Next Article in Journal
DNA Damage and Repair in Pancreatic Cancer—The Latest Findings
Previous Article in Journal
G4 Oligonucleotide-Based Chaperones of Heterogeneous Nuclear Ribonucleoprotein A1
Previous Article in Special Issue
Ocular Melanoma: A Comprehensive Review with a Focus on Molecular Biology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Beyond Hematologic Malignancies: Colorectal Cancer as a Solid Tumor Manifestation of Inherited Bone Marrow Failure Syndromes

1
Department of Health Sciences, Università Del Piemonte Orientale, 28100 Novara, Italy
2
Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40292, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(20), 10105; https://doi.org/10.3390/ijms262010105
Submission received: 27 August 2025 / Revised: 13 October 2025 / Accepted: 15 October 2025 / Published: 17 October 2025
(This article belongs to the Collection Latest Review Papers in Molecular Oncology)

Abstract

Inherited Bone Marrow Failure Syndromes (IBMFS) encompass a group of rare genetic disorders characterized by intrinsic hematopoietic stem cell defects, leading to impaired hematopoiesis and increased predisposition to malignancies, particularly hematologic cancers. As advances in supportive care and hematopoietic stem cell transplantation have extended patient survival, there is growing recognition of an elevated risk of solid tumors, including colorectal cancer (CRC), within this population. Epidemiologic data, although limited by small cohort sizes, suggest the need for earlier and more intensive CRC surveillance protocols tailored to IBMFS patients, who tend to develop CRC at younger ages compared to the general population. Among IBMFS, the most robust association with CRC has been reported in Diamond–Blackfan anemia syndrome (DBAS) and Fanconi anemia (FA), while emerging evidence suggests a potential link in dyskeratosis congenita (DC) and Shwachman–Diamond syndrome (SDS). The pathophysiological basis involves defective DNA repair mechanisms, telomere dysfunction, ribosomal protein abnormalities, and impaired cellular stress responses, each contributing to genomic instability and malignant transformation. The understanding of the molecular mechanisms underpinning the association between IBMFS and CRC may provide a foundation for future targeted prevention and surveillance strategies and offer broader insights into colorectal carcinogenesis.

1. Introduction

Inherited bone marrow failure syndromes (IBMFS) constitute a heterogeneous group of rare genetic disorders characterized by defective hematopoiesis, varying degrees of bone marrow failure, and a markedly increased predisposition to malignancy [1,2,3,4]. These syndromes, including Diamond–Blackfan anemia syndrome (DBAS), Fanconi anemia (FA), dyskeratosis congenita (DC) and Shwachman–Diamond syndrome (SDS), collectively affect approximately 1 in 100,000 individuals worldwide [5,6,7]. While historically recognized primarily for their hematological manifestations and their elevated risk of hematologic malignancies, mounting evidence reveals a concerning pattern of increased solid tumor incidence (Table 1) that warrants both clinical attention and mechanistic understanding [8,9,10,11]. The cancer predisposition in IBMFS appears to be intrinsic to the fundamental cellular processes disrupted in these conditions, with ribosomal dysfunction, telomere maintenance defects, and DNA repair pathway abnormalities each contributing distinct oncogenic vulnerabilities. In DBAS, haploinsufficiency of ribosomal protein (RP) genes leads to ribosomal stress and aberrant p53 activation, creating a cellular environment that paradoxically promotes both growth inhibition and oncogenic transformation [12]. Conversely, the genomic instability observed in Fanconi anemia and dyskeratosis congenita arises from defects in DNA repair and telomere maintenance, respectively, driving carcinogenesis through accumulated genetic damage [13,14].
This broad solid tumor susceptibility has transformed the clinical management paradigm for IBMFS patients, necessitating comprehensive cancer surveillance protocols that extend throughout the patient’s lifetime and encompass multiple organ systems. Importantly, the cancer predisposition associated with germline pathogenic variants in FA genes is not limited to individuals with classic biallelic mutations manifesting as Fanconi anemia. Heterozygous carriers of pathogenic variants in FA pathway genes—most notably FANCD1/BRCA2 and FANCN/PALB2—are at increased risk for various cancers, particularly breast, ovarian, and pancreatic malignancies [15,35]. Although the link with CRC remains less definitive, some studies suggest a potential association [36,37,38]. Emerging evidence also points to additional FA genes, such as RAD51D, FANCM, and FANCJ/BRIP1, as contributors to cancer risk in the heterozygous state [15,39]. The underlying mechanism likely involves haploinsufficiency or partial disruption of DNA interstrand crosslink (ICL) repair, leading to genomic instability even in the absence of overt marrow failure.
Among the spectrum of solid tumors associated with IBMFS, colorectal cancer (CRC) has emerged as a particularly significant clinical concern, especially in patients with DBAS. Patients with DBAS have a 4.8-fold increased relative risk of developing cancer compared to the general population, with a striking 45-fold increased relative risk for CRC [28]. This association is particularly remarkable given that CRC typically manifests in IBMFS patients at ages decades younger than the general population, often occurring in the third and fourth decades of life when standard screening protocols are not routinely implemented [27,40].
Colorectal cancer is the third most commonly diagnosed cancer worldwide and the second leading cause of cancer-related death [41]. As CRC incidence rises worldwide and survival of IBMFS patients improves, understanding cancer risk in this population becomes increasingly relevant.
The heightened susceptibility to CRC in IBMFS patients carries important and wide-ranging clinical consequences. First, the early onset of CRC in IBMFS patients necessitates a reevaluation of existing surveillance protocols, as standard age-based screening guidelines are inadequate for identifying high-risk individuals within this population. Second, elucidating the molecular pathways driving CRC in IBMFS may yield novel insights into the mechanisms of colorectal carcinogenesis more broadly, potentially uncovering new biomarkers or therapeutic targets [42]. Third, the treatment of CRC in IBMFS patients poses unique challenges, given their underlying hematologic fragility and potential hypersensitivity to DNA-damaging agents, which complicates the use of conventional chemotherapeutic regimens.
Despite growing recognition of the association between IBMFS and CRC, substantial knowledge gaps remain, particularly concerning the precise molecular mechanisms that connect bone marrow failure to colorectal carcinogenesis. As our understanding of IBMFS continues to evolve from isolated hematologic disorders to complex cancer predisposition syndromes, the development of comprehensive, multidisciplinary care strategies becomes essential to optimize outcomes for these patients throughout their lifespans. This narrative review synthesizes current evidence regarding the link between IBMFS and CRC risk, examining the epidemiological data, molecular mechanisms, and clinical implications for patient care. Only articles in English published between 1995 and July 2025 available on PubMed were considered. The literature search included keywords such as inherited bone marrow failure syndromes, Fanconi anemia, Diamond–Blackfan anemia, dyskeratosis congenita, and Shwachman–Diamond syndrome, alone or in combination with colorectal cancer. By integrating insights from registry observations, functional studies, and clinical research, we discuss the emerging understanding of how fundamental cellular processes disrupted in IBMFS contribute to colorectal carcinogenesis.

2. Overview of IBMFS

The four major IBMFS—FA, DC, DBAS and SDS—collectively account for approximately 30% of pediatric bone marrow failure cases [43]. With advancements in genetic testing and improved survival rates of affected individuals into adulthood, IBMFS are now increasingly recognized in adult populations. However, delayed diagnosis in adults remains a significant clinical challenge, often due to the heterogeneous presentations, subtle phenotypic features, and overlapping symptoms with acquired bone marrow disorders [44].
The National Cancer Institute (NCI) Inherited Bone Marrow Failure Syndromes (IBMFS) cohort study, initiated in 2002, is the largest prospective study of its kind, enrolling patients with FA, DC, DBAS, and SDS under a unified, comprehensive protocol. After fifteen years of follow-up, this cohort has provided crucial insights into cancer incidence patterns across IBMFS and has documented that patients with these syndromes have high risks of cancer development, with distinct patterns of malignancy for each syndrome [17]. Additional important registries include the Diamond–Blackfan Anemia Registry of North America, the International Fanconi Anemia Registry, and various national registries that have contributed to our understanding of cancer risks in IBMFS [21,27,45]. These registries have been instrumental in identifying CRC as an emerging cancer risk across multiple IBMFS, and in highlighting the need for specific cancer surveillance recommendations.

2.1. Fanconi Anemia

Fanconi anemia is caused by germline biallelic mutations in one of the 22 currently recognized FANC genes (FANCA-W), which encode proteins involved in the DNA ICL repair pathway [46,47]. Of these, mutations in 20 genes follow an autosomal recessive inheritance pattern, whereas FANCB is X-linked and FANCR/RAD51 may harbor a heterozygous variant that exerts a dominant-negative effect [48,49]. The most commonly affected gene is FANCA, accounting for approximately 60–70% of cases [50]. The FA pathway is essential for maintaining genomic stability by repairing DNA ICLs through homologous recombination. Defects in this pathway lead to chromosomal instability, hypersensitivity to DNA crosslinking agents, and characteristic chromosomal breakage patterns when cells are exposed to mitomycin C or diepoxybutane [51]. Patients with FA have a dramatically increased cancer risk, with a cumulative incidence of 20–30% by age 40 years [21,52]. The malignancy spectrum includes acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), and solid tumors, particularly squamous cell carcinomas of the head and neck, gynecologic tract, and gastrointestinal system [16,17,53,54]. Solid tumors typically occur later in life than hematological features, with head and neck squamous cell carcinoma (HNSCC) being the most frequently reported, showing a 500-fold increased risk [21,55,56]. Hepatocellular carcinoma and breast cancer are also reported with increased frequency [15,57]. Notably, biallelic mutations in certain FA pathway genes, such as FANCS/BRCA1, FANCO/RAD51C, FANCR/RAD51, and FANCM can cause an FA-like disorder that shares the cancer predisposition of classical FA and, in some cases, the associated developmental abnormalities, but lacks bone marrow failure [58]. FANCM is not essential for the formation or stabilization of the FA core complex, and both monoallelic and biallelic loss-of-function mutations have been linked to an increased risk of breast cancer, with biallelic mutations likely conferring a stronger effect [59,60].
While historically underrecognized, CRC has emerged as a significant solid tumor risk in FA patients. Large registry studies have documented CRC cases in FA cohorts, although precise incidence rates remain difficult to establish due to the rarity of the syndrome and competing mortality from bone marrow failure and other malignancies. Case reports describe CRC development in FA patients as early as the third and fourth decades of life, representing a substantially younger age of onset compared to sporadic CRC [61]. The defective DNA repair mechanisms characteristic of FA likely contribute to the increased CRC susceptibility, as ICL repair defects can lead to accumulation of mutations in colorectal epithelium.
Of particular relevance to CRC risk assessment is the emerging understanding of cancer susceptibility in heterozygous carriers of FA pathogenic variants. While most heterozygous carriers do not appear to have significantly increased cancer risks [62,63], susceptibility to cancer, including CRC, is well established for carriers of heterozygous pathogenic variants in FANCS/BRCA1, FANCD1/BRCA2, FANCN/PALB2, FANCJ/BRIP1, and FANCO/RAD51C [62,64]. Additional FA pathway genes, including FANCE and FANCM, have been implicated in familial CRC cohorts [61,65,66].

2.2. Dyskeratosis Congenita

Dyskeratosis congenita results from mutations in genes that encode components of the telomerase complex or telomere-associated proteins essential for telomere length maintenance [67]. The most commonly affected genes include DKC1 and POLA1 (X-linked), TERC, TERT, and TINF2 (autosomal dominant), and NOP10, NHP2, and WRAP53 (autosomal recessive) [23,24]. These mutations lead to accelerated telomere shortening, premature cellular senescence, and stem cell dysfunction. The resulting genomic instability and impaired cellular proliferation contribute to both the bone marrow failure phenotype and cancer predisposition [68,69]. Dyskeratosis congenita manifests with a classic triad of mucocutaneous features—reticulate skin pigmentation, nail dystrophy, and oral leukoplakia—but these features may be subtle or absent. The systemic nature of the disease reflects the impact of telomere dysfunction on high-turnover tissues. Bone marrow failure is the most common life-threatening complication, while other organ systems, including the lungs, liver, and gastrointestinal tract, may also be affected [70].
Patients with DC have a high propensity for developing malignancies, that occur in approximately 40–50% of patients [25,71]. The cancer spectrum includes both hematologic malignancies (AML, MDS, Hodgkin lymphoma) and solid tumors. Solid tumors characteristically include squamous cell carcinomas of the head and neck, lung, cervix, and anus [16,17,72]. Colorectal cancer has been reported in DC patients, though data remain limited due to the syndrome’s rarity. These cancers are often aggressive and difficult to treat due to the underlying tissue fragility and impaired tissue healing associated with DC [73]. One illustrative case involved a male patient with a confirmed DKC1 mutation who developed rectal adenocarcinomas at ages 16 and 18, without a family history of CRC [74]. This case highlights not only the early onset and aggressive nature of CRC in DC but also the importance of surveillance in at-risk individuals.

2.3. Diamond–Blackfan Anemia Syndrome

Diamond–Blackfan anemia syndrome (previously Diamond–Blackfan anemia, DBA) is a rare IBMFS with an estimated incidence of 5 to 7 cases per million live births [6,75]. Clinically, DBAS presents most often in infancy with macrocytic anemia, and is frequently associated with congenital anomalies (such as craniofacial, upper limb, and cardiac defects) and with an elevated lifetime risk of malignancies, most notably MDS and AML [76]. The pathophysiology of DBAS is fundamentally rooted in defective ribosome biogenesis, establishing it as the prototypical ribosomopathy. The disorder results from heterozygous mutations in genes encoding RPs, affecting both large and small ribosomal subunit components. The most commonly implicated genes include RPS19 (25% of cases), RPL5, RPL11, RPL35A, RPS10, RPS17, RPS24, and RPS26, among others [26]. These mutations lead to RP haploinsufficiency, which disrupts the normal assembly and function of ribosomes [26,77]. The molecular cascade begins with impaired ribosome biogenesis, which triggers nucleolar stress and activates the p53 pathway [78]. This activation results in enhanced apoptosis specifically in erythroid progenitor cells, leading to the characteristic erythroid failure observed in DBAS [79]. The tissue-specific manifestation of this ribosomal defect in erythropoiesis, despite ribosomes being essential for protein synthesis in all cells, remains an area of active investigation. Current hypotheses suggest that erythroid precursors may be particularly vulnerable to ribosomal stress due to their high proliferation rate and intensive protein synthesis requirements during hemoglobin production [80,81].
Registry data demonstrate that patients with DBAS have a 4.8-fold higher relative risk of developing malignancies compared to the general population, with an overall cumulative cancer incidence of 13.7% by age 45 years [9]. The two most prevalent solid tumors in DBAS patients are CRC and osteogenic sarcoma [82]. The Diamond–Blackfan Anemia Registry of North America (DBAR), the largest established DBA cohort with prospective follow-up since 1991, has provided the most comprehensive data on CRC incidence in DBA. Registry data demonstrate that CRC represents a significant and early-onset malignancy risk, with a median age of onset of 41 years (range 20–51 years) [28]. This early onset has prompted the development of preliminary CRC screening recommendations for DBAS patients, including consideration of colonoscopic screening beginning at age 20 years [6]. For patients with normal findings on initial colonoscopy, a follow-up interval of every five years is advised. In cases where abnormalities are detected, follow-up should adhere to established colorectal screening guidelines [83]. In patients who have undergone hematopoietic stem cell transplantation (HSCT) prior to age 20, it has been recommended that the first screening colonoscopy be performed within one year of HSCT, if clinically feasible, or prior to age 20 [6,28].
Among RP genes associated with DBAS, RPS20 represents a notable exception as germline heterozygous mutations in this gene are not typically associated with hematologic manifestations but rather confer a predisposition to cancer, particularly to hereditary nonpolyposis colorectal carcinoma without mismatch-repair deficiency. A truncating germline mutation (c.147dupA p.Val50Serfs*23) in RPS20 was identified in a four-generation Finnish pedigree with multiple cases of CRC [84]. No tumor showed loss of the wild-type allele, indicating that heterozygosity was sufficient to increase CRC risk. The mutation caused a defect in pre-ribosomal RNA maturation, as expected for DBAS genes [84]. Subsequent case series and screening studies identified additional RPS20 null and pathogenic missense mutations in early-onset CRC patients, but not in controls [85,86,87,88]. Interestingly, two missense variants affecting codon 84, both with a deleterious effect on protein function, have been reported in two DBAS patients who presented with transfusion-dependent anemia during the first month of life [89].

2.4. Shwachman–Diamond Syndrome

Shwachman–Diamond syndrome is a rare autosomal recessive disorder primarily characterized by exocrine pancreatic insufficiency, neutropenia, and skeletal abnormalities [90]. Approximately 90% of patients with SDS harbor biallelic mutations in the SBDS gene, which encodes a protein involved in ribosome biogenesis and mitotic spindle stabilization. SBDS mutations lead to defective ribosome maturation, impaired protein synthesis, and cellular dysfunction, placing SDS among the group of genetic diseases known as ribosomopathies. Additional genes, including DNAJC21, SRP54, and EFL1, have been identified in SBDS-negative cases [30]. Under normal conditions, SBDS, in cooperation with the GTPase EFL1, catalyzes the release of eIF6, which otherwise blocks the joining of 40S and 60S subunits. When SBDS is deficient or dysfunctional, eIF6 remains bound to the 60S subunit, preventing proper ribosome assembly and stalling translation initiation. This defect leads to inefficient ribosome formation and a global reduction in translation, resulting in hematopoietic failure [91,92].
Patients with SDS demonstrate a substantially increased predisposition to myeloid malignancies, particularly MDS or AML, with a cumulative lifetime risk probably exceeding 30% [93,94,95]. Data on gastrointestinal malignancies and other solid tumor risks have been less well-characterized due to the syndrome’s rarity and patient survival patterns [17,32,33,34,96].
Recent evidence suggests that germline pathogenic variants in SBDS may contribute to solid tumor development also in the heterozygous state. A large-scale study of 17,904 Chinese lung cancer patients identified SBDS as the most frequently mutated gene among cancer-susceptibility genes, with a prevalence of 1.37% [97]. The SBDS c.184A>T (p.Lys62*) pathogenic variant demonstrated a significant association with increased lung cancer risk, showing an odds ratio of 4.8 in case–control analysis compared to the gnomAD East Asian population [97]. The same germline variant was reported in one patients with malignant pleural mesothelioma [98] and in a pediatric patient with Ewing sarcoma [99]. These findings raise the possibility that heterozygous SBDS pathogenic variants may be predisposed to a broader range of solid tumors. In this context, preliminary data from our cohort of 292 patients with early-stage colorectal tumors identified two cases of heterozygous pathogenic variants in SBDS (manuscript in preparation). These findings indicate that heterozygous SBDS variants may contribute to the genetic predisposition to solid tumors, including CRC.

3. Molecular Mechanisms Underlying Colorectal Carcinogenesis in IBMFS

The pathophysiological mechanisms underpinning the increased CRC risk in IBMFS involve complex interactions between defective cellular pathways, genomic instability, and disrupted homeostatic processes. Each syndrome exhibits distinct molecular abnormalities that may contribute to colorectal carcinogenesis through overlapping yet syndrome-specific mechanisms (Figure 1).

3.1. DNA Repair Defects and Genomic Instability

Fanconi anemia represents a paradigmatic example of how inherited defects in DNA repair machinery can predispose individuals to cancer development through genomic instability. The FA pathway, comprising at least 22 complementation groups (FANCA-FANCW), constitutes a specialized DNA repair network that maintains genomic integrity primarily through the resolution of DNA ICLs [100]. The FA pathway is activated through a well-coordinated sequence of molecular events, initiated by the recognition of DNA ICLs during replication. The FA core complex, a multisubunit E3 ubiquitin ligase, is composed of FANCA, FANCB, FANCC, FANCE, FANCF, FANCG, and FANCL [101]. The catalytic activity resides in the FANCL subunit, which, in cooperation with the E2 ubiquitin-conjugating enzyme UBE2T, mediates the monoubiquitination of the FANCD2–FANCI (ID2) complex. This monoubiquitination event is a pivotal step in activating the DNA damage response to ICLs [102]. The monoubiquitinated ID2 complex serves as a platform for the recruitment of downstream effector proteins, including nucleases (e.g., FANCP/SLX4 and FANCQ/ERCC4), helicases (e.g., FANCJ/BRIP1 and FANCM), and homologous recombination factors (e.g., FANCD1/BRCA2, FANCN/PALB2, and FANCO/RAD51C) [102,103,104].
Recent genomic analyses have revealed that FA pathway deficiency creates a distinctive mutational signature characterized by increased numbers of structural variants, particularly small deletions [13]. This signature reflects the faulty response to unrepaired ICLs, which can lead to replication fork collapse and formation of DNA double-strand breaks. In FA cells these lesions are repaired through error-prone repair mechanisms, such as non-homologous end joining and microhomology-mediated end joining, which contribute to genomic instability [105,106]. Dysfunctions in the FA pathway compromise the cellular response to endogenous DNA damaging agents, particularly reactive aldehydes produced through normal metabolic processes. For example, acetaldehyde—generated during alcohol metabolism—can spontaneously react with DNA to form ICLs [107].
At the molecular level, deficiency in FA pathway components in colorectal epithelium creates a permissive environment for DNA damage that drives carcinogenesis. The continuous exposure to dietary and metabolic aldehydes, combined with the inability to repair resulting ICLs, leads to the accumulation of mutations in critical tumor suppressor genes and oncogenes [108]. Additionally, the chromosomal instability associated with FA may promote aneuploidy and loss of heterozygosity events that accelerate the progression from adenoma to carcinoma [109].
Dyskeratosis congenita is characterized by genomic instability arising from defective telomere maintenance rather than classical DNA repair pathways [70]. Critically shortened telomeres lose their protective function and are misrecognized by the DNA damage response machinery as double-strand breaks. This triggers inappropriate end-to-end chromosome fusions, resulting in dicentric chromosomes that undergo breakage during mitosis, creating a cycle of genomic rearrangements [110]. The accumulation of these chromosomal aberrations provides the foundation for neoplastic transformation. This mechanism explains the paradoxical observation that both very short and very long telomeres can be associated with cancer, depending on the cellular context and the ability to bypass senescence checkpoints [71].
Single-cell RNA sequencing studies in DBA cells indicate that the imbalance between rapid DNA replication and insufficient protein synthesis due to RP haploinsufficiency might trigger replication stress, leading to DNA damage, as reflected by anti-γH2AX positivity, and elevated pro-apoptotic gene expression [111].
In SDS, the deficiency in the SBDS gene leads to destabilization of the mitotic spindle, which can result in aberrant cell division processes, such as aneuploidy, where cells have an abnormal number of chromosomes, and centrosomal amplification, leading to multipolar spindles [112,113]. These events can promote the accumulation of further genetic damage over time. The connection between genomic instability and cancer in SDS is further evidenced by the observation of clonal cytogenetic abnormalities in the bone marrow of affected patients [114]. Leukemias that arise in individuals with SDS often exhibit aneuploidy and complex chromosomal aberrations [115]. The accumulation of such genetic alterations can lead to the loss of critical cellular checkpoints, such as the p53 tumor suppressor pathway, thereby promoting uncontrolled cell proliferation and tumorigenesis [93].

3.2. p53 Pathway Activation and Cellular Stress Response

Although DC, FA, DBAS, and SDS originate from disruptions in separate cellular processes, i.e., telomere biology, DNA repair, ribosome biogenesis and function, respectively, they converge on a shared pathogenic mechanism: chronic activation of the p53 stress response [116]. Increase in p53 level drives bone marrow failure by impairing self-renewal and promoting apoptosis or senescence of hematopoietic stem and progenitor cells (HSPCs) [79,117,118,119,120]. Theoretically, p53 activation should protect cells against malignant transformation by inducing cell cycle arrest or apoptosis. However, the selective pressure exerted by chronic p53 activation in IBMFS patients may favor the emergence of cells with p53 mutations or cells that have developed mechanisms to bypass p53-mediated growth arrest [121,122].
Cells with FA pathway deficiencies exhibit constitutive p53 activation due to their fundamental inability to properly repair DNA ICLs and other DNA lesions. The defective FA DNA repair pathway leads to accumulation of DNA damage that continuously triggers ATM-mediated p53 phosphorylation [123,124]. This persistent genotoxic stress results in elevated levels of p53 and its downstream effectors, and in increased production of reactive oxygen species (ROS), which create a feed-forward loop that further amplifies p53 signaling [125]. Importantly, FA cells demonstrate hypersensitivity to DNA crosslinking agents, which enhances p53-mediated apoptotic responses.
In DC, defective telomere maintenance leads to critically short telomeres that trigger persistent DNA damage responses. The shortest telomeres are recognized as double-strand breaks, activating ATM and ATR kinases that stabilize p53 [126]. This chronic telomere-induced DNA damage signaling results in sustained p53 activation, particularly pronounced in highly proliferative tissues such as the hematopoietic system [119]. Studies have demonstrated elevated p53 protein levels and increased expression of p53 target genes in DC patient cells [127,128].
Diamond–Blackfan Anemia Syndrome is characterized by RP haploinsufficiency that leads to the accumulation of unprocessed rRNA and impaired ribosomal subunit assembly within the nucleolus, activating a nucleolar stress response [129]. A key consequence is the stabilization and activation of p53, mediated by the sequestration of MDM2 by free RPs such as RPL5 and RPL11 [130]. Moreover, it has been recently reported that, under ribosomal or nucleolar stress, RPL22 promotes skipping of exon 6 in the MDM4 transcript by binding specific stem-loop RNA elements within intron 6. This exon skipping reduces full-length MDM4, thereby enhancing p53 activity and limiting proliferation [131,132]. DBAS patient cells show elevated p53 protein levels, increased p21 expression, and enhanced sensitivity to additional nucleolar stressors [133,134]. Loss or mutation of RPL22, which is rather frequent in CRC, interferes with the MDM4–p53 axis and leads to increased full-length MDM4, suppression of p53, and enhanced cell growth and drug resistance [131,135,136,137].
In SDS, dysfunction of the SBDS protein impairs ribosome maturation and leads to RPL5- and RPL11-mediated p53 stabilization similar to DBAS [138]. SDS cells demonstrate elevated p53 levels and increased expression of p53 target genes, particularly those involved in cell cycle control and apoptosis [138,139,140]. Furthermore, SDS cells show increased susceptibility to oxidative stress [141,142], which can further enhance p53 activation.
The constitutive stabilization and activation of p53 in IBMFS enforces cell cycle arrest, promotes apoptosis, and restricts cellular growth, thereby imposing a strong constraint on cellular proliferation, particularly within hematopoietic progenitors and other highly proliferative tissues. Under this pressure, cells that acquire somatic TP53 mutations, or otherwise lose p53 activity, are able to bypass the p53-mediated checkpoints and gain a competitive growth advantage. For instance, the p53-Arg273His pathogenic variant exhibits dominant-negative properties inhibiting wild-type p53 through oligomerization and confers a gain-of-function phenotype that promotes carcinogenesis. In CRC this mutant p53 has been shown to regulate specific long non-coding RNAs and transcriptional programs that sustain self-renewal, invasion, and therapeutic resistance [143,144,145]. Selection and expansion of cells harboring TP53 mutations is known to be responsible for clonal hematopoiesis and leukemic progression in IBMFS [146,147], and may also occur in non-hematopoietic tissues, including the intestinal epithelium.

3.3. Immune Dysfunction and Inflammation

Most IBMFS are characterized by constitutive activation of inflammatory pathways that can mediate stem cell exhaustion and clonal selection pressure. The pro-inflammatory microenvironment promotes genomic instability and suppresses normal hematopoiesis. Immune surveillance can also be impaired, facilitating immune evasion and malignant transformation. While there is no complete consensus on cytokine profiles, and studies report variable findings regarding inflammatory markers and interleukins, pro-inflammatory cytokines are generally increased across IBMFS subtypes [148,149] and may play a role in enhancing oxidative stress and DNA damage in IBMFS pathogenesis and carcinogenesis. The pro-inflammatory cytokine milieu associated with chronic p53 activation can promote a pro-tumorigenic microenvironment in the gastrointestinal tract.
In FA, abnormal production of pro-inflammatory cytokines has been observed in vitro, in vivo, and in patients [150,151]. Activation of TNF-α signaling represents the dominant inflammatory pathway. TNF-α is overexpressed due to dysregulated stress-response pathways such as NF-κB and MAPK [125]. Additionally, elevated levels of TGF-β3 suggest a hyperactivated TGF-β signaling pathway in the bone marrow microenvironment [152,153]. Both adults and children with FA show decreased B- and NK-cells compared to healthy individuals, and lower IgM [154,155].
For DC patients, the most consistently reported immunological abnormality in the literature is a marked reduction in B-cell and NK-cell counts, whereas T-cell lymphopenia is less common [154]. Dysgammaglobulinemia, characterized by decreased levels of IgG, IgM, or IgA, has been observed in some studies [156,157]. Elevated serum levels of G-CSF and Flt3L, along with reduced RANTES, have been linked to severe marrow failure in DC [154].
Unlike other IBMFS, DBAS has not shown significant changes in pro-inflammatory cytokines like TNF-α and IFN-γ in patients [154]. An earlier study reported no significant immunodeficiency in patients with DBAS [154]. However, subsequent research involving 128 children and adults with DBAS demonstrated reduced numbers of B-cells and NK-cells, along with decreased IgM levels; these findings were largely independent of steroid therapy [158]. Another study focusing on pediatric DBAS patients further confirmed low immunoglobulin levels in a subset of individuals [159].
Immune function in SDS patients is not severely affected [154], but neutrophils from SDS patients are reduced in number and have significantly impaired migratory activity [160]. SDS patients can exhibit elevated plasma levels of TGF-β, and upregulation of the TGF-β pathway has been observed [161], as well as elevated plasma levels of several pro-inflammatory cytokines involved in the mTOR/STAT3 pathway, such as IL-6, IL-8, and IL-12 [162].

3.4. Oxidative Stress

Oxidative stress represents another critical component of the cellular stress response in IBMFS. Reactive oxygen species are natural byproducts of mitochondrial energy metabolism and play a role in normal cellular signaling. However, excessive ROS can damage DNA, RNA, proteins, and cellular structures, contributing to genomic instability and cancer development. In FA, pathogenic variants in genes like FANCA, FANCC, and FANCD2 are associated with increased ROS production, impaired mitochondrial function, and diminished ATP generation [125]. Cells deficient in the FA pathway show elevated oxidative DNA damage, chromosomal breakage, and apoptosis [163]. The FA pathway plays a critical role in regulating oxidative stress, and its inactivation impairs systemic antioxidant defenses and mitochondrial ROS-scavenging capacity. Loss of mitochondrial integrity adds to redox imbalance. Furthermore, pro-inflammatory cytokines released by FA cells amplify ROS production both in vitro and in vivo [164].
Telomere dysfunction in DC leads to mitochondrial abnormalities and impaired antioxidant defenses, resulting in accumulation of oxidative damage [165]. Lymphocytes isolated from DC patients show elevated levels of ROS, DNA damage response and apoptotic markers, especially after exposure to cytotoxic agents [127]. Experimental models have shown that oxidative stress exacerbates telomere attrition, contributing to genomic instability and cellular senescence in DC [127,166].
Oxidative stress has also been implicated in DBAS pathophysiology. Mutations that impair ribosomal biogenesis and function cause defective synthesis of globin chains in erythroid progenitors, leading to a marked decrease in globin production. Since heme synthesis remains unaffected, there is an imbalance between globins and heme within the cell and the excess free heme contributes to oxidative stress and apoptosis or ferroptosis of erythroid progenitors, thereby exacerbating the anemia observed in DBAS [80,167]. Cells with RP deficiency show a downregulation in the expression of genes involved in the protection against oxidative stress [168]. Studies using Rp-deficient murine and patient-derived cells revealed that elevated ROS production exceeds antioxidant capacity, leading to oxidative DNA damage, cellular senescence, and activation of DNA damage response pathways [169,170].
Impaired protein synthesis in SDS cells results in decreased expression of antioxidant enzymes including superoxide dismutase (SOD), catalase, and glutathione peroxidase, creating an oxidative imbalance favoring ROS accumulation [139]. A reduction in mitochondrial SOD2 levels has also been reported in a yeast model of SDS [171].
Although increased oxidative stress has been extensively documented in hematopoietic cells and blood of patients with IBMFS [80,125,127,169,172], there is currently no direct evidence that ROS levels are elevated in the intestinal epithelium of these individuals. Nevertheless, given the known role of ROS in colorectal carcinogenesis [173] and the systemic oxidative imbalance observed in IBMFS, it is plausible that oxidative damage could also occur in the colonic mucosa, thereby contributing to cancer risk.

3.5. Other Mechanisms Possibly Involved in Colorectal Carcinogenesis

Alterations in the translation of specific mRNAs have been implicated in DBAS, SDS and DC pathophysiology. Reduction in ribosome levels profoundly impairs translation of a specific subset of transcripts [174]. Downregulation of Rps19 or Rpl11 in murine erythroblasts caused deregulated translation initiation of specific transcripts, and this result was confirmed in erythroblasts cultured from DBA patients [175]. RPS19 knockdown in erythroid cells reduced GATA1 protein expression without significantly affecting total cellular protein levels. Notably, total GATA1 mRNA levels were unchanged, consistent with the hypothesis that transcripts with complex 5′ UTR are inefficiently translated under conditions of limited ribosome abundance [176].
In SDS, the loss of SBDS function causes inefficient translation re-initiation, a mechanism that allows ribosomes to resume translation at a downstream start codon, of the mRNAs of the transcription factors C/EBPα and -β [177]. As a consequence, some protein isoforms of these transcripts are produced at reduced levels, leading to decreased MYC expression and impaired cellular proliferation that possibly contributes to the hematological phenotype of SDS [177].
A seminal study by Yoon et al. demonstrated that cap-dependent translation was not affected in a mouse model of DC, whereas Internal Ribosome Entry Site (IRES)-dependent translation was selectively impaired, reducing synthesis of mRNAs encoding key regulators such as p27(Kip1), Bcl-xL, and XIAP [178]. The same translational defects was observed in cells derived from DC patients [178]. Deficient translation of tumor suppressors and anti-apoptotic factors may create a permissive environment for genomic instability and uncontrolled proliferation that may drive oncogenesis [179].
Moreover, ribosomes purified from a cellular model of DC obtained by DKC1 knockdown, exhibited impaired translational fidelity [180], indicating an increased tendency to misincorporate amino acids or misread mRNAs during protein synthesis. This defect may contribute to the elevated cancer risk observed in DC.
Epigenetic dysregulation might be another contributor to the pathophysiology of IBMFS. FA cells exhibit significant alterations in their epigenetic regulatory machinery, characterized by marked downregulation of key DNA methyltransferases. Expression studies in peripheral blood mononuclear cells and bone marrow from FA patients show reduced levels of DNMT1, which maintains DNA methylation during replication, and DNMT3B, a tumor suppressor involved in establishing genomic methylation, compared to healthy controls [181]. The downregulation of epigenetic modifiers results in global DNA hypomethylation, particularly affecting tumor suppressor gene loci [181]. Interestingly, it has been proposed that vitamin D supplementation may offer a therapeutic strategy to slow disease progression and reduce cancer risk in FA patients, thanks to its ability to modulate immune responses and regulate the expression of genes like FANCE through epigenetic mechanisms [182].

4. Clinical Implications

The heightened CRC risk in IBMFS patients strongly support the need for tailored surveillance protocols, such as initiating colonoscopic screening well before standard age [6,28], and for management strategies based on precision oncology.
Treatment of CRC in IBMFS patients poses unique challenges. Patients with FA, in particular, exhibit severe hypersensitivity to DNA-damaging agents (e.g., alkylating chemotherapy, radiation), making conventional therapies highly toxic and often contraindicated. Surgery remains the primary curative modality for solid tumors in this population.
Poly (ADP-ribose) polymerase (PARP) inhibitors represent a promising class of therapeutic agents for CRC [183,184]. These inhibitors, originally developed for tumors with homologous recombination deficiencies such as BRCA-mutated breast and ovarian cancers, act by inducing synthetic lethality in cells with defects in the DNA damage repair (DDR) pathway. Such DDR alterations are frequently observed in CRC. Ongoing research is focused on defining the molecular subgroups of patients that are most likely to benefit from these therapies [183]. The use of PARP inhibitors in patients with FA raises serious concerns due to systemic toxicity. Since FA is caused by biallelic mutations in genes involved in homologous recombination (HR)-mediated DNA repair, all somatic cells are hypersensitive to DNA-damaging agents, including PARP inhibitors. As PARP inhibitors exploit HR deficiencies in tumors [184,185], their use in FA patients would likely result in widespread cytotoxic effects, making them clinically inappropriate in this context. In contrast, individuals who carry heterozygous germline mutations in FA genes without clinical FA, and who develop CRC with somatic loss of the wild-type allele, may represent a distinct subgroup in which PARP inhibitors could be therapeutically beneficial. In these patients, the tumor may exhibit synthetic lethality in response to PARP inhibition, while normal tissues retain sufficient FA pathway activity to tolerate treatment. This distinction underscores the importance of carefully differentiating between biallelic FA patients and heterozygous FA mutation carriers when considering therapies for CRC.

5. Conclusions

Inherited Bone Marrow Failure Syndromes are associated with a significantly increased risk of CRC, often manifesting at younger ages than in the general population. Future research priorities include prospective studies to better define CRC incidence and risk factors, establishment of surveillance protocols and development of personalized prevention and treatment strategies for optimal patient care.

Funding

This work was supported by grants from Associazione Italiana per la Ricerca sul Cancro (AIRC) (IG 2021-ID.25886, to I.D.); Fondazione Europea per la DBA (to A.A.); Diamond Blackfan Anemia Foundation (to A.A.).

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The authors acknowledge the use of ChatGPT (GPT-4o, OpenAI) and Claude (Anthropic’s AI assistant) for assistance with manuscript editing and language improvement. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AMLAcute myeloid leukemia
CRCColorectal cancer
DCDyskeratosis congenita
DDRDNA damage repair
DBASDiamond–Blackfan anemia syndrome
FAFanconi anemia
HRHomologous recombination
HSCTHematopoietic stem cell transplantation
IBMFSInherited bone marrow failure syndrome
ICLInterstrand crosslink
MDSMyelodysplastic syndrome
ROSReactive oxygen species
RPRibosomal protein
SDSShwachman–Diamond syndrome

References

  1. Park, M. Overview of Inherited Bone Marrow Failure Syndromes. Blood Res. 2022, 57, 49–54. [Google Scholar] [CrossRef] [PubMed]
  2. Wegman-Ostrosky, T.; Savage, S.A. The Genomics of Inherited Bone Marrow Failure: From Mechanism to the Clinic. Br. J. Haematol. 2017, 177, 526–542. [Google Scholar] [CrossRef] [PubMed]
  3. Crisà, E.; Boggione, P.; Nicolosi, M.; Mahmoud, A.M.; Essa, W.A.; Awikeh, B.; Aspesi, A.; Andorno, A.; Boldorini, R.; Dianzani, I.; et al. Genetic Predisposition to Myelodysplastic Syndromes: A Challenge for Adult Hematologists. Int. J. Mol. Sci. 2021, 22, 2525. [Google Scholar] [CrossRef] [PubMed]
  4. Hwang, S.M. Genomic Testing for Germline Predisposition to Hematologic Malignancies. Blood Res. 2024, 59, 12. [Google Scholar] [CrossRef]
  5. Willig, T.N.; Niemeyer, C.M.; Leblanc, T.; Tiemann, C.; Robert, A.; Budde, J.; Lambiliotte, A.; Kohne, E.; Souillet, G.; Eber, S.; et al. Identification of New Prognosis Factors from the Clinical and Epidemiologic Analysis of a Registry of 229 Diamond-Blackfan Anemia Patients. Pediatr. Res. 1999, 46, 553–561. [Google Scholar] [CrossRef]
  6. Wlodarski, M.W.; Vlachos, A.; Farrar, J.E.; Da Costa, L.M.; Kattamis, A.; Dianzani, I.; Belendez, C.; Unal, S.; Tamary, H.; Pasauliene, R.; et al. Diagnosis, Treatment, and Surveillance of Diamond-Blackfan Anaemia Syndrome: International Consensus Statement. Lancet Haematol. 2024, 11, e368–e382. [Google Scholar] [CrossRef] [PubMed]
  7. Tsangaris, E.; Klaassen, R.; Fernandez, C.V.; Yanofsky, R.; Shereck, E.; Champagne, J.; Silva, M.; Lipton, J.H.; Brossard, J.; Michon, B.; et al. Genetic Analysis of Inherited Bone Marrow Failure Syndromes from One Prospective, Comprehensive and Population-Based Cohort and Identification of Novel Mutations. J. Med. Genet. 2011, 48, 618–628. [Google Scholar] [CrossRef]
  8. Gianferante, D.M.; Mendez, K.J.W.; Cole, S.; Gadalla, S.M.; Alter, B.P.; Savage, S.A.; Giri, N. Genotype-phenotype Associations in Individuals with Diamond Blackfan Anaemia. EJHaem 2024, 5, 1117–1124. [Google Scholar] [CrossRef]
  9. Vlachos, A.; Rosenberg, P.S.; Atsidaftos, E.; Alter, B.P.; Lipton, J.M. Incidence of Neoplasia in Diamond Blackfan Anemia: A Report from the Diamond Blackfan Anemia Registry. Blood 2012, 119, 3815–3819. [Google Scholar] [CrossRef]
  10. Altintas, B.; Giri, N.; McReynolds, L.J.; Best, A.; Alter, B.P. Genotype-Phenotype and Outcome Associations in Patients with Fanconi Anemia: The National Cancer Institute Cohort. Haematologica 2023, 108, 69–82. [Google Scholar] [CrossRef]
  11. Aspesi, A.; Ellis, S.R. Rare Ribosomopathies: Insights into Mechanisms of Cancer. Nat. Rev. Cancer 2019, 19, 228–238. [Google Scholar] [CrossRef]
  12. De Keersmaecker, K.; Sulima, S.O.; Dinman, J.D. Ribosomopathies and the Paradox of Cellular Hypo- to Hyperproliferation. Blood 2015, 125, 1377–1382. [Google Scholar] [CrossRef] [PubMed]
  13. Webster, A.L.H.; Sanders, M.A.; Patel, K.; Dietrich, R.; Noonan, R.J.; Lach, F.P.; White, R.R.; Goldfarb, A.; Hadi, K.; Edwards, M.M.; et al. Genomic Signature of Fanconi Anaemia DNA Repair Pathway Deficiency in Cancer. Nature 2022, 612, 495–502. [Google Scholar] [CrossRef] [PubMed]
  14. Roka, K.; Solomou, E.; Kattamis, A.; Stiakaki, E. Telomere Biology Disorders: From Dyskeratosis Congenita and Beyond. Postgrad. Med. J. 2024, 100, 879–889. [Google Scholar] [CrossRef] [PubMed]
  15. Gianni, P.; Matenoglou, E.; Geropoulos, G.; Agrawal, N.; Adnani, H.; Zafeiropoulos, S.; Miyara, S.J.; Guevara, S.; Mumford, J.M.; Molmenti, E.P.; et al. The Fanconi Anemia Pathway and Breast Cancer: A Comprehensive Review of Clinical Data. Clin. Breast Cancer 2022, 22, 10–25. [Google Scholar] [CrossRef]
  16. Alter, B.P.; Giri, N.; Savage, S.A.; Peters, J.A.; Loud, J.T.; Leathwood, L.; Carr, A.G.; Greene, M.H.; Rosenberg, P.S. Malignancies and Survival Patterns in the National Cancer Institute Inherited Bone Marrow Failure Syndromes Cohort Study. Br. J. Haematol. 2010, 150, 179–188. [Google Scholar] [CrossRef] [PubMed]
  17. Alter, B.P.; Giri, N.; Savage, S.A.; Rosenberg, P.S. Cancer in the National Cancer Institute Inherited Bone Marrow Failure Syndrome Cohort after Fifteen Years of Follow-Up. Haematologica 2017, 103, 30–39. [Google Scholar] [CrossRef]
  18. McReynolds, L.J.; Biswas, K.; Giri, N.; Sharan, S.K.; Alter, B.P. Genotype-Cancer Association in Patients with Fanconi Anemia Due to Pathogenic Variants in FANCD1 (BRCA2) or FANCN (PALB2). Cancer Genet. 2021, 258–259, 101–109. [Google Scholar] [CrossRef]
  19. Namikawa, T.; Tanaka, T.; Utsunomiya, M.; Yokota, K.; Munekage, M.; Maeda, H.; Kitagawa, H.; Kurioka, Y.; Satake, H.; Kobayashi, M.; et al. Gastric Cancer with Fanconi Anemia in Adolescent and Young Adult Patient Diagnosed by Comprehensive Genome Profiling Using Next-Generation Sequencing. Clin. J. Gastroenterol. 2024, 17, 12–17. [Google Scholar] [CrossRef] [PubMed]
  20. Rosenberg, P.S.; Greene, M.H.; Alter, B.P. Cancer Incidence in Persons with Fanconi Anemia. Blood 2003, 101, 822–826. [Google Scholar] [CrossRef]
  21. Kutler, D.I.; Singh, B.; Satagopan, J.; Batish, S.D.; Berwick, M.; Giampietro, P.F.; Hanenberg, H.; Auerbach, A.D. A 20-Year Perspective on the International Fanconi Anemia Registry (IFAR). Blood 2003, 101, 1249–1256. [Google Scholar] [CrossRef] [PubMed]
  22. Nepal, M.; Che, R.; Zhang, J.; Ma, C.; Fei, P. Fanconi Anemia Signaling and Cancer. Trends Cancer 2017, 3, 840–856. [Google Scholar] [CrossRef]
  23. Tummala, H.; Walne, A.M.A.J.; Badat, M.; Patel, M.; Walne, A.M.A.J.; Alnajar, J.; Chow, C.C.; Albursan, I.; Frost, J.M.; Ballard, D.; et al. The Evolving Genetic Landscape of Telomere Biology Disorder Dyskeratosis Congenita. EMBO Mol. Med. 2024, 16, 2560–2582. [Google Scholar] [CrossRef] [PubMed]
  24. Savage, S.A.; Niewisch, M.R. Dyskeratosis Congenita and Related Telomere Biology Disorders. In GeneReviews®; University of Washington: Seattle, WA, USA, 2023. [Google Scholar]
  25. Alter, B.P.; Giri, N.; Savage, S.A.; Rosenberg, P.S. Cancer in Dyskeratosis Congenita. Blood 2009, 113, 6549–6557. [Google Scholar] [CrossRef]
  26. Liu, Y.; Karlsson, S. Perspectives of Current Understanding and Therapeutics of Diamond-Blackfan Anemia. Leukemia 2023, 38, 1–9, Erratum in Leukemia 2024, 38, 228. https://doi.org/10.1038/s41375-023-02101-w. [Google Scholar] [CrossRef] [PubMed]
  27. Lipton, J.M.; Molmenti, C.L.S.; Hussain, M.; Desai, P.; Florento, M.; Atsidaftos, E.; Vlachos, A. Colorectal Cancer Screening and Surveillance Strategy for Patients with Diamond Blackfan Anemia: Preliminary Recommendations from the Diamond Blackfan Anemia Registry. Pediatr. Blood Cancer 2021, 68, e28984. [Google Scholar] [CrossRef]
  28. Lipton, J.M.; Molmenti, C.L.S.; Desai, P.; Lipton, A.; Ellis, S.R.; Vlachos, A. Early Onset Colorectal Cancer: An Emerging Cancer Risk in Patients with Diamond Blackfan Anemia. Genes 2022, 13, 56. [Google Scholar] [CrossRef] [PubMed]
  29. Quarello, P.; Garelli, E.; Carando, A.; Cillario, R.; Brusco, A.; Giorgio, E.; Ferrante, D.; Corti, P.; Zecca, M.; Luciani, M.; et al. A 20-Year Long Term Experience of the Italian Diamond-Blackfan Anaemia Registry: RPS and RPL Genes, Different Faces of the Same Disease? Br. J. Haematol. 2020, 190, 93–104. [Google Scholar] [CrossRef] [PubMed]
  30. Kawashima, N.; Oyarbide, U.; Cipolli, M.; Bezzerri, V.; Corey, S.J. Shwachman-Diamond Syndromes: Clinical, Genetic, and Biochemical Insights from the Rare Variants. Haematologica 2023, 108, 2594–2605. [Google Scholar] [CrossRef]
  31. Thompson, A.S.; Giri, N.; Gianferante, D.M.; Jones, K.; Savage, S.A.; Alter, B.P.; McReynolds, L.J. Shwachman Diamond Syndrome: Narrow Genotypic Spectrum and Variable Clinical Features. Pediatr. Res. 2022, 92, 1671–1680. [Google Scholar] [CrossRef]
  32. Bou Mitri, F.; Beaupain, B.; Flejou, J.F.; Patient, M.; Okhremchuck, I.; Blaise, D.; Izadifar-Legrand, F.; Martignoles, J.A.; Delhommeau, F.; Bellanne-Chantelot, C.; et al. Shwachman-Diamond Syndrome and Solid Tumors: Three New Patients from the French Registry for Severe Chronic Neutropenia and Literature Review. Pediatr. Blood Cancer 2021, 68, e29071. [Google Scholar] [CrossRef]
  33. Singh, S.A.; Vlachos, A.; Morgenstern, N.J.; Ouansafi, I.; Ip, W.; Rommens, J.M.; Durie, P.; Shimamura, A.; Lipton, J.M. Breast Cancer in a Case of Shwachman Diamond Syndrome. Pediatr. Blood Cancer 2012, 59, 945–946. [Google Scholar] [CrossRef] [PubMed]
  34. Dhanraj, S.; Manji, A.; Pinto, D.; Scherer, S.W.; Favre, H.; Loh, M.L.; Chetty, R.; Wei, A.C.; Dror, Y. Molecular Characteristics of a Pancreatic Adenocarcinoma Associated with Shwachman-Diamond Syndrome. Pediatr. Blood Cancer 2013, 60, 754–760. [Google Scholar] [CrossRef]
  35. Li, S.; Silvestri, V.; Leslie, G.; Rebbeck, T.R.; Neuhausen, S.L.; Hopper, J.L.; Nielsen, H.R.; Lee, A.; Yang, X.; Mcguffog, L.; et al. Cancer Risks Associated With BRCA1 and BRCA2 Pathogenic Variants. J. Clin. Oncol. 2022, 40, 1529–1541. [Google Scholar] [CrossRef]
  36. Feng, Z.; Yang, X.; Tian, M.; Zeng, N.; Bai, Z.; Deng, W.; Zhao, Y.; Guo, J.; Yang, Y.; Zhang, Z.; et al. BRCA Genes as Candidates for Colorectal Cancer Genetic Testing Panel: Systematic Review and Meta-Analysis. BMC Cancer 2023, 23, 807. [Google Scholar] [CrossRef] [PubMed]
  37. Xu, P.; Sun, D.; Gao, Y.; Jiang, Y.; Zhong, M.; Zhao, G.; Chen, J.; Wang, Z.; Liu, Q.; Hong, J.; et al. Germline Mutations in a DNA Repair Pathway Are Associated with Familial Colorectal Cancer. JCI Insight 2021, 6, e148931. [Google Scholar] [CrossRef] [PubMed]
  38. AlDubayan, S.H.; Giannakis, M.; Moore, N.D.; Han, G.C.; Reardon, B.; Hamada, T.; Mu, X.J.; Nishihara, R.; Qian, Z.; Liu, L.; et al. Inherited DNA-Repair Defects in Colorectal Cancer. Am. J. Hum. Genet. 2018, 102, 401–414. [Google Scholar] [CrossRef]
  39. Susswein, L.R.; Marshall, M.L.; Nusbaum, R.; Vogel Postula, K.J.; Weissman, S.M.; Yackowski, L.; Vaccari, E.M.; Bissonnette, J.; Booker, J.K.; Cremona, M.L.; et al. Pathogenic and Likely Pathogenic Variant Prevalence among the First 10,000 Patients Referred for next-Generation Cancer Panel Testing. Genet. Med. 2016, 18, 823–832, Erratum in Genet. Med. 2016, 18, 531–532. https://doi.org/10.1038/gim.2016.21. [Google Scholar] [CrossRef] [PubMed]
  40. Hossain, M.S.; Karuniawati, H.; Jairoun, A.A.; Urbi, Z.; Ooi, D.J.; John, A.; Lim, Y.C.; Kaderi Kibria, K.M.; Mohiuddin, A.K.M.; Ming, L.C.; et al. Colorectal Cancer: A Review of Carcinogenesis, Global Epidemiology, Current Challenges, Risk Factors, Preventive and Treatment Strategies. Cancers 2022, 14, 1732. [Google Scholar] [CrossRef] [PubMed]
  41. Morgan, E.; Arnold, M.; Gini, A.; Lorenzoni, V.; Cabasag, C.J.; Laversanne, M.; Vignat, J.; Ferlay, J.; Murphy, N.; Bray, F. Global Burden of Colorectal Cancer in 2020 and 2040: Incidence and Mortality Estimates from GLOBOCAN. Gut 2023, 72, 338–344. [Google Scholar] [CrossRef]
  42. Li, Q.; Geng, S.; Luo, H.; Wang, W.; Mo, Y.Q.; Luo, Q.; Wang, L.; Song, G.B.; Sheng, J.P.; Xu, B. Signaling Pathways Involved in Colorectal Cancer: Pathogenesis and Targeted Therapy. Signal Transduct. Target. Ther. 2024, 9, 266. [Google Scholar] [CrossRef] [PubMed]
  43. Moore, C.A.; Krishnan, K. Bone Marrow Failure. In StatPearls [Internet]; StatPearls: Petersburg, FL, USA, 2023. [Google Scholar]
  44. Gutierrez-Rodrigues, F.; Patel, B.A.; Groarke, E.M. When to Consider Inherited Marrow Failure Syndromes in Adults. Hematol. Am. Soc. Hematol. Educ. Progr. 2023, 2023, 548–555. [Google Scholar] [CrossRef]
  45. Vlachos, A.; Klein, G.W.; Lipton, J.M. The Diamond Blackfan Anemia Registry: Tool for Investigating the Epidemiology and Biology of Diamond-Blackfan Anemia. J. Pediatr. Hematol. Oncol. 2001, 23, 377–382. [Google Scholar] [CrossRef]
  46. Kolinjivadi, A.M.; Crismani, W.; Ngeow, J. Emerging Functions of Fanconi Anemia Genes in Replication Fork Protection Pathways. Hum. Mol. Genet. 2020, 29, R158–R164. [Google Scholar] [CrossRef] [PubMed]
  47. Peake, J.D.; Noguchi, E. Fanconi Anemia: Current Insights Regarding Epidemiology, Cancer, and DNA Repair. Hum. Genet. 2022, 141, 1811–1836. [Google Scholar] [CrossRef] [PubMed]
  48. Jung, M.; Ramanagoudr-Bhojappa, R.; van Twest, S.; Rosti, R.O.; Murphy, V.; Tan, W.; Donovan, F.X.; Lach, F.P.; Kimble, D.C.; Jiang, C.S.; et al. Association of Clinical Severity with FANCB Variant Type in Fanconi Anemia. Blood 2020, 135, 1588–1602. [Google Scholar] [CrossRef]
  49. Ameziane, N.; May, P.; Haitjema, A.; Van De Vrugt, H.J.; Van Rossum-Fikkert, S.E.; Ristic, D.; Williams, G.J.; Balk, J.; Rockx, D.; Li, H.; et al. A Novel Fanconi Anaemia Subtype Associated with a Dominant-Negative Mutation in RAD51. Nat. Commun. 2015, 6, 8829. [Google Scholar] [CrossRef]
  50. Zhong, L.; Zhang, W.; Zhang, K.; Li, C.; Mu, X.; Chu, Y.; Gai, Z.; Wei, H. Clinical and Genetic Features of Fanconi Anemia Associated with a Variant of FANCA Gene: Case Report and Literature Review. Medicine 2024, 103, e39358. [Google Scholar] [CrossRef] [PubMed]
  51. Farkas, G.; Székely, G.; Goda, V.; Kállay, K.M.; Kocsis, Z.S.; Szakszon, K.; Benyó, G.; Erdélyi, D.; Liptai, Z.; Csordás, K.; et al. Chromosomal Breakage Tests in the Differential Diagnosis of Fanconi Anemia and Aplastic Anemia. Eur. J. Haematol. 2023, 111, 254–262. [Google Scholar] [CrossRef] [PubMed]
  52. Woodward, E.R.; Meyer, S. Fanconi Anaemia, Childhood Cancer and the BRCA Genes. Genes 2021, 12, 1520. [Google Scholar] [CrossRef] [PubMed]
  53. Beddok, A.; Velleuer, E.; Sicre de Fontbrune, F.; Brakenhoff, R.H.; Dalle, J.H.; Dufour, C.; Faivre, S.; Genet, C.; Klijanienko, J.; Krieg, C.; et al. Strategies for Early Detection and Detailed Characterization of Oral Lesions and Head and Neck Squamous Cell Carcinoma in Fanconi Anemia Patients. Cancer Lett. 2025, 617, 217529. [Google Scholar] [CrossRef]
  54. Rosenberg, P.S.; Alter, B.P.; Ebell, W. Cancer Risks in Fanconi Anemia: Findings from the German Fanconi Anemia Registry. Haematologica 2008, 93, 511–517. [Google Scholar] [CrossRef]
  55. Lee, R.H.; Kang, H.; Yom, S.S.; Smogorzewska, A.; Johnson, D.E.; Grandis, J.R. Treatment of Fanconi Anemia–Associated Head and Neck Cancer: Opportunities to Improve Outcomes. Clin. Cancer Res. 2021, 27, 5168–5187. [Google Scholar] [CrossRef]
  56. de Pablo García-Cuenca, A.; Rofín-Fontanet, P.; Lorente-Guerrero, J.; Balmaña-Gelpí, J.; Carrasco-López, E.; Temprana-Salvador, J.; Granado-Carrasco, R.; Braña-García, I.; Vaquero-Martínez, P.; Pujol-Pina, R.; et al. Head and Neck Cancer in Fanconi Anemia: Report of 11 Cases and a Systematic Review. Cancers 2025, 17, 349. [Google Scholar] [CrossRef] [PubMed]
  57. Masserot-Lureau, C.; Adoui, N.; Degos, F.; de Bazelaire, C.; Soulier, J.; Chevret, S.; Socié, G.; Leblanc, T. Incidence of Liver Abnormalities in Fanconi Anemia Patients. Am. J. Hematol. 2012, 87, 547–549. [Google Scholar] [CrossRef] [PubMed]
  58. Bogliolo, M.; Surrallés, J. Fanconi Anemia: A Model Disease for Studies on Human Genetics and Advanced Therapeutics. Curr. Opin. Genet. Dev. 2015, 33, 32–40. [Google Scholar] [CrossRef]
  59. Bogliolo, M.; Bluteau, D.; Lespinasse, J.; Pujol, R.; Vasquez, N.; D’Enghien, C.D.; Stoppa-Lyonnet, D.; Leblanc, T.; Soulier, J.; Surrallés, J. Biallelic Truncating FANCM Mutations Cause Early-Onset Cancer but Not Fanconi Anemia. Genet. Med. 2018, 20, 458–463. [Google Scholar] [CrossRef] [PubMed]
  60. Catucci, I.; Osorio, A.; Arver, B.; Neidhardt, G.; Bogliolo, M.; Zanardi, F.; Riboni, M.; Minardi, S.; Pujol, R.; Azzollini, J.; et al. Individuals with FANCM Biallelic Mutations Do Not Develop Fanconi Anemia, but Show Risk for Breast Cancer, Chemotherapy Toxicity and May Display Chromosome Fragility. Genet. Med. 2018, 20, 452–457. [Google Scholar] [CrossRef]
  61. Parsa, F.G.; Nobili, S.; Karimpour, M.; Aghdaei, H.A.; Nazemalhosseini-Mojarad, E.; Mini, E. Fanconi Anemia Pathway in Colorectal Cancer: A Novel Opportunity for Diagnosis, Prognosis and Therapy. J. Pers. Med. 2022, 12, 396. [Google Scholar] [CrossRef] [PubMed]
  62. Deng, J.; Altintas, B.; Haley, J.S.; Kim, J.; Ramos, M.; Carey, D.J.; Stewart, D.R.; McReynolds, L.J. Most Fanconi Anemia Heterozygotes Are Not at Increased Cancer Risk: A Genome-First DiscovEHR Cohort Population Study. Genet. Med. 2024, 26, 101042. [Google Scholar] [CrossRef]
  63. McReynolds, L.J.; Giri, N.; Leathwood, L.; Risch, M.O.; Carr, A.G.; Alter, B.P. Risk of Cancer in Heterozygous Relatives of Patients with Fanconi Anemia. Genet. Med. 2022, 24, 245–250. [Google Scholar] [CrossRef] [PubMed]
  64. Rahman, N.; Seal, S.; Thompson, D.; Kelly, P.; Renwick, A.; Elliott, A.; Reid, S.; Spanova, K.; Barfoot, R.; Chagtai, T.; et al. PALB2, Which Encodes a BRCA2-Interacting Protein, Is a Breast Cancer Susceptibility Gene. Nat. Genet. 2007, 39, 165–167. [Google Scholar] [CrossRef] [PubMed]
  65. Esteban-Jurado, C.; Franch-Expósito, S.; Muñoz, J.; Ocaña, T.; Carballal, S.; López-Cerón, M.; Cuatrecasas, M.; Vila-Casadesús, M.; Lozano, J.J.; Serra, E.; et al. The Fanconi Anemia DNA Damage Repair Pathway in the Spotlight for Germline Predisposition to Colorectal Cancer. Eur. J. Hum. Genet. 2016, 24, 1501–1505. [Google Scholar] [CrossRef]
  66. Cannon-Albright, L.A.; Teerlink, C.C.; Stevens, J.; Snow, A.K.; Thompson, B.A.; Bell, R.; Nguyen, K.N.; Sargent, N.R.; Kohlmann, W.K.; Neklason, D.W.; et al. FANCM C5791C>T Stopgain Mutation (Rs144567652) Is a Familial Colorectal Cancer Risk Factor. Mol. Genet. Genom. Med. 2020, 8, e1532. [Google Scholar] [CrossRef]
  67. Norris, K.; Walne, A.J.; Ponsford, M.J.; Cleal, K.; Grimstead, J.W.; Ellison, A.; Alnajar, J.; Dokal, I.; Vulliamy, T.; Baird, D.M. High-Throughput STELA Provides a Rapid Test for the Diagnosis of Telomere Biology Disorders. Hum. Genet. 2021, 140, 945–955. [Google Scholar] [CrossRef]
  68. Liao, P.; Yan, B.; Wang, C.; Lei, P. Telomeres: Dysfunction, Maintenance, Aging and Cancer. Aging Dis. 2023, 15, 2595–2631. [Google Scholar] [CrossRef]
  69. Carvalho, V.S.; Gomes, W.R.; Calado, R.T. Recent Advances in Understanding Telomere Diseases. Fac. Rev. 2022, 11, 31. [Google Scholar] [CrossRef] [PubMed]
  70. Tummala, H.; Walne, A.; Dokal, I. The Biology and Management of Dyskeratosis Congenita and Related Disorders of Telomeres. Expert Rev. Hematol. 2022, 15, 685–696. [Google Scholar] [CrossRef] [PubMed]
  71. Savage, S.A. Telomere Length and Cancer Risk: Finding Goldilocks. Biogerontology 2024, 25, 265–278. [Google Scholar] [CrossRef]
  72. Tometten, M.; Beier, F.; Kirschner, M.; Schumacher, Y.; Walter, J.; Vieri, M.; Kricheldorf, K.; Röth, A.; Platzbecker, U.; Radsak, M.; et al. Late-Onset Telomere Biology Disorders in Adults: Clinical Insights and Treatment Outcomes from a Retrospective Registry Cohort. Blood Adv. 2025, 9, 2183–2191. [Google Scholar] [CrossRef] [PubMed]
  73. Ono, K.; Hiyoshi, Y.; Ono, A.; Ouchi, M.; Kosumi, K.; Eto, K.; Ida, S.; Iwatsuki, M.; Baba, Y.; Miyamoto, Y.; et al. Locally Advanced Rectal Cancer in a Young Adult Affected with Dyskeratosis Congenita (Zinsser–Cole–Engman Syndrome): A Case Report. Surg. Case Rep. 2024, 10, 206. [Google Scholar] [CrossRef]
  74. Watanabe, M.; Yamamoto, G.; Fujiyoshi, K.; Akagi, Y.; Kakuta, M.; Nishimura, Y.; Akagi, K. Development of Metachronous Rectal Cancers in a Young Man with Dyskeratosis Congenita: A Case Report. J. Med. Case Rep. 2019, 13, 117. [Google Scholar] [CrossRef] [PubMed]
  75. Da Costa, L.; Leblanc, T.; Mohandas, N. Diamond-Blackfan Anemia. Blood 2020, 136, 1262–1273. [Google Scholar] [CrossRef]
  76. Iskander, D.; Roy, N.B.A.; Payne, E.; Drasar, E.; Hennessy, K.; Harrington, Y.; Christodoulidou, C.; Karadimitris, A.; Batkin, L.; de la Fuente, J. Diamond-Blackfan Anemia in Adults: In Pursuit of a Common Approach for a Rare Disease. Blood Rev. 2023, 61, 101097. [Google Scholar] [CrossRef] [PubMed]
  77. Flygare, J.; Aspesi, A.; Bailey, J.C.; Miyake, K.; Caffrey, J.M.; Karlsson, S.; Ellis, S.R. Human RPS19, the Gene Mutated in Diamond-Blackfan Anemia, Encodes a Ribosomal Protein Required for the Maturation of 40S Ribosomal Subunits. Blood 2007, 109, 980–986. [Google Scholar] [CrossRef]
  78. Lindström, M.S.; Bartek, J.; Maya-Mendoza, A. P53 at the Crossroad of DNA Replication and Ribosome Biogenesis Stress Pathways. Cell Death Differ. 2022, 29, 972–982. [Google Scholar] [CrossRef]
  79. Bhoopalan, S.V.; Yen, J.S.; Mayuranathan, T.; Mayberry, K.D.; Yao, Y.; Lillo Osuna, M.A.; Jang, Y.; Liyanage, J.S.S.; Blanc, L.; Ellis, S.R.; et al. An RPS19-Edited Model for Diamond-Blackfan Anemia Reveals TP53-Dependent Impairment of Hematopoietic Stem Cell Activity. JCI Insight 2023, 8, e161810. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, Z.; Keel, S.B.; Shimamura, A.; Liu, L.; Gerds, A.T.; Li, H.Y.; Wood, B.L.; Scott, B.L.; Abkowitz, J.L. Delayed Globin Synthesis Leads to Excess Heme and the Macrocytic Anemia of Diamond Blackfan Anemia and Del(5q) Myelodysplastic Syndrome. Sci. Transl. Med. 2016, 8, 338ra67. [Google Scholar] [CrossRef] [PubMed]
  81. Doty, R.T.; Yan, X.; Meng, C.; Lausted, C.; Tian, Q.; Abkowitz, J.L. Single-Cell Analysis of Erythropoiesis in Rpl11 Haploinsufficient Mice Reveals Insight into the Pathogenesis of Diamond–Blackfan Anemia. Exp. Hematol. 2021, 97, 66–78.e6. [Google Scholar] [CrossRef]
  82. Vlachos, A.; Rosenberg, P.S.; Atsidaftos, E.; Kang, J.; Onel, K.; Sharaf, R.N.; Alter, B.P.; Lipton, J.M. Increased Risk of Colon Cancer and Osteogenic Sarcoma in Diamond Blackfan Anemia. Blood 2018, 132, 2205–2208. [Google Scholar] [CrossRef] [PubMed]
  83. Gupta, S.; Lieberman, D.; Anderson, J.C.; Burke, C.A.; Dominitz, J.A.; Kaltenbach, T.; Robertson, D.J.; Shaukat, A.; Syngal, S.; Rex, D.K. Recommendations for Follow-Up After Colonoscopy and Polypectomy: A Consensus Update by the US Multi-Society Task Force on Colorectal Cancer. Gastroenterology 2020, 158, 1131–1153.e5. [Google Scholar] [CrossRef]
  84. Nieminen, T.T.; O’Donohue, M.F.; Wu, Y.; Lohi, H.; Scherer, S.W.; Paterson, A.D.; Ellonen, P.; Abdel-Rahman, W.M.; Valo, S.; Mecklin, J.P.; et al. Germline Mutation of RPS20, Encoding a Ribosomal Protein, Causes Predisposition to Hereditary Nonpolyposis Colorectal Carcinoma without DNA Mismatch Repair Deficiency. Gastroenterology 2014, 147, 595–598. [Google Scholar] [CrossRef]
  85. Broderick, P.; Dobbins, S.E.; Chubb, D.; Kinnersley, B.; Dunlop, M.G.; Tomlinson, I.; Houlston, R.S. Validation of Recently Proposed Colorectal Cancer Susceptibility Gene Variants in an Analysis of Families and Patients—A Systematic Review. Gastroenterology 2017, 152, 75–77.e4. [Google Scholar] [CrossRef]
  86. Thompson, B.A.; Snow, A.K.; Koptiuch, C.; Kohlmann, W.K.; Mooney, R.; Johnson, S.; Huff, C.D.; Yu, Y.; Teerlink, C.C.; Feng, B.J.; et al. A Novel Ribosomal Protein S20 Variant in a Family with Unexplained Colorectal Cancer and Polyposis. Clin. Genet. 2020, 97, 943–944. [Google Scholar] [CrossRef] [PubMed]
  87. Amiot, J.; Gubeljak, L.; Fontaine, A.; Smith, D.; Mortemousque, I.; Parodi, N.; Mauillon, J.; Kasper, E.; Baert-Desurmont, S.; Tinat, J.; et al. New RPS20 Gene Variant in Colorectal Cancer Diagnosis: Insight from a Large Series of Patients. Fam. Cancer 2025, 24, 22. [Google Scholar] [CrossRef]
  88. Djursby, M.; Madsen, M.B.; Frederiksen, J.H.; Berchtold, L.A.; Therkildsen, C.; Willemoe, G.L.; Hasselby, J.P.; Wikman, F.; Okkels, H.; Skytte, A.B.; et al. New Pathogenic Germline Variants in Very Early Onset and Familial Colorectal Cancer Patients. Front. Genet. 2020, 11, 566266. [Google Scholar] [CrossRef]
  89. Bhar, S.; Zhou, F.; Reineke, L.C.; Morris, D.K.; Khincha, P.P.; Giri, N.; Mirabello, L.; Bergstrom, K.; Lemon, L.D.; Williams, C.L.; et al. Expansion of Germline RPS20 Mutation Phenotype to Include Diamond–Blackfan Anemia. Hum. Mutat. 2020, 41, 1918–1930. [Google Scholar] [CrossRef]
  90. Han, X.; Lu, S.; Gu, C.; Bian, Z.; Xie, X.; Qiao, X. Clinical Features, Epidemiology, and Treatment of Shwachman-Diamond Syndrome: A Systematic Review. BMC Pediatr. 2023, 23, 503. [Google Scholar] [CrossRef]
  91. Hamabata, T.; Umeda, K.; Kouzuki, K.; Tanaka, T.; Daifu, T.; Nodomi, S.; Saida, S.; Kato, I.; Baba, S.; Hiramatsu, H.; et al. Pluripotent Stem Cell Model of Shwachman–Diamond Syndrome Reveals Apoptotic Predisposition of Hemoangiogenic Progenitors. Sci. Rep. 2020, 10, 14859, Erratum in Sci. Rep. 2021, 11, 2107. https://doi.org/10.1038/s41598-021-81066-1. [Google Scholar] [CrossRef] [PubMed]
  92. Oyarbide, U.; Bezzerri, V.; Staton, M.; Boni, C.; Shah, A.; Cipolli, M.; Calo, E.; Corey, S.J. Reduced EIF6 Dosage Attenuates TP53 Activation in Models of Shwachman-Diamond Syndrome. J. Clin. Investig. 2025, 135, e187778. [Google Scholar] [CrossRef] [PubMed]
  93. Reilly, C.R.; Shimamura, A. Predisposition to Myeloid Malignancies in Shwachman-Diamond Syndrome: Biological Insights and Clinical Advances. Blood 2022, 141, 1513–1523. [Google Scholar] [CrossRef]
  94. Furutani, E.; Liu, S.; Galvin, A.; Steltz, S.; Malsch, M.M.; Loveless, S.K.; Mount, L.; Larson, J.H.; Queenan, K.; Bertuch, A.A.; et al. Hematologic Complications with Age in Shwachman-Diamond Syndrome. Blood Adv. 2022, 6, 297–306. [Google Scholar] [CrossRef] [PubMed]
  95. Donadieu, J.; Fenneteau, O.; Beaupain, B.; Beaufils, S.; Bellanger, F.; Mahlaoui, N.; Lambilliotte, A.; Aladjidi, N.; Bertrand, Y.; Mialou, V.; et al. Classification of and Risk Factors for Hematologic Complications in a French National Cohort of 102 Patients with Shwachman-Diamond Syndrome. Haematologica 2012, 97, 1312–1319. [Google Scholar] [CrossRef]
  96. Morecroft, R.; Logothetics, C.N.; Tarnawsky, S.P.; Davis, A.A. Chemotherapy-Induced Neutropenia Management in a Patient with Metastatic Breast Cancer and Shwachman-Diamond Syndrome (SDS): A Case Report. Transl. Breast Cancer Res. 2024, 5, 26. [Google Scholar] [CrossRef]
  97. Yu, Z.; Zhang, Z.; Liu, J.; Wu, X.; Fan, X.; Pang, J.; Bao, H.; Yin, J.; Wu, X.; Shao, Y.; et al. Identification of Pathogenic Germline Variants in a Large Chinese Lung Cancer Cohort by Clinical Sequencing. Mol. Oncol. 2024, 18, 1301–1315. [Google Scholar] [CrossRef]
  98. Sculco, M.; La Vecchia, M.; Aspesi, A.; Pinton, G.; Clavenna, M.G.; Casalone, E.; Allione, A.; Grosso, F.; Libener, R.; Muzio, A.; et al. Malignant Pleural Mesothelioma: Germline Variants in DNA Repair Genes May Steer Tailored Treatment. Eur. J. Cancer 2022, 163, 44–54. [Google Scholar] [CrossRef]
  99. Alonso-Luna, O.; Mercado-Celis, G.E.; Melendez-Zajgla, J.; Barquera, R.; Zapata-Tarres, M.; Juárez-Villegas, L.E.; Mendoza-Caamal, E.C.; Rey-Helo, E.; Borges-Yañez, S.A. Germline Mutations in Pediatric Cancer Cohort with Mixed-Ancestry Mexicans. Mol. Genet. Genom. Med. 2024, 12, e2332. [Google Scholar] [CrossRef]
  100. García-De-teresa, B.; Rodríguez, A.; Frias, S. Chromosome Instability in Fanconi Anemia: From Breaks to Phenotypic Consequences. Genes 2020, 11, 1528. [Google Scholar] [CrossRef]
  101. Wang, S.; Wang, R.; Peralta, C.; Yaseen, A.; Pavletich, N.P. Structure of the FA Core Ubiquitin Ligase Closing the ID Clamp on DNA. Nat. Struct. Mol. Biol. 2021, 28, 300–309. [Google Scholar] [CrossRef]
  102. Renaudin, X.; Rosselli, F. The FANC/BRCA Pathway Releases Replication Blockades by Eliminating DNA Interstrand Cross-Links. Genes 2020, 11, 585. [Google Scholar] [CrossRef] [PubMed]
  103. Longerich, S.; Li, J.; Xiong, Y.; Sung, P.; Kupfer, G.M. Stress and DNA Repair Biology of the Fanconi Anemia Pathway. Blood 2014, 124, 2812–2819. [Google Scholar] [CrossRef]
  104. Sumpter, R.; Levine, B. Emerging Functions of the Fanconi Anemia Pathway at a Glance. J. Cell Sci. 2017, 130, 2657–2662. [Google Scholar] [CrossRef]
  105. Michl, J.; Zimmer, J.; Tarsounas, M. Interplay between Fanconi Anemia and Homologous Recombination Pathways in Genome Integrity. EMBO J. 2016, 35, 909–923. [Google Scholar] [CrossRef] [PubMed]
  106. Hosea, R.; Hillary, S.; Naqvi, S.; Wu, S.; Kasim, V. The Two Sides of Chromosomal Instability: Drivers and Brakes in Cancer. Signal Transduct. Target. Ther. 2024, 9, 75. [Google Scholar] [CrossRef] [PubMed]
  107. Langevin, F.; Crossan, G.P.; Rosado, I.V.; Arends, M.J.; Patel, K.J. Fancd2 Counteracts the Toxic Effects of Naturally Produced Aldehydes in Mice. Nature 2011, 475, 53–58. [Google Scholar] [CrossRef]
  108. Pawlikowska, P.; Delestré, L.; Gregoricchio, S.; Oppezzo, A.; Esposito, M.; Diop, M.B.; Rosselli, F.; Guillouf, C. FANCA Deficiency Promotes Leukaemic Progression by Allowing the Emergence of Cells Carrying Oncogenic Driver Mutations. Oncogene 2023, 42, 2764–2775. [Google Scholar] [CrossRef]
  109. Pino, M.S.; Chung, D.C. The chromosomal instability pathway in colon cancer. Gastroenterology 2010, 138, 2059–2072. [Google Scholar] [CrossRef]
  110. Lazzerini-Denchi, E.; Sfeir, A. Stop Pulling My Strings-What Telomeres Taught Us about the DNA Damage Response. Nat. Rev. Mol. Cell. Biol. 2016, 17, 364–378. [Google Scholar] [CrossRef]
  111. Wang, B.; Wang, C.; Wan, Y.; Gao, J.; Ma, Y.; Zhang, Y.; Tong, J.; Zhang, Y.; Liu, J.; Chang, L.; et al. Decoding the Pathogenesis of Diamond–Blackfan Anemia Using Single-Cell RNA-Seq. Cell Discov. 2022, 8, 41. [Google Scholar] [CrossRef]
  112. Austin, K.M.; Gupta, M.L.; Coats, S.A.; Tulpule, A.; Mostoslavsky, G.; Balazs, A.B.; Mulligan, R.C.; Daley, G.; Pellman, D.; Shimamura, A. Mitotic Spindle Destabilization and Genomic Instability in Shwachman-Diamond Syndrome. J. Clin. Investig. 2008, 118, 1511–1518. [Google Scholar] [CrossRef]
  113. Sera, Y.; Imanaka, T.; Iguchi, Y.; Yamaguchi, M. Mitotic Abnormalities and Spindle Assembly Checkpoint Inactivation in a Cell Model of Shwachman-Diamond Syndrome with Mutations in the Shwachman-Bodian-Diamond Syndrome Gene, 258+2T > C. Drug Discov. Ther. 2024, 18, 283–289. [Google Scholar] [CrossRef]
  114. Valli, R.; De Paoli, E.; Nacci, L.; Frattini, A.; Pasquali, F.; Maserati, E. Novel Recurrent Chromosome Anomalies in Shwachman–Diamond Syndrome. Pediatr. Blood Cancer 2017, 64, e26454. [Google Scholar] [CrossRef] [PubMed]
  115. Maserati, E.; Pressato, B.; Valli, R.; Minelli, A.; Sainati, L.; Patitucci, F.; Marletta, C.; Mastronuzzi, A.; Poli, F.; Lo Curto, F.; et al. The Route to Development of Myelodysplastic Syndrome/Acute Myeloid Leukaemia in Shwachman-Diamond Syndrome: The Role of Ageing, Karyotype Instability, and Acquired Chromosome Anomalies. Br. J. Haematol. 2009, 145, 190–197. [Google Scholar] [CrossRef] [PubMed]
  116. Tsai, Y.Y.; Su, C.H.; Tarn, W.Y. P53 Activation in Genetic Disorders: Different Routes to the Same Destination. Int. J. Mol. Sci. 2021, 22, 9307. [Google Scholar] [CrossRef]
  117. Ceccaldi, R.; Parmar, K.; Mouly, E.; Delord, M.; Kim, J.M.; Regairaz, M.; Pla, M.; Vasquez, N.; Zhang, Q.S.; Pondarre, C.; et al. Bone Marrow Failure in Fanconi Anemia Is Triggered by an Exacerbated P53/P21 DNA Damage Response That Impairs Hematopoietic Stem and Progenitor Cells. Cell Stem Cell 2012, 11, 36–49. [Google Scholar] [CrossRef]
  118. Dutt, S.; Narla, A.; Lin, K.; Mullally, A.; Abayasekara, N.; Megerdichian, C.; Wilson, F.H.; Currie, T.; Khanna-Gupta, A.; Berliner, N.; et al. Haploinsufficiency for Ribosomal Protein Genes Causes Selective Activation of P53 in Human Erythroid Progenitor Cells. Blood 2011, 117, 2567–2576. [Google Scholar] [CrossRef]
  119. Fok, W.C.; Niero, E.L.d.O.; Dege, C.; Brenner, K.A.; Sturgeon, C.M.; Batista, L.F.Z. P53 Mediates Failure of Human Definitive Hematopoiesis in Dyskeratosis Congenita. Stem Cell Rep. 2017, 9, 409–418. [Google Scholar] [CrossRef] [PubMed]
  120. Marion, W.; Boettcher, S.; Ruiz-Torres, S.; da Rocha, E.L.; Lundin, V.; Morris, V.; Chou, S.; Zhao, A.M.; Kubaczka, C.; Aumais, O.; et al. An Induced Pluripotent Stem Cell Model of Fanconi Anemia Reveals Mechanisms of P53-Driven Progenitor Cell Differentiation. Blood Adv. 2020, 4, 4679–4692. [Google Scholar] [CrossRef]
  121. Choijilsuren, H.B.; Park, Y.; Jung, M. Mechanisms of Somatic Transformation in Inherited Bone Marrow Failure Syndromes. Hematolgy 2021, 2021, 390–398. [Google Scholar] [CrossRef] [PubMed]
  122. Kennedy, A.L.; Myers, K.C.; Bowman, J.; Gibson, C.J.; Camarda, N.D.; Furutani, E.; Muscato, G.M.; Klein, R.H.; Ballotti, K.; Liu, S.; et al. Distinct Genetic Pathways Define Pre-Malignant versus Compensatory Clonal Hematopoiesis in Shwachman-Diamond Syndrome. Nat. Commun. 2021, 12, 1334. [Google Scholar] [CrossRef]
  123. Yamamoto, K.; Nihrane, A.; Aglipay, J.; Sironi, J.; Arkin, S.; Lipton, J.M.; Ouchi, T.; Liu, J.M. Upregulated ATM Gene Expression and Activated DNA Crosslink-Induced Damage Response Checkpoint in Fanconi Anemia: Implications for Carcinogenesis. Mol. Med. 2008, 14, 167–174. [Google Scholar] [CrossRef]
  124. Biller, M.; Kabir, S.; Boado, C.; Nipper, S.; Saffa, A.; Tal, A.; Allen, S.; Sasanuma, H.; Dréau, D.; Vaziri, C.; et al. REV7-P53 Interaction Inhibits ATM-Mediated DNA Damage Signaling. Cell Cycle 2024, 23, 339–352. [Google Scholar] [CrossRef] [PubMed]
  125. Marano, L.; Nedoszytko, B.; Kalinowski, L.; Repczynska, A.; Ciastek, B.; Haus, O. New Insights into the Fanconi Anemia Pathogenesis: A Crosstalk Between Inflammation and Oxidative Stress. Int. J. Mol. Sci. 2024, 25, 11619. [Google Scholar] [CrossRef] [PubMed]
  126. He, H.; Wang, Y.; Guo, X.; Ramchandani, S.; Ma, J.; Shen, M.-F.; Garcia, D.A.; Deng, Y.; Multani, A.S.; You, M.J.; et al. Pot1b Deletion and Telomerase Haploinsufficiency in Mice Initiate an ATR-Dependent DNA Damage Response and Elicit Phenotypes Resembling Dyskeratosis Congenita. Mol. Cell. Biol. 2009, 29, 229–240. [Google Scholar] [CrossRef]
  127. Pereboeva, L.; Westin, E.; Patel, T.; Flaniken, I.; Lamb, L.; Klingelhutz, A.; Goldman, F. DNA Damage Responses and Oxidative Stress in Dyskeratosis Congenita. PLoS ONE 2013, 8, e76473. [Google Scholar] [CrossRef] [PubMed]
  128. Pereboeva, L.; Hubbard, M.; Goldman, F.D.; Westin, E.R. Robust DNA Damage Response and Elevated Reactive Oxygen Species in TINF2-Mutated Dyskeratosis Congenita Cells. PLoS ONE 2016, 11, e0148793. [Google Scholar] [CrossRef]
  129. Hannan, K.M.; Soo, P.; Wong, M.S.; Lee, J.K.; Hein, N.; Poh, P.; Wysoke, K.D.; Williams, T.D.; Montellese, C.; Smith, L.K.; et al. Nuclear Stabilization of P53 Requires a Functional Nucleolar Surveillance Pathway. Cell Rep. 2022, 41, 111571. [Google Scholar] [CrossRef]
  130. Donati, G.; Peddigari, S.; Mercer, C.A.; Thomas, G. 5S Ribosomal RNA Is an Essential Component of a Nascent Ribosomal Precursor Complex That Regulates the Hdm2-P53 Checkpoint. Cell Rep. 2013, 4, 87–98. [Google Scholar] [CrossRef]
  131. Weinstein, H.N.W.; Hu, K.; Fish, L.; Chen, Y.A.; Allegakoen, P.; Pham, J.H.; Hui, K.S.F.; Chang, C.H.; Tutar, M.; Benitez-Rivera, L.; et al. RPL22 Is a Tumor Suppressor in MSI-High Cancers and a Splicing Regulator of MDM4. Cell Rep. 2024, 43, 114622. [Google Scholar] [CrossRef]
  132. Jansen, J.; Bohnsack, K.E.; Böhlken-Fascher, S.; Bohnsack, M.T.; Dobbelstein, M. The Ribosomal Protein L22 Binds the MDM4 Pre-MRNA and Promotes Exon Skipping to Activate P53 upon Nucleolar Stress. Cell Rep. 2024, 43, 114610. [Google Scholar] [CrossRef] [PubMed]
  133. Aspesi, A.; Monteleone, V.; Betti, M.; Actis, C.; Morleo, G.; Sculco, M.; Guarrera, S.; Wlodarski, M.W.; Ramenghi, U.; Santoro, C.; et al. Lymphoblastoid Cell Lines from Diamond Blackfan Anaemia Patients Exhibit a Full Ribosomal Stress Phenotype That Is Rescued by Gene Therapy. Sci. Rep. 2017, 7, 12010, Erratum in Sci. Rep. 2018, 8, 17227. https://doi.org/10.1038/s41598-018-35522-0. [Google Scholar] [CrossRef]
  134. Ellis, S.R. Nucleolar Stress in Diamond Blackfan Anemia Pathophysiology. Biochim. Biophys. Acta 2014, 1842, 765–768. [Google Scholar] [CrossRef] [PubMed]
  135. Jansen, J.; Dobbelstein, M. MDM4 Exon Skipping upon Dysfunctional Ribosome Assembly. Trends Cell Biol. 2025, 35, 544–547. [Google Scholar] [CrossRef] [PubMed]
  136. Lindström, M.S. The Central Role of Ribosomal Proteins in P53 Regulation. Cancers 2025, 17, 1597. [Google Scholar] [CrossRef] [PubMed]
  137. Fan, W.; Liu, H.; Stachelek, G.C.; Begum, A.; Davis, C.E.; Dorado, T.E.; Ernst, G.; Reinhold, W.C.; Ozbek, B.; Zheng, Q.; et al. Ribosomal RNA Transcription Regulates Splicing through Ribosomal Protein RPL22. Cell Chem. Biol. 2025, 32, 908–925.e9. [Google Scholar] [CrossRef]
  138. Hao, Q.; Wang, J.; Chen, Y.; Wang, S.; Cao, M.; Lu, H.; Zhou, X. Dual Regulation of P53 by the Ribosome Maturation Factor SBDS. Cell Death Dis. 2020, 11, 197. [Google Scholar] [CrossRef] [PubMed]
  139. Sera, Y.; Yamamoto, S.; Mutou, A.; Koba, S.; Kurokawa, Y.; Imanaka, T.; Yamaguchi, M. SBDS Gene Mutation Increases ROS Production and Causes DNA Damage as Well as Oxidation of Mitochondrial Membranes in the Murine Myeloid Cell Line 32Dcl3. Biol. Pharm. Bull. 2024, 47, 1376–1382. [Google Scholar] [CrossRef]
  140. Frattini, A.; Bolamperti, S.; Valli, R.; Cipolli, M.; Pinto, R.M.; Bergami, E.; Frau, M.R.; Cesaro, S.; Signo, M.; Bezzerri, V.; et al. Enhanced P53 Levels Are Involved in the Reduced Mineralization Capacity of Osteoblasts Derived from Shwachman–Diamond Syndrome Subjects. Int. J. Mol. Sci. 2021, 22, 13331. [Google Scholar] [CrossRef] [PubMed]
  141. Ambekar, C.; Das, B.; Yeger, H.; Dror, Y. SBDS-Deficiency Results in Deregulation of Reactive Oxygen Species Leading to Increased Cell Death and Decreased Cell Growth. Pediatr. Blood Cancer 2010, 55, 1138–1144. [Google Scholar] [CrossRef]
  142. Ravera, S.; Dufour, C.; Cesaro, S.; Bottega, R.; Faleschini, M.; Cuccarolo, P.; Corsolini, F.; Usai, C.; Columbaro, M.; Cipolli, M.; et al. Evaluation of Energy Metabolism and Calcium Homeostasis in Cells Affected by Shwachman-Diamond Syndrome. Sci. Rep. 2016, 6, 25441. [Google Scholar] [CrossRef]
  143. Zhao, Y.; Li, Y.; Sheng, J.; Wu, F.; Li, K.; Huang, R.; Wang, X.; Jiao, T.; Guan, X.; Lu, Y.; et al. P53-R273H Mutation Enhances Colorectal Cancer Stemness through Regulating Specific LncRNAs. J. Exp. Clin. Cancer Res. 2019, 38, 379. [Google Scholar] [CrossRef]
  144. Wong, R.P.C.; Tsang, W.P.; Chau, P.Y.; Co, N.N.; Tsang, T.Y.; Kwok, T.T. P53-R273H Gains New Function in Induction of Drug Resistance through down-Regulation of Procaspase-3. Mol. Cancer Ther. 2007, 6, 1054–1061. [Google Scholar] [CrossRef]
  145. Cooks, T.; Pateras, I.S.; Tarcic, O.; Solomon, H.; Schetter, A.J.; Wilder, S.; Lozano, G.; Pikarsky, E.; Forshew, T.; Rozenfeld, N.; et al. Mutant P53 Prolongs NF-ΚB Activation and Promotes Chronic Inflammation and Inflammation-Associated Colorectal Cancer. Cancer Cell 2013, 23, 634–646, Erratum in Cancer Cell 2013, 24, 272. [Google Scholar] [CrossRef]
  146. Testa, U.; Castelli, G.; Pelosi, E. Clonal Hematopoiesis, a Risk Condition for Developing Myeloid Neoplasia. Hemato 2025, 6, 10. [Google Scholar] [CrossRef]
  147. Patnaik, M.M. Clonal Hematopoiesis in Inherited Bone Marrow Failure Syndromes: Adaptive Origins With Maladaptive Consequences? Hematology 2023, 20, 1615–1622. [Google Scholar] [CrossRef]
  148. Neoman, N.; Kim, H.N.; Viduya, J.; Goyal, A.; Liu, Y.L.; Sakamoto, K.M. Inflammatory Pathways and the Bone Marrow Microenvironment in Inherited Bone Marrow Failure Syndromes. Stem Cells 2025, 43, sxaf021. [Google Scholar] [CrossRef] [PubMed]
  149. Kawashima, N.; Bezzerri, V.; Corey, S.J. The Molecular and Genetic Mechanisms of Inherited Bone Marrow Failure Syndromes: The Role of Inflammatory Cytokines in Their Pathogenesis. Biomolecules 2023, 13, 1249. [Google Scholar] [CrossRef] [PubMed]
  150. Dufour, C.; Corcione, A.; Svahn, J.; Haupt, R.; Poggi, V.; Béka’ssy, A.N.; Scimè, R.; Pistorio, A.; Pistoia, V. TNF-α and IFN-γ Are Overexpressed in the Bone Marrow of Fanconi Anemia Patients and TNF-α Suppresses Erythropoiesis in Vitro. Blood 2003, 102, 2053–2059. [Google Scholar] [CrossRef] [PubMed]
  151. Helbling-Leclerc, A.; Garcin, C.; Rosselli, F. Beyond DNA Repair and Chromosome Instability—Fanconi Anaemia as a Cellular Senescence-Associated Syndrome. Cell Death Differ. 2021, 28, 1159–1173. [Google Scholar] [CrossRef]
  152. Zhang, H.; Kozono, D.E.; O’Connor, K.W.; Vidal-Cardenas, S.; Rousseau, A.; Hamilton, A.; Moreau, L.; Gaudiano, E.F.; Greenberger, J.; Bagby, G.; et al. TGF-β Inhibition Rescues Hematopoietic Stem Cell Defects and Bone Marrow Failure in Fanconi Anemia. Cell Stem Cell 2016, 18, 668–681. [Google Scholar] [CrossRef]
  153. Rodríguez, A.; Yang, C.; Furutani, E.; García de Teresa, B.; Velázquez, M.; Filiatrault, J.; Sambel, L.A.; Phan, T.; Flores-Guzmán, P.; Sánchez, S.; et al. Inhibition of TGFβ1 and TGFβ3 Promotes Hematopoiesis in Fanconi Anemia. Exp. Hematol. 2020, 93, 70–84.e4. [Google Scholar] [CrossRef]
  154. Giri, N.; Alter, B.P.; Penrose, K.; Falk, R.T.; Pan, Y.; Savage, S.A.; Williams, M.; Kemp, T.J.; Pinto, L.A. Immune Status of Patients with Inherited Bone Marrow Failure Syndromes. Am. J. Hematol. 2015, 90, 702–708. [Google Scholar] [CrossRef]
  155. Myers, K.C.; Sauter, S.; Zhang, X.; Bleesing, J.J.; Davies, S.M.; Wells, S.I.; Mehta, P.A.; Kumar, A.; Marmer, D.; Marsh, R.; et al. Impaired Immune Function in Children and Adults with Fanconi Anemia. Pediatr. Blood Cancer 2017, 64, 26599. [Google Scholar] [CrossRef]
  156. Jyonouchi, S.; Forbes, L.; Ruchelli, E.; Sullivan, K.E. Dyskeratosis Congenita: A Combined Immunodeficiency with Broad Clinical Spectrum—A Single-Center Pediatric Experience. Pediatr. Allergy Immunol. 2011, 22, 313–319. [Google Scholar] [CrossRef]
  157. Knight, S.W.; Heiss, N.S.; Vulliamy, T.J.; Aalfs, C.M.; McMahon, C.; Richmond, P.; Jones, A.; Hennekam, R.C.M.; Poustka, A.; Mason, P.J.; 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, 335–339. [Google Scholar] [CrossRef]
  158. Iskander, D.; Roberts, I.; Rees, C.; Szydlo, R.; Alikian, M.; Neale, M.; Harrington, Y.; Kelleher, P.; Karadimitris, A.; de la Fuente, J. Impaired Cellular and Humoral Immunity Is a Feature of Diamond-Blackfan Anaemia; Experience of 107 Unselected Cases in the United Kingdom. Br. J. Haematol. 2019, 186, 321–326. [Google Scholar] [CrossRef]
  159. Ragab, I.; Makkeyah, S.; Hassan, N.; Botros, M.; Da Costa, L.; Aly, N.H. Immunodeficiency in Children with Diamond Blackfan and Diamond Blackfan like Anemia. Blood Cells Mol. Dis. 2025, 111, 102911. [Google Scholar] [CrossRef] [PubMed]
  160. Rochowski, A.; Sun, C.; Glogauer, M.; Alter, B.P. Neutrophil Functions in Patients with Inherited Bone Marrow Failure Syndromes. Pediatr. Blood Cancer 2010, 57, 306–309. [Google Scholar] [CrossRef] [PubMed]
  161. Joyce, C.E.; Saadatpour, A.; Ruiz-Gutierrez, M.; Bolukbasi, O.V.; Jiang, L.; Thomas, D.D.; Young, S.; Hofmann, I.; Sieff, C.A.; Myers, K.C.; et al. TGF-β Signaling Underlies Hematopoietic Dysfunction and Bone Marrow Failure in Shwachman-Diamond Syndrome. J. Clin. Investig. 2019, 129, 3821–3826. [Google Scholar] [CrossRef] [PubMed]
  162. Sabbioni, G.; D’Aversa, E.; Breveglieri, G.; Altieri, M.T.; Boni, C.; Pegoraro, A.; Finotti, A.; Gambari, R.; D’Amico, G.; Vella, A.; et al. Constitutive Systemic Inflammation in Shwachman-Diamond Syndrome. Mol. Med. 2025, 31, 81. [Google Scholar] [CrossRef] [PubMed]
  163. Kulanuwat, S.; Jungtrakoon, P.; Tangjittipokin, W.; Yenchitsomanus, P.T.; Plengvidhya, N. Fanconi Anemia Complementation Group C Protection against Oxidative Stress-Induced ß-Cell Apoptosis. Mol. Med. Rep. 2018, 18, 2485–2491. [Google Scholar] [CrossRef]
  164. Cappelli, E.; Bertola, N.; Bruno, S.; Degan, P.; Regis, S.; Corsolini, F.; Banelli, B.; Dufour, C.; Ravera, S. A Multidrug Approach to Modulate the Mitochondrial Metabolism Impairment and Relative Oxidative Stress in Fanconi Anemia Complementation Group A. Metabolites 2021, 12, 6. [Google Scholar] [CrossRef] [PubMed]
  165. Gao, X.; Yu, X.; Zhang, C.; Wang, Y.; Sun, Y.; Sun, H.; Zhang, H.; Shi, Y.; He, X. Telomeres and Mitochondrial Metabolism: Implications for Cellular Senescence and Age-Related Diseases. Stem Cell Rev. Rep. 2022, 18, 2315–2327. [Google Scholar] [CrossRef] [PubMed]
  166. Gavia-García, G.; Rosado-Pérez, J.; Arista-Ugalde, T.L.; Aguiñiga-Sánchez, I.; Santiago-Osorio, E.; Mendoza-Núñez, V.M. Telomere Length and Oxidative Stress and Its Relation with Metabolic Syndrome Components in the Aging. Biology 2021, 10, 253. [Google Scholar] [CrossRef]
  167. Doty, R.T.; Phelps, S.R.; Shadle, C.; Sanchez-Bonilla, M.; Keel, S.B.; Abkowitz, J.L. Coordinate Expression of Heme and Globin Is Essential for Effective Erythropoiesis. J. Clin. Investig. 2015, 125, 4681–4691. [Google Scholar] [CrossRef] [PubMed]
  168. Aspesi, A.; Pavesi, E.; Robotti, E.; Crescitelli, R.; Boria, I.; Avondo, F.; Moniz, H.; Da Costa, L.; Mohandas, N.; Roncaglia, P.; et al. Dissecting the Transcriptional Phenotype of Ribosomal Protein Deficiency: Implications for Diamond-Blackfan Anemia. Gene 2014, 545, 282–289. [Google Scholar] [CrossRef]
  169. Kapralova, K.; Jahoda, O.; Koralkova, P.; Gursky, J.; Lanikova, L.; Pospisilova, D.; Divoky, V.; Horvathova, M. Oxidative DNA Damage, Inflammatory Signature, and Altered Erythrocytes Properties in Diamond-Blackfan Anemia. Int. J. Mol. Sci. 2020, 21, 9652. [Google Scholar] [CrossRef] [PubMed]
  170. Kampen, K.R.; Sulima, S.O.; Vereecke, S.; de Keersmaecker, K. Hallmarks of Ribosomopathies. Nucleic Acids Res. 2021, 48, 1013–1028. [Google Scholar] [CrossRef]
  171. Jensen, L.T.; Phyu, T.; Jain, A.; Kaewwanna, C.; Jensen, A.N. Decreased Accumulation of Superoxide Dismutase 2 within Mitochondria in the Yeast Model of Shwachman-Diamond Syndrome. J. Cell. Biochem. 2019, 120, 13867–13880. [Google Scholar] [CrossRef]
  172. Richardson, C.; Yan, S.; Vestal, C.G. Oxidative Stress, Bone Marrow Failure, and Genome Instability in Hematopoietic Stem Cells. Int. J. Mol. Sci. 2015, 16, 2366–2385. [Google Scholar] [CrossRef]
  173. Bardelčíková, A.; Šoltys, J.; Mojžiš, J. Oxidative Stress, Inflammation and Colorectal Cancer: An Overview. Antioxidants 2023, 12, 901. [Google Scholar] [CrossRef] [PubMed]
  174. Khajuria, R.K.; Munschauer, M.; Ulirsch, J.C.; Fiorini, C.; Ludwig, L.S.; McFarland, S.K.; Abdulhay, N.J.; Specht, H.; Keshishian, H.; Mani, D.R.; et al. Ribosome Levels Selectively Regulate Translation and Lineage Commitment in Human Hematopoiesis. Cell 2018, 173, 90–103.e19. [Google Scholar] [CrossRef]
  175. Horos, R.; Ijspeert, H.; Pospisilova, D.; Sendtner, R.; Andrieu-Soler, C.; Taskesen, E.; Nieradka, A.; Cmejla, R.; Sendtner, M.; Touw, I.P.; et al. Ribosomal Deficiencies in Diamond-Blackfan Anemia Impair Translation of Transcripts Essential for Differentiation of Murine and Human Erythroblasts. Blood 2012, 119, 262–272. [Google Scholar] [CrossRef] [PubMed]
  176. Ludwig, L.S.; Gazda, H.T.; Eng, J.C.; Eichhorn, S.W.; Thiru, P.; Ghazvinian, R.; George, T.I.; Gotlib, J.R.; Beggs, A.H.; Sieff, C.A.; et al. Altered Translation of GATA1 in Diamond-Blackfan Anemia. Nat. Med. 2014, 20, 748–753. [Google Scholar] [CrossRef]
  177. In, K.; Zaini, M.A.; Müller, C.; Warren, A.J.; Von Lindern, M.; Calkhoven, C.F. Shwachman-Bodian-Diamond Syndrome (SBDS) Protein Deficiency Impairs Translation Re-Initiation from C/EBPα and C/EBPβ MRNAs. Nucleic Acids Res. 2016, 44, 4134–4146. [Google Scholar] [CrossRef]
  178. Yoon, A.; Peng, G.; Brandenburg, Y.; Zollo, O.; Xu, W.; Rego, E.; Ruggero, D. Impaired Control of IRES-Mediated Translation in X-Linked Dyskeratosis Congenita. Science 2006, 312, 902–906. [Google Scholar] [CrossRef] [PubMed]
  179. Ruggero, D.; Pandolfi, P.P. Does the Ribosome Translate Cancer? Nat. Rev. Cancer 2003, 3, 179–192. [Google Scholar] [CrossRef] [PubMed]
  180. Penzo, M.; Rocchi, L.; Brugiere, S.; Carnicelli, D.; Onofrillo, C.; Coute, Y.; Brigotti, M.; Montanaro, L. Human Ribosomes from Cells with Reduced Dyskerin Levels Are Intrinsically Altered in Translation. FASEB J. 2015, 29, 3472–3482. [Google Scholar] [CrossRef] [PubMed]
  181. Belo, H.; Silva, G.; Cardoso, B.A.; Porto, B.; Minguillon, J.; Barbot, J.; Coutinho, J.; Casado, J.A.; Benedito, M.; Saturnino, H.; et al. Epigenetic Alterations in Fanconi Anaemia: Role in Pathophysiology and Therapeutic Potential. PLoS ONE 2015, 10, e0139740. [Google Scholar] [CrossRef] [PubMed]
  182. Velleuer, E.; Carlberg, C. Impact of Epigenetics on Complications of Fanconi Anemia: The Role of Vitamin d-Modulated Immunity. Nutrients 2020, 12, 1355. [Google Scholar] [CrossRef]
  183. Skouteris, N.; Papageorgiou, G. PARP Inhibitors in Colorectal Malignancies: A 2023 Update. Rev. Recent Clin. Trials 2023, 19, 101–108. [Google Scholar] [CrossRef] [PubMed]
  184. Wu, F.H.; Wei, H.Z.; Deng, H.Y.; Xiao, G.H.; Zhang, Y.C. PARP in Colorectal Cancer: Molecular Mechanisms, Immunity, Clinical Trials, and Drug Combinations. Neoplasma 2023, 70, 1–14. [Google Scholar] [CrossRef] [PubMed]
  185. Lord, C.J.; Ashworth, A. PARP Inhibitors: Synthetic Lethality in the Clinic. Science 2017, 355, 1152–1158. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular mechanisms underlying IBMFS pathophysiology and CRC risk (created with BioRender.com).
Figure 1. Molecular mechanisms underlying IBMFS pathophysiology and CRC risk (created with BioRender.com).
Ijms 26 10105 g001
Table 1. Tumor types described in patients with IBMFS.
Table 1. Tumor types described in patients with IBMFS.
DiseaseAffected GenesAssociated Solid Tumors
Fanconi AnemiaFANCA, FANCB, FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCJ, FANCL, FANCM, FANCN, FANCO, FANCP, FANCQ, FANCR, FANCS, FANCT, FANCU, FANCV, FANCW [15]Breast, ovarian, colorectal, lung, thyroid, head and neck, brain, bladder, kidney, vulval, vaginal, cervical, prostate, testicular, esophageal, gastric, pancreatic, liver, anal cancer; neuroblastoma, squamous cell carcinoma, basal cell carcinoma, melanoma, soft-tissue sarcoma, osteosarcoma [10,16,17,18,19,20,21,22].
Dyskeratosis CongenitaDKC1, TERC, TERT, TINF2, NAF1, NHP2, NOP10, PARN, POT1, RTEL1, STN1, CTC1, WRAP53, POLA1, ACD, USB1, ZCCHC8, NPM1, MDM4, RPA1, DCLRE1B, TYMS-ENOSF1 [23,24]Head and neck, colorectal, thyroid, liver, lung, esophageal, gastric, anal, pancreatic, cervical cancer; squamous cell carcinoma, basal cell carcinoma [16,17,25].
Diamond–Blackfan Anemia SyndromeRPS19, RPL5, RPS26, RPL11, RPL35a, RPS7, RPL8, RPL35, RPS10, RPS24, RPS17, RPL15, RPS20, RPS28, RPS29, RPS15a, RPS27, RPL9, RPL17, RPL18, RPL26, RPL27, RPL31.
TSR2, GATA1, HEATR3 (non-RP genes) [26]
Colorectal cancer, osteosarcoma; liver, breast, lung, thyroid, kidney, testicular, uterine, cervical, vaginal, gastroesophageal, esophageal, gastric cancer; melanoma, squamous cell carcinoma, basal cell carcinoma, rhabdomyosarcoma, soft-tissue sarcoma [16,17,27,28,29].
Shwachman–Diamond SyndromeSBDS, EFL1, DNAJC21, SRP54 [30]Ovarian, breast, pancreatic cancer; dermatofibrosarcoma, esophageal squamous cell carcinoma, peritoneal carcinoma [17,31,32,33,34].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cagliano, S.; Potenza, M.; La Vecchia, M.; Ellis, S.R.; Dianzani, I.; Aspesi, A. Beyond Hematologic Malignancies: Colorectal Cancer as a Solid Tumor Manifestation of Inherited Bone Marrow Failure Syndromes. Int. J. Mol. Sci. 2025, 26, 10105. https://doi.org/10.3390/ijms262010105

AMA Style

Cagliano S, Potenza M, La Vecchia M, Ellis SR, Dianzani I, Aspesi A. Beyond Hematologic Malignancies: Colorectal Cancer as a Solid Tumor Manifestation of Inherited Bone Marrow Failure Syndromes. International Journal of Molecular Sciences. 2025; 26(20):10105. https://doi.org/10.3390/ijms262010105

Chicago/Turabian Style

Cagliano, Sara, Marta Potenza, Marta La Vecchia, Steven R. Ellis, Irma Dianzani, and Anna Aspesi. 2025. "Beyond Hematologic Malignancies: Colorectal Cancer as a Solid Tumor Manifestation of Inherited Bone Marrow Failure Syndromes" International Journal of Molecular Sciences 26, no. 20: 10105. https://doi.org/10.3390/ijms262010105

APA Style

Cagliano, S., Potenza, M., La Vecchia, M., Ellis, S. R., Dianzani, I., & Aspesi, A. (2025). Beyond Hematologic Malignancies: Colorectal Cancer as a Solid Tumor Manifestation of Inherited Bone Marrow Failure Syndromes. International Journal of Molecular Sciences, 26(20), 10105. https://doi.org/10.3390/ijms262010105

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop