia is nearly complete, with most affected individuals developing hematologic abnormalities by or early adulthood. However, the disorder exhibits variable expressivity, leading to a wide range of clinical severity even among individuals with similar genotypes; for instance, those with biallelic pathogenic variants in FANCD1 () often present with a severe including early-onset malignancies, though milder manifestations have been reported in some cases. The frequency for Fanconi anemia pathogenic variants in the general population is estimated at 1 in 100 to 300, with higher rates in specific founder populations such as (approximately 1 in 90) and . pathogenic variants are rare, occurring in fewer than 5% of cases overall, though they are more common in the autosomal dominant RAD51-related form. is essential for families affected by Fanconi anemia, particularly emphasizing the 25% recurrence risk to siblings in autosomal recessive forms, which underscores the importance of testing, prenatal diagnosis, and when pathogenic variants are identified. For X-linked cases, females face a 50% risk of transmitting the variant to offspring, necessitating tailored counseling strategies.

Genes and complementation groups

Fanconi anemia (FA) is genetically heterogeneous, with pathogenic variants identified in 23 complementation groups, designated A through Y, each corresponding to a specific in the FA pathway. These groups were defined through somatic cell hybridization studies that demonstrated functional complementation between cells from different FA patients, revealing the distinct genetic defects underlying the disorder. The most prevalent group is FANCA, accounting for 60-70% of cases worldwide, followed by FANCC (10-15%) and FANCG (approximately 10%); the remaining groups collectively represent less than 20% of cases, with some (such as FANCD1, FANCD2, and FANCE) each comprising 1-3%. Ethnic variations influence distribution; for instance, FANCC mutations predominate in Ashkenazi Jewish populations, while FANCA is ubiquitous across ethnicities. Key genes associated with these groups include FANCA, located on chromosome 16q24.3, which encodes a essential for the FA core complex assembly; FANCC at 9q22.3, encoding a protein involved in the stability of this core complex; and (also known as FANCD1) at 13q13.1, which plays a role in DNA recombination processes. Other notable genes are FANCB (Xp22.2), FANCD2 (3p25.3), FANCE (6p21.2), and FANCF (11p15), each defining rarer complementation groups. The full spectrum encompasses genes such as FANCI (15q25-26), FANCL (2p16.1), FANCM (14q21), (FANCN, 16p12), RAD51C (FANCO, 17q22), and (FANCS, 17q21), among others, with the most recent addition being FAAP100 (FA-Y, 11p15.1). Pathogenic variants in FA genes are predominantly biallelic, consistent with the autosomal recessive inheritance pattern of the disorder, though FANCB exhibits X-linked inheritance. —where an individual inherits two different pathogenic variants in the same gene—is common, particularly in FANCA and FANCC. Mutation types include , frameshift, and splicing defects, with gross deletions also frequent in FANCA (about 30% of cases). For example, the splicing mutation IVS38-6G>A in FANCA is recurrent among patients. Approximately 20-30% of FA cases involve mutations, many of which are potentially amenable to translational therapies using drugs like . Monoallelic carriers of pathogenic variants in certain FA genes, such as , , , RAD51C, and BRIP1 (FANCJ at 17q22.3), face a mildly elevated of cancers, including and ovarian malignancies, highlighting an overlap with hereditary cancer syndromes. BRIP1 variants, in particular, were linked to this cancer predisposition in complementation group J, with key studies reinforcing the connection around 2016.
Complementation GroupGeneChromosomal LocationApproximate Frequency (%)
AFANCA16q24.360-70
CFANCC9q22.310-15
GFANCG9p1310
D2FANCD23p25.33
BFANCBXp22.22
Others (D1, E, F, etc.)VariousVarious<2 each

Pathophysiology

Defective DNA repair pathway

Fanconi anemia (FA) is characterized by defects in a specialized DNA repair pathway known as the , which primarily functions to resolve DNA interstrand cross-links (ICLs) that impede DNA replication and transcription. The pathway involves a multi-subunit core complex composed of eight proteins: FANCA, FANCB, FANCC, FANCE, FANCF, FANCG, FANCL, and FANCM. This complex acts as an E3 ubiquitin ligase, with FANCL serving as the catalytic subunit, to monoubiquitinate the (the ID2 complex) in response to DNA damage. FANCM functions as a sensor that recruits the core complex to sites of stalled replication forks induced by ICLs, while the other subunits assemble to facilitate ubiquitination. The pathway is activated by various genotoxic stresses, including ICLs generated by endogenous sources such as aldehydes (e.g., formaldehyde and acetaldehyde from metabolic processes) or exogenous agents like chemotherapy drugs (e.g., cisplatin and mitomycin C). Upon activation, the core complex monoubiquitinates the FANCD2-FANCI dimer at lysine residues (K561 on FANCD2 and K523 on FANCI), which promotes the recruitment of downstream effectors to the lesion site. This monoubiquitination is essential for coordinating ICL repair, involving initial unhooking of the cross-link by structure-specific nucleases such as the FANCP/SLX4 complex, which incises the DNA to create a double-strand break. Subsequent steps include translesion synthesis (TLS) to bypass the lesion using specialized polymerases (e.g., POLζ) and template switching via homologous recombination (HR) to restore the genome. The FA pathway overlaps significantly with the BRCA-dependent HR pathway, integrating ICL repair with error-free double-strand break resolution. Specifically, downstream FA proteins include FANCD1/BRCA2, which facilitates RAD51 loading for HR; FANCN/PALB2, a BRCA2 binding partner that bridges BRCA1 and BRCA2; and FANCJ/BRIP1, a DNA helicase that unwinds DNA structures and interacts with BRCA1. These connections explain the shared genomic instability phenotypes between FA and BRCA-related disorders, such as increased sensitivity to DNA damaging agents. Cells from individuals with FA exhibit profound hypersensitivity to ICL-inducing agents, including mitomycin C (MMC) and diepoxybutane (DEB), with survival rates reduced by 100- to 1000-fold compared to normal cells at equivalent doses. This vulnerability arises from the inability to efficiently monoubiquitinate , leading to persistent replication fork stalling and accumulation of chromosomal aberrations. Recent studies (as of 2024) highlight how inflammation and oxidative stress exacerbate FA pathway dysfunction, creating a vicious cycle that amplifies DNA damage, with crosstalk involving disrupted mitochondrial metabolism impairing core complex assembly and upregulating pro-apoptotic signaling. These insights underscore the pathway's broader role in cellular stress responses beyond ICL repair.

Hematopoietic stem cell dysfunction

In Fanconi anemia (FA), the core defect in the FA DNA repair pathway results in the accumulation of interstrand crosslinks and other DNA lesions in hematopoietic stem cells (HSCs), which profoundly impairs their function and leads to bone marrow failure. This accumulation triggers a heightened DNA damage response, characterized by G2/M cell cycle arrest mediated by ATR and CHK1 kinases, followed by p53-dependent activation of p21, promoting apoptosis and replicative senescence in HSCs. Consequently, FA HSCs exhibit increased sensitivity to genotoxic stress, resulting in their progressive depletion and exhaustion, as evidenced by reduced colony-forming potential in vitro. FA HSCs also fail to maintain quiescence, the dormant state essential for long-term self-renewal, leading to premature entry into the cell cycle and accelerated exhaustion. In murine models, such as Fanca-deficient mice, this loss of quiescence is associated with a approximately 10-fold reduction in long-term repopulating HSC numbers compared to wild-type controls. The inability to preserve dormancy exacerbates DNA damage during replication, further compromising HSC maintenance and contributing to stem cell pool attrition over time. Cytokine dysregulation amplifies HSC dysfunction in FA, with elevated levels of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) in the bone marrow microenvironment promoting inflammation-driven apoptosis. These proinflammatory cytokines, overexpressed in FA patients, directly impair HSC survival and proliferation by activating death receptor pathways, such as TNF-α-induced caspase signaling, which synergizes with unresolved DNA damage to accelerate cell loss. The progression of HSC dysfunction in FA typically begins with early dysplastic changes in the bone marrow, evolving into hypocellular marrow and overt aplastic anemia, with approximately 90% of patients developing bone marrow failure by age 40. Recent studies (as of 2024) highlight how mitochondrial dysfunction and reactive oxygen species (ROS) overload exacerbate this process, as impaired mitochondrial metabolism in FA cells leads to excessive ROS production that further damages HSCs and promotes their senescence.

Oncogenic mechanisms

Fanconi anemia (FA) arises from defects in the DNA interstrand crosslink (ICL) repair pathway, leading to genomic instability that predisposes individuals to malignancy. Unrepaired ICLs trigger replication fork stalling and collapse, resulting in chromosomal aberrations such as radial formations and breakage, as well as loss of heterozygosity (LOH) at fragile sites. This instability fosters the accumulation of oncogenic mutations, including frequent TP53 alterations in FA-associated cancers, which impair cell cycle checkpoints and apoptosis, thereby promoting tumorigenesis. Hematologic malignancies, particularly acute myeloid leukemia (AML), represent a primary oncogenic outcome in FA, with patients facing a 500- to 1000-fold increased risk compared to the general population and a median onset age of 16 years. Myelodysplastic syndrome (MDS) often serves as a precursor, evolving from clonal hematopoiesis driven by FA pathway defects. Somatic mutations in genes such as GATA2 and RUNX1 further contribute to leukemogenesis by disrupting hematopoietic transcription and differentiation. Solid tumors also emerge as a significant risk, with a substantially increased risk of head and neck squamous cell carcinoma () following hematopoietic stem cell transplantation (), alongside gynecologic cancers in females, and a median onset age of 23 years for these malignancies. Underlying mechanisms include hyperactivity of error-prone repair pathways, such as non-homologous end joining (), which introduces deleterious mutations during ICL resolution. Additionally, ALDH2 deficiency exacerbates aldehyde-induced DNA damage, particularly from acetaldehyde, amplifying genotoxicity in FA cells. Recent studies (as of 2024) highlight the role of inflammation and the tumor microenvironment in FA oncogenesis, where chronic proinflammatory cytokine production (e.g., TNF-α, IL-1β) and oxidative stress from reactive oxygen species (ROS) create a permissive niche for malignant transformation. This interplay, involving NF-κB activation and mitochondrial dysfunction, bridges DNA repair defects to enhanced leukemogenesis and solid tumor progression.

Diagnosis

Clinical criteria

Fanconi anemia (FA) should be suspected in individuals with a family history of the disorder, related cancers such as or , or consanguinity, particularly when accompanied by early-onset bone marrow failure before age 18 years. Additional triggers include unexplained cytopenias, congenital anomalies, or excessive sensitivity to chemotherapy or radiation, which may prompt initial clinical evaluation. Key physical examination findings that raise suspicion include radial ray defects such as hypoplastic or absent thumbs and radii, skin hyperpigmentation with café-au-lait spots or generalized hyperpigmentation, short stature, and microphthalmia or other ophthalmic anomalies. Microcephaly, genitourinary malformations, and skeletal abnormalities, such as hypoplastic or absent thumbs and radii or lower limb issues like hip dysplasia, observed in approximately 50% of affected individuals, primarily involving the upper extremities, are also common hallmarks. FA is syndromically associated with VACTERL-H, encompassing vertebral defects, anal atresia, cardiac malformations, tracheoesophageal fistula, esophageal atresia, renal anomalies, limb abnormalities, and hydrocephalus, particularly in cases involving certain genetic variants. The PHENOS phenotype, featuring pigmentation changes, small head, small eyes, nervous system issues, otologic problems, and short stature, further supports clinical suspicion when multiple features are present. Presentation is often age-dependent, with congenital anomalies evident in infancy, progressive cytopenias typically emerging by school age around 7-10 years, and increased cancer risk manifesting in adolescence or early adulthood. The median age at diagnosis is 7 years, though some cases remain undetected until adulthood due to milder features. Differential diagnosis includes other inherited bone marrow failure syndromes such as , characterized by nail dystrophy and oral leukoplakia, or , featuring exocrine pancreatic insufficiency and neutropenia. Distinguishing features like specific congenital anomalies or family history guide further evaluation, often leading to confirmatory laboratory tests such as chromosome breakage analysis.

Laboratory investigations

Laboratory investigations for Fanconi anemia begin with routine hematologic evaluations to assess for progressive bone marrow failure, followed by specialized tests to confirm the diagnosis through evidence of genomic instability. A complete blood count (CBC) typically reveals trilineage cytopenias, including anemia with hemoglobin levels below 10 g/dL, thrombocytopenia with platelet counts under 50,000/μL, and neutropenia with absolute neutrophil counts less than 1,000/μL, often progressing over time from childhood. Macrocytosis is a hallmark finding, with mean corpuscular volume (MCV) exceeding 100 fL in over 90% of patients, even prior to overt aplasia, alongside low reticulocyte counts below 1% indicating impaired erythropoiesis. Bone marrow aspiration and biopsy demonstrate hypocellularity, often less than 30% cellularity, with fatty replacement, reduced myeloid and erythroid precursors, and decreased megakaryocytes; dysplasia may be present, such as erythroid multinuclearity or ringed sideroblasts, raising suspicion for myelodysplastic syndrome. Elevated fetal hemoglobin levels, greater than 1% in adults, reflect stress erythropoiesis and are commonly observed in conjunction with these marrow changes. The gold standard for diagnosis is chromosomal breakage analysis, performed on peripheral blood lymphocytes stimulated with phytohemagglutinin and exposed to DNA crosslinking agents such as diepoxybutane (DEB) at 0.1 μg/mL or mitomycin C (MMC) at 0.1 μg/mL for 72 hours. This test shows markedly increased chromosomal aberrations, including breaks, gaps, rearrangements, and especially radial figures, with typically ≥30% of cells showing radials in DEB-treated cultures indicating a positive result in non-mosaic cases; the test is highly sensitive for detecting FA, though negative results may necessitate testing skin fibroblasts to detect mosaicism. Additional supportive tests include measurement of erythrocyte adenosine deaminase (eAD) activity, which is typically normal in but elevated in about 80% of cases of , aiding differential diagnosis. These investigations, prompted by clinical suspicion of congenital anomalies or family history, provide essential evidence of the disorder's hematologic and chromosomal hallmarks.

Genetic confirmation

Genetic confirmation of Fanconi anemia (FA) involves molecular testing to identify biallelic pathogenic variants in one of the 22 known FA genes, providing a definitive diagnosis following initial screening such as chromosomal breakage analysis. Targeted next-generation sequencing (NGS) panels are the primary method, analyzing all 22 FA genes (FANCA through FANS2) for sequence variants, small insertions/deletions, and copy number variations, with detection rates exceeding 95% in confirmed cases. These panels typically achieve high analytical sensitivity (>99%) for single nucleotide variants and structural changes greater than 1 . If NGS identifies potential causative variants but complementation group assignment is unclear, functional complementation testing is performed using retroviral or lentiviral of patient-derived fibroblasts with wild-type cDNA from candidate FA genes; restoration of resistance to DNA crosslinking agents like (MMC) confirms the affected gene and group, as seen with FANCA reducing chromosomal aberrations to normal levels. Prenatal diagnosis is available for at-risk pregnancies via (typically 10-14 weeks gestation) or (15-20 weeks), involving direct mutation analysis or chromosomal breakage testing on fetal cells; postnatal confirmation follows similar molecular approaches, while carrier screening in populations with founder mutations (e.g., IVS4+4A>T in FANCA among ) uses targeted . Identified variants are interpreted according to American College of Medical Genetics and Genomics (ACMG) guidelines, requiring biallelic pathogenic or likely pathogenic variants for diagnosis; challenges arise with variants of uncertain significance (VUS), which occur in approximately 5-10% of cases and may necessitate functional assays or family segregation studies for resolution.

Management

Supportive care

Supportive care in Fanconi anemia focuses on alleviating symptoms of progressive bone marrow failure, mitigating risks, and addressing secondary complications through non-curative interventions. This approach includes transfusions, prophylactic therapies, iron management, and routine surveillance to improve while awaiting definitive treatment. Blood product support is a cornerstone for managing cytopenias. transfusions are indicated for symptomatic , typically when levels drop below 7-8 g/dL, using leukoreduced and irradiated units to reduce the risks of febrile reactions, alloimmunization, and . Platelet transfusions are provided prophylactically for counts below 10,000-20,000/µL or in cases of active bleeding, with similar precautions including avoidance of cytomegalovirus-positive products and family donors to prevent . These measures provide immediate hematologic relief but require careful monitoring to avoid . Infection prophylaxis is essential given the high susceptibility to bacterial, fungal, and opportunistic infections from . Trimethoprim-sulfamethoxazole is routinely prescribed at 5 mg/kg/day (trimethoprim component) three times weekly for prevention in patients with absolute neutrophil counts below 500/µL. Antifungal agents like (3-6 mg/kg/day) are used prophylactically during periods of profound or following intensive therapies. (G-CSF) at doses of 5 µg/kg/day subcutaneously can be administered cautiously for severe (absolute neutrophil count <200/µL) to temporarily boost counts and reduce infection rates, though its prolonged use is limited due to potential leukemogenic effects and increased risk of myelodysplastic syndrome transformation. Transfusion-related iron overload necessitates chelation therapy to prevent organ damage from hemosiderosis. Deferasirox, an oral iron chelator, is initiated when serum ferritin levels exceed 1000 ng/mL or after approximately 10-20 units of red blood cell transfusions, at a starting dose of 20-30 mg/kg/day, with adjustments based on ferritin trends and renal function monitoring. This intervention has been shown to reduce hepatic iron concentration and improve outcomes in chronically transfused patients. Androgen therapy serves as a hematopoietic stimulant for patients with moderate to severe bone marrow failure. Oxymetholone, dosed at 2-5 mg/kg/day orally, or danazol at 200-600 mg/day, promotes erythropoiesis and can stabilize hemoglobin and platelet counts in about 50% of treated individuals, often within 1-3 months of initiation. Therapy is typically started when hemoglobin is below 8 g/dL or platelets below 30,000/µL, but it requires vigilant oversight due to adverse effects including virilization, cholestatic hepatitis, peliosis hepatis, and an elevated risk of progression to myelodysplastic syndrome or hepatocellular carcinoma; liver function tests are monitored every 3 months, with hepatic ultrasounds every 6-12 months. A multidisciplinary monitoring protocol is critical to detect early complications and malignancies, given the disorder's predisposition to solid tumors. Complete blood counts are checked every 3-4 months, with annual bone marrow aspiration and biopsy recommended after age 2 to assess for cytogenetic abnormalities or myelodysplasia. Cancer surveillance includes biannual oral-dental examinations starting at age 10, annual gynecologic evaluations with Pap smears from age 16 for females, and dermatologic assessments every 6-12 months to screen for squamous cell carcinomas of the head, neck, aerodigestive tract, and anogenital regions. Human papillomavirus vaccination is strongly recommended by age 9-12 to mitigate oncogenic risks. Endocrine and nutritional evaluations are also integrated annually to address growth and metabolic issues.

Hematopoietic stem cell transplantation

Hematopoietic stem cell transplantation (HSCT) serves as the primary curative therapy for bone marrow failure in , particularly when severe aplastic anemia develops or high-risk myelodysplastic syndrome emerges. Transplantation is ideally performed before age 10 to optimize outcomes, as younger patients exhibit lower rates of transplant-related complications and improved engraftment. Prior to HSCT, supportive care such as transfusions and growth factors may stabilize patients, but it does not alter the underlying hematopoietic defect. Donor selection prioritizes an HLA-matched sibling, which yields the highest success rates with minimal graft-versus-host disease (GVHD) risk. In the absence of a matched sibling, an HLA-matched unrelated donor is the next preferred option, offering comparable outcomes when high-resolution typing is used. Since the 2010s, haploidentical donors with post-transplant cyclophosphamide (PTCy) have become viable alternatives, reducing GVHD incidence through selective T-cell depletion while promoting engraftment. Due to the inherent DNA repair deficiency in Fanconi anemia, which heightens sensitivity to alkylating agents and radiation, reduced-intensity conditioning regimens are standard to minimize toxicity. A typical protocol includes low-dose cyclophosphamide (20 mg/kg total), fludarabine (typically 120-150 mg/m²), and low-dose total body irradiation (2-3 Gy), often combined with antithymocyte globulin for GVHD prophylaxis. This approach achieves sufficient immunosuppression for donor engraftment without excessive regimen-related mortality. With matched donors, overall survival reaches 80-90% in pediatric patients, reflecting advances in supportive measures and refined protocols. Graft failure occurs in 10-20% of cases overall, though rates are lower (under 10%) with matched siblings and modern conditioning. Acute GVHD affects 20-30% of recipients, with chronic GVHD rarer when T-cell depletion is employed. Following HSCT, immunosuppression is gradually tapered over months to years to prevent GVHD while monitoring for infections and chimerism. Despite successful hematopoietic reconstitution, the risk of solid tumors persists lifelong due to the underlying genetic instability, necessitating vigilant surveillance for head and neck, skin, and gynecologic cancers.

Emerging therapies

Gene therapy approaches for Fanconi anemia primarily target the most common subtype, FANCA, by introducing a functional copy of the gene into patient-derived CD34+ hematopoietic stem cells using lentiviral vectors. In the phase 1/2 FANCOLEN-1 trial (recruitment 2016-2019), gene therapy was administered without genotoxic conditioning; among 8 evaluable patients, 5 (62.5%) met the primary endpoint of sustained engraftment (≥0.1 vector copies per nucleated cell at year 2), with follow-up up to 7 years showing hematopoietic reconstitution and reversal of bone marrow failure in responders. In the global RP-L102 program, updated results as of 2024 from 14 treated patients (aged 1-6 years) showed genetic and phenotypic correction with improved blood counts and reduced transfusion dependence in several cases; however, as of October 2025, Rocket Pharmaceuticals paused further investment in the program following withdrawal of the FDA Biologics License Application. CRISPR-Cas9-based gene editing represents a precise alternative to viral addition, focusing on correcting specific mutations in Fanconi anemia genes, particularly nonsense mutations like FANCA c.190T>C that account for a subset of cases. Preclinical studies have achieved efficient correction of these mutations in patient-derived hematopoietic stem cells using base and techniques, restoring DNA repair function and improving cell survival under genotoxic stress. As of 2025, these approaches remain in , with in vivo editing demonstrated in models harboring FA mutations. Translational read-through agents, such as and gentamicin, aim to suppress premature stop codons prevalent in about 20% of Fanconi anemia mutations, allowing production of full-length proteins. and studies published in 2025 show that induces in FANCA-, FANCC-, and FANCF-mutant lines, resulting in a 1.4- to 2.1-fold increase in viability upon exposure to DNA crosslinking agents like and a reduction in micronuclei formation indicative of genomic instability. Induced pluripotent stem cell (iPSC)-derived hematopoietic stem cells offer a potential autologous transplant strategy by patient fibroblasts, correcting the genetic defect, and into functional blood progenitors. Early-stage in 2024 has generated corrected iPSCs from Fanconi anemia patients, with models demonstrating successful engraftment of derivative hematopoietic cells and restoration of multilineage blood production without tumorigenicity. These proof-of-concept studies highlight the feasibility for clinical translation, though challenges in scalable and immune compatibility persist. Small-molecule inhibitors targeting the ATR pathway, such as berzosertib, seek to alleviate replication inherent to Fanconi anemia's deficiency, potentially preventing failure progression and . Preclinical shows berzosertib enhances of Fanconi anemia cells under by modulating checkpoint responses, while related Chk1 inhibitors like SRA737 (ceralasertib) are in phase I trials for associated cancers, exploiting in FA-deficient tumors.

Prognosis

Survival rates

Survival rates for individuals with Fanconi anemia have improved significantly over time due to advances in supportive care and (HSCT). Historically, prior to 2000, the survival was approximately 20 years, primarily limited by failure and malignancies; by 2025, cohort studies report a overall survival of 29 to 37 years, reflecting better early and strategies. With supportive care but without HSCT, median survival is approximately 29-30 years as of 2025, with patients achieving approximately 70-80% survival to age 20, though long-term deteriorates due to progressive failure and cancer risks. In contrast, HSCT substantially enhances outcomes, with 5-year overall survival rates reaching 83-85% in pediatric cohorts using reduced-intensity conditioning, compared to about 50% with alone. Cause-specific mortality in Fanconi anemia cohorts highlights failure as the leading contributor, followed by malignancies (including and solid tumors) and infections. Data from the Fanconi Anemia Research Fund registry and related cohorts indicate that about 70% of patients remain alive at 30 years post-HSCT, underscoring the procedure's role in mitigating hematologic progression. Recent trends from 2024-2025 show further gains, with transplant-related mortality declining to 14.5% post-2010 due to optimized regimens and earlier interventions, potentially extending median survival with HSCT to 30-40 years. Emerging therapies, such as gene correction, may further improve these rates by addressing underlying defects.

Long-term complications

Survivors of (FA) who undergo (HSCT) face significant long-term risks, including chronic (GVHD), which occurs in approximately 35% of patients within two years post-transplant. This condition arises due to the of donor cells against the recipient's tissues and is exacerbated in FA by the underlying defect, leading to prolonged and organ damage. is also prevalent, affecting up to 70-80% of evaluable patients, primarily from the gonadotoxic effects of conditioning regimens involving alkylating agents or total body irradiation (TBI). In females, ovarian failure is common, with only rare spontaneous pregnancies reported post-HSCT. Secondary malignancies represent a major threat, with head and neck squamous cell carcinoma (HNSCC) showing a cumulative incidence of 14% by age 40 in FA patients overall, rising to 25% by age 50, and further elevated post-HSCT due to chronic GVHD and prior exposures. This heightened susceptibility stems from FA's impaired , making cells vulnerable to oncogenic mutations from environmental carcinogens like and HPV. In post-HSCT cohorts, s emerge in about 8% at 10 years and 14% at 15 years, often with poor prognosis. Endocrine dysfunction is widespread in FA survivors, independent of but compounded by HSCT. Hypothyroidism affects 30-60% of children and 37-57% of adults, often requiring lifelong replacement, and is linked to both the disease and TBI-containing regimens. Diabetes mellitus develops in 5-10% of patients, while impaired glucose tolerance impacts up to 68%, contributing to risks. Osteoporosis and are prevalent, occurring in over 90% of adults, exacerbated by use for GVHD, therapy for anemia, and . Neurologic and cognitive complications, though less dominant than physical manifestations, affect a notable subset of FA survivors. Learning disabilities and developmental delays occur in approximately 40% of patients, often manifesting as attention deficits, processing delays, or academic challenges, potentially worsened by chronic illness burdens or subtle neuroinflammatory processes. A progressive neuroinflammatory known as Fanconi anemia neurological syndrome (FANS) has been identified in some pediatric and patients as of 2024, involving lesions and symptoms such as seizures, motor , and cognitive deficits. Heightened sensitivity to HPV further elevates HNSCC risk in the head and neck region, underscoring the need for vigilant oral surveillance. Neurological dysfunction, including or seizures, is reported in about 16% post-HSCT. Quality of life in FA survivors is profoundly impacted by persistent , stemming from ongoing endocrine imbalances, prior , or treatment sequelae, alongside growth impairment affecting over 50% with . Approximately 50% of patients require lifelong multidisciplinary care involving endocrinologists, oncologists, and psychologists to manage these chronic burdens. Recent 2025 cohort analyses indicate that enhanced solid tumor surveillance protocols have reduced mortality from malignancies by up to 15% through early detection and intervention.

References

  1. [1]
    Fanconi anemia: MedlinePlus Genetics
    ### Summary of Fanconi Anemia
  2. [2]
    Fanconi Anemia - GeneReviews® - NCBI Bookshelf
    Feb 14, 2002 · Fanconi anemia (FA) is characterized by physical abnormalities, bone marrow failure, and increased risk for malignancy.
  3. [3]
    Study Maps Clinical Evolution of Fanconi Anemia Using Large ...
    Feb 26, 2025 · The results show that patients diagnosed with FA have a cumulative cancer incidence of 86% by the age of 50, including all solid tumor and blood ...Missing: prevalence | Show results with:prevalence<|separator|>
  4. [4]
    Fanconi Anemia - StatPearls - NCBI Bookshelf
    Fanconi anemia is a rare genetic disorder affecting bone marrow, characterized by impaired DNA repair. Clinicians should recognize physical anomalies, ...
  5. [5]
    The concept and practice of Fanconi Anemia: from the clinical ...
    Jul 2, 2013 · FA was first described and named as a disease in 1927 by the Swiss pediatrician-Guido Fanconi, 3 brothers with a specific combination of bone ...
  6. [6]
    Guido Fanconi (1892-1979): a jack of all trades - PubMed
    In 1927 the Swiss paediatrician Guido Fanconi described a family in which three boys had physical birth defects and died of a condition that resembled ...
  7. [7]
    Fanconi Anemia: Background, Etiology, Epidemiology
    Apr 22, 2025 · Fanconi anemia accounts for approximately 25% of the cases of aplastic anemia seen at large referral centers. Approximately 25% of known ...Background · Etiology · Epidemiology · Prognosis
  8. [8]
    Evidence for at least four Fanconi anaemia genes including FACC ...
    Jun 1, 1992 · These results suggest that mutations in at least four different genes lead to FA, a degree of genetic heterogeneity comparable to that of other DNA repair ...
  9. [9]
    Cytogenetics in Fanconi Anemia: The Importance of Follow-Up and ...
    The recognition of FA as a chromosomal instability disorder was made by the pioneer observations of Schroeder in 1964, where he noticed that lymphocytes of ...
  10. [10]
    Chromosome fragility in patients with Fanconi anaemia - PubMed
    Jan 7, 2011 · This study proposes a new chromosome fragility index and suggests that genome instability during embryo development may be related to malformations in FA.Missing: discovery 1960s
  11. [11]
    The Genetic Basis of Fanconi Anemia - NCBI - NIH
    Seventy-five years ago, Dr. Guido Fanconi reported three siblings who exhibited both congenital defects and aplastic anemia.
  12. [12]
    Association of biallelic BRCA2/FANCD1 mutations with ...
    Apr 1, 2004 · Fanconi anemia diagnosis and the diepoxybutane (DEB) test [editorial]. ... Spontaneous chromosomal breakage and high incidence of leukemia ...
  13. [13]
    The Fanconi Anemia and Breast Cancer Susceptibility Pathways - NIH
    In families carrying a BRCA2/FANCD1 mutation, the peak age of onset of breast or ovarian cancer among heterozygote carriers is in the fourth decade 89, whereas ...
  14. [14]
    An update on Fanconi anemia: Clinical, cytogenetic and molecular ...
    FA has an incidence of 1 in 300,000 live births and a prevalence of 1-9 per million (4). ... In general, the male:female ratio of the presentation of the disease ...
  15. [15]
    [PDF] Fanconi anemia (FA), named for the Swiss pediatrician Guido ...
    Fanconi anemia (FA), named for the Swiss pediatrician Guido Fanconi, is an inherited disorder that can lead to bone marrow failure (aplastic anemia), leukemia ...
  16. [16]
  17. [17]
    A common founder mutation in FANCA underlies the world's highest ...
    A common founder mutation in FANCA underlies the world's highest prevalence of Fanconi anemia in Gypsy families from Spain. Blood. 2005 Mar 1;105(5):1946-9.
  18. [18]
    Molecular and genealogical evidence for a founder effect in Fanconi ...
    We then identified the FANCA gene by both functional complementation and positional cloning (2, 3). We have now screened this gene for mutations in ...
  19. [19]
    Diagnosis of Fanconi Anemia: Mutation Analysis by Multiplex ... - NIH
    FANCA mutations were found in 31 of the patients (57%), FANCC mutations in ... mutation of the Fanconi anemia gene FAC in the Ashkenazi Jewish population.Missing: rates | Show results with:rates
  20. [20]
    Fanconi anemia in Ashkenazi Jews - PubMed - NIH
    Fanconi anemia (FA) should be included among the genetic diseases that occur at high frequency in the Ashkenazi Jewish population.
  21. [21]
    Fanconi anemia associated with moyamoya disease in Saudi Arabia
    As FA is highly prevalent in Saudi Arabia, mainly due to a high rate of consanguinity, and inherited marriage as a result, more than 120 allogenic SCT have ...Missing: prevalence | Show results with:prevalence
  22. [22]
    Clinical characteristics and genetic subtypes of Fanconi anemia in ...
    FA is the most frequent indication for bone marrow transplantation among patients with inherited bone marrow failure syndromes in Saudi Arabia (4). However, the ...
  23. [23]
    The Ashkenazi Jewish carrier screening panel: evolution, status quo ...
    Jul 3, 2014 · Prenatal/preconception screening for Tay-Sachs disease in the Ashkenazi Jewish (AJ) population has reduced the incidence of these diseases by 90 ...
  24. [24]
    Carrier screening in individuals of Ashkenazi Jewish descent - PMC
    The overall carrier rate in this population is significant, with between 1 in 4 and 1 in 5 Ashkenazi Jews carrying a mutation for any one of these disorders.
  25. [25]
    Patterns and frequency of renal abnormalities in Fanconi anaemia
    Apr 12, 2018 · Our analysis identifies a prevalence of 50%, confirming the high frequency of earlier observations in what is, to our knowledge, the largest ...
  26. [26]
    Treatment of the bone marrow failure in Fanconi anemia patients ...
    Feb 15, 2012 · More than 90% of Fanconi anemia (FA) patients experience progressive bone marrow failure during life with a median onset at 8 years of age.
  27. [27]
    Fanconi Anemia: a DNA Repair Disorder Characterized by ...
    Fanconi Anemia (FA) is a rare autosomal genetic disorder characterized by progressive bone marrow failure (BMF), endocrine dysfunction, cancer, and other ...
  28. [28]
    Hematologic abnormalities in Fanconi anemia - PubMed - NIH
    Of those, 332 developed hematologic abnormalities at a median age of 7 years (range, birth to 31 years).Missing: incidence | Show results with:incidence
  29. [29]
    Reticulocytopenia (Concept Id: C0858867) - NCBI
    Fanconi anemia (FA) is characterized by physical abnormalities, bone marrow failure, and increased risk for malignancy. Physical abnormalities, present in ...
  30. [30]
    An experience with 124 cases of fanconi anemia: clinical spectrum ...
    Fanconi anemia is an inherited bone marrow failure syndrome characterized by somatic abnormalities and an increased predisposition to malignancies.
  31. [31]
    Diagnosis of Myelodysplastic Syndrome Among a Cohort of 119 ...
    Analysis of the data for 754 patients from International Fanconi Anemia Registry demonstrated a cumulative incidence of 90% for BMF, 33% for hematologic ...
  32. [32]
    Leukemia and chromosomal instability in aged Fancc−/− mice - PMC
    The overall risk of leukemia in FA is increased 600-fold. Young mice ... acute myeloid leukemia in patients with Fanconi anemia. Cancer Genet ...
  33. [33]
    Endocrine Disorders in Fanconi Anemia - PubMed Central - NIH
    About 80% of children and adults with FA have at least one endocrine abnormality, including short stature, GH deficiency, abnormal glucose or insulin metabolism ...
  34. [34]
    Hearing loss and speech perception in noise difficulties in Fanconi ...
    Conductive hearing loss was present in 24.1%, sensorineural in 20.7%, and mixed in 10.3%.
  35. [35]
  36. [36]
    Oesophageal atresia, tracheo‐oesophageal fistula, and the ...
    Gastro‐intestinal atresias have been reported in 14% of cases of Fanconi anaemia. Other malformations falling within the VACTERL spectrum also occur: skeletal ( ...
  37. [37]
    Impaired immune function in children with Fanconi anaemia - PMC
    Our data provide evidence of immune dysfunction in a significant number of patients with Fanconi anaemia, particularly involving NK cells. Further understanding ...Missing: dysregulation | Show results with:dysregulation
  38. [38]
    Liver abnormalities are frequent and persistent in patients with ... - NIH
    Our data show that liver disease is common in patients with FA, and because most children with FA now reach adulthood, end-stage liver disease in young ...Missing: prevalence | Show results with:prevalence
  39. [39]
    Fanconi anemia - Orphanet
    Fanconi anemia ; Prevalence: 1-9 / 1 000 000 ; Inheritance: Autosomal recessive, X-linked recessive ; Age of onset: Childhood.
  40. [40]
    How high are carrier frequencies of rare recessive syndromes ...
    Jul 7, 2011 · One such syndrome is Fanconi Anemia (FA). FA is a model disease in cancer research, yet there are no contemporary data on carrier frequency or ...
  41. [41]
    The FANCA Gene in Japanese Fanconi Anemia - PubMed
    This report describes mutations of the FANCA gene, which we studied by direct sequencing of cDNA with confirmation on genomic DNA in 15 unclassified Japanese FA ...Missing: IVS38- 6G>
  42. [42]
    Development of translational read-through-inducing drugs for ...
    Development of translational read-through-inducing drugs for Fanconi Anemia caused by nonsense mutations ... nonsense mutations (20-30% of individuals with FA).
  43. [43]
    The Fanconi anemia pathway and DNA interstrand cross-link repair
    Here, we review our current understanding of how these FA proteins participate in ICL repair and discuss the molecular mechanisms that regulate the FA pathway ...Missing: FANCB | Show results with:FANCB
  44. [44]
    Fanconi Anemia Proteins, DNA Interstrand Crosslink Repair ...
    Here we will review current research directed at identifying FA genes and understanding the function of FA proteins in DNA damage responses.Fa-Brca Pathway: Components... · Fa Nuclear Core Complex · Fancd2, Fanci, Brca2/fancd1...<|control11|><|separator|>
  45. [45]
    Fanconi Anemia Pathway - Reactome
    FA core complex assembles at DNA interstrand crosslinks (ICLs) (Homo sapiens) · FANCD2 binds FANCI (Homo sapiens) · FANCD2:FANCI complex and UBE2T bind ICL-DNA ...
  46. [46]
    Structural insight into FANCI–FANCD2 monoubiquitination
    Jul 29, 2020 · The Fanconi anemia (FA) pathway coordinates a faithful repair mechanism for DNA damage that blocks DNA replication, such as interstrand ...
  47. [47]
    Expanded roles of the Fanconi anemia pathway in preserving ...
    A central event in the pathway is the monoubiquitination of FANCD2 and FANCI upon DNA damage, which is mediated by a group of upstream FA proteins (FANCA, FANCB ...
  48. [48]
    The Fanconi Anemia Pathway in Cancer - PMC - PubMed Central
    Fanconi anemia (FA) is a complex genetic disorder characterized by bone marrow failure (BMF), congenital defects, inability to repair DNA interstrand ...The Fanconi Anemia Pathway... · 3.1. Dna Resection And The... · 5.1. Germline Monoallelic Fa...
  49. [49]
    Fanconi Anemia Pathway: Mechanisms of Breast Cancer ... - Frontiers
    Apr 1, 2020 · Truncating mutations in the Fanconi anemia J gene BRIP1 are low-penetrance breast cancer susceptibility alleles. Nat. Genet. 38, 1239–1241 ...Missing: paper | Show results with:paper
  50. [50]
    Defects in homologous recombination repair behind the human ...
    Hereditary breast and ovarian cancer (HBOC) syndrome and a rare childhood disorder Fanconi anemia (FA) are caused by homologous recombination (HR) defects.Genome Integrity Is... · The Nucleases In The Fa... · The Core Hr Genes In The Fa...
  51. [51]
    Regulation of DNA cross-link repair by the Fanconi anemia/BRCA ...
    Accordingly, FA patient-derived cells are hypersensitive to DNA cross-linking agents, such as mitomycin C (MMC) or diepoxybutane (DEB), with a dramatic increase ...
  52. [52]
    New Insights into the Fanconi Anemia Pathogenesis - PubMed Central
    Oct 29, 2024 · Recent findings indicate the interplay of inflammation, oxidative stress, disrupted mitochondrial metabolism, and impaired intracellular signaling in the FA ...
  53. [53]
    Stress and DNA repair biology of the Fanconi anemia pathway
    The FANCF structure suggests a flexible protein that links the FANCC-FANCE and FANCA-FANCG subcomplexes and mediates recruitment of the entire complex to sites ...
  54. [54]
    Bone marrow failure in Fanconi anemia is triggered by an ...
    Bone marrow failure in Fanconi anemia is triggered by an exacerbated p53/p21 DNA damage response that impairs hematopoietic stem and progenitor cells.
  55. [55]
    Fanconi anemia cells with unrepaired DNA damage activate ...
    Sep 18, 2015 · Upon DNA damage, normal and FA cells inhibit the cell cycle progression, until the G2/M checkpoint is turned off by the checkpoint recovery, ...
  56. [56]
    Cell-Cycle-Specific Function of p53 in Fanconi Anemia ... - NIH
    Jan 4, 2018 · Given that loss of p53 alone reduced quiescence without causing significant HSC exhaustion, we postulated that the FA proteins cooperate ...Results · Loss Of P53 In Fanca Mice... · P53 Deficiency Leads To...
  57. [57]
    Studies in an Early Development Window Unveils a Severe HSC ...
    Oct 25, 2018 · Keywords: HSC, Fanconi anemia, mouse embryonic development, human ... LTC-IC assays showed that the LTC-IC frequency was 10-fold lower ...
  58. [58]
    Fancb deficiency impairs hematopoietic stem cell function - Nature
    Dec 11, 2015 · Indeed, we found that loss of Fancb protein compromises HSC repopulation ability and increases proportion of cycling cells in the SLAM ...
  59. [59]
    Disrupted Signaling through the Fanconi Anemia Pathway Leads to ...
    Macrocytosis (enlargement of red blood cells) is usually the first to be detected, followed by thrombocytopenia and aplastic anemia (insufficient production ...<|control11|><|separator|>
  60. [60]
    TNF-α signaling in Fanconi anemia - PMC - PubMed Central - NIH
    The patients with FA are often found overproducing TNF-α, which may directly affect hematopoietic stem cell (HSC) function by impairing HSC survival, homing and ...
  61. [61]
    Fanconi Anemia and its Diagnosis - PMC - PubMed Central
    Fanconi anemia (FA) is a genetically and phenotypically heterogeneous recessive disorder characterized by diverse congenital malformations, progressive ...Fanconi Anemia And Its... · Major Congenital... · Diagnostic Tests
  62. [62]
    Fanconi Anemia and the Underlying Causes of Genomic Instability
    Fanconi anemia (FA) is a rare genetic disorder, characterized by birth defects, progressive bone marrow failure, and a predisposition to cancer.Fanconi Anemia And The... · 2.2. Fancd2... · 5. Dna-Protein Cross-Link...Missing: TP53 | Show results with:TP53
  63. [63]
    Fanconi Anemia Signaling and Cancer - PMC - NIH
    Here, we review how aberrant function of the twenty-two FA genes and their signaling network contributes to malignancy.
  64. [64]
    Secondary leukemia in patients with germline transcription factor ...
    Mutations in ASXL1 are the most frequent concurrent somatic mutation in GATA2-associated MDS/AML, with NRAS/KRAS, WT1, STAG2, and SETBP1 among the other ...
  65. [65]
    Head and Neck Cancer in Patients with Fanconi Anemia
    Head and neck squamous cell carcinoma (HNSCC) is the most common solid tumor in patients with FA. The incidence of HNSCC in FA patients is 500- to 700-fold ...
  66. [66]
    Aldehyde Dehydrogenase 2 in Aplastic Anemia, Fanconi Anemia ...
    This review discusses the unexpected interrelationship between aldehydes, ALDH2 and hematopoietic stem cell biology, and in particular its relationship to ...
  67. [67]
    [PDF] Fanconi Anemia Clinical Care Guidelines_5th Edition 2020_web
    The Fanconi Anemia Research Fund (FARF) is the world leader in advancing research for better treatments and a cure for Fanconi anemia (FA). Founded in.
  68. [68]
    Fanconi Anemia Workup: Approach Considerations, CBC Count ...
    Apr 22, 2025 · The diagnosis of Fanconi anemia is not made using routine laboratory tests; it must be tested for using chromosome breakage in blood or fibroblasts, or ...Missing: criteria | Show results with:criteria
  69. [69]
    [PDF] Hematologic Abnormalities in Patients with Fanconi Anemia
    Chang YH, Shaw CF, Wu KH, Hsieh KH, Su YN, Lu PJ (2009) Treatment with deferiprone for iron overload alleviates bone marrow failure in a. Fanconi anemia patient ...
  70. [70]
    Diagnosis of Fanconi Anemia: Chromosomal Breakage Analysis - NIH
    Cells derived from FA patients are—by definition—hypersensitive to DNA cross-linking agents, such as mitomycin C, diepoxybutane, or cisplatinum, which becomes ...
  71. [71]
  72. [72]
    Diagnosis of Fanconi Anemia: Testing and Genetic Counseling
    This chapter describes the importance of early diagnosis and the role of genetic counseling and cytogenetic and molecular tests used to diagnose FA.
  73. [73]
    Comprehensive laboratory diagnosis of Fanconi anaemia
    Background Fanconi anaemia (FA) is a rare inherited bone marrow failure disease caused by germline pathogenic variants in any of the 22 genes involved in ...
  74. [74]
    Characterization and genotype-phenotype correlation of patients ...
    Jul 1, 2020 · A common founder mutation in FANCA underlies the ... Fanconi anaemia group A (FANCA) mutations in Israeli non-Ashkenazi Jewish patients.
  75. [75]
    Fanconi Anemia Panel - Prevention Genetics
    Genome platform: PCR-free next-generation sequencing (NGS) is used to sequence the coding regions of nearly all genes as well as intronic and intergenic regions ...
  76. [76]
    High Sensitivity and Specificity of Retroviral Complementation ...
    Nov 16, 2004 · Fanconi anemia (FA) is a heterogeneous autosomal recessive DNA repair disorder with at least 11 complementation groups (ABCD1D2EFGIJL) ...
  77. [77]
    Fanconi anemia: prenatal diagnosis in 30 fetuses at risk - PubMed
    We report our experience, since 1978, with prenatal diagnosis in fetuses at risk for Fanconi anemia. Amniotic fluid cells from 30 fetuses from 24 families ...Missing: amniocentesis analysis carrier founder mutations
  78. [78]
    Specifications of the ACMG/AMP variant curation guidelines ... - NIH
    Sep 17, 2025 · Fanconi Anemia (FA) of any subtype is generally considered an exceedingly rare, severe, early-onset disease with variable features. In the case ...
  79. [79]
    The Fanconi anemia pathway induces chromothripsis and ecDNA ...
    Oct 17, 2024 · FA-pathway-induced chromothripsis generates complex genomic rearrangements and extrachromosomal DNA that confer acquired resistance to anti-cancer therapies.
  80. [80]
  81. [81]
    Management of Fanconi anemia beyond childhood - PMC - NIH
    Dec 8, 2023 · Fanconi anemia (FA) is an inherited bone marrow failure (BMF) disorder caused by pathogenic variants in 1 of 23 genes that result in defective ...Introduction · Table 1 · Hematologic Malignancy...
  82. [82]
    Predictors of outcomes in hematopoietic cell transplantation for ...
    Oct 17, 2023 · Allogeneic hematopoietic cell transplantation (HCT) remains the only cure for the hematologic manifestations of Fanconi anemia (FA).
  83. [83]
    HEMATOPOIETIC CELL TRANSPLANTATION IN FANCONI ANEMIA
    While an healthy HLA matched sibling donor certainly remains the donor of choice, and an HLA matched unrelated volunteer donor is the next best alternative ...2.0 Stem Cell Source · 2.1 Hla Matched Sibling Bm /... · 3.0 Allohct And Fa...
  84. [84]
    Outcomes of hematopoietic stem cell transplantation in 813 pediatric ...
    Sep 19, 2024 · Allogeneic hematopoietic stem cell transplantation (HSCT) is the only established curative option for Fanconi anemia (FA)–associated bone ...Key Points · Introduction · Results · Discussion
  85. [85]
    Improving Outcomes with Haploidentical Stem Cell Transplantation ...
    Sep 28, 2023 · Haplo-identical SCT with post-transplant cyclophosphamide is a reasonable option for children who do not have a matched sibling donor.
  86. [86]
    Bone Marrow Transplantation for Fanconi Anemia Using ...
    Aker, et al. Fludarabine-based reduced intensity conditioning for stem cell transplantation of Fanconi anemia patients from fully matched related and unrelated ...
  87. [87]
    Radiation-free, alternative-donor HCT for Fanconi anemia patients
    Survival was superior in younger patients (OS 92.3% in <10 years vs 63.2% ≥10 years; P = .02) (Figure 4A). Patients transplanted for marrow failure fared better ...
  88. [88]
    [Advancements in CRISPR-Cas9 for Fanconi anemia] - PubMed
    Mar 14, 2025 · This article reviews the latest research progress of CRISPR technology in FA treatment and analyzes the advantages and disadvantages of this technology in FA ...Missing: emerging | Show results with:emerging
  89. [89]
    Development of translational read-through-inducing drugs as novel ...
    Jun 21, 2025 · Fanconi anemia (FA) is a genetically diverse disorder caused by mutations in at least 22 identified Fanconi Anemia Complementation group (FANC) ...
  90. [90]
    Development of translational read-through-inducing drugs as novel ...
    Jun 21, 2025 · Ataluren showed a 2.1-fold increase in cell viability upon genotoxic challenge (MMC) in FANCF-mutant cells, and a 1.4-fold increase in FANCA ...
  91. [91]
    Inducible pluripotent stem cell models to study bone marrow failure ...
    Nov 2, 2024 · This review synthesizes recent advances in iPSC-based disease modeling for various inherited BMF/MDS disorders, including Fanconi anemia, ...
  92. [92]
    Inducible pluripotent stem cell models to study bone marrow failure ...
    This review synthesizes recent advances in iPSC-based disease modeling for various inherited BMF/MDS disorders, including Fanconi anemia, dyskeratosis congenita ...
  93. [93]
    The enrichment of Fanconi anemia/homologous recombination ...
    Dec 1, 2023 · For example, Berzosertib is a highly potent and first-in-class ATR inhibitor that has demonstrated promising results during phases I and/or II ...
  94. [94]
    Fanconi Anemia Pipeline 2025: MOA, ROA, and Clinical Trial ...
    Jul 10, 2025 · Emerging Fanconi Anemia therapies in the different phases of clinical trials are- FP-045, RP-L102, pembrolizumab, Abatacept, ICL670, SRA737, and ...<|separator|>
  95. [95]
    Long‐Term Outcome of Fanconi Anemia Patients From the Italian ...
    Jun 6, 2025 · The cumulative overall survival from birth was 91.0% at age 10 years, 71.6% at age 20, and 47.4% at age 30 years; the median survival age was ...
  96. [96]
    Outcomes of hematopoietic stem cell transplantation in 813 pediatric ...
    Sep 19, 2024 · Excellent survival and low incidence of graft failure was obtained in children with FA given HSCT using MFD and MUD.
  97. [97]
    HSCT Is Effective in Pediatric Patients With Fanconi Anemia
    Oct 17, 2024 · The overall survival (OS) was 87% at 1 year and 83% at 5 years. The event-free survival (EFS) was 82% and 78% at 1 and 5 years, respectively.
  98. [98]
  99. [99]
    Late effects in patients with Fanconi anemia following allogeneic ...
    In a series of 120 patients with FA, 48% of patients who had not undergone HSCT and 53% of patients who had undergone transplant were found to have ...Missing: fibrosis | Show results with:fibrosis
  100. [100]
    High Incidence of Head and Neck Squamous Cell Carcinoma in ...
    Results Of the 754 patients in the International Fanconi Anemia Registry, 19 (3%) had HNSCC. This is a significantly higher incidence of HNSCC compared with ...
  101. [101]
    Head and Neck Cancer in Fanconi Anemia: Clinical Challenges and ...
    Fanconi anemia is an inherited bone marrow failure syndrome with an increased incidence of cancers due to DNA repair deficiency. Head and neck cancer is the ...
  102. [102]
    Endocrine Disorders in Patients with Fanconi Anemia
    Patients with FA experience endocrine disorders in myriad ways, including short stature, challenges with weight, abnormal glucose and insulin metabolism, ...
  103. [103]
    Endocrine Abnormalities in Patients with Fanconi Anemia
    Many of the endocrine disorders to which FA patients are susceptible (e.g. GHD, hypogonadism, glucose intolerance, hypothyroidism, dyslipidemia, osteopenia) ...
  104. [104]
    [PDF] Fanconi Anemia
    Fanconi anemia is commonly associated with a range of neurological and developmental issues characterized by mild to significant impairment, such as attention ...
  105. [105]
    A Deadly Duet: Fanconi Anemia (FA) With Head and Neck Cancer
    Oct 26, 2025 · FA patients are 500 times more likely than the general population to acquire solid and hematological malignancies such as head and neck squamous ...
  106. [106]
    Late Effects Following Hematopoietic Stem Cell Transplantation ...
    Feb 28, 2025 · This study was planned to evaluate long-term post-transplant complications in patients who underwent transplantation with the diagnosis of Fanconi anemia (FA) ...
  107. [107]
  108. [108]
    Long‐Term Outcome of Fanconi Anemia Patients From the Italian ...
    Jun 6, 2025 · We analyzed 193 Fanconi anemia patients from the Italian Registry, focusing on hematological outcome, cancer risk, and mortality, ...4 Results · 4.5 Solid Tumors And... · 5 Discussion<|control11|><|separator|>