ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Dyskeratosis congenita and other telomere biology disorders

Dyskeratosis congenita and other telomere biology disorders
Literature review current through: Jan 2024.
This topic last updated: Apr 15, 2022.

INTRODUCTION — Dyskeratosis congenita (DC) is an inherited disorder characterized by bone marrow failure (BMF), cancer predisposition, and somatic (nonhematologic) abnormalities. DC and related telomere biology disorders (TBD) are caused by mutations that interfere with normal maintenance of telomeres, the regions at the ends of the chromosomes that protect nucleated cells from the loss or gain of genetic material.

This topic discusses the evaluation, diagnosis, and management of DC and related TBDs.

Other inherited BMF syndromes including Fanconi anemia (FA), Shwachman-Diamond syndrome (SDS), Diamond-Blackfan anemia (DBA), and a general approach to the diagnostic evaluation of unexplained BMF are presented separately.

FA – (See "Clinical manifestations and diagnosis of Fanconi anemia" and "Management and prognosis of Fanconi anemia".)

SDS – (See "Shwachman-Diamond syndrome".)

DBA – (See "Overview of causes of anemia in children due to decreased red blood cell production", section on 'Diamond-Blackfan anemia'.)

General approach – (See "Aplastic anemia: Pathogenesis, clinical manifestations, and diagnosis" and "Approach to the adult with pancytopenia".)

PATHOPHYSIOLOGY

Role of telomeres — Telomeres are specialized structures at the end of chromosomes, comprised of nucleic acid and protein components that maintain the integrity of chromosome ends, protecting the natural ends of chromosomes from loss of DNA, abnormal fusion to other chromosomes, and from activation of DNA damage pathway responses that normally would occur in response at free ends of DNA created by strand breaks [1]. Telomeric DNA consists of tandem repeats of the six-base TTAGGG sequence. Most of the telomeric DNA exists as duplex DNA, with a terminal single-stranded overhang of typically approximately 150 to 200 nucleotides of the G-rich strand [2]. The shortening of the duplex DNA portion of telomeres is most characteristic of telomere biology disorders (TBDs).

At birth, the length of telomeric DNA from somatic cells such as lymphocytes, ranges from 8 to 14 kilobases (kb) [3]. With each cell division, 50 to 100 base pairs of this telomeric DNA is removed due to incomplete replication of the 3' ends. When telomeres reach a critically short threshold, the cell can no longer divide properly and undergoes apoptosis or senescence [4]. In most somatic cells, shortening of telomere length is a normal consequence of aging. As an example, average lymphocyte telomere length declines from approximately 11 kb at birth to only approximately 4 kb in centenarians. Thus, telomeres have been referred to as the "biological clock" within a cell that determines the number of possible cell divisions. (See "Basic genetics concepts: Chromosomes and cell division", section on 'Mitosis' and "Basic genetics concepts: Chromosomes and cell division", section on 'Parts of the chromosome'.)

In contrast to the limited normal replicative capacity of most somatic cells, certain cell types that require constant replicative regeneration are capable of restoring and maintain longer telomere length to enable a greater number of divisions [5]. Examples include embryonic stem cells, multipotent stem cells in high-turnover tissues such as epithelial tissues, and bone marrow, and malignant tumors [6,7]. This increased replicative capacity is made possible by the action of the telomerase complex, a ribonuclear protein complex (RNA and proteins) that counteracts telomere shortening by adding back DNA to the ends of chromosomes. The ability of telomerase to lengthen telomeres is regulated by several other mechanisms. The nucleoprotein factors contributing to these mechanisms, and consequently the genes encoding these factors in which mutations lead to TBDs (figure 1), have been subdivided into five categories [8]:

Telomerase activity – Telomerase is a reverse transcriptase that adds TTAGGG repeats to the chromosome end. It consists of TERT, a catalytic protein; and TR, an RNA template. Assembly and stability of telomerase requires the action of several small nucleolar ribonucleoproteins (snoRNPs) including dyskerin, NHP2, and NOP10. The protein encoded by NAF1 is a box H/ACA RNA biogenesis factor similar to dyskerin that may regulate TR levels [9].

Telomerase trafficking and recruitment to telomeres – Once telomerase is assembled, it must be recruited to the telomere. Trafficking mediated by the protein TCAB1, which binds to TR directly, is critical to this process [10,11]. The shelterin complex, which consists of the proteins TRF1, TRF2, RAP1, TIN2, TPP1, and POT1, serves the function of creating a "t-loop" (TRF2), resulting in the recruitment of additional proteins that create a stable telomere "cap." TRF1 and TRF2 recruit TNF2 to the telomere, which in turn recruits a heterodimer of TPP1/POT1. TPP1 then recruits telomerase to the telomere via its TEL patch motif [12].

Telomere replication – The CTC1/STN1/TEN1 (CST) complex promotes extension of the C-rich strand of telomere DNA after telomerase has elongated the G-rich strand of telomere DNA, creating extension of the duplex telomere DNA component.

Telomere stability – RTEL1 (regulator of telomere length 1) is a DNA helicase that contributes both to integrity of duplex telomere DNA replication as well as to the dismantling of structures called D-Loops that could otherwise result in telomere DNA loss through excision repair mechanisms.

Unknown or multifactorial roles – While TIN2 contributes to telomerase recruitment, mutations in the gene encoding TIN2 appear to play a dominant-negative role in telomere maintenance through as yet undetermined mechanisms. In addition, the exoribonuclease PARN may impact RNA transcript stability of other telomerase associated factors [13].

In TBDs (also called short telomere syndromes or telomeropathies), mutations in the genes encoding any of the above factors implicated in telomere function lead to abnormally short telomeres (figure 1). Mutations known to cause DC and related TBDs encode the proteins involved in telomerase activity and trafficking, formation of the shelterin "capping" complex, and telomere stability. These mutations and their associated inheritance patterns and named syndromes are listed below. (See 'Genetics' below and 'Inheritance patterns' below.)

Compared with the normal rate of telomere shortening in unaffected individuals of approximately 60 bp per year, individuals with telomere disorders lose telomeric DNA at approximately 120 bp per year [14]. Furthermore, successive generations of affected individuals may be born with progressively shorter telomeres (a phenomenon known as disease anticipation) [15,16]. Premature telomere shortening in TBDs leads to premature cell death, senescence, or genomic instability, which in turn leads to impaired organ and tissue function, altered homeostasis, or inappropriate growth [17]. Clinical consequences are described below. (See 'Clinical features' below.)

DC is the prototypic TBD [18,19]. It is one of several inherited bone marrow failure syndromes (IBMFS), each of which is characterized by its own spectrum of associated clinical findings. However, the initial descriptions of DC, referred to at the time as Zinsser-Cole-Engman syndrome, focused on its skin findings [20]. Subsequently, the classic triad of abnormal skin pigmentation, nail dystrophy, and oral leukoplakia have come to define DC. However, other well-recognized complications are often the major causes of morbidity in DC, particularly bone marrow failure (BMF).

In addition to DC and other TBDs, certain inherited and acquired cancer syndromes are associated with mutations in telomerase complex and shelterin complex without BMF. Examples include familial melanoma, leukemias, and others [21]. (See "Inherited susceptibility to melanoma" and "Familial disorders of acute leukemia and myelodysplastic syndromes".)

In contrast to the TBDs, the abnormally short telomeres in certain progeria (premature aging) syndromes appear to affect different populations of cells; there is virtually no clinical overlap between premature aging syndromes and TBDs, with the exception of increased mortality due to disease complications in both cases. (See "Emery-Dreifuss muscular dystrophy", section on 'Laminopathies' and "Hutchinson-Gilford progeria syndrome".)

Familial syndromes associated with abnormally long telomeres have not been reported, although there may be an association between long telomeres and certain acquired malignancies such as chronic lymphocytic leukemia (CLL) [22].

Genetics — Mutations in genes encoding components of the telomerase complex, shelterin proteins, and other telomerase regulators have been described individuals with DC and other TBDs. Causative gene mutations continue to be identified.

In patients meeting diagnostic criteria for DC, it is estimated that up to 70 percent will have a pathogenic mutation in one of the following genes (table 1):

ACD (adrenocortico dysplasia homolog) – Autosomal dominant DC, autosomal recessive DC, Hoyeraal-Hreidarsson syndrome. ACD encodes the TPP1 protein. (See 'Hoyeraal-Hreidarsson syndrome' below.)

CTC1 (conserved telomere maintenance component 1) – Autosomal recessive DC, autosomal recessive Coats plus syndrome, also known as cerebroretinal microangiopathy with calcifications and cysts (CRMCC). (See 'Coats plus syndrome' below.)

DKC1 – X-linked DC, Hoyeraal-Hreidarsson syndrome. (See 'Hoyeraal-Hreidarsson syndrome' below.)

NAF1 (NEF-associated factor 1; also called TNFAIP3-interacting protein 1 [TNIP1]) – Autosomal dominant TBD features including pulmonary fibrosis [9].

NHP2 (non-histone protein 2; also called nucleolar protein family A, member 2 [NOLA2]) – Autosomal recessive DC or autosomal dominant TBD features including pulmonary fibrosis.

NOP10 (nucleolar protein 10; also called nucleolar protein family A, member 3 [NOLA3]) – Autosomal recessive DC.

PARN (polyadenylate-specific ribonuclease) – Autosomal recessive DC, pulmonary fibrosis, Hoyeraal-Hreidarsson syndrome. (See 'Hoyeraal-Hreidarsson syndrome' below.)

RTEL1 (regulator of telomere elongation helicase 1) – Autosomal dominant DC, autosomal recessive DC, pulmonary fibrosis, Hoyeraal-Hreidarsson syndrome [23]. (See 'Hoyeraal-Hreidarsson syndrome' below.)

STN1 – Autosomal recessive Coats plus syndrome [24]. (See 'Coats plus syndrome' below.)

TERC – Autosomal dominant DC, pulmonary fibrosis [25,26].

TERT (telomerase reverse transcriptase) – Autosomal dominant DC, autosomal recessive DC, familial melanoma, pulmonary fibrosis.

TINF2 (TRF1-interacting nuclear factor 2) – Autosomal dominant DC, Hoyeraal-Hreidarsson syndrome, Revesz syndrome. (See 'Other specific syndromes' below.)

WRAP53 (WD repeat-containing protein antisense to TP53; encodes the TCAB1 protein) – Autosomal recessive DC.

ZCCHC8 – (required for telomerase RNA maturation) – Autosomal dominant pulmonary fibrosis [27].

USB1 (previously C16orf57; required for snRNA maturation) – Biallelic mutations are associated with Rothmund Thomson syndrome, poikiloderma with neutropenia, and some patients with a dyskeratosis congenita phenotype, albeit with normal telomere lengths [28].

MDM4 (increases p53 activity) – Germline missense mutation associated with BMF and short telomeres in a family with features of DC, including tongue squamous cell carcinoma and acute myeloid leukemia [29].

NPM1 – Heterozygous germline mutations that disrupted protein translation without modulating telomeres [30].

Mutations in specific genes have been demonstrated to segregate with clinical effects in the TBDs and genotype-phenotype correlations for specific mutations within individual genes are increasingly becoming recognized [31]. In general, the more clinically severe variants of DC are associated with the greatest reduction in telomere length [19].

Inheritance patterns — Various inheritance patterns have been observed for TBDs, depending on the affected gene. (See 'Genetics' above.)

Autosomal dominant – Autosomal dominant disease has been observed with mutations in ACD, PARN, RTEL1, TERC, TERT, TINF2, and NAF1 [16,32-40]. Disease manifestations may become more severe over generations (disease anticipation) in individuals with TERC or TERT mutations, which has been attributed to the transmission of progressively shorter telomeres from one generation to the next [15,16]. Patients with TINF2 mutations have extremely short telomeres and frequently present with BMF before five years of age [36].

Autosomal recessive – Autosomal recessive disease has been observed with mutations in ACD, CTC1, NHP2, NOP10, PARN, RTEL1, STN1, TERT, and WRAP53 [11,41-44].

X-linked – The only X-linked DC syndrome that has been reported involves mutations in the DKC1 gene [45]. Heterozygous females show skewed X-chromosome inactivation patterns, suggesting a survival advantage for cells that express the normal DCK1 allele [46]. These females are considered unaffected carriers; however, a retrospective analysis of six heterozygous individuals found that five had nail dystrophy and skin pigmentation changes characteristic of DC, and two had clinically significant delays in wound healing [47].

EPIDEMIOLOGY — The true prevalence of DC and other TBDs is unknown, in large part due to the incomplete penetration of disease features in many individuals. Patients with subtle findings may receive other diagnoses such as idiopathic aplastic anemia (AA), idiopathic pulmonary fibrosis, idiopathic cirrhosis, or sporadic congenital abnormalities. Additionally, it is likely that many other individuals with gene mutations affecting telomere length may have a more subtle clinical phenotype. At the other extreme, it is conceivable that severe forms of DC may be underdiagnosed due to high mortality rates.

With these caveats, the prevalence of DC has been estimated to be approximately 1 in 1 million people in the general population [48]. Approximately 2 to 5 percent of patients with bone marrow failure are identified to have DC. Children are more likely to be diagnosed with DC than adults, however other TBDs resulting in pulmonary fibrosis and liver disease that present in adulthood are increasingly being recognized [49,50]. Initial reports suggested an increased frequency in males, but this may be related to an earlier belief that DC was solely an X-linked disorder [20].

Certain forms of DC appear to be more common in certain populations, such as the increased incidence of Hoyerall-Hreidarsson syndrome due to RTEL1 mutations in Ashkenazi Jews [51].

CLINICAL FEATURES — DC can affect the bone marrow, immune system, skin, lung, liver, and teeth. Patients are also at increased risk of a number of malignancies. These features can be variable and include the classic presentations of DC or atypical presentations in which only a subset of findings are present (table 2). Common findings include mucocutaneous changes (see 'Classic DC presentation' below), bone marrow failure (BMF) (see 'Bone marrow failure' below), a variety of somatic abnormalities (see 'Additional somatic features' below), and increased risks of certain malignancies (see 'Cancer predisposition' below).

Classic DC presentation — Initially, DC was recognized clinically by a triad of mucocutaneous findings (picture 1):

Abnormal skin pigmentation – Lacy reticular hyperpigmentation involving the upper chest and neck.

Nail dystrophy – Small, thin nail plates with longitudinal ridges that disappear with age. Nails of the hands and feet are affected.

Oral leukoplakia – Oral leukoplakia involves the oral mucosa and tongue.

This classic presentation is helpful for diagnosis, when present, but many individuals do not have all three features upon presentation. In a review of approximately 500 reported cases up to the year 2000 (ie, when diagnosis was based solely on clinical features, before the genetic basis for DC was identified), it was determined that approximately three-fourths of patients had at least one of these three classic manifestations, and slightly fewer than half had all three findings [20]. Notably, the median age at diagnosis was 14 years (range, birth to 75 years), which was approximately seven years later than that of Fanconi anemia in the same analysis. However, with increasing recognition of the features of DC, many patients are being diagnosed at younger ages. In some families, findings may become more severe in successive generations due to genetic anticipation. (See 'Pathophysiology' above.)

Bone marrow failure — BMF can present at any age, and in many cases, it may be the presenting sign of DC. Nearly half of patients will develop signs of BMF by age 40. In the London DC Registry, the median age of onset of BMF was 10 years [17,52].

Thrombocytopenia and anemia are often the first signs of BMF, although cytopenias in early childhood are not necessarily due to bone marrow aplasia. Cytopenias may also be immune-mediated, due to immune dysregulation; or caused by organ dysfunction and/or bleeding. In some cases, patients have presented early in life with what are thought to be isolated immune cytopenias within a single lineage, only to develop true pancytopenia in later childhood or adolescence.

Additional somatic features — Patients with DC and other telomere syndromes are also at risk of developing other clinical features, including a wide array of organ manifestations [17,53].

Some features typically appear in childhood, although the age of onset is also highly variable. Early in life, patients may begin to develop features including esophageal strictures, lacrimal duct destruction, severe dental or periodontal disease, recurrent infections due to immune deficiency, enteropathy/enterocolitis, short stature, hypogonadism, urethral strictures, retinopathy, pulmonary and gastrointestinal vascular changes, and cognitive developmental impairment [53]. However, many of these somatic abnormalities may be absent in early childhood. As patients enter the second decade of life and beyond, they are at increasing risk for pulmonary fibrosis; liver disease including hepatopulmonary syndrome and cirrhosis; vascular malformations, osteoporosis and other bone abnormalities; premature graying of the hair; and neuropsychiatric disorders [53].

In a summary of findings from 118 patients in the London DC Registry, the following features were present [17,52]:

BMF – 86 percent

Classic mucocutaneous and additional dermatologic findings

Skin dyspigmentation – 89 percent

Nail irregularities – 88 percent

Leukoplakia – 78 percent

Premature graying/hair loss – 16 percent

Hyperhidrosis – 15 percent

Ophthalmologic/Epiphora (excessive tearing/lacrimal duct stenosis) – 31 percent

Neurologic/Cognitive

Developmental delay – 25 percent

Ataxia/cerebellar hypoplasia – approximately 7 percent

Microcephaly – 6 percent

Pulmonary disease (pulmonary fibrosis, diffuse arteriovenous malformations due to hepatopulmonary syndrome [49]) – 20 percent

Endocrine/Growth/Urologic features

Short stature – 20 percent

Intrauterine growth retardation – 8 percent

Hypogonadism/Undescended testes – 6 percent

Urethral stricture/phimosis – 5 percent

Osteoporosis and related complications – 5 percent

Unlike Fanconi anemia, individuals with DC do not appear to have impaired fertility [20,54].

Dental manifestations (caries) – 17 percent

Gastroenterologic/Hepatologic manifestations [55]

Esophageal strictures – 17 percent

Liver disease (cirrhosis, fibrosis) or gastroenteropathy – 7 percent

Intestinal vascular malformations

Cancer – 10 percent (see 'Cancer predisposition' below)

Other series have reported similar findings [20,56-60].

Additionally, a number of other more uncommon manifestations are found in subsets of patients with telomere disorders [61]. These include:

Abnormal eyelash/eyelids, leading to corneal abrasions and infection

Exudative retinopathy (in Revesz syndrome)

Hearing loss/deafness

Pulmonary or hepatic arteriovenous malformations and vascular ectasias [55,62,63]

Hepatopulmonary syndrome

Neuropsychiatric disorders including schizophrenia and bipolar disorder

Pulmonary fibrosis and liver cirrhosis deserve specific attention. In contrast to many of the organ abnormalities listed above, these complications typically present in adulthood (fourth or fifth decade) and may be the first presenting feature of TBDs, other than DC [64]. Pulmonary fibrosis is seen in approximately one-fifth of individuals with DC [65]. Further, up to 15 percent of familial and 5 percent of sporadic cases of idiopathic pulmonary fibrosis are associated with mutations in TERT, TERC, PARN, or RTEL1 [66,67].

The combination of pulmonary fibrosis and bone marrow hypoplasia is emerging as a strong predictor of a telomere disorder [68]. Retrospective study of 38 consecutive individuals referred for evaluation of pulmonary fibrosis and/or BMF who lacked mucocutaneous findings of DC found that 10 had a germline mutation in TERT or TERC and evidence of impaired telomerase activity [26].

Cancer predisposition — Patients with classic DC are at high risk for developing many types of cancer. The incidence of cancer in DC is less than 10 percent by age 20 years, but rises to 20 to 30 percent by age 50, with the first cancer diagnosis at a median age of 29 years.

Studies from the National Cancer Institute (NCI) prospective DC cohort and from a literature review involving over 775 DC cases have defined the increased incidence of a number of specific malignancies in DC [69-71]. These common types of cancers reported in DC, and the proportion of all cancers in DC that they account for, are as follows:

Head/Neck squamous cell carcinoma (40 percent)

Stomach/Esophageal (approximately 17 percent)

Anorectal (12 percent)

Skin (12 percent)

Acute leukemia (8 percent; myeloid more common than lymphoid)

Liver (5 percent)

Hematopoietic neoplasms, such as MDS and acute myeloid leukemia (AML), and solid tumors, including squamous cell cancers of the head and neck, have a similar incidence to that seen in Fanconi anemia (FA), although these appear to develop at a later age in DC than they do in FA [72]. Other reported cancers include Hodgkin lymphoma and cancers of the lung, pancreas, colon, and cervix. Given the numerous vascular malformations and ectasias seen in DC, it is also notable that two adolescent/young adult patients with DC have been reported to have angiosarcomas involving the liver, a tumor that is extremely rare in this age group [73,74].

Management of malignancy in patients with DC is discussed below and in separate topic reviews:

Hematologic neoplasms – (See 'AML or MDS' below and "Familial disorders of acute leukemia and myelodysplastic syndromes".)

Solid tumors – (See 'Solid tumors' below and "Treatment and prognosis of low-risk cutaneous squamous cell carcinoma (cSCC)" and "Locally advanced squamous cell carcinoma of the head and neck: Approaches combining chemotherapy and radiation therapy".)

Other specific syndromes — The following syndromes are considered to be forms of DC:

Hoyeraal-Hreidarsson syndrome — Hoyeraal-Hreidarsson syndrome (HHS) is a clinically severe form of DC with disease manifestations beginning in early childhood [75].

HHS was originally associated with X-linked mutations in DKC1, referred to as X-linked dyskeratosis congenita (DKCX; also called Zinsser-Cole-Engman syndrome); the syndrome was subsequently found to occur with recessive mutations in ACD, RTEL1, TERT, and PARN, along with autosomal dominant mutations in TINF2 [31,39,76,77]. (See 'Genetics' above.)

In addition to the classic mucocutaneous and somatic features of DC, patients with HHS have the following clinical features [51,75,78]:

Intrauterine growth retardation

Cerebellar hypoplasia

Microcephaly

Developmental delay

Severe immunodeficiency (worse than classic DC)

Early-onset progressive BMF

As the diagnosis of HHS has evolved over time, cerebellar hypoplasia has become a diagnostic requirement [31].

As noted above, HHS is seen at increased frequency in individuals of Ashkenazi Jewish ancestry, due to a founder mutation (c.3791G>A [p.R1264H]). (See 'Epidemiology' above.)

Revesz syndrome — In addition to having often severe features of DC, patients with Revesz syndrome (RS) are defined based on the presence of bilateral exudative retinopathy; often there are also intracranial calcifications and intrauterine growth restriction. Mutations in TINF2 are the most commonly identified genetic cause of Revesz syndrome, though many cases have yet to have a genetic cause identified. (See 'Genetics' above.)

Coats plus syndrome — Coats plus syndrome is an autosomal recessive disorder characterized by retinal telangiectasias with exudates, intracranial calcifications, cerebellar movement disorder, osteopenia, leukodystrophy, poor growth, and bone marrow abnormalities [79]. It is caused by autosomal recessive mutations in CTC1 or STN1, components of the CST telomere replication complex involved in duplex telomere DNA elongation [24,80]. Telomeres may be quite short in patients with Coats plus syndrome caused by CTC1 mutation, though some patients may have normal telomere lengths, suggesting telomere dysfunction apart from telomere shortening may drive disease features in Coats plus syndrome [81,82].

CTC1 mutations are also seen in patients with classic DC features, highlighting the overlap between Coats plus syndrome and other short telomere syndromes [81,83].

DIAGNOSIS

Overview of diagnosis — The diagnosis of DC is evolving from a purely clinical diagnosis based on classic mucocutaneous findings with or without bone marrow failure (BMF) to a broader range of clinical findings and more subtle presentations. This broadening of diagnosis has been spurred by research demonstrating the role of telomere dysfunction in the disease (see 'Role of telomeres' above), assays for telomere length, and genetic testing for specific abnormalities known to affect telomeres. Previously defined syndromes including Hoyeraal-Hreidarsson and Revesz syndromes are now thought to be subcategories of DC.

However, the nomenclature regarding certain patients lacking classic mucocutaneous features of DC, but who have short telomeres and mutations in telomere disorder-related genes has yet to be universally defined. Some experts consider most of these patients to have DC, whereas others will refer to these patients as having a telomere biology disorder (TBD), restricting the diagnosis of DC to patients who have classic mucocutaneous findings. (See 'Classic DC presentation' above.)

Diagnosis of a new proband is generally made using a combination of clinical findings and laboratory testing. Diagnosis of additional affected family members can then take into account the known familial mutation and clinical features. The diagnosis of DC is most easily suspected in individuals with the classic presentation of mucocutaneous findings, BMF, and other organ system involvement; or in those from a family with a known DC mutation. Additional clues to the underlying diagnosis may include isolated cytopenias, pulmonary fibrosis, hepatic disease, immunodeficiencies, and/or early graying of the hair. The co-occurrence of BMF and pulmonary fibrosis is highly suggestive of an underlying telomere disorder, as discussed above. (See 'Additional somatic features' above.)

Patients in whom the diagnosis of DC is considered should have a thorough family history, personal medical history, and physical examination to assess for features of DC. It is especially important to identify DC when present because patients may benefit from surveillance for additional clinical manifestations including cancer and organ dysfunction; family members with a shared mutation but with less penetrant disease features may also be identified; and siblings who could serve as potential donors for hematopoietic cell transplantation (HCT) must be screened for DC. Therapeutic options for BMF are very specific to the DC diagnosis.

Clinical criteria — Clinical criteria for the diagnosis of telomere biology disorders have evolved and are not universally agreed upon. Initially, registration in the DC registry required a family to have an index case with the classic triad of mucocutaneous findings that define DC. (See 'Classic DC presentation' above.)

Subsequently, two different sets of diagnostic criteria have been adopted:

In the first set of criteria, patients with any of the following combinations of features are defined as having DC [84]:

All three classic mucocutaneous findings of abnormal skin pigmentation, nail dystrophy, and leukoplakia (see 'Classic DC presentation' above)

One of three mucocutaneous features plus BMF and at least two other somatic features known to occur in DC (see 'Additional somatic features' above)

Four or more features of the Hoyeraal-Hreidarsson syndrome (eg, intrauterine growth retardation [IUGR], developmental delay, severe immune deficiency, BMF, cerebellar hypoplasia)

Aplastic anemia, myelodysplastic syndrome, or pulmonary fibrosis in the setting of a known pathogenic genetic variant affecting telomerase function

Two or more features of DC and laboratory evidence of short telomeres (ie, less than the 1st percentile for age by multicolor flow cytometry with fluorescence in situ hybridization [flow-FISH] in several subsets of lymphocytes)

The second set of criteria differentiates findings suggestive of DC versus testing required to definitively establish the diagnosis. In this scheme, suggestive findings that lead to a suspected diagnosis of DC include the following [61]:

At least two of the three classic mucocutaneous features

One classic mucocutaneous feature plus two somatic features

Progressive BMF, MDS, or AML

Solid tumors associated with DC and that are otherwise atypical for age

Pulmonary fibrosis

Short lymphocyte telomeres (below the 1st percentile for age)

According to these criteria, a pathogenic variant must be identified in a DC-associated gene to definitively establish the DC diagnosis.

In addition to the above criteria, a family member of an index patient may be diagnosed with DC or a TBD if they share the same genetic variant with the proband and are identified to have short telomeres.

Laboratory testing and bone marrow — The diagnostic testing for DC consists of telomere length analysis and genetic testing for specific mutations. (See 'Pathophysiology' above.)

The approach used depends on the patient's presentation:

Telomere length analysis – In all individuals with a de novo presentation and/or a family history consistent with a telomere disorder, flow-FISH (multi-color flow cytometry with fluorescence in situ hybridization) is performed on peripheral blood lymphocytes using peptide nucleic acid probes for telomeric DNA [4,85]. Average telomere length below the 1st percentile for age is considered indicative of abnormally short telomeres and is consistent with DC or a related TBD. Even in cases with a known familial mutation, telomere length analysis is recommended to help predict the degree to which an individual family member may be affected by DC-related complications.

Molecular sequencing – All patients who are suspected to have DC based on clinical criteria and telomere length analysis should have genetic testing performed to identify a causative mutation. This testing is critical for definitive establishment of a genetic diagnosis and enabling testing of first degree relatives for potential carrier or disease status, and to determine family member eligibility to be an HCT donor.

There are three general ways in which sequencing can be performed:

Sequential single gene testing – Single gene testing was initially the only method available for molecular testing in patients with DC. For the most part, this approach has been replaced by multi-gene next generation sequencing panels, which are more cost-effective and take less time than sequential single gene testing in most cases. However, for patients in whom mutations in a specific gene are highly suspected based on clinical presentation (eg, p.R1264H variant in RTEL1 in individuals with Ashkenazi Jewish ancestry, CTC1 mutations in patients presenting with features of Coats plus syndrome), this approach may be optimal.

Next generation sequencing (NGS) panels – An NGS gene panel is generally the most recommended approach for identifying gene mutations in patients with DC. A number of commercial panels are available that include 11 genes with mutations associated with TBDs (see 'Genetics' above). Results from these panels are typically available within six to eight weeks. Notably, though, these panels may vary in sensitivity, genes tested, and whether specific deletion duplication analysis is included.

Whole exome sequencing (WES) – WES, which uses NGS methods to sequence the entire exome (coding regions of genes), is also readily commercially available. WES has the advantage of being able to identify for mutations in novel genes not previously associated with telomere disorders. The disadvantages of WES are that coverage is often not 100 percent for all DC-associated genes (eg, TERC mutations cannot be identified by WES); insurance coverage may be hard to obtain and the turnaround time on testing is typically longer than for NGS panels.

A list of clinical laboratories that perform telomere length testing and molecular genetic testing for DC is available on the Genetic Testing Registry website, and additional information about NGS panels and WES is presented separately. (See "Next-generation DNA sequencing (NGS): Principles and clinical applications".)

Identification of a pathogenic DC mutation is considered diagnostic in the appropriate clinical setting, as noted above (see 'Clinical criteria' above). However, negative results from genetic testing do not eliminate the possibility of DC, since a significant proportion of patients lack identifiable mutations. It is likely that additional genes involved in telomere maintenance will be discovered.

In addition to the above testing, determining whether a patient has BMF or myelodysplastic syndrome is often useful in the diagnosis of DC. BMF is defined as a hypocellular marrow, typically <25 percent of the age expected cellularity, along with the presence of peripheral blood cytopenias. Myelodysplastic syndrome (MDS) is defined the presence of morphologic abnormalities along with characteristic cytogenetic abnormalities. (See "Clinical manifestations, diagnosis, and classification of myelodysplastic syndromes (MDS)".)  

Assessments for BMF and MDS should include:

Complete blood count (CBC) with mean corpuscular volume (MCV) and absolute reticulocyte count (ARC). Patients with DC typically have elevated MCV and low ARC.

Unilateral bone marrow aspirate and biopsy (>1 cm) for morphologic review. Typical bone marrow findings in DC include hypocellularity, decreased megakaryocytes, and some degree of dyspoiesis [20].

Cytogenetic studies on the bone marrow aspirate including:

G-banding analysis of at least 20 metaphases to assess for acquired chromosomal aberrations.

FISH analysis for specific aberrations including 5q-, monosomy 7, 7q-, trisomy 8, and 20q-.

Next generation sequencing (NGS) panel assessing for somatic gene mutation commonly associated with MDS.

Notably, there are no pathognomonic bone marrow findings for DC; the bone marrow abnormalities may be indistinguishable from other inherited BMF syndromes, and the CBC abnormalities are seen in a number of conditions.

In contrast to the use of bone marrow examination for diagnosis, which is not required in all individuals, bone marrow for surveillance is an important part of the management of DC. (See 'Management' below.)

DIFFERENTIAL DIAGNOSIS — The differential diagnosis of DC depends on the presenting clinical features and may include other inherited or acquired bone marrow failure (BMF) syndromes or hematologic neoplasms, acquired/de novo hematologic malignancies, other causes of congenital anomalies, and other causes of interstitial lung disease (ILD).

Other inherited BMF syndromes – Other inherited BMF syndromes include Fanconi anemia (FA), Shwachman-Diamond syndrome (SDS), Diamond-Blackfan anemia (DBA), and congenital amegakaryocytic thrombocytopenia (CAMT). Like DC, most of these syndromes can have variable presentations with variable cytopenias and degrees of bone marrow hypocellularity. Unlike DC, these disorders are not associated with pulmonary fibrosis or hepatic fibrosis, most of these patients do not have short telomeres, and they do not have pathogenic mutations in genes that encode telomerase components. These other syndromes and an approach to distinguishing among them is presented separately. (See "Clinical manifestations and diagnosis of Fanconi anemia" and "Shwachman-Diamond syndrome" and "Overview of causes of anemia in children due to decreased red blood cell production", section on 'Diamond-Blackfan anemia'.)

Familial MDS or AML – Familial myelodysplastic syndromes (MDS) and familial acute myeloid leukemia (AML) are inherited syndromes associated with an increased risk of hematologic neoplasms. Like DC, they can present in childhood with multiple affected family members. Like DC, patients with these syndromes who have developed MDS or AML may have very short telomeres on diagnostic testing, but these short telomeres are due to the underlying clone and are only present in blood and bone marrow cells. Unlike DC, these syndromes are not associated with organ system involvement typical of DC or pathogenic mutations affecting telomerase components. (See "Familial disorders of acute leukemia and myelodysplastic syndromes".)

Acquired BMF – Acquired causes of BMF include a number of infectious and toxic exposures, idiopathic acquired aplastic anemia and paroxysmal nocturnal hemoglobinuria (PNH). Like DC, these conditions are associated with cytopenias and bone marrow abnormalities. Unlike DC, these conditions are not associated with congenital anomalies, organ system involvement (with the exception of hepatitis in virally-induced aplastic anemia), or pathogenic mutations affecting telomerase components. Most of these acquired BMF conditions are not associated with short telomeres, with the exception of hepatitis-associated acquired aplastic anemia [86]. The causes of acquired BMF are discussed separately. (See "Aplastic anemia: Pathogenesis, clinical manifestations, and diagnosis" and "Treatment of acquired aplastic anemia in children and adolescents".)

Genetic syndromes with overlapping dermatologic features – Rothmund-Thomson syndrome is a disease associated with thin, brittle hair, telangiectasias, thin skin and somatic features including enteropathy, poor dentition, and poor growth/short stature. It is caused by mutations in RECQL4. Patients who have poikiloderma with neutropenia have mutations in USB1 (previously C16orf57), and present with skin dyspigmentation, telangiectasias, and atrophy, along with immune deficiency and neutropenia. Patients with pachyonychia congenita, caused by mutations in genes that encode keratin proteins, present with nail dystrophy, hypertrophic skin on palms/soles, oral leukokeratosis that mimics leukoplakia, and hyperhidrosis. Unlike DC, patients with these conditions have normal telomere length, do not develop hypocellular BMF, and do not have mutations in genes that affect telomerase function. (See "Kindler epidermolysis bullosa", section on 'Differential diagnosis'.)

Other causes of ILD – Interstitial lung disease (ILD) is a broad term that applies to conditions that diffusely affect the lung parenchyma. There are numerous underlying causes, including infection, immune defects, hypersensitivities, and systemic disorders. Telomere disorders have been estimated to account for 1 to 3 percent of sporadic cases of pulmonary fibrosis and 8 to 15 percent of familial cases of pulmonary fibrosis [25,87]. As noted above, families with pulmonary fibrosis and bone marrow hypoplasia have a very high likelihood of DC. (See 'Additional somatic features' above.)

Like DC, other causes of ILD may present in childhood and show evidence of pulmonary fibrosis. Unlike DC, these disorders are not associated with bone marrow hypoplasia, abnormally short telomeres, or pathogenic mutations affecting telomerase components. Discussions of the other causes of ILD are presented separately. (See "Approach to the infant and child with diffuse lung disease (interstitial lung disease)" and "Approach to the adult with interstitial lung disease: Clinical evaluation".)

MANAGEMENT — Patients with DC and telomere biology disorders (TBDs) should be managed at a center with expertise in these disorders.

Initial and ongoing screening — All patients diagnosed with DC, based on the above studies (see 'Diagnosis' above) should have a comprehensive assessment, including family and medical history and a thorough physical examination, performed in a dedicated center with expertise in management of patients with TBDs. Specific aspects of initial and ongoing screening include the following:

Assessment for bone marrow failure/myelodysplastic syndrome – Initial screening for bone marrow failure (BMF) and myelodysplastic syndrome (MDS) should include a complete blood count (CBC) with differential and mean corpuscular volume (MCV, which is typically elevated in patients with DC); reticulocyte count; and bone marrow aspirate/biopsy studies with cytogenetic assessments including G-banding metaphase karyotype analysis and fluorescence in situ hybridization (FISH) for specific abnormalities including 5q-, 7q-/monosomy 7, trisomy 8, and 20q-.

Ongoing screening and monitoring depends on the results of the initial screening:

For patients with normal CBC, normal bone marrow cellularity, and no cytogenetic abnormality, ongoing screening with annual CBC with differential is sufficient, with any significant change in the CBC warranting repeat bone marrow studies and more frequent monitoring.

For patients with moderate cytopenias not requiring transfusion support, CBC monitoring every three to six months and annual bone marrow studies are recommended. Any patient with cytogenetic abnormalities, morphologic features suggestive of worsening MDS such as excess blasts (see 'AML or MDS' below) or with severe cytopenias approaching transfusion thresholds, more intensive monitoring is required, with planning for therapies directed at cytopenias.

Screening and preventive care for solid tumors – Screening guidelines for solid tumors were established in 2015 for patients with DC and TBDs [71]. Of note, screening may need to begin earlier than general guidelines indicate for patients who have previously undergone hematopoietic cell transplantation (HCT), particularly for those affected by post-HCT graft-versus-host disease (GVHD). Guidelines for specific cancers include the following:

Squamous cell carcinoma of the head and neck – Thorough dental examinations (or oral surgery consultation as indicated, depending on the dentist's degree of experience with oral cancer) every six months beginning in early childhood (eg, age three to five years). Nasolaryngoscopy should be performed every one to three years beginning at age 10 by an otolaryngologist with specific experience in head and neck cancer. Preventive care education should focus on good oral hygiene as well as avoidance of smoking, oral tobacco products, and alcohol.

Anorectal cancer – Physical examination and stool for occult blood annually, beginning at age 12. In some centers, anorectal endoscopy screening begins at age 18. Human papilloma virus (HPV) vaccination should be given, because HPV may play a role in the increased anorectal cancer incidence in DC.

Esophageal cancer – Esophagoscopy screening is done every three to five years beginning by age 18. For patients with symptoms of esophageal dysfunction, screening should start earlier and should be repeated annually.

Liver cancer – Initial screening should include liver enzyme testing, and bilirubin testing, and right upper quadrant ultrasound. Ongoing screening should consist of annual liver enzyme and bilirubin assessments, with liver ultrasound repeated every three to five years. Additional assessments for other hepatic manifestations are noted below.

Preventative measures include avoidance of all alcohol consumption, and use of chelation therapy if iron overload from red blood cell transfusions has developed. (See "Iron chelators: Choice of agent, dosing, and adverse effects".)

Skin cancer – Annual examinations by a dermatologist. Preventative education includes training of the patient and family in skin evaluations and sun protection measures such as sun avoidance and use of sunscreen.

Gynecologic cancer – Annual gynecologic examinations beginning at age 16. As noted above, HPV vaccination should be administered due to the possible role of HPV in gynecologic cancers in individuals with DC [88,89] and is recommended for all children in the United States starting at nine years of age.

Lung cancer – Avoidance of smoking and other exposures that increase the risk of pulmonary fibrosis should be emphasized, as discussed below. Some experts offer chest radiography after age 40.

Pulmonary manifestations – Initial screening for pulmonary fibrosis should include pulse oximetry for Sp02 assessment, and, for patients over age seven who are able to comply, formal pulmonary function testing (PFT), including assessment of the diffusion capacity of the lung for carbon monoxide (DLCO). For patients younger than age seven in whom pulmonary abnormalities are suspected, a six-minute walk test may be performed. Contrast echocardiography ("bubble echo"), which assesses for right-to-left shunting, should be performed in any patient with cyanosis, clubbing, or PFT/DLCO testing suspicious for pulmonary arteriovenous malformation and/or hepatopulmonary syndrome.

PFTs should be repeated every three to five years if normal or annually if significant abnormalities are identified. The use of routine computed tomography (CT) scans for screening remains controversial in DC, given the concerns for radiation exposure; however, in patients with abnormal PFT/DLCO testing, CT may be useful to further define the extent of pulmonary fibrosis present. In patients who have undergone HCT, initial PFT/DLCO assessment is recommended one year after HCT to determine the frequency of ongoing follow-up.

Education to avoid exposures that increase the risk of pulmonary fibrosis is highly recommended for patients with DC, as well as for family members who are asymptomatic but possess telomere disorder-associated familial gene mutations. Exposures for which avoidance is recommended include the following [90]:

Tobacco or cannabis smoking and exposure to second-hand smoke

Drugs associated with lung dysfunction (eg, nitrofurantoin, amiodarone, busulfan)

Elective general anesthesia when possible, due to associated injury of lung epithelium

Occupational environments where exposure to high particulate air concentrations is likely, or use of filtering respirator masks in such environments

Respiratory illnesses, through adherence to immunization schedules and good hand-hygiene practices

Hepatic manifestations – Due to risks for portal hypertension and liver cirrhosis, all patients require an annual examination for liver size and measurement of liver enzymes, bilirubin, albumin, and prothrombin time. A baseline liver ultrasound should be obtained at the time of diagnosis and reassessed every three to five years. Ultrasound or magnetic resonance imaging (MRI) may be useful for detecting liver fibrosis. Doppler assessment of portal venous flow should be performed in patients with splenomegaly or with other clinical features that could indicate the presence of portal hypertension. Liver biopsy may be appropriate if significant abnormalities are identified by laboratory testing or imaging studies. Notably, patients receiving androgen therapy are at increased risk of developing hepatic dysfunction and should be monitored more closely. Preventative management focuses on avoidance of alcohol consumption and medications with known hepatic toxicities.

Endocrine manifestations Hormone deficiencies are not common in DC/TBD's and monitoring should be individualized. Patients who have undergone HCT and those are receiving androgen therapy should have an annual assessment by an endocrinologist because of the high risk of endocrine complications in these patients.

Thyroid – Some experts obtain baseline assessments of thyroid hormone function (including TSH), especially if HCT is anticipated.

Bone health – Poor bone health/osteopenia is common in patients with DC, and we obtain annual screening of serum calcium, magnesium, and 25-hydroxy vitamin D levels, and supplement any deficiencies.

At age 12 to 14, we perform initial dual-energy x-ray absorptiometry (DXA) screening and repeat the assessment every three to five years, depending on coexistent risk factors. Patients should be educated about signs and symptoms of fracture and avascular necrosis of the hip and shoulder joints.

Short stature – Patients with short stature should have assessment of growth hormone axis function by an endocrinologist, although most patients with DC who have short stature do not have growth hormone deficiency [91].

Gonadal function/fertility – While infertility is not a common feature of TBDs, patients with hypogonadism and/or pubertal delay should have an assessment by an endocrinologist for bone age, testosterone, LH, and FSH levels.

Women with DC or TBD typically have normal menarche, but they may need fertility assistance and they have a higher rate of pregnancy complications. A multi-disciplinary approach, including maternal-fetal medicine and hematology is helpful [54].

Additional screening – We also perform the following screening tests:

Eyes – Annual ophthalmology evaluations, with initial screening for epiphora and retinal changes including exudative retinopathy and neovascularization.

Hearing – Formal hearing examination, with follow-up audiologic assessment of any abnormalities.

Gastrointestinal – Upper and lower endoscopy, both for cancer screening as indicated above as well as for any patient with dysphagia suggestive of esophageal stricture or evidence of upper or lower gastrointestinal bleeding.

Genito-urinary – Urology consultation for any patient with urethral or hymenal strictures, phimosis, or hypogonadism.

Immunity – Assessment for immune deficiency, with initial screens that include quantitative immunoglobulin levels (IgE, IgA, IgM, and IgG), cellular immunology panel assessing absolute, T, B, and NK cell numbers/subsets, and lymphocyte proliferation to mitogens. The need for ongoing screening is determined by the initial screening results.

Neurology – Neurologic assessment, with initial brain MRI for any patient suspected of having HH syndrome (see 'Hoyeraal-Hreidarsson syndrome' above), and head CT to assess for intracranial calcifications in a patient suspected of having Revesz syndrome. (See 'Revesz syndrome' above.)

Mental health – Psychologic/psychiatric/social work assessment, with an initial assessment for all patients newly diagnosed with DC to determine the need for ongoing services to help with coping strategies related to the diagnosis of a chronic medical disease; formal assessment for neuropsychiatric disorders associated with disease; and assistance in developing an individualized education plan to maximize educational and occupational achievements. Ongoing screening should be determined based on individual needs.

Screening of family members and relatives — If a pathogenic gene mutation has been identified in the proband, first degree family members (especially siblings) should be tested as soon as feasible. This is important for identifying clinically occult disease that may require interventions, and for determining eligibility as an HCT donor should HCT be necessary for the index patient or other affected family members. Testing for known familial mutations can also be pursued for other interested family members in addition to siblings. Testing should be accompanied by counseling with a genetic counselor or clinician with expertise in DC.  

Prenatal testing is possible if needed, using cells obtained by chorionic villus sampling, amniocentesis, or cordocentesis. In vitro fertilization with prenatal genetic diagnosis (PGD) is another method utilized to detect disease or carrier status prior to implantation of sibling embryos. (See "Preimplantation genetic testing".)

If a pathogenic mutation has not been identified and the proband's diagnosis is based on short telomeres along with characteristic clinical features, we recommend telomere length analysis in both parents, along with in any first-degree relative with shared clinical features of DC. Notably, this testing should be done with the assistance of a genetic counselor to facilitate active discussion with the involved family members regarding the implications of test results. (See "Genetic counseling: Family history interpretation and risk assessment" and "Genetic testing", section on 'Ethical, legal, and psychosocial issues'.)

Treatment of specific complications

Treatment of bone marrow failure — As noted above, individuals with DC should have a bone marrow evaluation at the time of diagnosis (see 'Laboratory testing and bone marrow' above and 'Initial and ongoing screening' above). This may show hypocellularity and some degree of dyspoiesis [20]. Some individuals may have clonal cytogenetic abnormalities, myelodysplasia and/or acute leukemia.

For those with evidence of bone marrow hypocellularity and moderate to severe cytopenias, cytogenetic abnormalities, or dysplasia, referral to a transplant center with expertise in managing inherited bone marrow failure syndromes (IBMFS) is appropriate, in order to facilitate planning and review the indications for HCT. (See 'Hematopoietic cell transplantation (HCT)' below.)

Supportive care — Transfusions may be required for severe anemia or thrombocytopenia. We use a judicious approach, as extensive transfusions may be associated with worse outcomes from HCT, due to transfusional iron overload and/or alloimmunization against red blood cell or HLA antigens. Erythropoietin is generally not used due to concerns it may increase the risk of MDS or AML in this susceptible population. We limit the use of granulocyte colony-stimulating factor (G-CSF; filgrastim) to individuals with an absolute neutrophil count (ANC) below 200/microL or those with known active invasive bacterial or fungal infection and an ANC <1000/microL, due to concerns that G-CSF may also increase the risk of MDS and AML in patients with BMF syndromes.

As mentioned below, danazol may improve blood counts, although this improvement is not permanent and is lost with drug cessation; we might use danazol to improve blood counts if we anticipated a delay in starting HCT for a patient with DC, or if a patient were ineligible to receive HCT (see 'Therapies to enhance telomere function' below). Notably, the combination of growth factors with androgens should be avoided, as this led to splenic peliosis and rupture in two individuals with DC [20].

Hematopoietic cell transplantation (HCT) — Allogeneic HCT remains the only curative option for BMF in patients with DC, and our approach is to pursue HCT with the best available donor for all pediatric patients with DC who have BMF and who are transplant-eligible, based on organ function criteria. However, this approach has many risks [92]. Additionally, HCT does not treat the extra-hematopoietic complications of DC and related syndromes. HCT may actually increase the risk of non-hematologic malignancies. Importantly, potential sibling (or other related) donors must be evaluated for DC and demonstrated not to have the familial mutation. (See 'Screening of family members and relatives' above.)

Early studies of HCT in patients with DC used myeloablative preparative regimens, resulting in very poor short-term and long-term survival, with fewer than 30 percent of patients surviving 10 to 15 years after HCT [93]. While the sensitivity to genotoxic agents is similar to patients with Fanconi anemia (FA), the adverse sequelae differ. Individuals with DC who received standard myeloablative conditioning regimens had high rates of hepatic and pulmonary fibrosis, with late onset veno-occlusive disease of the liver [20]. As a result, reduced intensity conditioning (RIC) regimens are universally used for HCT in patients with DC.  

DC-specific RIC regimens have been published [94-96]. However, a 2016 systematic review of the published literature that included 109 patients who underwent HCT for DC showed that even for patients receiving conventional RIC regimens after the year 2000, five-year overall survival remained only 70 percent [97]. HCT using an unaffected matched sibling donor conveyed a significant survival advantage over receiving alternative donor HCT (92 to 58 percent), and performing HCT at age <20 years may also convey a survival advantage. Causes of death have included graft failure, sepsis, pulmonary fibrosis, hepatic veno-occlusive disease, GVHD, and solid tumors.

In 2014, a pilot study involving four patients with DC who underwent HCT from an unrelated donor demonstrated the efficacy of an alkylator- and radiation-free conditioning regimen consisting of fludarabine and alemtuzumab [98]. All patients experienced engraftment, there were no deaths, no patient developed significant organ toxicities, and only one patient developed mild chronic GVHD. This promising approach has been extended into a multicenter clinical trial, for which results are awaited [99].

Importantly, dermatologic, hepatic, and enteropathy manifestations of DC may be mistaken for GVHD in patients who have undergone HCT [93]. Biopsies of the affected tissues may help to distinguish these entities based on the extent of inflammatory infiltrates and pathognomonic features of GVHD such as crypt drop-out in gut biopsies or portal tract loss in liver biopsies. However the sensitivity and specificity of these findings in DC patients is not well established. Most modern regimens, including the fludarabine and alemtuzumab regimen, use T cell depletion of donor grafts to mitigate the risks of GVHD in patients with DC. This may be done in vivo (with alemtuzumab or anti-thymocyte globulin) or ex vivo (prior to graft infusion).

Additional details of HCT for patients with DC and other TBDs and transplant outcomes are presented separately. (See "Hematopoietic cell transplantation (HCT) for inherited bone marrow failure syndromes (IBMFS)", section on 'Dyskeratosis congenita'.)

Alternatives for those who do not have access to HCT (eg, due to comorbidities that preclude transplant, lack of a suitable donor, cost) or prefer not to pursue HCT include treatment with androgenic steroids and supportive care with transfusions and growth factors. (See 'Therapies to enhance telomere function' below and 'Supportive care' above.)

AML or MDS — DC is too rare to have high quality evidence supporting a disease-specific approach to treating AML or MDS. As with other inherited BMF conditions, pre-HCT chemotherapy is unlikely to be tolerated due to prolonged myelosuppression. Patients with low-grade MDS should proceed to HCT directly, although the optimal HCT regimen for such patients is yet to be defined. General management of AML or MDS in patients with BMF is covered in a separate topic. (See "Management and prognosis of Fanconi anemia", section on 'Hematologic neoplasms'.)

Solid tumors — There are no specific guidelines that suggest how to adjust the intensity of cytotoxic chemotherapy or radiation therapy in patients with DC, but anecdotal evidence suggests that reduced intensity is indicated to prevent prolonged cytopenias, pulmonary toxicity, hepatotoxicity, and, in the case of radiation therapy, severe local toxicity at the site of treatment [61]. Oncologists treating patients with DC for solid tumors should develop an individualized treatment plan in coordination with a BMF specialist with experience in treating patients with DC. Further information on these solid tumors including oral leukoplakia is presented separately. (See "Oral lesions".)

Treatment of non-malignant skin, nail, and hair lesions — Hyperkeratotic skin lesions of the palms and soles have been treated effectively with agents such as urea creams and/or salicylic acid creams. There are no treatments known to improve the skin dyspigmentation seen in many patients with DC, but moisturizers may be helpful in reducing the roughened texture of some of these skin areas. Patients with hyperhidrosis may benefit from anticholinergic therapy, although this should be done in conjunction with an experienced dermatologist. Minoxidil may help with thinning hair or alopecia, and the ophthalmic solution bimatoprost may help with eyelash regrowth [100]. Normal nail health is unlikely, but breakage can be reduced using nail lacquers to strengthen nails, avoidance of excessive detergent exposure/hand washing, and avoidance of artificial nails. Biotin may be of benefit in strengthening nails and hair. Additional information is provided separately:

Nail disorders – (See "Overview of nail disorders" and "Principles and overview of nail surgery".)

Skin hyperpigmentation – (See "Acquired hyperpigmentation disorders".)

Treatment of pulmonary fibrosis — Pulmonary fibrosis affects approximately one-fifth of individuals with DC, and 5 to 15 percent of individuals with pulmonary fibrosis have mutations in TERT or TERC. A whole exome sequencing study of 262 individuals with idiopathic pulmonary fibrosis reported that 11 percent of case subjects had an identifiable pathogenic variant in TERT, RTEL1, or PARN [67]. (See 'Additional somatic features' above.)

One of the most important aspects of treating patients with pulmonary fibrosis is ensuring that testing is sent to diagnose a telomere disorder, as this may have profound implications for treatment, as discussed separately. (See "Approach to the adult with interstitial lung disease: Diagnostic testing".)  

All individuals with DC and findings on PFTs or lung CT that suggest pulmonary fibrosis should be referred to a pulmonologist for further evaluation and management. Reports of patients with DC who have undergone lung transplantation for pulmonary fibrosis suggest that this approach is feasible, although complications may be increased compared to individuals without DC [101]. Other aspects of the management of pulmonary fibrosis are discussed in detail separately. (See "Treatment of idiopathic pulmonary fibrosis".)

Therapies to enhance telomere function — Therapy that restores normal telomere function is appealing because it has the potential to treat all organ system manifestations of DC. A variety of approaches to increasing telomerase activity are under investigation.

Androgen therapy – Patients with DC and related telomere biology disorders have long been treated with androgen hormone therapies, which have been observed to increase blood counts in patients with cytopenias. In a 2016 study, the androgen danazol was shown to be associated with consistent telomere elongation [14]. The study involved a prospective series of 27 individuals ages 17 and older who had at least one cytopenia associated with a telomere disorder other than classic DC, diagnosed based on short telomere length or genetic testing. Danazol (400 mg) was administered twice daily for two years. The study was stopped early after the first 11 patients reached the two-year evaluation because the effect on the prespecified primary endpoint (20 percent reduction in telomere shortening) was much more dramatic than expected. Telomere elongation was documented in 89 percent of patients at 12 months and 92 percent at 24 months. Hematologic responses were seen in 79 percent of patients at three months and 83 percent at 24 months. Stabilization of the DLCO occurred in all seven patients for whom pre- and post-treatment results were available. The most common adverse events were liver enzyme elevations (41 percent), muscle cramps (33 percent), edema (26 percent), and lipid abnormalities (26 percent). Among eight individuals for whom telomere measurements were performed after danazol was stopped, all reverted to telomere shortening, suggesting this therapy would need to be given indefinitely.

In a 2014 observational cohort study that included 16 younger patients (median age, 11 years), many of whom had classic DC features, the impact of androgen therapy was less pronounced. While 69 percent showed a hematologic response, none had changes in the expected, age-related telomere decline. Additionally, one patient developed a concerning hyperechoic liver lesion, and two patients developed splenic peliosis, which led to splenic rupture in one patient [102].

The difference in outcomes of these two studies suggests that more studies are needed to define whether favorable outcomes for androgen therapy are dependent upon the specific androgen used, with danazol showing the most benefit thus far, or whether benefit depends upon the population studied, with androgen therapy perhaps having a more favorable risk/benefit profile in older patients with milder genetic defects in telomerase function.

For patients who have BMF and meet eligibility criteria for HCT, including having a suitably matched related or unrelated donor, we use HCT rather than androgen therapy. Danazol use may be appropriate in patients with transfusion-dependent cytopenias in whom HCT cannot be performed safely if a long delay prior to HCT is expected. (See 'Treatment of bone marrow failure' above.)

For patients who have other organ dysfunction (including pulmonary fibrosis) associated with a TBD, androgen therapy should be restricted to prospective clinical trials. (See 'Treatment of pulmonary fibrosis' above.)

On danazol, blood counts need to be monitored closely (at least monthly initially), and close monitoring for adverse effects (eg, hepatotoxicity, lipid abnormalities, accelerated growth, splenic peliosis) is required [102,103]. Patients on androgen therapy require liver function assessments every three months, liver ultrasounds yearly, and yearly screens for dyslipidemia. Androgens should not be used in combination with growth factors, as this led to splenic peliosis and rupture in two individuals with DC [20]. (See 'Supportive care' above.)

Therapies targeting Wnt signaling – Preclinical studies using induced pluripotent stem (iPS) cells derived from patients with DC or animal models have suggested that upregulating signaling through the Wnt pathway in tissue-specific stem cells may ameliorate telomerase dysfunction in DC [104-106]. Known agonists of Wnt signaling, including lithium chloride, are being studied in these preclinical models, but clinical trials testing this approach in patients with telomere disorders have not been initiated.

Gene therapy – The possibility of gene therapy has been suggested for a number of conditions associated with loss of telomerase function, as well as aging; however, no strategy has reached the stage of clinical trials for gene correction of DC-related gene mutations [107]. Challenges of applying gene therapy to the TBDs include the multigenic origins of the disease that necessitate unique targeting strategies for each affected gene, the experience with most clinical gene correction strategies in targeting autosomal recessive or X-linked disorders (whereas DC is often an AD disease), and the reliance of most strategies on autologous gene correction in specific tissues such as liver and hematopoietic cells, which will not lead to gene correction in other tissues affected by telomerase dysfunction.

PROGNOSIS — The overall survival of individuals with DC has been reported at 49 years in the early 21st century [20]. This represents a significant improvement over survival rates for the 20th century (eg, age approximately mid-30s), which has been attributed to better management of organ system involvement and improved outcomes with hematopoietic cell transplantation (HCT). Additionally, the prognosis may be improved due to the inclusion of individuals with milder phenotypes who were diagnosed using telomere measurements and genetic testing. However overall survival continues to lag behind that seen in patients with other bone marrow failure (BMF) syndromes.

The primary cause of mortality in individuals with DC and related telomere disorders is BMF, its consequences (eg, bleeding, infection), or morbidity associated with its treatment using HCT [17]. Solid tumors, pulmonary fibrosis, and hepatic disease account for significant mortality as well.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Bone marrow failure syndromes".)

SUMMARY AND RECOMMENDATIONS

Telomeres – Telomeres are specialized structures at the ends of chromosomes that shorten with each cell division. In dyskeratosis congenita (DC) and related telomere biology disorders (TBDs), mutations in genes that maintain telomere length (table 1 and figure 1) lead to premature cell death, senescence, genomic instability, and impaired function in many organs and tissues. (See 'Pathophysiology' above.)

Clinical features – The spectrum of findings with DC and other TBDs is highly variable and can affect bone marrow, immunity, skin, lung, liver, and teeth (table 2 and picture 1). Some features typically appear in childhood, but pulmonary fibrosis and liver cirrhosis typically present in adulthood and may be the first presenting feature of TBDs other than DC. (See 'Clinical features' above.)

Diagnosis – Diagnosis of a new proband is generally based on clinical findings suggestive of a TBD, followed by identification of a pathogenic gene variant, using telomere length analysis, genetic testing, and bone marrow evaluation. (See 'Diagnosis' above.)

Differential diagnosis – The differential diagnosis includes other causes of inherited or acquired bone marrow failure (BMF) syndromes, hematologic malignancies, congenital anomalies, or interstitial lung disease. (See 'Differential diagnosis' above.)

Management – All patients with DC should have a comprehensive assessment in a center with expertise in management of patients with TBDs.

Surveillance – Patients require ongoing surveillance for BMF, myelodysplastic syndrome (MDS), certain solid tumors, pulmonary fibrosis, liver disease, thyroid function, osteopenia, and other manifestations. If a pathogenic variant was identified, siblings and other first degree family members should be tested promptly to identify clinically occult disease and to determine eligibility as a donor for hematopoietic cell transplantation (HCT). Prenatal testing is also possible, if needed. (See 'Initial and ongoing screening' above and 'Screening of family members and relatives' above.)

Bone marrow failure – Transfusions may be required for managing severe anemia or thrombocytopenia. We pursue HCT for all medically-eligible pediatric patients with DC who have BMF. However, HCT is associated with significant adverse effects, it does not treat the extra-hematopoietic complications of DC, and it may increase the risk of non-hematologic malignancies. (See 'Treatment of bone marrow failure' above.)

Danazol may be helpful for selected individuals who cannot undergo HCT, but androgens should not be used in combination with hematopoietic growth factors due to the risk of splenic rupture. (See 'Therapies to enhance telomere function' above.)

Other management – Treatment for hematopoietic neoplasms, solid tumors, skin and nail findings, and pulmonary fibrosis are discussed above and in separate topic reviews listed above. (See 'AML or MDS' above and 'Solid tumors' above and 'Treatment of non-malignant skin, nail, and hair lesions' above and 'Treatment of pulmonary fibrosis' above.)

  1. Barbaro PM, Ziegler DS, Reddel RR. The wide-ranging clinical implications of the short telomere syndromes. Intern Med J 2016; 46:393.
  2. Makarov VL, Hirose Y, Langmore JP. Long G tails at both ends of human chromosomes suggest a C strand degradation mechanism for telomere shortening. Cell 1997; 88:657.
  3. Aubert G, Baerlocher GM, Vulto I, et al. Collapse of telomere homeostasis in hematopoietic cells caused by heterozygous mutations in telomerase genes. PLoS Genet 2012; 8:e1002696.
  4. Alter BP, Baerlocher GM, Savage SA, et al. Very short telomere length by flow fluorescence in situ hybridization identifies patients with dyskeratosis congenita. Blood 2007; 110:1439.
  5. Lansdorp PM. Telomeres, stem cells, and hematology. Blood 2008; 111:1759.
  6. Allsopp RC, Morin GB, DePinho R, et al. Telomerase is required to slow telomere shortening and extend replicative lifespan of HSCs during serial transplantation. Blood 2003; 102:517.
  7. Goldman FD, Aubert G, Klingelhutz AJ, et al. Characterization of primitive hematopoietic cells from patients with dyskeratosis congenita. Blood 2008; 111:4523.
  8. Bertuch AA. The molecular genetics of the telomere biology disorders. RNA Biol 2016; 13:696.
  9. Stanley SE, Gable DL, Wagner CL, et al. Loss-of-function mutations in the RNA biogenesis factor NAF1 predispose to pulmonary fibrosis-emphysema. Sci Transl Med 2016; 8:351ra107.
  10. Venteicher AS, Abreu EB, Meng Z, et al. A human telomerase holoenzyme protein required for Cajal body localization and telomere synthesis. Science 2009; 323:644.
  11. Zhong F, Savage SA, Shkreli M, et al. Disruption of telomerase trafficking by TCAB1 mutation causes dyskeratosis congenita. Genes Dev 2011; 25:11.
  12. Nandakumar J, Bell CF, Weidenfeld I, et al. The TEL patch of telomere protein TPP1 mediates telomerase recruitment and processivity. Nature 2012; 492:285.
  13. Tummala H, Walne A, Collopy L, et al. Poly(A)-specific ribonuclease deficiency impacts telomere biology and causes dyskeratosis congenita. J Clin Invest 2015; 125:2151.
  14. Townsley DM, Dumitriu B, Liu D, et al. Danazol Treatment for Telomere Diseases. N Engl J Med 2016; 374:1922.
  15. Vulliamy T, Marrone A, Szydlo R, et al. Disease anticipation is associated with progressive telomere shortening in families with dyskeratosis congenita due to mutations in TERC. Nat Genet 2004; 36:447.
  16. Armanios M, Chen JL, Chang YP, et al. Haploinsufficiency of telomerase reverse transcriptase leads to anticipation in autosomal dominant dyskeratosis congenita. Proc Natl Acad Sci U S A 2005; 102:15960.
  17. Dokal I. Dyskeratosis congenita. Hematology Am Soc Hematol Educ Program 2011; 2011:480.
  18. Nelson ND, Bertuch AA. Dyskeratosis congenita as a disorder of telomere maintenance. Mutat Res 2012; 730:43.
  19. Alter BP, Rosenberg PS, Giri N, et al. Telomere length is associated with disease severity and declines with age in dyskeratosis congenita. Haematologica 2012; 97:353.
  20. Shimamura A, Alter BP. Pathophysiology and management of inherited bone marrow failure syndromes. Blood Rev 2010; 24:101.
  21. Jones M, Bisht K, Savage SA, et al. The shelterin complex and hematopoiesis. J Clin Invest 2016; 126:1621.
  22. Ojha J, Codd V, Nelson CP, et al. Genetic Variation Associated with Longer Telomere Length Increases Risk of Chronic Lymphocytic Leukemia. Cancer Epidemiol Biomarkers Prev 2016; 25:1043.
  23. Ballew BJ, Yeager M, Jacobs K, et al. Germline mutations of regulator of telomere elongation helicase 1, RTEL1, in Dyskeratosis congenita. Hum Genet 2013; 132:473.
  24. Simon AJ, Lev A, Zhang Y, et al. Mutations in STN1 cause Coats plus syndrome and are associated with genomic and telomere defects. J Exp Med 2016; 213:1429.
  25. Armanios MY, Chen JJ, Cogan JD, et al. Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med 2007; 356:1317.
  26. Parry EM, Alder JK, Qi X, et al. Syndrome complex of bone marrow failure and pulmonary fibrosis predicts germline defects in telomerase. Blood 2011; 117:5607.
  27. Gable DL, Gaysinskaya V, Atik CC, et al. ZCCHC8, the nuclear exosome targeting component, is mutated in familial pulmonary fibrosis and is required for telomerase RNA maturation. Genes Dev 2019; 33:1381.
  28. Walne AJ, Vulliamy T, Beswick R, et al. Mutations in C16orf57 and normal-length telomeres unify a subset of patients with dyskeratosis congenita, poikiloderma with neutropenia and Rothmund-Thomson syndrome. Hum Mol Genet 2010; 19:4453.
  29. Toufektchan E, Lejour V, Durand R, et al. Germline mutation of MDM4, a major p53 regulator, in a familial syndrome of defective telomere maintenance. Sci Adv 2020; 6:eaay3511.
  30. Nachmani D, Bothmer AH, Grisendi S, et al. Germline NPM1 mutations lead to altered rRNA 2'-O-methylation and cause dyskeratosis congenita. Nat Genet 2019; 51:1518.
  31. Glousker G, Touzot F, Revy P, et al. Unraveling the pathogenesis of Hoyeraal-Hreidarsson syndrome, a complex telomere biology disorder. Br J Haematol 2015; 170:457.
  32. Vulliamy TJ, Marrone A, Knight SW, et al. Mutations in dyskeratosis congenita: their impact on telomere length and the diversity of clinical presentation. Blood 2006; 107:2680.
  33. Savage SA, Stewart BJ, Weksler BB, et al. Mutations in the reverse transcriptase component of telomerase (TERT) in patients with bone marrow failure. Blood Cells Mol Dis 2006; 37:134.
  34. Vulliamy T, Marrone A, Goldman F, et al. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature 2001; 413:432.
  35. Xin ZT, Beauchamp AD, Calado RT, et al. Functional characterization of natural telomerase mutations found in patients with hematologic disorders. Blood 2007; 109:524.
  36. Walne AJ, Vulliamy T, Beswick R, et al. TINF2 mutations result in very short telomeres: analysis of a large cohort of patients with dyskeratosis congenita and related bone marrow failure syndromes. Blood 2008; 112:3594.
  37. Savage SA, Giri N, Baerlocher GM, et al. TINF2, a component of the shelterin telomere protection complex, is mutated in dyskeratosis congenita. Am J Hum Genet 2008; 82:501.
  38. Guo Y, Kartawinata M, Li J, et al. Inherited bone marrow failure associated with germline mutation of ACD, the gene encoding telomere protein TPP1. Blood 2014; 124:2767.
  39. Kocak H, Ballew BJ, Bisht K, et al. Hoyeraal-Hreidarsson syndrome caused by a germline mutation in the TEL patch of the telomere protein TPP1. Genes Dev 2014; 28:2090.
  40. Stuart BD, Choi J, Zaidi S, et al. Exome sequencing links mutations in PARN and RTEL1 with familial pulmonary fibrosis and telomere shortening. Nat Genet 2015; 47:512.
  41. Marrone A, Walne A, Tamary H, et al. Telomerase reverse-transcriptase homozygous mutations in autosomal recessive dyskeratosis congenita and Hoyeraal-Hreidarsson syndrome. Blood 2007; 110:4198.
  42. Gramatges MM, Qi X, Sasa GS, et al. A homozygous telomerase T-motif variant resulting in markedly reduced repeat addition processivity in siblings with Hoyeraal Hreidarsson syndrome. Blood 2013; 121:3586.
  43. Walne AJ, Vulliamy T, Marrone A, et al. Genetic heterogeneity in autosomal recessive dyskeratosis congenita with one subtype due to mutations in the telomerase-associated protein NOP10. Hum Mol Genet 2007; 16:1619.
  44. Vulliamy T, Beswick R, Kirwan M, et al. Mutations in the telomerase component NHP2 cause the premature ageing syndrome dyskeratosis congenita. Proc Natl Acad Sci U S A 2008; 105:8073.
  45. Heiss NS, Knight SW, Vulliamy TJ, et al. X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions. Nat Genet 1998; 19:32.
  46. Vulliamy TJ, Knight SW, Dokal I, Mason PJ. Skewed X-inactivation in carriers of X-linked dyskeratosis congenita. Blood 1997; 90:2213.
  47. Alder JK, Parry EM, Yegnasubramanian S, et al. Telomere phenotypes in females with heterozygous mutations in the dyskeratosis congenita 1 (DKC1) gene. Hum Mutat 2013; 34:1481.
  48. https://www.dcoutreach.org/ (Accessed on March 16, 2017).
  49. Gorgy AI, Jonassaint NL, Stanley SE, et al. Hepatopulmonary syndrome is a frequent cause of dyspnea in the short telomere disorders. Chest 2015; 148:1019.
  50. Kannengiesser C, Borie R, Ménard C, et al. Heterozygous RTEL1 mutations are associated with familial pulmonary fibrosis. Eur Respir J 2015; 46:474.
  51. Fedick AM, Shi L, Jalas C, et al. Carrier screening of RTEL1 mutations in the Ashkenazi Jewish population. Clin Genet 2015; 88:177.
  52. Kirwan M, Dokal I. Dyskeratosis congenita: a genetic disorder of many faces. Clin Genet 2008; 73:103.
  53. Dyskeratosis Congenita and Telomere Biology Disorders: Diagnosis and Management Guidelines, 1st edition, Savage SA, Cook EF. (Eds), Dyskeratosis Congenita Outreach, Inc, 2015. (Available online at www.dcoutreach.org/guidelines)
  54. Giri N, Alter BP, Savage SA, Stratton P. Gynaecological and reproductive health of women with telomere biology disorders. Br J Haematol 2021; 193:1238.
  55. Himes RW, Chiou EH, Queliza K, et al. Gastrointestinal Hemorrhage: A Manifestation of the Telomere Biology Disorders. J Pediatr 2021; 230:55.
  56. Tsilou ET, Giri N, Weinstein S, et al. Ocular and orbital manifestations of the inherited bone marrow failure syndromes: Fanconi anemia and dyskeratosis congenita. Ophthalmology 2010; 117:615.
  57. Giri N, Lee R, Faro A, et al. Lung transplantation for pulmonary fibrosis in dyskeratosis congenita: Case Report and systematic literature review. BMC Blood Disord 2011; 11:3.
  58. Atkinson JC, Harvey KE, Domingo DL, et al. Oral and dental phenotype of dyskeratosis congenita. Oral Dis 2008; 14:419.
  59. Jyonouchi S, Forbes L, Ruchelli E, Sullivan KE. Dyskeratosis congenita: a combined immunodeficiency with broad clinical spectrum--a single-center pediatric experience. Pediatr Allergy Immunol 2011; 22:313.
  60. Jonassaint NL, Guo N, Califano JA, et al. The gastrointestinal manifestations of telomere-mediated disease. Aging Cell 2013; 12:319.
  61. Savage SA. Dyskeratosis Congenita. 2009 Nov 12 [Updated 2016 May 26]. In: Pagon RA, Adam MP, Ardinger HH, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2017. Available from: https://www.ncbi.nlm.nih.gov/sites/books/NBK22301/
  62. Higgs C, Crow YJ, Adams DM, et al. Understanding the evolving phenotype of vascular complications in telomere biology disorders. Angiogenesis 2019; 22:95.
  63. Khincha PP, Bertuch AA, Agarwal S, et al. Pulmonary arteriovenous malformations: an uncharacterised phenotype of dyskeratosis congenita and related telomere biology disorders. Eur Respir J 2017; 49.
  64. Kelmenson DA, Hanley M. Dyskeratosis Congenita. N Engl J Med 2017; 376:1460.
  65. Knight S, Vulliamy T, Copplestone A, et al. Dyskeratosis Congenita (DC) Registry: identification of new features of DC. Br J Haematol 1998; 103:990.
  66. Calado RT. Telomeres in lung diseases. Prog Mol Biol Transl Sci 2014; 125:173.
  67. Petrovski S, Todd JL, Durheim MT, et al. An Exome Sequencing Study to Assess the Role of Rare Genetic Variation in Pulmonary Fibrosis. Am J Respir Crit Care Med 2017; 196:82.
  68. Richeldi L, Collard HR, Jones MG. Idiopathic pulmonary fibrosis. Lancet 2017.
  69. Alter BP, Giri N, Savage SA, et al. Malignancies and survival patterns in the National Cancer Institute inherited bone marrow failure syndromes cohort study. Br J Haematol 2010; 150:179.
  70. Alter BP, Giri N, Savage SA, Rosenberg PS. Cancer in dyskeratosis congenita. Blood 2009; 113:6549.
  71. Alter BP, Savage SA. Cancer in Dyskeratosis Congenita. In: Dyskeratosis Congenita and Telomere Biology Disorders: Diagnosis and Management Guidelines, 1st edition, Savage SA, Cook EF (Eds), Dyskeratosis Congenita Outreach, Inc, 2015.
  72. Alter BP, Giri N, Savage SA, Rosenberg PS. Cancer in the National Cancer Institute inherited bone marrow failure syndrome cohort after fifteen years of follow-up. Haematologica 2018; 103:30.
  73. Horiguchi N, Kakizaki S, Iizuka K, et al. Hepatic Angiosarcoma with Dyskeratosis Congenita. Intern Med 2015; 54:2867.
  74. Olson TS, Chan ES, Paessler ME, et al. Liver failure due to hepatic angiosarcoma in an adolescent with dyskeratosis congenita. J Pediatr Hematol Oncol 2014; 36:312.
  75. Khincha PP, Savage SA. Neonatal manifestations of inherited bone marrow failure syndromes. Semin Fetal Neonatal Med 2016; 21:57.
  76. Knight SW, Heiss NS, Vulliamy TJ, et al. Unexplained aplastic anaemia, immunodeficiency, and cerebellar hypoplasia (Hoyeraal-Hreidarsson syndrome) due to mutations in the dyskeratosis congenita gene, DKC1. Br J Haematol 1999; 107:335.
  77. Burris AM, Ballew BJ, Kentosh JB, et al. Hoyeraal-Hreidarsson Syndrome due to PARN Mutations: Fourteen Years of Follow-Up. Pediatr Neurol 2016; 56:62.
  78. Lim BC, Yoo SK, Lee S, et al. Hoyeraal-Hreidarsson syndrome with a DKC1 mutation identified by whole-exome sequencing. Gene 2014; 546:425.
  79. Crow YJ, McMenamin J, Haenggeli CA, et al. Coats' plus: a progressive familial syndrome of bilateral Coats' disease, characteristic cerebral calcification, leukoencephalopathy, slow pre- and post-natal linear growth and defects of bone marrow and integument. Neuropediatrics 2004; 35:10.
  80. Savage SA. Connecting complex disorders through biology. Nat Genet 2012; 44:238.
  81. Walne AJ, Bhagat T, Kirwan M, et al. Mutations in the telomere capping complex in bone marrow failure and related syndromes. Haematologica 2013; 98:334.
  82. Polvi A, Linnankivi T, Kivelä T, et al. Mutations in CTC1, encoding the CTS telomere maintenance complex component 1, cause cerebroretinal microangiopathy with calcifications and cysts. Am J Hum Genet 2012; 90:540.
  83. Keller RB, Gagne KE, Usmani GN, et al. CTC1 Mutations in a patient with dyskeratosis congenita. Pediatr Blood Cancer 2012; 59:311.
  84. Dokal I, Vulliamy T, Mason P, Bessler M. Clinical utility gene card for: Dyskeratosis congenita - update 2015. Eur J Hum Genet 2015; 23.
  85. Baerlocher GM, Lansdorp PM. Telomere length measurements in leukocyte subsets by automated multicolor flow-FISH. Cytometry A 2003; 55:1.
  86. Babushok DV, Grignon AL, Li Y, et al. Disrupted lymphocyte homeostasis in hepatitis-associated acquired aplastic anemia is associated with short telomeres. Am J Hematol 2016; 91:243.
  87. Armanios M. Telomeres and age-related disease: how telomere biology informs clinical paradigms. J Clin Invest 2013; 123:996.
  88. Alter BP, Giri N, Savage SA, et al. Squamous cell carcinomas in patients with Fanconi anemia and dyskeratosis congenita: a search for human papillomavirus. Int J Cancer 2013; 133:1513.
  89. Alter BP, Giri N, Pan Y, et al. Antibody response to human papillomavirus vaccine in subjects with inherited bone marrow failure syndromes. Vaccine 2014; 32:1169.
  90. Garcia CK, Armanios M. Pulmonary Fibrosis. In: Dyskeratosis Congenita and Telomere Biology Disorders: Diagnosis and Management Guidelines, 1st edition, Savage SA, Cook EF (Eds), Dyskeratosis Congenita Outreach, Inc, 2015.
  91. Kossiva L, Vartzelis G, Harisi M, et al. Too short stature, too many stigmata. BMJ Case Rep 2010; 2010.
  92. Fioredda F, Iacobelli S, Korthof ET, et al. Outcome of haematopoietic stem cell transplantation in dyskeratosis congenita. Br J Haematol 2018; 183:110.
  93. Dietz AC, Mehta PA, Vlachos A, et al. Current Knowledge and Priorities for Future Research in Late Effects after Hematopoietic Cell Transplantation for Inherited Bone Marrow Failure Syndromes: Consensus Statement from the Second Pediatric Blood and Marrow Transplant Consortium International Conference on Late Effects after Pediatric Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2017; 23:726.
  94. Dietz AC, Orchard PJ, Baker KS, et al. Disease-specific hematopoietic cell transplantation: nonmyeloablative conditioning regimen for dyskeratosis congenita. Bone Marrow Transplant 2011; 46:98.
  95. Nishio N, Takahashi Y, Ohashi H, et al. Reduced-intensity conditioning for alternative donor hematopoietic stem cell transplantation in patients with dyskeratosis congenita. Pediatr Transplant 2011; 15:161.
  96. Ayas M, Nassar A, Hamidieh AA, et al. Reduced intensity conditioning is effective for hematopoietic SCT in dyskeratosis congenita-related BM failure. Bone Marrow Transplant 2013; 48:1168.
  97. Barbaro P, Vedi A. Survival after Hematopoietic Stem Cell Transplant in Patients with Dyskeratosis Congenita: Systematic Review of the Literature. Biol Blood Marrow Transplant 2016; 22:1152.
  98. Lehmann LE, Williams DA, London WB, et al. Full Donor Myeloid Engraftment with Minimal Toxicity in Dyskeratosis Congenita Patients Undergoing Allogeneic Bone Marrow Transplantation without Radiation or Alkylating Agents. Blood 2014; 124:2941.
  99. https://clinicaltrials.gov/ct2/results?term=NCT01659606 (Accessed on March 22, 2017).
  100. Nambudiri VE, Cowen EW. Dermatologic Manifestations in Dyskeratosis Congenita. In: Dyskeratosis Congenita and Telomere Biology Disorders: Diagnosis and Management Guidelines, 1st edition, Savage SA, Cook EF (Eds), Dyskeratosis Congenita Outreach, Inc, 2015.
  101. Silhan LL, Shah PD, Chambers DC, et al. Lung transplantation in telomerase mutation carriers with pulmonary fibrosis. Eur Respir J 2014; 44:178.
  102. Khincha PP, Wentzensen IM, Giri N, et al. Response to androgen therapy in patients with dyskeratosis congenita. Br J Haematol 2014; 165:349.
  103. Islam A, Rafiq S, Kirwan M, et al. Haematological recovery in dyskeratosis congenita patients treated with danazol. Br J Haematol 2013; 162:854.
  104. Yang TB, Chen Q, Deng JT, et al. Mutual reinforcement between telomere capping and canonical Wnt signalling in the intestinal stem cell niche. Nat Commun 2017; 8:14766.
  105. Woo DH, Chen Q, Yang TL, et al. Enhancing a Wnt-Telomere Feedback Loop Restores Intestinal Stem Cell Function in a Human Organotypic Model of Dyskeratosis Congenita. Cell Stem Cell 2016; 19:397.
  106. Gu BW, Apicella M, Mills J, et al. Impaired Telomere Maintenance and Decreased Canonical WNT Signaling but Normal Ribosome Biogenesis in Induced Pluripotent Stem Cells from X-Linked Dyskeratosis Congenita Patients. PLoS One 2015; 10:e0127414.
  107. Jäger K, Walter M. Therapeutic Targeting of Telomerase. Genes (Basel) 2016; 7.
Topic 100663 Version 21.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟