Fanconi Anemia

Earn CME/CE in your profession:


Continuing Education Activity

Fanconi anemia is the most common cause of inherited bone marrow failure due to a rare autosomal recessive genetic disorder involving all 3 blood cell lines in which homozygous or heterozygous mutations result in pathogenic alleles, including point mutations, duplications, splicing defects, and deletions. These genetic mutations of Fanconi anemia genes cause an accruement of chromosomal damage due to the cell's inability to conduct repairs. These genetic mutations in the Fanconi anemia pathway lead to cells that cannot properly repair DNA damage, resulting in genomic instability, subsequent pancytopenia, and an increased susceptibility to cytotoxic agents, ultraviolet radiation, spontaneous deformation, and predisposition to malignancies. Additionally, Fanconi anemia affects almost all organs of the body.

Fanconi anemia is also thought of as an inherited form of aplastic anemia. The disorder may also predispose patients to the development of hematologic and solid tumors. A Fanconi anemia diagnosis is typically confirmed with a chromosomal fragility test, which remains the gold standard. Management of Fanconi anemia primarily includes supportive therapy, hematopoeitic stem cell transplantation, and androgen treatment. This activity for healthcare professionals aims to enhance learners' competence in selecting appropriate diagnostic tests, managing Fanconi anemia, and fostering effective interprofessional teamwork to improve outcomes.

Objectives:

  • Identify the etiology of Fanconi anemia and what is to be expected in terms of disease progression.

  • Determine the appropriate evaluation of Fanconi anemia, including special genetic testing and the advantages and shortcomings of available treatments.

  • Compare the management options available for Fanconi anemia with special attention to the age groups involved.

  • Determine the best interprofessional team strategies for improving care coordination to improve patient outcomes.

Introduction

Fanconi anemia is the most common cause of inherited bone marrow failure due to a rare autosomal recessive genetic disorder involving all 3 blood cell lines in which homozygous or heterozygous mutations result in pathogenic alleles, including point mutations, duplications, splicing defects, and deletions.[1] These genetic mutations of Fanconi anemia genes cause an accruement of chromosomal damage due to the cell's inability to conduct repairs.[2][3] Typically, Fanconi anemia proteins maintain genomic integrity and replicative capacity through DNA interstrand crosslinks (ICLs) repair. ICLs prevent DNA strand separation and maintain DNA integrity. These genetic mutations in the Fanconi anemia pathway lead to cells that cannot properly repair DNA damage, resulting in genomic instability, subsequent pancytopenia, and an increased susceptibility to cytotoxic agents, ultraviolet radiation, spontaneous deformation, and predisposition to malignancies. Additionally, Fanconi anemia affects almost all organs of the body.[4]

Fanconi anemia is also thought of as an inherited form of aplastic anemia. Extensive studies of other bone marrow failure syndromes and chromosome fragility diseases have enhanced the scientific understanding of bone marrow failure in Fanconi anemia. Common clinical symptoms of Fanconi anemia include shortness of breath, chest pain, dizziness, and fatigue. Additionally, a clinical history of epistaxis, petechiae, and excessive bleeding from a wound site is common due to thrombocytopenia. The condition is mainly associated with other congenital deformities and is usually more common during childhood, with the average age of diagnosis being 7 years. Structural extremity abnormalities are more commonly observed on physical exams in patients with Fanconi anemia. The disorder may also predispose patients to the development of hematologic and solid tumors. Pancytopenia characteristic of Fanconi anemia is usually evident in serum laboratory studies demonstrating a decrease in all 3 blood cell lines, including red blood cells (RBCs), platelets, and leukocytes. 

Fanconi anemia should be evaluated in patients presenting with signs and symptoms of pancytopenia with or without characteristic malformations and in patients with a family history of bone marrow failure or unbalanced translocations identified during a diagnostic leukemia evaluation. Furthermore, Fanconi anemia should be considered in patients with early-onset tumors or excessive toxicity after standard-dose chemotherapy. A Fanconi anemia diagnosis is typically confirmed with a chromosomal fragility test, which remains the gold standard. Management of Fanconi anemia primarily includes supportive therapy, hematopoeitic stem cell transplantation, and androgen treatment.[5] 

Etiology

Fanconi anemia is an inherited autosomal recessive disease, but about 2% is inherited in an X-linked recessive manner, with either homozygous or heterozygous mutations. DNA sequencing has identified pathogenic alleles, including point mutations, duplications, splicing defects, and deletions.[1] Mutation of Fanconi anemia genes results in impaired double-stranded DNA repair. More than 23 Fanconi anemia complementation genes (FANC) have been recognized, and all of them are involved in the DNA repair pathway.[6] Most of them are autosomal recessive except a few, such as FANCB, which is X-linked, and FANCR (RAD51), which is autosomal dominant.[7][8] 

Epidemiology

Fanconi anemia is a very rare type of anemia. Overall, an average of 1 out of 136,000 newborns has Fanconi anemia, ranging from 1 in 100,000 to 250,000 births.[9] European registries and data reveal the prevalence of Fanconi anemia is just 4 to 7 per million live births.[10] Fanconi anemia has been found in all races.[11] However, the rate is higher in South Africans, sub-Saharan Africans, and Spanish Gitanos, with rates of 1 in 40,000 births.[12] A higher carrier frequency among US Ashkenazi Jews of 1 case per 100 people has been reported. There is also a birthrate of approximately 1 per 30,000 live births.[13] Fanconi anemia has a slight predilection for men more than women, but overall, 99% of cases are equal in both sexes. 

Pathophysiology

Fanconi anemia is characterized by chromosomal damage due to the cell's inability to conduct repairs.[2][3] With biallelic mutations in the genes associated with the Fanconi anemia core complex, dysfunctionality occurs in the entire Fanconi anemia pathway. Biallelic mutations of FANCA, FANCC, and FANCG account for over 80% of cases.[14] The bone marrow failure of Fanconi anemia is thought to occur from the selective destruction of CD34+ stem cells. The primary processes affected are related to DNA repair, including homologous recombination, nucleotide excision repair, mutagenic translational synthesis, and alternate end joining. Replication processes include the stabilization of replicative forks as well as the regulation of cytokines. 

The purpose of Fanconi anemia proteins is to maintain genomic integrity and replicative capacity. Various Fanconi anemia proteins maintain genomic stability by repairing DNA interstrand crosslinks (ICLs). ICLs prevent DNA strand separation and maintain DNA integrity. Genetic defects in the Fanconi anemia pathway lead to cells that cannot properly repair DNA damage, resulting in genomic instability, subsequent pancytopenia, and an increased susceptibility to cytotoxic agents, ultraviolet radiation, spontaneous deformation, and predisposition to malignancies. Studies have shown that there is a high frequency of solid tumors (eg, head and neck squamous cell carcinomas) with FANCA variants affecting exons 27 to 30.[15]

Proteins encoded by Fanconi anemia genes include ubiquitin ligase, monoubiquitinated protein, helicase, and breast and ovarian cancer susceptibility proteins. These proteins are responsible for DNA repair and resistance against DNA damage insults. Some Fanconi anemia proteins are similar to the BRCA2 protein, such as FANCD1, which works in the Fanconi anemia-BRCA network. An interactive network of these Fanconi anemia proteins with other proteins is responsible for other rare genetic syndromes (eg, ataxia-telangiectasia, Bloom syndrome, and breast and ovarian cancer).[16][17][18] These Fanconi anemia proteins also have multiple other functions. The Fanconi anemia pathway involves FANC genes and their products to maintain genomic integrity. Fanconi cells show hypersensitivity to crosslinking agents (eg, mitomycin C, diepoxybutane, and cisplatin. Fanconi anemia proteins are also involved in other stress response pathways. The inability of the affected cells to withstand normal oxidative stress and oxygen-free radicals causes oxidative damage.[19]

Fanconi Anemia Genetics and Genomic Instability

Several Fanconi anemia genes have been identified including FANCA, FANCB, FANCC, FANCD1/BRCA2, FANCD2, FANCE, FANCI, FANCM, FANCN/PALB2, FANCO/RAD51C, FANCP/SLX4, FANCR/RAD51, FANCS/BRCA1, FANCV/REV7 and FANCW/RFWD3.[20] Proteins encoded by these genes have various roles in the DNA repair Fanconi anemia pathway. The formation of ICLs leads to Fanconi anemia core complex arrangements. Among them, the FANCD2 protein has a central role in this pathway. Core proteins FANC (A, B, C, E, F, G, L, M), more specifically, the FANCL, component acts as E3 ligase responsible for monoubiquitination, and other core proteins for phosphorylation and translocation of FANCD2 proteins in nuclear foci.[9] Monoubiquitination and phosphorylation are the activation processes after various agents damage DNA in the normal DNA repair pathway. Monoubiquitinated FANCD2 accumulates in nuclear foci with other proteins such as BRCA1, FANCD1/BRCA2, RAD51, and FANCN/PALB2, which are involved in DNA repair and resistance against DNA crosslinking and damaging agents like ionizing radiation, hydroxyurea, and UV light irradiation.

FANCI is monoubiquitylated and phosphorylated after DNA damage, but its role is not prominent. Genetic defects of any component of this pathway result in decreased resistance against damaging agents, ultimately leading to chromosome fragility. Fanconi anemia core complex proteins FANCC and FANCE localizes to nuclear foci along with FANCD2, FANCG with BRCA2, and RAD51. This nuclear foci formation and the monoubiquitination of FANCD2 proteins are the primary steps in the Fanconi anemia DNA repair pathway. The Fanconi anemia core protein complex, with the help of the FANCM protein, stimulates translocase and ATPase, which causes ATP hydrolysis and energizes translocase to move the Fanconi anemia core complex along the DNA strand. Another FAAP24 protein binds to ssDNA and the branching structures at the damaged site. In addition, FANCM contains helicase and endonuclease domains, which separate the double-stranded DNA and cleave the phosphodiester bond at the damaged site to repair the damaged part. FANCJ is also a helicase that binds and interacts with BRCA1.

Fanconi anemia core complex with other proteins (eg, BLM, RPA, and topoisomerase III-alpha) are also involved in the DNA repair pathway. Some of the Fanconi anemia proteins, for instance, FANCD1 with BRCA2 and FANCJ with BACH1 and BRIP1, work downstream on homologous recombination DNA repair. Ultimately, bone marrow failure and other complications are due to defects in the DNA repair pathway, increased oxygen-reactive species, and inflammatory cytokines.[3] Cancer predisposition has the same common reason for defective DNA repair systems.[21] 

Histopathology

Microscopic examination of a bone marrow biopsy shows hypoplasia and hypocellularity with fatty replacement characteristic of aplastic anemia. Hypocellularity is often out of proportion with cytopenias. Bone marrow biopsy at infancy may be normocellular. Some islands of hyperplastic erythroblasts and erythroid dysplasia can be seen. 

History and Physical

Clinical History

Common clinical symptoms of Fanconi anemia include shortness of breath, chest pain, dizziness, and fatigue. Additionally, a clinical history of epistaxis, petechiae, and excessive bleeding from a wound site is common due to thrombocytopenia. The risk of recurrent infections increases with the severity of leukopenia, which presents with fever and flu-like illness. A history of low birth weight is present in some cases, and a history of weight loss is essential to elicit from patients for cases complicated by cancer. Family and marriage history are crucial, primarily in populations with a high prevalence of consanguinity marriages.

Physical Examination Findings

Approximately 75% of Fanconi anemia is associated with congenital disabilities. On physical examination, the child's stature may be short, and light-brown skin pigmentation (ie, cafe-au-lait) lesions in more than 50% of cases.[22] Structural abnormalities of extremities are more common in patients with Fanconi anemia. Common upper limb abnormalities include absent, bifid, supernumerary, low set, short or hypoplastic thumb, absent or hypoplastic radii, and dysplastic ulna. Lower limb abnormalities include polydactyly, short toes, club foot, flat feet, hip dislocation, abnormal femur, and thigh osteoma. Other skeletal abnormalities consist of head and face anomalies, microcephaly and hydrocephaly, frontal bossing, flathead, micrognathia, sloped forehead, webbed and short neck, low hairline, spina bifida, scoliosis, abnormal ribs, extra vertebrae. Hypogonadism is common in both sexes and includes less genitalia development, undescended and absent testis, phimosis, hypospadias, micropenis, and vaginal atresia. Fanconi anemia complicates pregnancies. 

Other anomalies include abnormal epicanthal folds, proptosis, ptosis, cataracts, blindness, epiphora, and ear anomalies such as absent eardrums, small or large pinnae, and atresia of the ear canal. Physical signs of pancytopenia are sometimes visible, including pallor, petechiae, bruising, and coldness of the hands and feet. Gastrointestinal abnormalities such as imperforate anus, tracheoesophageal fistula, atresia of the intestine, Meckel diverticulum, megacolon, hepatocellular adenoma, and umbilical hernia are less common. Proximal renal transport impairment may occur, unlike Fanconi syndrome, which has no primary glomerular involvement. Cardiac effects include ventricular septal defects. 

Evaluation

Fanconi anemia leads to pancytopenia, the consequence of bone marrow failure. Pancytopenia characteristic of Fanconi anemia is usually evident in serum laboratory studies demonstrating a decrease in all 3 blood cell lines, including red blood cells (RBCs), platelets, and leukocytes. Diagnosis may be delayed until the bone marrow failure develops. Different studies have summed up the average age of diagnosis as 7 years, although earlier diagnosis has been increased due to disease awareness and prenatal screening diagnostic enhancement. Early diagnosis may prevent severe complications [23]

Fanconi anemia should be evaluated in patients presenting with signs and symptoms of pancytopenia with or without characteristic malformations and in patients with a family history of bone marrow failure or unbalanced translocations identified during a diagnostic leukemia evaluation. Furthermore, Fanconi anemia should be considered in patients with early-onset tumors or excessive toxicity after standard-dose chemotherapy. A Fanconi anemia diagnosis is typically confirmed with a chromosomal fragility test, which remains the gold standard.[5]

Laboratory Studies

A complete blood count reveals the level of RBCs, white blood cells (WBCs), and platelets. Raised mean corpuscular volume (MCV) indicates macrocytosis, and there can be high fetal hemoglobin levels due to increased stress. Serum erythropoietin is increased due to the low level of blood cells and the low response of hematopoietic stem cells. Bone marrow aspiration and biopsy reveal hypocellularity, aplasia with fatty marrow, and absence of myeloid, erythroid, and megakaryocyte stem cell lines. Myelodysplastic cases show hypo- or hyper granularity, hypopigmentation of myeloid precursors, and hypo-lobulated or hyper-lobulated megakaryocytes. Clonic malformations are seen in leukemic transformations.

The chromosomal breakage/stress cytogenetics test is a diagnostic test indicated in those with severe pancytopenia defined as an absolute neutrophil count of <100 mL, hemoglobin <10 g/dL, reticulocyte <40,000 mL, bone marrow cellularity <25%, and a platelet count of <50,000 mL. Testing with DNA cross-linkers such as diepoxybutane or mitomycin C stimulates the breakage of DNA in the absence of a DNA repair system. These agents increase the chromatids' breaks, gaps, reduplications, or rearrangements. Cultured fibroblasts also show chromosome fragility and are preferred in patients with negative T-lymphocyte chromosome fragility testing or those who have already undergone hematopoietic cell transplantation.[24]

Cell cycle analysis/flow cytometry is an alternative to the chromosomal breakage test. In this test, those cells with impaired DNA repair undergo G2 arrest following DNA cross-linking agent exposure. Immunoblotting of GANCD2 mutation is less commonly used. Fanconi anemia gene sequencing is recommended in patients with positive chromosomal breakage testing. The identification of genetic defects is confirmatory and excludes other chromosome breakage syndromes. Prenatally, abnormal chromosome breakage can be analyzed with amniotic fluid cells or chorionic villous biopsy. An elevated level of serum alpha-protein is a rapid screening diagnostic test, but it may not be identified in many cases.[25]

Additional imaging surveys may be done to evaluate for other structural and congenital disability abnormalities. A skeletal x-ray can detect the size and type of bone defects. The x-ray of the head shows a crewcut appearance. An abdominal ultrasound can assess liver and kidney abnormalities. Additionally, magnetic resonance imaging (MRI) is crucial to identify CNS abnormalities (eg, absence of the corpus callosum, cerebellar and pituitary hypoplasia). 

Treatment / Management

Supportive Therapy

Blood transfusions are the best supportive therapy for Fanconi anemia. Packed RBCs and platelet transfusions have an immediate effect. RBC transfusion from family members should be avoided due to alloimmunization and graft-versus-host disease. Leukopenia responds well to granulocyte-colony-stimulating factor, but this is reserved for those patients with an ANC<200/mL. Extensive transfusion has poor outcomes in patients with hematopoietic cell transplantation.

Hematopoeitic Stem Cell Transplantation

Cure of aplastic anemia and prevention of myelodysplastic syndrome can be achieved through bone marrow, peripheral blood cells, and cord blood transplantation. Bone marrow transplantation from an HLA-matched sibling is the preferred method. Recent clinical trials utilized a low-intensity, fludarabine-based preparative regimen.[26] The 5-year overall survival was higher with Fanconi patients having aplastic anemia as compared with those having underlying myelodysplasia or acute promyelocytic leukemia. Chromosome breakage testing of siblings or other related donors should be performed to exclude Fanconi anemia from donors. However, hematopoeitic stem cell transplantation (HCT) is not accessible to everyone and is typically reserved for patients with severe myelodysplastic syndrome, leukemia, or treatment failure. Approximately 50% to 75% of patients respond, and it is a more permanent treatment.[27] Alternate approaches using autologous transplants are being investigated to prevent the adverse effects of allogeneic hematopoietic stem cell transplant (eg, graft versus host disease).[28][29] One methodology utilizes "digital" genomic editing, or correction, of the FANCA mutation (Fanconi Anemia Complementation Group A) through cytosine and adenosine base editors. 

Androgen Therapy

Androgen therapy may be considered in patients who are not good candidates for hematopoeitic stem cell transplantation. Oxymetholone is the most commonly used androgen. Other androgens less widely used are danazol and oxandrolone. Androgens stimulate hematopoietic stem cell proliferation, but it is not curative. Oxymetholone is initiated with a starting dose of 2 to 5mg/kg/day and then tapered to avoid toxicity. Patients with severe bone marrow hypocellularity frequently have a poor response to androgen therapy. Red blood cells respond well, but platelets and leukocytes are less likely to respond. Some sources advocate using androgens only in children as a bridge therapy to transplantation or in patients ineligible for transplant.[14]

Surgical Therapy

Surgery is performed only for the management of structural deformities. Splitting of anomalies of the hand should be repaired early in life to avoid functional delays. Other surgeries include congenital heart defect surgery, repair of trachea-esophagus fistulas, and imperforate anus. Surgery may be required for cancers as well.

Gene Therapy

Gene therapy is a developing treatment that replaces an abnormal gene with a normal gene. The correction of CD34+ in affected cells is now feasible.[30] However, gene therapy reportedly does not fully correct the problem.[14]

Differential Diagnosis

Fanconi anemia resembles a large variety of diseases. Other hematological problems that manifest clinical features of Fanconi anemia and congenital structural defects associated with Fanconi anemia should be excluded.[23][31]

Acquired aplastic anemia: This condition is due to acquired hematopoietic stem cells' destruction of bone marrow following various toxicogenic agents. Pancytopenia is the cardinal feature of Fanconi anemia due to bone marrow failure. Hypocellularity of the bone marrow is sufficient for diagnosing acquired aplastic anemia, which does not show chromosome fragility by chromosome breakage test. Fanconi anemia shows chromosome fragility test positive, and gene sequencing is required to confirm the diagnosis.[32][33]

Other inherited bone marrow failure syndromes: Because bone marrow failure is distinct in Fanconi anemia, differentiation of other causes of bone marrow failure is essential. Diseases with a shortened telomerase, such as dyskeratosis congenital, are associated with bone marrow aplasia. Bone marrow aplasia caused by reticular dysgenesis is a combined immunodeficiency disease due to AK2 gene mutation. However, sensorineural hearing loss and adaptive immune deficiency in infancy are prominent features of reticular dysgenesis. Diamond-Blackfan anemia is pure red cell aplasia that typically presents with bone marrow abnormalities.[34] Platelets and white blood cells are generally normal, but reticulocytes are decreased. Schwachman-Diamond syndrome is bone marrow aplasia primarily associated with neutropenia in 88% to 100% of cases.[35] There is also associated pancreatic insufficiency. Congenital amegakaryocytic thrombocytopenia principally affects platelets.[36] All these diseases may show trilineage aplasia in the bone marrow after an extended period.

Denovo myelodysplastic syndrome: This condition is a clonal myelodysplastic disease characterized by ineffective hematopoiesis and peripheral cytopenias. Denovo myelodysplastic syndrome (MDS) is also associated with Fanconi anemia, but de novo MDS is not susceptible to chromosome breakage tests and does not have FA mutation or congenital anomalies. Suspected MDS patients with associated FA should be tested with skin fibroblast chromosome breakage tests rather than peripheral lymphocytes.

Drug-induced or infection-associated pancytopenia: Exposure to many cytotoxic drugs, mainly cancer chemotherapy, viral and bacterial infections, and chemicals cause transient bone marrow hypoplasia and pancytopenia in children, but unlike FA, this pancytopenia lacks congenital anomalies and also pancytopenia is reversible with removal of the agents. The chromosomal breakage test is negative.

Paroxysmal nocturnal hemoglobinuria: Paroxysmal nocturnal hemoglobinuria results from mutations of the PIGA gene encoding anchoring protein glycosylphosphatidylinositol (GPI), mainly in hematopoietic progenitor cells, which leads to nocturnal hemolysis due to activation of the complement system in the acidic environment during shallow breathing at night. Patients are anemic and have an increased risk of thrombosis, hemoglobin in the urine, and jaundice. Bone marrow failure is mainly due to acquired autoimmunity rather than being inherited. Chromosomal breakage testing results are negative.

Other rare chromosomal breakage syndromes: Many chromosomal instability diseases in the presence of irradiation, DNA cross-linkers, and chemicals are evident but rare, which mimic FA and necessitate their exclusion for an accurate diagnosis and treatment plan. These disorders include Bloom syndrome,[37] LIG4 syndrome, Ataxia-telangiectasia, Nijmegen breakage syndrome, Seckel syndrome, NHEJ1 deficiency, Warsaw breakage syndrome, and cohesinopathies (eg, Robert syndrome).[38][39][38][40][41][42] Similar to Fanconi anemia, these diseases show an abnormal chromosomal breakage test and congenital anomalies (eg, short stature, microcephaly, and malignancy). They can be quite difficult to differentiate. However, bone marrow failure is not typical of rare chromosomal breakage syndromes, and there are also spectral differences in the congenital anomalies and malignancies associated with Fanconi anemia and other chromosomal breakage syndromes. Genomic sequencing of mutation may be utilized to differentiate these conditions. 

Pertinent Studies and Ongoing Trials

Several clinical trials are ongoing to investigate the therapy and prevention of Fanconi anemia. Clinicians should be aware of these trials and refer their patients to ones they may qualify for.

Prognosis

The prognosis of Fanconi's anemia is poor.[2] Severe aplastic anemia is the main cause of mortality that leads to death before 10 years of age in the absence of a diagnosis. Many children with Fanconi Anemia die of bacterial and fungal infections.[3] Survival has been improved in developed countries due to reduced mortality by bleeding or infection complications. However, living well into adulthood has increased the chances of cancer development. Most individuals eventually develop acute myelogenous leukemia. Regular monitoring of patients is needed for cancer in those who have undergone a bone marrow transplant. Most of the patients have other associated birth defects, including developmental delays, kidney problems, and microcephaly. 

Complications

The major complications of Fanconi anemia are aplastic anemia, myelodysplastic syndrome (MDS), acute myeloid leukemia, and specific solid tumors [2][43]

Fanconi anemia is associated with various cancers, as the defective FA gene is associated with various cancers.[44] Defects of genes of the Fanconi anemia pathway responsible for DNA repair and cell cycle checkpoints are involved in the hypersensitivity of cancer cells to chemotherapeutic drugs, viruses, and radiation. Impairment of the DNA repair system and lack of stability in cell cycle checkpoints leads to the uncontrolled proliferation of cells. Common variants of cancer include squamous cell carcinomas of the head, neck, and upper esophagus and carcinomas of the vulva, anus, and cervix, which have a 50-fold higher risk as compared to the absence of Fanconi anemia association. Myelodysplastic syndrome is the most common malignancy, which has a 6000-fold higher chance as compared to the general population. Acute myelogenous leukemia is the second most common cancer, which has a 700-fold higher chance as compared to the general population. Clonal mosaicism in Fanconi anemia has been demonstrated in aging and cancers.[45] The presence of +1q or 7Q- are harbingers of AML transformation.[14]

Additionally, Fanconi anemia registries reveal around 90% of patients suffer from bone marrow failure by the age of 40 years.[46] Bone marrow failure ultimately leads to pancytopenia.[47] A wide range of endocrine disorders are either intrinsic to molecular defects or associated HCT and androgen therapies. Structural disruption of the hypothalamus-pituitary axis may lead to short stature due to growth hormone deficiency, hypothyroidism is found in around 60% of patients, adrenal dysfunction that responds to exogenous ACTH, and dysfunction of pancreatic islets associated with glucose intolerance, dyslipidemia, and infertility due to hypogonadism. Androgen therapy causes benign and malignant liver tumors and peliosis hepatis.[48][49] 

Deterrence and Patient Education

Information and understanding of Fanconi anemia are essential for family members because of the genetic inheritance pattern of the disease, and the patient is usually a child at the time of clinical presentation and diagnosis. Genetic counseling for the disease helps the family members have enough patient education to increase compliance and the patient-clinician relationship. Fanconi anemia in a child necessitates the testing of the disease in other family members, most importantly, siblings, to either catch the disease in the early stage or to prevent future childbirth with genetic defects.[50] Recent studies have shown that, although the biallelic patient is at risk for cancer, the heterozygote relatives do not share the same disadvantage [51]. However, marriages within cultures or communities do not guarantee that this is always the case. 

A chromosomal breakage test and HLA matching of siblings is essential to evaluate for possible hematopoietic stem cell transplantation between the siblings. A level of knowledge regarding the disease process, signs and symptoms, and the treatment plan should be a part of parents' education. Parents should be informed about any interventions, the benefits of one over other options, and health hazards. After thorough counseling, patients may choose the best treatment plan. 

Enhancing Healthcare Team Outcomes

Fanconi anemia is a rare genetic disorder affecting bone marrow, characterized by impaired DNA repair. Clinicians should recognize physical anomalies, pancytopenia, and an increased risk of malignancies. Diagnosis involves genetic testing. Management includes hematopoietic stem cell transplant, supportive care, and surveillance for complications. Genetic counseling is vital due to the hereditary nature of Fanconi anemia. Regular monitoring and early intervention are essential to mitigate potential complications and improve patient outcomes in this complex disorder.

Fanconi anemia requires an interprofessional team approach for comprehensive care. Healthcare outcomes are enhanced by collaboration between healthcare clinicians. The extent of the disease determines the need for many health professionals from different specialties. Fanconi anemia, although mainly a hematological problem, is associated with multiple diseases, including birth defects and solid as well as hematological cancers. The condition requires the care of primary clinicians, oncologists, hematologists, gynecologists, pathologists, radiologists, nurses, genetic analysts, pharmacists, physiotherapists, and especially the support and care of the patient's family and friends. 


Details

Editor:

Yana Puckett

Updated:

2/25/2024 2:26:01 PM

References


[1]

Chandrasekharappa SC, Lach FP, Kimble DC, Kamat A, Teer JK, Donovan FX, Flynn E, Sen SK, Thongthip S, Sanborn E, Smogorzewska A, Auerbach AD, Ostrander EA, NISC Comparative Sequencing Program. Massively parallel sequencing, aCGH, and RNA-Seq technologies provide a comprehensive molecular diagnosis of Fanconi anemia. Blood. 2013 May 30:121(22):e138-48. doi: 10.1182/blood-2012-12-474585. Epub 2013 Apr 23     [PubMed PMID: 23613520]


[2]

Moldovan GL, D'Andrea AD. How the fanconi anemia pathway guards the genome. Annual review of genetics. 2009:43():223-49. doi: 10.1146/annurev-genet-102108-134222. Epub     [PubMed PMID: 19686080]


[3]

Green AM, Kupfer GM. Fanconi anemia. Hematology/oncology clinics of North America. 2009 Apr:23(2):193-214. doi: 10.1016/j.hoc.2009.01.008. Epub     [PubMed PMID: 19327579]


[4]

Bagby GC, Alter BP. Fanconi anemia. Seminars in hematology. 2006 Jul:43(3):147-56     [PubMed PMID: 16822457]


[5]

Dufour C, Pierri F. Modern management of Fanconi anemia. Hematology. American Society of Hematology. Education Program. 2022 Dec 9:2022(1):649-657. doi: 10.1182/hematology.2022000393. Epub     [PubMed PMID: 36485157]


[6]

Velleuer E, Carlberg C. Impact of Epigenetics on Complications of Fanconi Anemia: The Role of Vitamin D-Modulated Immunity. Nutrients. 2020 May 9:12(5):. doi: 10.3390/nu12051355. Epub 2020 May 9     [PubMed PMID: 32397406]


[7]

Wang W. Emergence of a DNA-damage response network consisting of Fanconi anaemia and BRCA proteins. Nature reviews. Genetics. 2007 Oct:8(10):735-48     [PubMed PMID: 17768402]


[8]

Meetei AR, Levitus M, Xue Y, Medhurst AL, Zwaan M, Ling C, Rooimans MA, Bier P, Hoatlin M, Pals G, de Winter JP, Wang W, Joenje H. X-linked inheritance of Fanconi anemia complementation group B. Nature genetics. 2004 Nov:36(11):1219-24     [PubMed PMID: 15502827]


[9]

Che R, Zhang J, Nepal M, Han B, Fei P. Multifaceted Fanconi Anemia Signaling. Trends in genetics : TIG. 2018 Mar:34(3):171-183. doi: 10.1016/j.tig.2017.11.006. Epub 2017 Dec 16     [PubMed PMID: 29254745]


[10]

Gulbis B, Eleftheriou A, Angastiniotis M, Ball S, Surrallés J, Castella M, Heimpel H, Hill A, Corrons JL. Epidemiology of rare anaemias in Europe. Advances in experimental medicine and biology. 2010:686():375-96. doi: 10.1007/978-90-481-9485-8_22. Epub     [PubMed PMID: 20824457]

Level 3 (low-level) evidence

[11]

Schroeder TM, Tilgen D, Krüger J, Vogel F. Formal genetics of Fanconi's anemia. Human genetics. 1976 Jun 29:32(3):257-88     [PubMed PMID: 939547]


[12]

Tipping AJ, Pearson T, Morgan NV, Gibson RA, Kuyt LP, Havenga C, Gluckman E, Joenje H, de Ravel T, Jansen S, Mathew CG. Molecular and genealogical evidence for a founder effect in Fanconi anemia families of the Afrikaner population of South Africa. Proceedings of the National Academy of Sciences of the United States of America. 2001 May 8:98(10):5734-9     [PubMed PMID: 11344308]


[13]

Rosenberg PS, Tamary H, Alter BP. How high are carrier frequencies of rare recessive syndromes? Contemporary estimates for Fanconi Anemia in the United States and Israel. American journal of medical genetics. Part A. 2011 Aug:155A(8):1877-83. doi: 10.1002/ajmg.a.34087. Epub 2011 Jul 7     [PubMed PMID: 21739583]


[14]

Olson TS. Management of Fanconi anemia beyond childhood. Hematology. American Society of Hematology. Education Program. 2023 Dec 8:2023(1):556-562. doi: 10.1182/hematology.2023000489. Epub     [PubMed PMID: 38066849]


[15]

Altintas B, Giri N, McReynolds LJ, Best A, Alter BP. Genotype-phenotype and outcome associations in patients with Fanconi anemia: the National Cancer Institute cohort. Haematologica. 2023 Jan 1:108(1):69-82. doi: 10.3324/haematol.2021.279981. Epub 2023 Jan 1     [PubMed PMID: 35417938]


[16]

Taniguchi T, D'Andrea AD. Molecular pathogenesis of Fanconi anemia: recent progress. Blood. 2006 Jun 1:107(11):4223-33     [PubMed PMID: 16493006]


[17]

Kennedy RD, Chen CC, Stuckert P, Archila EM, De la Vega MA, Moreau LA, Shimamura A, D'Andrea AD. Fanconi anemia pathway-deficient tumor cells are hypersensitive to inhibition of ataxia telangiectasia mutated. The Journal of clinical investigation. 2007 May:117(5):1440-9     [PubMed PMID: 17431503]


[18]

D'Andrea AD, Grompe M. The Fanconi anaemia/BRCA pathway. Nature reviews. Cancer. 2003 Jan:3(1):23-34     [PubMed PMID: 12509764]


[19]

Longerich S, Li J, Xiong Y, Sung P, Kupfer GM. Stress and DNA repair biology of the Fanconi anemia pathway. Blood. 2014 Oct 30:124(18):2812-9. doi: 10.1182/blood-2014-04-526293. Epub 2014 Sep 18     [PubMed PMID: 25237197]


[20]

Del Valle J, Rofes P, Moreno-Cabrera JM, López-Dóriga A, Belhadj S, Vargas-Parra G, Teulé À, Cuesta R, Muñoz X, Campos O, Salinas M, de Cid R, Brunet J, González S, Capellá G, Pineda M, Feliubadaló L, Lázaro C. Exploring the Role of Mutations in Fanconi Anemia Genes in Hereditary Cancer Patients. Cancers. 2020 Mar 30:12(4):. doi: 10.3390/cancers12040829. Epub 2020 Mar 30     [PubMed PMID: 32235514]


[21]

Kennedy RD, D'Andrea AD. DNA repair pathways in clinical practice: lessons from pediatric cancer susceptibility syndromes. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2006 Aug 10:24(23):3799-808     [PubMed PMID: 16896009]


[22]

Ogilvie P, Hofmann UB, Bröcker EB, Hamm H. [Skin manifestations of Fanconi anemia]. Der Hautarzt; Zeitschrift fur Dermatologie, Venerologie, und verwandte Gebiete. 2002 Apr:53(4):253-7     [PubMed PMID: 12053693]


[23]

Sharma P, Sharma N, Sharma D. A Narrative Review on Fanconi Anemia: Genetic and Diagnostic Considerations. Global medical genetics. 2022 Sep:9(3):237-241. doi: 10.1055/s-0042-1751303. Epub 2022 Sep 5     [PubMed PMID: 36071913]

Level 3 (low-level) evidence

[24]

Auerbach AD. Fanconi anemia and its diagnosis. Mutation research. 2009 Jul 31:668(1-2):4-10. doi: 10.1016/j.mrfmmm.2009.01.013. Epub 2009 Feb 28     [PubMed PMID: 19622403]


[25]

Alter BP, Giri N. Serum alpha fetoprotein levels in Fanconi anaemia. British journal of haematology. 2019 Mar:184(6):1074-1076. doi: 10.1111/bjh.15517. Epub 2018 Aug 20     [PubMed PMID: 30125993]


[26]

Chattopadhyay S, Lionel S, Selvarajan S, Devasia AJ, Korula A, Kulkarni U, Na F, Sindhuvi E, Lakshmi KM, Srivastava A, Abraham A, Mathews V, George B. Fludarabine-Based Low-Intensity Conditioning for Fanconi Anemia is Associated with Good Outcomes in Aplastic Anemia but not in MDS - a Single-Center Experience. Mediterranean journal of hematology and infectious diseases. 2023:15(1):e2023039. doi: 10.4084/MJHID.2023.039. Epub 2023 Jul 1     [PubMed PMID: 37435039]


[27]

Ebens CL, MacMillan ML, Wagner JE. Hematopoietic cell transplantation in Fanconi anemia: current evidence, challenges and recommendations. Expert review of hematology. 2017 Jan:10(1):81-97. doi: 10.1080/17474086.2016.1268048. Epub 2016 Dec 21     [PubMed PMID: 27929686]


[28]

Sipe CJ, Kluesner MG, Bingea SP, Lahr WS, Andrew AA, Wang M, DeFeo AP, Hinkel TL, Laoharawee K, Wagner JE, MacMillan ML, Vercellotti GM, Tolar J, Osborn MJ, McIvor RS, Webber BR, Moriarity BS. Correction of Fanconi Anemia Mutations Using Digital Genome Engineering. International journal of molecular sciences. 2022 Jul 29:23(15):. doi: 10.3390/ijms23158416. Epub 2022 Jul 29     [PubMed PMID: 35955545]


[29]

Siegner SM, Ugalde L, Clemens A, Garcia-Garcia L, Bueren JA, Rio P, Karasu ME, Corn JE. Adenine base editing efficiently restores the function of Fanconi anemia hematopoietic stem and progenitor cells. Nature communications. 2022 Nov 12:13(1):6900. doi: 10.1038/s41467-022-34479-z. Epub 2022 Nov 12     [PubMed PMID: 36371486]


[30]

Río P, Navarro S, Bueren JA. Advances in Gene Therapy for Fanconi Anemia. Human gene therapy. 2018 Oct:29(10):1114-1123. doi: 10.1089/hum.2018.124. Epub     [PubMed PMID: 30117331]

Level 3 (low-level) evidence

[31]

Deviren A, Yalman N, Hacihanefioglu S. Differential diagnosis of Fanconi anemia by nitrogen mustard and diepoxybutane. Annals of hematology. 2003 Apr:82(4):223-7     [PubMed PMID: 12707724]


[32]

Hou JW, Wang TR. Differentiation of Fanconi anemia from aplastic anemia by chromosomal breakage test. Zhonghua Minguo xiao er ke yi xue hui za zhi [Journal]. Zhonghua Minguo xiao er ke yi xue hui. 1997 Mar-Apr:38(2):121-6     [PubMed PMID: 9151465]


[33]

Talmoudi F, Kilani O, Ayed W, Ben Halim N, Mellouli F, Torjmane L, Aissaoui L, Ben Youssef Y, Kammoun L, Ben Othmane T, Bejaoui M, Ben Romdhane N, Elloumi M, Hadiji S, Hentati S, Chemkhi I, Abidli N, Guermani H, Abdelhak S, Amouri A. Differentiation of Fanconi anemia and aplastic anemia using mitomycin C test in Tunisia. Comptes rendus biologies. 2013 Jan:336(1):29-33. doi: 10.1016/j.crvi.2013.02.001. Epub 2013 Mar 11     [PubMed PMID: 23537767]


[34]

Gadhiya K, Wills C. Diamond Blackfan Anemia. StatPearls. 2024 Jan:():     [PubMed PMID: 31424886]


[35]

Burroughs L, Woolfrey A, Shimamura A. Shwachman-Diamond syndrome: a review of the clinical presentation, molecular pathogenesis, diagnosis, and treatment. Hematology/oncology clinics of North America. 2009 Apr:23(2):233-48. doi: 10.1016/j.hoc.2009.01.007. Epub     [PubMed PMID: 19327581]


[36]

Al-Qahtani FS. Congenital amegakaryocytic thrombocytopenia: a brief review of the literature. Clinical medicine insights. Pathology. 2010 Jun 4:3():25-30     [PubMed PMID: 21151552]


[37]

Arora H, Chacon AH, Choudhary S, McLeod MP, Meshkov L, Nouri K, Izakovic J. Bloom syndrome. International journal of dermatology. 2014 Jul:53(7):798-802. doi: 10.1111/ijd.12408. Epub 2014 Mar 6     [PubMed PMID: 24602044]


[38]

Collins NB, Wilson JB, Bush T, Thomashevski A, Roberts KJ, Jones NJ, Kupfer GM. ATR-dependent phosphorylation of FANCA on serine 1449 after DNA damage is important for FA pathway function. Blood. 2009 Mar 5:113(10):2181-90. doi: 10.1182/blood-2008-05-154294. Epub 2008 Dec 24     [PubMed PMID: 19109555]


[39]

Tsuchida K, Komatsu K. Impaired removal of DNA interstrand cross-link in Nijmegen breakage syndrome and Fanconi anemia, but not in BRCA-defective group. Cancer science. 2008 Nov:99(11):2238-43. doi: 10.1111/j.1349-7006.2008.00915.x. Epub 2008 Sep 1     [PubMed PMID: 18771529]


[40]

van der Lelij P, Oostra AB, Rooimans MA, Joenje H, de Winter JP. Diagnostic Overlap between Fanconi Anemia and the Cohesinopathies: Roberts Syndrome and Warsaw Breakage Syndrome. Anemia. 2010:2010():565268. doi: 10.1155/2010/565268. Epub 2010 Jul 18     [PubMed PMID: 21490908]


[41]

Abou-Zahr F, Bejjani B, Kruyt FA, Kurg R, Bacino C, Shapira SK, Youssoufian H. Normal expression of the Fanconi anemia proteins FAA and FAC and sensitivity to mitomycin C in two patients with Seckel syndrome. American journal of medical genetics. 1999 Apr 23:83(5):388-91     [PubMed PMID: 10232749]


[42]

Lilleyman JS. Constitutional hypoplastic anemia associated with familial "bird-headed" dwarfism (Seckel syndrome). The American journal of pediatric hematology/oncology. 1984 Summer:6(2):207-9     [PubMed PMID: 6465473]


[43]

Alter BP. Fanconi anemia and the development of leukemia. Best practice & research. Clinical haematology. 2014 Sep-Dec:27(3-4):214-21. doi: 10.1016/j.beha.2014.10.002. Epub 2014 Oct 15     [PubMed PMID: 25455269]


[44]

Nepal M, Che R, Zhang J, Ma C, Fei P. Fanconi Anemia Signaling and Cancer. Trends in cancer. 2017 Dec:3(12):840-856. doi: 10.1016/j.trecan.2017.10.005. Epub 2017 Nov 10     [PubMed PMID: 29198440]


[45]

Reina-Castillón J, Pujol R, López-Sánchez M, Rodríguez-Santiago B, Aza-Carmona M, González JR, Casado JA, Bueren JA, Sevilla J, Badel I, Català A, Beléndez C, Dasí MÁ, Díaz de Heredia C, Soulier J, Schindler D, Pérez-Jurado LA, Surrallés J. Detectable clonal mosaicism in blood as a biomarker of cancer risk in Fanconi anemia. Blood advances. 2017 Jan 24:1(5):319-329. doi: 10.1182/bloodadvances.2016000943. Epub 2017 Jan 23     [PubMed PMID: 29296947]

Level 3 (low-level) evidence

[46]

Kutler DI, Singh B, Satagopan J, Batish SD, Berwick M, Giampietro PF, Hanenberg H, Auerbach AD. A 20-year perspective on the International Fanconi Anemia Registry (IFAR). Blood. 2003 Feb 15:101(4):1249-56     [PubMed PMID: 12393516]

Level 3 (low-level) evidence

[47]

Kelaidi C, Makis A, Petrikkos L, Antoniadi K, Selenti N, Tzotzola V, Ioannidou ED, Tsitsikas K, Kitra V, Kalpini-Mavrou A, Fryssira H, Polychronopoulou S. Bone Marrow Failure in Fanconi Anemia: Clinical and Genetic Spectrum in a Cohort of 20 Pediatric Patients. Journal of pediatric hematology/oncology. 2019 Nov:41(8):612-617. doi: 10.1097/MPH.0000000000001549. Epub     [PubMed PMID: 31259830]


[48]

Rose SR, Myers KC, Rutter MM, Mueller R, Khoury JC, Mehta PA, Harris RE, Davies SM. Endocrine phenotype of children and adults with Fanconi anemia. Pediatric blood & cancer. 2012 Oct:59(4):690-6. doi: 10.1002/pbc.24095. Epub 2012 Jan 31     [PubMed PMID: 22294495]


[49]

Petryk A, Kanakatti Shankar R, Giri N, Hollenberg AN, Rutter MM, Nathan B, Lodish M, Alter BP, Stratakis CA, Rose SR. Endocrine disorders in Fanconi anemia: recommendations for screening and treatment. The Journal of clinical endocrinology and metabolism. 2015 Mar:100(3):803-11. doi: 10.1210/jc.2014-4357. Epub 2015 Jan 9     [PubMed PMID: 25575015]


[50]

Repczynska A, Julga K, Skalska-Sadowska J, Kacprzak MM, Bartoszewska-Kubiak A, Lazarczyk E, Loska D, Drozniewska M, Czerska K, Wachowiak J, Haus O. Next-generation sequencing reveals novel variants and large deletion in FANCA gene in Polish family with Fanconi anemia. Orphanet journal of rare diseases. 2022 Jul 19:17(1):282. doi: 10.1186/s13023-022-02424-4. Epub 2022 Jul 19     [PubMed PMID: 35854323]


[51]

McReynolds LJ, Giri N, Leathwood L, Risch MO, Carr AG, Alter BP. Risk of cancer in heterozygous relatives of patients with Fanconi anemia. Genetics in medicine : official journal of the American College of Medical Genetics. 2022 Jan:24(1):245-250. doi: 10.1016/j.gim.2021.08.013. Epub 2021 Nov 30     [PubMed PMID: 34906449]