Open Access

Cribriform morular thyroid carcinoma: Clinicopathological and molecular basis for both a preventive and therapeutic approach for a rare tumor (Review)

  • Authors:
    • Soledad Cameselle‑García
    • Ihab Abdulkader‑Nallib
    • María Sánchez‑Ares
    • José Manuel Cameselle‑Teijeiro
  • View Affiliations

  • Published online on: July 17, 2024     https://doi.org/10.3892/or.2024.8778
  • Article Number: 119
  • Copyright: © Cameselle‑García et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cribriform morular thyroid carcinoma (CMTC) has been included within the group of thyroid tumors of uncertain histogenesis in the recent World Health Organization classification of endocrine tumors. Most CMTCs occur in young euthyroid women with multiple (and bilateral) thyroid nodules in cases associated with familial adenomatous polyposis (FAP) or as single nodules in sporadic cases. CMTC generally behaves indolently, while aggressiveness and mortality are associated with high‑grade CMTC. This tumor histologically displays a distinctive combination of growth patterns with morular structures. Strong diffuse nuclear and cytoplasmic immunostaining for β‑catenin is the hallmark of CMTC. Tumor cells are also positive for thyroid transcription factor‑1 and for estrogen and progesterone receptors, but negative for thyroglobulin and calcitonin. It is possible that the CMTC phenotype could result from blockage in the terminal/follicular differentiation of follicular cells (or their precursor cells) secondary to the permanent activation of the Wnt/β‑catenin pathway. In CMTC, the activation of the Wnt/β‑catenin pathway is the central pathogenetic event, which in FAP‑associated cases results from germline mutations of the APC regulator of WNT signaling pathway (APC) gene, and in sporadic cases from somatic inactivating mutations in the APC, AXIN1 and CTNNB1 genes. Estrogens appear to play a tumor‑promoting role by stimulating both the PI3K/AKT/mTOR and the RAS/RAF/MAPK signaling pathways. Additional somatic mutations (i.e. RET rearrangements, or KRAS, phosphatidylinositol‑4,5‑bisphosphate 3‑kinase catalytic subunit α, telomerase reverse transcriptase or tumor protein 53 mutations) may further potentiate the development and progression of CMTC. While hemithyroidectomy would be the treatment of choice for sporadic cases without high‑risk data, total thyroidectomy would be indicated in FAP‑associated cases. There is insufficient clinical data to propose therapies targeting the Wnt/β‑catenin pathway, but multikinase or selective inhibitors could be used in a manner analogous to that of conventional thyroid tumors. It is also unknown whether adjuvant antiestrogenic therapy could be useful in the subgroup of women undergoing surgery with high‑risk CMTC, as well as when there is tumor recurrence and/or metastasis.

Introduction

Cribriform morular thyroid carcinoma (CMTC) is a type of TC associated with familial adenomatous polyposis (FAP), which can also at times occur sporadically (1). This rare tumor was first identified in patients with FAP as a distinctive follicular neoplasm different from conventional follicular TC (FTC) and papillary TC (PTC), usually presenting with multiple and bilateral tumor foci (2). Later, the sporadic counterpart of this neoplasm was reported as a subtype of PTC (cribriform morular variant) (3). In fact, both the familial and sporadic forms of this tumor were designated as the cribriform-morular variant of PTC in the 4th edition of the classification of thyroid tumors of the World Health Organization (WHO) (4). More recently, the peculiar combination of growth patterns of CMTC has been associated with the permanent activation of the Wnt/β-catenin signaling pathway (5). Consequently, this tumor has been included as a distinct TC in the most recent WHO classification of endocrine and neuroendocrine tumors, within the group of thyroid tumors of uncertain histogenesis (1).

As this new tumor entity remains poorly understood (1,69), for the present review, the MEDLINE-PubMed database (https://pubmed.ncbi.nlm.nih.gov) was searched for entries added up to January 2024 with the search terms ‘cribriform’, ‘morular’ and ‘thyroid’ to evaluate the epidemiological, clinicopathological, immunohistochemical and molecular features of CMTC as well as its possible histogenesis. Only papers written in English were reviewed. At the same time, the study aimed to underscore the molecular characteristics of this peculiar neoplasm as a basis for proposing an approach that is both preventive and therapeutic.

Epidemiological characteristics

CMTC accounts for 0.16–0.5% of all cases of TC (10,11) and 0.16–0.66% of the cases of PTC (3,12,13). Although a case of CMTC has been reported in a 15-year-old female with hypothyroidism secondary to neck radiotherapy due to Hodgkin's disease (14), no increased prevalence of CMTC was detected in a population exposed to ionizing radiation post-Chernobyl (11). SARS-CoV-2 infection has been reported in one sporadic case of CMTC and lymphocytic thyroiditis (15).

CMTC has been reported in 0.4–2.6% of patients with FAP (16,17), but these percentages increased to 3–12% when ultrasonography (US) was used (17,18), and it was detected in up to 16% of patients with FAP when US in combination with fine needle aspiration cytology (FNAC) was carried out (19). A higher risk and incidence of CMTC in Hispanic patients with FAP have also been reported (20). About 60% of cases of CMTC occur in the setting of FAP and in 40% of these cases, it is the first clinical manifestation of FAP (13,16,18,2123). Cases of CMTC and a conventional subtype of PTC in different members of the same family sharing the same adenomatous polyposis coli (APC) germline mutation have been described (18,24,25). It is possible that cases of conventional PTC in these families are actually cases of incidental PTC not related to the APC germline mutation.

Clinical findings

Most patients with CMTC are euthyroid young women (female-to-male ratio, ~61:1), and the mean age at diagnosis was determined to be 25 years (range, 8–69 years), both for sporadic and for FAP-associated CMTC (5,12,26). CMTC may be incidentally detected during physical and/or US examination with or without FNAC (19) or present as a painless mass, dysphagia and/or hoarseness (14,27,28). During US screening, most nodules are observed to be well-defined, oval to round, hypoechoic and solid without calcifications (with benign-looking features) (17,18,29,30). While most cases of familial CMTC are multifocal (and bilateral) at diagnosis, most sporadic cases present as single nodules (5,26,31). Because congenital hypertrophy of the retinal pigment epithelium (CHRPE) is reported to occur in up to 80% of individuals with FAP (32,33), and ophthalmoscopic examination can help to clinically confirm hereditary cases of CMTC (34).

In addition to colorectal adenomatous polyps and cancer (3335), the coexistence of TC with medulloblastoma (36), stomach polyps (37), duodenal polyps (36,38), ampullary neoplasms (25,37), hepatoblastoma (39), adrenal adenoma (25), endometrial cancer (40), osteomas (35), lipomas (35), sebaceous and epidermoid cysts (35), desmoid tumors (25,35,38) as well as bilateral breast fibromatosis in the context of silicone prosthetics (41) have been described in patients with FAP.

Pathological features

CMTC is encapsulated or well demarcated and often partially lobulated by sclerotic septa (2,3). Histologically, the tumor exhibits ‘an intricate blending of cribriform, follicular, papillary, trabecular and solid patterns of growth, with morular (squamoid) areas’ (3) (Fig. 1). In different cases of CMTC and even within the same tumor, the percentage of these different growth patterns is variable. The cribriform pattern is composed of tumor cells without interposed stroma that merge with tubulo-glandular follicles (devoid of colloid), as well as with papillae. The papillae are lined by cuboidal or tall cells, often mimicking the tall-cell or columnar-cell subtypes of PTC (3,8). There is also continuity with areas of the trabecular pattern, reminiscent of the hyalinizing trabecular tumor and areas of solid patterns with spindle-like cells (1,3,5). Psammoma bodies are rare. Nuclei are round to oval, clear and can show irregular contours, grooves and pseudoinclusions. Morular structures are nests of nonkeratinized cells having biotin-rich nuclei with characteristic chromatin clearance (1,3,5). Ultrastructurally, these clear morular nuclei have numerous aligned quasiparallel filaments (10,42,43).

There are usually <5 mitotic figures per 2 mm2. Vascular and capsular invasion have been reported in 30 and 40% of tumors, respectively (2,3,5,4447). Isolated cases with marked stromal hyalinization and calcification (48), adamantinous-like pattern (44), adenoid cystic carcinoma-like areas (49) or focal squamous differentiation (9) have been reported.

According to the criteria of the new edition of the WHO classification of thyroid tumors (1,6), rare cases of ‘high-grade CMTC’ with necrosis and/or high proliferative activity have also been reported (7,9,46,5052).

Immunohistochemical profile

Strong diffuse nuclear and cytoplasmic immunostaining for β-catenin is the hallmark of CMTC (1,5,7) (Fig. 1). The immunohistochemical profile of CMTC is summarized in Table I. Tumor cells are always immunoreactive for thyroid transcription factor-1 (TTF1)/NK2 homeobox 1 (NKX2-1) (clones 8G3G7/1 and SPT24) and keratin (KRT) (7), as well as negative for thyroglobulin (Tg), calcitonin and KRT20 (1,5,53). Variable staining for paired box-8 (PAX8) (clones MRQ-50 and SP348) has been detected (5,7,8). Strong positivity for estrogen receptor (ER) (Fig. 1) and progesterone receptor (PR) is highly characteristic, but focal reactivity for androgen receptors has also been detected (1,5,7,11,53). The Ki-67 proliferative index is usually <5% (1,5).

Table I.

Immunohistochemical profile of morular cribriform thyroid carcinoma.

Table I.

Immunohistochemical profile of morular cribriform thyroid carcinoma.

MarkerMain tumor cellsMorular structures(Refs.)
β-catenin++(1,5,7,8,56)
TTF1/NKX2-1+-(1,5,79,56)
PAX8-/+a-(1,5,7,8)
Thyroglobulin--(1,5,8,56)
Thyroperoxidase--(8)
Calcitonin--(1,5)
CEA--(5,8)
Chromogranin-b-(5,44)
Synaptophysin-b-(5,44)
β-estrogen receptors+-(1,5,8,11)
Progesterone receptors+-(1,5,8,11)
Androgen receptors+/--(5)
KRT5-+(1,7)
KRT7++/-(5,8)
KRT19+-(5)
KRT20--(1,5,8)
KRT5/6++(1,8)
KRT (clone AE1/AE3)++(5)
KRT (clone 34βE12)++(1,5)
KRT (clone CAM5.2)++(5)
EMA+NS(1,3,5)
Vimentin+-(5)
CA 19.9-+(5)
CA 125--(5)
CDX2-+(1,5,8,56,57)
CD5-+(1,79)
CD10-/++(1,5,79)
CD117 (c-KIT)-/+-(5,8)
E-cadherin+-/+(1,5)
HBME1+/--(5,7)
Galectin-3+-/+(5,8)
APC+NS(51)
LEF-1+NS(54)
BCL-2++(5,44)
p27+NS(5,44)
Rb+NS(3,5)
β-hCG+NS(5,55)
NIS--(8)
EGFR--(5,7,8,44)
p53+/-cNS(7,8)
p63--(5,7,8)
p40-d-(79)
Calretinin--(5)
WT1--(1,5,44)
SATB2--(8)

a Negativity and positivity have been described with both monoclonal and polyclonal antibodies;

b an isolated case has been described with tumor areas positive for chromogranin and synaptophysin (44);

c diffuse nuclear positivity has been reported to be associated with high tumor grade;

d an isolated case with strong nuclear positivity in poorly differentiated areas has been described (9). TTF1/NKX2-1, NK2 homeobox 1; PAX8, paired box 8; CEA, carcinoembryonic antigen; KRT, keratin; EMA, epithelial membrane antigen; CA, carbohydrate antigen; CD, cluster of differentiation; HBME1, Hector Battifora mesothelial cell-1; APC, adenomatous polyposis coli; LEF-1, lymphoid-enhancing factor-1; Rb, retinoblastoma; β-hCG, human chorionic gonadotropin beta; NIS, sodium-iodide symporter; EGFR; epidermal growth factor receptor; WT1, Wilms tumor protein; SATB2, SATB homeobox 2; NS, not specified.

Morular structures also show aberrant nuclear and cytoplasmic positivity for β-catenin (Fig. 1) and can be easily identified in the tumor by their characteristic positivity for caudal type homeobox 2 (CDX2), CA19.9, CD10, CD5 and KRT5 (3,5,7,56,57).

Follicular lineage

CMTC has been initially considered a distinct type of follicular cell neoplasm associated with FAP (2). According to its nuclear characteristics and the immunohistochemical data (particularly the KRT profile), this tumor was later considered a variant of PTC (3,4). Of note, RET rearrangements, a molecular alteration typical of PTC, have also been described in certain cases of CMTC (44,40,58). Despite the features shared by PTC and CMTC, our group postulated that the latter deserves to be considered its own tumor category, CMTC, based on its ‘peculiar primitive endodermal (intestinal-like) type phenotype and permanent activation of the wingless (Wnt/β-catenin) signaling pathway’ (5). Different researchers have proposed that CMTC arises due to germline or somatic APC mutations or because of somatic mutations in functionally equivalent genes related to the Wnt/β-catenin signaling pathway (5,5860).

The immunohistochemical profile (KRT pattern, positivity for TTF1, variable staining for PAX8 and negativity for calcitonin and CEA), fits with a primary non-neuroendocrine epithelial neoplasm of the thyroid gland. A conclusive follicular cell derivation has not been demonstrated, however, due to negativity for Tg (both at the protein and mRNA levels), thyroperoxidase and solute carrier family 5 member 5 (NIS) (8).

A putative thymic/ultimobranchial pouch-related differentiation has also been proposed due to the lack of Tg in the tumor cells, the occasional negativity for PAX8 and the phenotype of the morular component, positivity for KRT5 and CD5 and negativity for p63, p40, TTF1 and PAX8 (7). Others (8), however, considered that the negativity for p40 and p63 described by Boyraz et al (7) in the same series contradicts their own hypothesis due to the recognized positivity for p40 and p63 in both thymic tissue and intrathyroidal remains of the ultimobranchial body (solid cell nests) (61). A relation to the ultimobranchial body has also been proposed in a unique case of CMTC based on positivity for p40 in the poorly differentiated component (9); in this case, however, the negativity for p40 in the differentiated component of the tumor (including the morulae) as well as the total negativity for CD5 argue against both the ultimobranchial and the thymic derivations, respectively. Another case, however, supports an origin from follicular cells (or their endodermal precursors) based on the cytopathological similarities of this tumor with classic PTC (8).

Morular structures (positive for CD10 and CDX2) have been the subject of different interpretations (6264). Of note, they are characteristically present in a series of tumors such as fetal adenocarcinoma of the lung (65), pancreatoblastoma (66), mesonephric-like adenocarcinoma of the ovary (67), colorectal polyps (68) and others (69), all of which share the permanent activation of the Wnt/β-catenin pathway. It is recognized that the activation of Wnt/β-catenin signaling, through transcription factor CDX2, activates small intestine gene expression at low levels and colonic gene expression at higher levels (70). These mechanisms of embryonic intestinal induction would explain both the blockage in the terminal/follicular differentiation of follicular cells (or their precursor cells) in CMTC, as well as its phenotype. CMTC would be another example of TC with endodermal/intestinal-like (non-committed) differentiation (71). In fact, the immunophenotype of CMTC cells fits into the progenitor stem cell phase in its continuum toward thyroid follicular cells, according to a recent study (72).

In summary, the molecular alterations linked to the constitutive activation of the Wnt/β-catenin pathway are consistent with the development and phenotype of the CMTC from follicular cells (5). Additional points supporting an origin of CMTC from follicular cells (or their precursor cells) are as follows (8): a) The cytoarchitectural and immunohistochemical similarities with other neoplasms of follicular lineage, particularly with PTC; and b) the multicentricity of CMTC associated with patients with FAP (usually with molecularly different tumors), which is easier to explain when they are derived from different follicular cells (or precursor thyroid cells) than from multiple intrathyroid thymic or branchial remnants. Of note, a light increase in Tg levels along with a slight iodine uptake in pulmonary metastases of CMTC after treatment with 131I has been described (73). Furthermore, a rapid increase in serum Tg levels parallel to histologically confirmed lung metastases has also been reported in another case of CMTC after treatment with 131I and a selective inhibitor of receptor tyrosine kinases (lenvatinib) (74). It is still necessary to carry out functional studies on Wnt/β-catenin pathway inactivation in primary CMTC cell cultures to confirm (or discard) this hypothesis (8).

Pathogenesis

FAP is an autosomal dominant genetic disorder caused by germline mutations in the APC gene (34). The constitutive activation of the Wnt/β-catenin signaling pathway has a central role in the pathogenesis of CMTC, mainly through inactivating mutations in the APC, CTNNB1 and AXIN1 genes (5) (Fig. 2). Additional molecular alterations may have a synergistic effect on this pathway, while sex hormones appear to play a promoting role in CMTC development, which is elaborated on further below.

Figure 2.

Schematic representation of the WNT signaling pathway in CMTC. (A) In normal thyroid cells, the canonical (β-catenin-dependent) WNT signaling is in the ‘off’ state because the APC protein, together with the scaffolding protein Axin, serine/threonine kinases CK1 and GSK3β, and β-catenin, form a ‘destruction complex’ that phosphorylates β-catenin, thus promoting its ubiquitination and proteasomal degradation. (B) In CMTC cells, different somatic mutations in APC, CTNNB1 and/or AXIN1 genes alter the destruction complex, impairing β-catenin phosphorylation and degradation in the proteasome. β-catenin accumulates in the cytoplasm and subsequently translocates to the nucleus, where it binds to the TCF/LEF family to activate the transcription of target genes involved in proliferation and loss of differentiation, such as MYC and CCND1. (C) Localization of β-catenin in CMTC. Tumor cells (right) show characteristic nuclear and cytoplasmic positivity for β-catenin, while non-tumor cells (left) show membrane positivity with discrete cytoplasmic immunostaining (original magnification, ×400). CMTC, cribriform morular thyroid carcinoma; TCF/LEF, T-cell factor/lymphoid enhancer factor; CCND1, cyclin D1; APC, APC regulator of WNT signaling pathway; CK1, casein kinase 1; GSK3β, glycogen synthase kinase 3β; DVL, disheveled; CBP, cyclic adenosine monophosphate responsive element binding protein 1 binding protein; LRP5/6, low-density lipoprotein receptor-related protein 5/6.

CMTC and FAP

More than 85% of germline APC mutations in patients with CMTC have been detected in exon 15 (codons 463 to 1,387), in the same genomic location usually associated with CHRPE (39). This area also includes a hotspot (codon 1,061) for CMTC and hepatoblastoma (25,35,37,39), but mutations in codons 140, 159, 161, 213, 278, 302, 313, 325, 332, 418, 471, 499, 554, 578, 582, 593, 625, 654, 698, 704, 737, 769, 778, 804, 834, 848, 935, 937, 938, 964, 976, 977, 979, 993, 1,062, 1,068, 1,073, 1,105, 1,110, 1,157, 1,275, 1,307, 1,309, 1,394, 1,465 and 2,092 have also been described (33,56,75,76). When comparing the prevalence of APC mutations in patients with FAP and TC in relation to the prevalence of such mutations in unselected individuals with FAP, a higher risk of CMTC exists in the population harboring APC mutations proximal to the 5′ end (proximal to codon 528) as well as in the established high-risk group with mutation at codon 1,061 (75). It is noteworthy that part of the β-catenin binding sites and the axin binding sites are outside the mutation cluster region (codons 1,286 to 1,513) of the APC gene (7).

When Wnt/β-catenin signaling is not activated, the APC protein, together with the scaffolding protein Axin, serine/threonine kinases CK1 and glycogen synthase kinase (GSK)3β and β-catenin, form a ‘destruction complex’ that phosphorylates β-catenin, thus promoting its ubiquitination and proteosomal degradation (Fig. 2). APC mutations lead to a truncated APC protein unable to bind to the destruction complex, which prevents β-catenin phosphorylation and leads to its accumulation in the cytoplasm (77) (Fig. 2). Therefore, the accumulated β-catenin translocates to the nucleus where it binds to T-cell factor (TCF)/lymphoid enhancer factor family DNA-binding proteins triggering the constitutive expression of Wnt target genes (MYC, cyclin D1, AXIN2 and Dickkopf 1) involved in proliferation, invasion and loss of differentiation, as well as in oncogenesis (77). The Wnt/β-catenin pathway plays a key role in the maintenance of the intestinal stem cell niche (78) and it is well known that activation of this signaling pathway by APC mutation is sufficient to induce intestinal epithelial hyperproliferation and polyposis (78), matching both the phenotype and the risk of CMTC in patients with FAP.

As the function-inactivating germline mutation of the APC gene can be partially compensated for by the other allele, an additional somatic mutation of the APC gene or another phenotypically equivalent gene (second hit), is required for tumor development (25,52). In fact, in patients with FAP, CMTC is frequently multifocal and shows different somatic APC mutations in each tumor (79). A missense AXIN1 somatic mutation in exon 7 was detected by our group in a case of CMTC associated with FAP (80). In other cases of familial CMTCs, second-hit somatic mutations in the lysine methyltransferase 2D (KMT2D) and KMT2C genes have been reported (25). Of note, both KMT2D and KMT2C have been shown to be transcriptional regulators of the ER gene (81).

Sporadic CMTC

In cases of CMTC without APC germline mutation, the coexistence of two different oncogenic somatic variants of the APC gene has been detected (8,82). In a sporadic case of CTCM with a single somatic APC mutation at codon 1,309, the dominant negative effect of this mutation agrees with the two-hit Knudson hypothesis (59).

The existence of sporadic cases of CMTC with missense somatic mutations of exon 3 of the β-catenin gene (CTNNB1) lacking mutations in APC or loss of heterozygosity near the APC gene (83), have confirmed the key role of the constitutive activation of the Wnt/β-catenin pathway in the development of CMTC. In these cases, mutations in exon 3 of CTNNB1 were located at codons 29, 22, 39, 44, 49, 54 and 56. In a way similar to that described for the APC gene, the multicentric CMTCs showed different somatic mutations of CTNNB1 (83), additionally supporting an independent origin of each of the foci of these multifocal tumors.

In another sporadic case of CMTC, a missense AXIN1 somatic mutation, has been identified in exon 1 (80).

Additional molecular alterations in FAP-associated CMTC

In addition to germline APC mutations, certain cases of familial CMTC showed molecular alterations typically described in follicular-derived TC. In fact, coiled-coil domain containing 6::RET and nuclear receptor coactivator 4::RET rearrangements have been reported in two CMTC cases (III-1 and III-11) from a family (kindred #1) with FAP-associated CMTC (40), and RET translocations have also been described in other series (44,58). A somatic K-RAS mutation (p.Gln61Lys) was detected in FAP-associated CMTC (84). An activating phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) mutation and copy number gains of BRCA2, fibroblast growth factor (FGF23), FGFR1 and PIK3CB have been detected exclusively in the high-grade component of a FAP-associated CMTC, with tumor protein (TP)53 mutation in the well-differentiated component and concurrent APC and telomerase reverse transcriptase (TERT) promoter mutations in both tumor components (9).

Additional molecular alterations in sporadic CMTC

Somatic KRAS mutation was reported in one case of sporadic CMTC (80). The same mutation in exon 9, codon 545 of the PIK3CA gene (p.Glu545Ala) was detected in 3 cases of sporadic CMTC (85). TERT promoter mutation was described in sporadic CMTC (47). As has been confirmed in conventional follicular cell differentiated TC (86), as well as in cases with FAP, sporadic CMTC cases with TERT promoter mutations have been associated with aggressive behavior (47). Alterations of TP53 do not appear to be an early event in CMTC tumorigenesis (8,40).

Sex hormones as a growth promoter of CMTC

ERs may have a role in follicular-lineage TC through genomic (ER-α and ER-β isoforms) and non-genomic pathways, stimulating both the PI3K/AKT/mTOR and the RAS/RAF/MAPK signaling pathways, resulting in increased reactive oxygen species production and cell cycle progression/proliferation through the modulation of cyclin D1, as well as angiogenesis and migration (87). Since genetic alterations in the PI3K/AKT/mTOR pathway mainly promote the transformation of TC with follicular growth pattern (FTC and follicular subtype of PTC) and alterations in the MAPK pathway are associated with PTC with a papillary growth pattern (88), the activation of both pathways by estrogens is consistent with the mixed (papillary and follicular) growth pattern of CMTC. Circulating estrogen levels would produce activation of PI3K/AKT/mTOR and the RAS/RAF/MAPK pathways, which, acting synergistically with the Wnt/β-catenin pathway, would explain the notable predominance of CMTC in women, even among patients with FAP (5).

Recent studies using human colonic epithelial cells demonstrated that activation of ERα but not ERβ increased the protein levels of cyclin D1, proliferating cell nuclear antigen and β-catenin, indicating increased proliferation and migration (89). Consequently, an analogous activation mechanism may be hypothesized to be present in CMTC cells, given their ‘intestinal/non-committed’ phenotype and their richness in ERα, producing and thus further increasing the levels of β-catenin. Since ERβ is overexpressed in PTC stem cells (90), estrogens could also participate in the maintenance of the non-committed status of CMTC cells through the effect on ERβ.

Estrogen-related receptor γ (ERRγ) acts as a tumor suppressor in gastrointestinal cancers through inhibition of the Wnt/β-catenin pathway (91). In preclinical models, it was recently shown that an inverse agonist of ERRγ, GSK5182, enhances the function of NIS protein via the modulation of ERRγ and MAPK signaling, thereby leading to increased responsiveness to radioiodine (RAI) in previously RAI-refractory PTC cells (92). This suggests that estrogens could also participate in the lack of expression of follicular differentiation markers in CMTC cells.

Prognostic markers

CMTC generally behaves more indolently than PTC (26,93), with ~12% of cases having lymph node metastases at diagnosis (3,16,18,27,36,4547,55,58,93,94) and 5% have distant metastases (mainly to the lung, bone and brain) (10,36,44,46,47,51,55,73,74,93) (Table II). To our knowledge, mortality due to CMTC has been reported in only 4 patients (10,16,36,44).

Table II.

Main clinicopathological features of patients with cribriform morular thyroid carcinoma and distant metastasis.

Table II.

Main clinicopathological features of patients with cribriform morular thyroid carcinoma and distant metastasis.

Author(s), yearSex/age, yearsFAPRelevant tumor findingsDistant metastasisFollow-up, monthsOutcome(Refs.)
Okamoto et al, 1995F/29NSTS: 50 mmBone and lungs45.6Died with widespread metastases(10)
Okamoto et al, 1995F/36NSTS: 17 mmLungs92.4Alive with metastases(10)
Perrier et al, 1998F/36YesTwo local recurrences at 6 and 84 months after the initial surgeryExtensive mediastinal adenopathy85Treatment after TT with 131I and T4 suppression. Died of cardiac tamponade due to tumor infiltration.(16)
Fenton et al, 2001F/20YesRecurrence in the right supraclavicular fossaa 29 years after TTExtensive metastasis in the cervical spine (C2-C4, T1 and T2), 360 months after TT360Initial TT followed by 131I. After radiotherapy sessions on the metastases, the patient developed basal pneumonia and died.(36)
Cameselle-Teijeiro et al, 2009M/42YesAngioinvasive neoplasm with positivity for chromogranin and synaptophysin in 40% of tumor cells. 7 MF/10 HPF. Ki-67 index: 60%. APC p.Thr1493Thr RET rearrangementsBilateral lung metastases at diagnosis23After diagnosis, radiolabeled somatostatin analogue therapy (dotatoc-90Yttrium, 5 GBq) was attempted with partial response to treatment. Developed multiple brain metastases treated with palliative whole-brain radiotherapy. The patient continued to deteriorate and died.(44)
Nakazawa et al, 2013F/35NoTS: 90 mm. Poorly differentiated features (50%). Necrosis and prominent venous invasion. 4 MF/10 HPF. Ki-67 index: 15–20%. APC p.Cys520Tyr_fsX534Multiple osteolytic metastases (pelvis and left femur), bilateral lung metastasesa and hilar lympha-denopathya at diagnosis9TT followed by external radiotherapy (23 Gy) and therapy with 131I. Alive with multiple bone and pulmonary metastases.(46)
Alikhan et al, 2015F/25YesTumor infiltration of surgical margins. Positivity for β-hCG in tumor cells. Tumor recurrence and cervical lymph node metastasis 12 months after TT.Left pleural effusion and pleural metastases 36 months after TT49TT followed by 30 mCi 131I. An additional dose of 100 mCi 131I 12 months later. Palliative surgical debulking of the tumor in the neck was performed 48 months after TT.(55)
Oh et al, 2017F/45NoTS: 27 mm. Lymphatic and venous tumor invasion with perithyroid soft tissue infiltration. Metastasis in 5 central compartment lymph nodes. TERTp (C228T)Bone metastasis (right humerus, left seventh rib, C7, L2 and L5 vertebrae, and left iliac bone 16 months after diagnosis48TT with central lymphadenectomy followed by 150 mCi 131I. Partial resection of left 7th rib and curettage of the right distal humerus and palliative radiotherapy (3,000 cGy) for bone metastases. Second ablation with 200 mCi 131I. Patient is alive multiple metastases and continues to be with followed up.(47)
Tsuji et al, 2018F/28NoTS: 91 mm. 6 MF/10 HPF. Ki-67 index: 40·2% (primary tumor) and 22.1% (lung metastases)Multiple bilateral metastases 36 months after TT36TT with cervical lymph node dissection followed by thyrotropin suppression, radioactive iodine ablation and sorafenib therapies.(51)
Akaishi et al, 2018F/37NSTS: 95 mm. Presence of lymphatic and venous tumor invasionLung metastases84Treated with 3 sessions of high doses of 131I. Alive with lung metastases.(93)
Ito et al, 2019F/24YesTS: 87 mm. Ki-67 index: 50% (primary tumor) and 70% (lung metastases). No lymph node metastases.Multiple bilateral metastases 42 months after TT70TT with prophylactic central node dissection and right modified radical neck dissection followed by 100 mCi 131I 45 months after surgery. Subsequently, treatment with lenvatinib for 24 months with partial and sustained response of lung metastases during follow-upb.(74)
Laforga et al, 2020F/45NSTS: 55 mm.Multiple bilateral metastases 84 months after TT84TT followed by 99.5 mCi 131I. Sixty months later, ultrasound findings and slightly increased Tg levels led to a right neck dissection that identified metastasis in 2 lymph nodes. Then, repeated treatment with 131I (147.5 mC1) resulted in slight iodine uptake in pulmonary metastases (histologically confirmed).(73)

a The octeotride- 111Indium scan showed intense uptake.

b At the same time, lenvatinib was also associated with a reduction in the number and size of colon polyps, as well as an increase in Tg levels. FAP, familial adenomatous polyposis; F, female; M, male; NS, not specified; TS, tumor size; TT, total thyroidectomy; 131I, iodine-131; T4, levothyroxine; MF, mitotic figures; HPF, high-power field; β-hCG, human chorionic gonadotropin; Tg, thyroglobulin.

Although cases of encapsulated CMTC with both a high Ki-67 percentage and apoptotic cell count have a favorable prognosis (95), tumors with necrosis and/or a high proliferative index (high-grade CMTC) usually behave aggressively (1,44,46,51). In agreement with the immunohistochemical negativity of CMTC cells for Tg, radioactive iodine whole-body scans show no iodine-avid lesions (47,74), and the serum Tg levels without anti-Tg antibodies are not reliable for following up CMTC patients (47,55,73,96). Curiously, in a case with lung metastases, Tg levels were low for tumor volume, but the associated Tg doubling time was short (74). Uptake of octreotide-111Indium by the CMTC cells has been described in two cases (36,44).

Therapeutic and preventive approaches

This review confirms that the permanent activation of the Wnt/β-catenin signaling pathway constitutes the key pathogenetic event for the development of CMTC. High expression of sex hormone receptors (mainly ERα) in tumor cells would act synergistically in young women promoting tumor development through the PI3K/AKT/mTOR and the RAS/RAF/MAPK signaling pathways. Different somatic mutational events (e.g., RET rearrangements, or KRAS, PIK3CA, TERT or TP53 mutations) may further potentiate the development and/or progression of CMTC. These molecular events constitute the basis for the establishment of targeted therapies but given their current state of development, it is reasonable to propose surgical treatment as an initial approach. This treatment should follow the same surgical approach as that currently recommended for PTC (97), with the specifications described below. The role of radioactive isotopes, hormone therapy and the so-called precision medicine treatments are also discussed.

At the same time, all patients with CMTC should be informed and investigated in relation to the FAP syndrome (97), including genetic counseling with screening for colonic tumors and other extracolonic manifestations of the disease in FAP-positive cases.

Surgical treatment

Hemithyroidectomy would be sufficient in cases of sporadic CMTC without high-risk data (Fig. 3). Total thyroidectomy would be indicated in high-risk cases (high-grade tumors, widely invasive tumors, those with extensive lymphatic or venous vascular invasion and/or with extrathyroidal extension), as well as in all cases of CMTC associated with FAP due to their association with multicentricity and bilaterality. Prophylactic and therapeutic central neck dissection should be performed with the same criteria as those currently used for the treatment of differentiated TC (DTC) of a follicular lineage (97).

Radiopharmaceutical therapy

RAI with 131I has occasionally been used in CMTC (see Table II). However, the use of this isotope in CMTC is controversial, given its uncertain histogenesis and the consistent immunohistochemical negativity of tumor cells for Tg. A few isolated cases with elevation of serum Tg levels (73,74), as well as with slight iodine uptake in pulmonary metastases, have been described (73). Recent data suggest that multikinase inhibitors (MKIs) have the potential for re-establishing 131I uptake in selected patients with RAI refractory metastatic DTC (mDTC) of follicular lineage (98,99). These drugs may also enhance 131I uptake in these mDTCs on a diagnostic and/or post-therapy radioiodine scan, as well as increase the likelihood of a better therapeutic effect for the administered 131I activity (99). Therefore, a divergence between increasing Tg and structural response may be a reliable biomarker of redifferentiation in certain TCs of follicular lineage refractory to radioactive iodine (100). Whether the increase in Tg associated with lenvatinib treatment in a patient with metastatic CMTC (74) represents an example of redifferentiation deserves further investigation. If these findings could be demonstrated in more patients with CMTC, both the follicular lineage of the CMTC and an opportunity for additional 131I therapy would be confirmed.

A partial response to radiolabeled somatostatin analogue therapy (90Yttrium-dotatoc) was reported in a patient with an aggressive CMTC who showed neuroendocrine differentiation in 40% of the tumor cells (44) (Table II). Intense uptake of 111Indium-labelled octreotide was detected in lung and lymph node metastases from another patient with chromogranin- and synaptophysin-negative CMTC (46). The expression of somatostatin receptors in cell cultures and tumor tissue of non-medullary thyroid cancers has been described (101), but limited information is available about these receptors and their possible clinical utility in CMTC (44).

Hormone therapy
Levothyroxine and thyroid stimulating hormone (TSH)

There is no rational basis for TSH suppression in patients with CMTC until their follicular lineage has been demonstrated and/or the existence of TSH receptors in their tumor cells has been confirmed. Replacement treatment with levothyroxine, however, should be indicated in patients with CMTC after total thyroidectomy to prevent hypothyroidism; in addition, TSH suppression could be considered given the existence of isolated cases with elevated Tg and/or uptake of 131I in metastases (73,74,93) (Table II).

Antiestrogens

The use of drugs with antiestrogenic activity widely used in hormone-dependent breast cancer, such as tamoxifen and aromatase inhibitors (102), could imply therapeutic repositioning in patients undergoing surgery with high-risk CMTC, as well as when there is tumor recurrence and/or metastasis (Fig. 4); however, there are currently insufficient clinical data in this regard.

Figure 4.

Schematic illustration of the rationale for possible targeted therapies in CMTC. Blocking the constitutive activation of the Wnt/β-catenin signaling pathway could play an essential role in the treatment of CMTC through the degradation of β-catenin by stabilizing Axin2 (tankyrase inhibitors), CK1 agonists (SSTC3), inhibitors targeting β-catenin-TFC transcription complex (LF3), CBP inhibitors, as well as porcupine inhibitors, but data on clinical efficacy and safety are still insufficient. E2 via ER would have a tumor-promoting role through the PI3K/AKT/mTOR and the RAS/RAF/MAPK signaling pathways, which would justify treatment with both antiestrogens (tamoxifen and aromatase inhibitors) and MKI inhibitors. The detection of additional somatic events (e.g. RET rearrangements) could be subsidiary to treatment with specific inhibitors. CMTC, morular cribriform thyroid carcinoma; E2, estrogens; ER, estrogen receptors; MKI, multikinase; TCF/LEF, T-cell factor/lymphoid enhancer factor; CTNNB1, β-catenin 1; CCND1, cyclin D1; APC, APC regulator of WNT signaling pathway; CK1, casein kinase 1; GSK3β, glycogen synthase kinase 3β; DVL, disheveled; CBP, cyclic adenosine monophosphate responsive element binding protein 1 binding protein; LRP5/6, low-density lipoprotein receptor-related protein 5/6; PI3K, phosphatidylinositol 3-kinase, AKT, serine/threonine kinase; mTOR, mechanistic target of rapamycin kinase; RAS, small GTPase; RAF, Raf-1 proto-oncogene, serine/threonine kinase; MAPK, mitogen-activated protein kinase.

The efficacy of preventive therapy with selective ER modulators or aromatase inhibitors in women with germline APC gene mutation (FAP) may be questionable, however. The preventive benefits in these patients do not seem clear, since they would mainly be young women who need long periods of treatment with drugs that have serious adverse effects (102104).

Targeted therapy
Targeting the Wnt/β-catenin pathway

Blocking the constitutive activation of the Wnt/β-catenin pathway constitutes the theoretical cornerstone of targeted treatment of CMTC (Fig. 4). These therapies should be aimed at restoring the β-catenin destruction complex and/or neutralizing the TCF/β-catenin transcription complex, since the initial component of this signaling cascade, i.e., the Wnt ligand/receptor interface, usually does not participate in the development of CMTC (see above). An antineoplastic effect of tankyrase inhibitors has been demonstrated through the degradation of β-catenin by stabilizing axin 2 (105), and CK1 agonists such as SSTC3 have been shown to inhibit the growth of colorectal cancer in mice (106). Inhibitors targeting β-catenin-TFC transcription complex such as LF3 and ICG-001 (107) as well as CREB binding protein inhibitors have shown antineoplastic efficacy with minimal off-target effects (108), but most of these data still come from preclinical models. Small-molecule compounds targeting the Wnt/β-catenin cascade have been used to inhibit cancer stem cells (109). Unfortunately, phase I/II studies using some of these molecules, such as WNT9748 (a first-in-class Porcupine inhibitor) in colorectal cancer have not shown improved antitumor efficacy (110), and data from another study with niclosamide, an anti-helminthic agent inhibitor of the Wnt/β-catenin pathway in colorectal cancer (111) were not published. Therefore, insufficient data exist to support the therapeutic efficacy and safety of therapies targeting the Wnt/β-catenin pathway in CMTC.

Cyclooxygenase (COX)1 and COX2 inhibitors may inhibit the Wnt/β-catenin pathway in cancer cells; they could therefore be used for adjuvant therapy or chemoprevention of CMTC in a manner analogous to that used in Lynch syndrome and FAP (112), but clinical data are still lacking.

Unfortunately, despite these preclinical data, randomized clinical trials have not demonstrated a clear impact of vitamin D supplementation on colorectal cancer incidence, progression or mortality (113). Vitamin D receptor increases the levels of E-cadherin in the plasma membrane and decreases the levels of β-catenin in the nucleus in differentiated TC (114). Therefore, it is possible that vitamin D could play a chemopreventive role in women with FAP.

Other precision therapy

Although the evidence is scarce, a sustained response to treatment with lenvatinib was observed in a patient with lung metastases of CMTC during follow-up of the patient (74) (Table II). The clinical response to this MKI is consistent with the signaling of the PI3K/AKT/mTOR and the RAS/RAF/MAPK pathways induced by estrogens in CMTC. Lenvatinib, sorafenib, cabozantinib and other MKIs have shown antiangiogenic activity and could be used as treatments for CMTC with progression after surgery in a manner analogous to that of differentiated TC (114) (Fig. 4). Furthermore, selective RET inhibitors (i.e. selpercatinib and pralsetinib) may be used in cases of CMTC with RET rearrangements (115). Thus, although the clinical evidence is still insufficient, the detection of somatic mutations through next-generation sequencing could represent a precision therapeutic alternative in CMTC.

Limitations

In this review, hypotheses are proposed in an attempt to understand the nature of CMTC and establish a therapeutic approach. As most of the information comes from isolated cases or small series, the level of evidence is limited. Therefore, additional validations and appropriate clinical trials are still necessary.

Conclusions

CMTC is the TC associated with FAP. This rare tumor usually occurs in young women with multiple lesions or as a single nodule in sporadic cases. Constitutive activation of the WNT/β-catenin signaling pathway plays a key role in the distinctive growth pattern of CMTC and its immunohistochemical profile. Strong nuclear and cytoplasmic positivity for β-catenin is the hallmark of CMTC. Tumor cells are also positive for ER and PR and for TTF1/NKX2-1, but negative for Tg and calcitonin. Although the histogenesis of CMTC is controversial, the presence of a follicular or follicular precursor cell lineage may be postulated, whose block in differentiation would be secondary to WNT/β-catenin signaling. In FAP cases, total thyroidectomy is the treatment of choice due to the frequent multifocality and bilaterality of CMTC. Despite the central role of the Wnt/β-catenin signaling pathway in the pathogenesis of this tumor, more studies seem necessary to be able to translate the targeting of this signaling pathway into clinical practice. Although there is still little clinical evidence, in cases of CMTC recurrence and metastasis, targeted treatments with MKIs or more selective inhibitors could be useful. And although sufficient clinical data are also lacking, adjuvant antiestrogenic therapy could also be useful in women undergoing surgery with high-risk CMTC as well as when there is tumor recurrence and/or metastasis. The chemopreventive role of vitamin D and COX inhibitors in women with FAP requires additional investigation.

Acknowledgements

Not applicable.

Funding

This work was supported by the Instituto de Salud Carlos III (ISCIII), Spain (grant nos. PI19/01316 and PI23/00722), co-funded by the European Union (EU).

Availability of data and materials

Not applicable.

Authors' contributions

All authors contributed to the conception of the review and interpretation of the data. SCG: Review of the literature, writing-original draft and editing. IAN and MSA: critical review and editing. JMCT: Review of the literature, writing-original draft and editing, and funding acquisition. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

The images in Figs. 1 and 2C were obtained from cases in the TIROCHUS collection. TIROCHUS is a registered collection of human tissues at the ISCIII national biobank (no. c.0003960 to JMCT) of the Department of Pathology of the Hospital Clínico Universitario (Santiago de Compostela, Spain). Informed consent was obtained from each patient with a specific signed document updated to the current legislation.

Competing interests

The authors declare that they have no competing interests.

References

1 

Erickson LA, Mete O, Cameselle-Teijeiro JM, LiVolsi V, Sobrinho-Simoes M and Jung CK: Cribriform morular thyroid carcinoma. WHO Classification of Tumours Editorial Board. Endocrine and neuroendocrine tumours [Internet] Lyon (France): International Agency for Research on Cancer; 2022, [2023 03 07]. (WHO classification of tumours series, 5th edition. Vol. 10). Available from:. https://tumourclassification.iarc.who.int/chapters/53

2 

Harach HR, Williams GT and Williams ED: Familial adenomatous polyposis associated thyroid carcinoma: A distinct type of follicular cell neoplasm. Histopathology. 25:549–561. 1994. View Article : Google Scholar : PubMed/NCBI

3 

Cameselle-Teijeiro J and Chan JK: Cribriform-morular variant of papillary carcinoma: A distinctive variant representing the sporadic counterpart of familial adenomatous polyposis-associated thyroid carcinoma? Mod Pathol. 12:400–411. 1999.PubMed/NCBI

4 

Lloyd RV, Osamura RY, Klöppel G and Rosai J: WHO classification of tumours of endocrine organs. 4th edition. IARC; Lyon: 2017

5 

Cameselle-Teijeiro JM, Peteiro-González D, Caneiro-Gómez J, Sánchez-Ares M, Abdulkader I, Eloy C, Melo M, Amendoeira I, Soares P and Sobrinho-Simões M: Cribriform-morular variant of thyroid carcinoma: A neoplasm with distinctive phenotype associated with the activation of the WNT/β-catenin pathway. Mod Pathol. 31:1168–1179. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Baloch ZW, Asa SL, Barletta JA, Ghossein RA, Juhlin CC, Jung CK, LiVolsi VA, Papotti MG, Sobrinho-Simões M, Tallini G and Mete O: Overview of the 2022 WHO classification of thyroid neoplasms. Endocr Pathol. 33:27–63. 2022. View Article : Google Scholar : PubMed/NCBI

7 

Boyraz B, Sadow PM, Asa SL, Dias-Santagata D, Nosé V and Mete O: Cribriform-morular thyroid carcinoma is a distinct thyroid malignancy of uncertain cytogenesis. Endocr Pathol. 32:327–335. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Echegoyen-Silanes A, Pineda-Arribas JJ, Sánchez-Ares M, Cameselle-García S, Sobrino B, Ruíz-Ponte C, Piso-Neira M, Anda E and Cameselle-Teijeiro JM: Cribriform morular thyroid carcinoma: A case report with pathological, immunohistochemical, and molecular findings suggesting an origin from follicular cells (or their endodermal precursors). Virchows Arch. 482:615–623. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Dettmer MS, Hürlimann S, Scheuble L, Vassella E, Perren A and Wicke C: Cribriform morular thyroid carcinoma-ultimobranchial pouch-related? Deep molecular insights of a unique case. Endocr Pathol. 34:342–348. 2023. View Article : Google Scholar : PubMed/NCBI

10 

Okamoto Y, Kashima K, Daa T, Yokoyama S, Nakayama I and Noguchi S: Morule with biotin-containing intranuclear inclusions in thyroid carcinoma. Pathol Int. 45:573–579. 1995. View Article : Google Scholar : PubMed/NCBI

11 

Lam AK and Fridman M: Characteristics of cribriform morular variant of papillary thyroid carcinoma in post-Chernobyl affected region. Hum Pathol. 74:170–177. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Hirokawa M, Maekawa M, Kuma S and Miyauchi A: Cribriform-morular variant of papillary thyroid carcinoma-cytological and immunocytochemical findings of 18 cases. Diagn Cytopathol. 38:890–896. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Levy RA, Hui VW, Sood R, Fish S, Markowitz AJ, Wong RJ and Guillem JG: Cribriform-morular variant of papillary thyroid carcinoma: An indication to screen for occult FAP. Fam Cancer. 13:547–551. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Or Koca A and Güler Şimşek G: Post-radiotherapy cribriform-morular thyroid carcinoma. J Clin Lab Anal. 37:e248192023. View Article : Google Scholar : PubMed/NCBI

15 

Lahbacha B, Chaabane A, Nechi S, Mfarrej MK, Douggaz A, Kharrat G and Chelbi E: Cribriform-morular thyroid carcinoma: A case report with review of the literature. Ear Nose Throat J. 14556132311523322023.(Epub ahead of print). PubMed/NCBI

16 

Perrier ND, van Heerden JA, Goellner JR, Williams ED, Gharib H, Marchesa P, Church JM, Fazio VW and Larson DR: Thyroid cancer in patients with familial adenomatous polyposis. World J Surg. 22:738–743. 1998. View Article : Google Scholar : PubMed/NCBI

17 

Jarrar AM, Milas M, Mitchell J, Laguardia L, O'Malley M, Berber E, Siperstein A, Burke C and Church JM: Screening for thyroid cancer in patients with familial adenomatous polyposis. Ann Surg. 253:515–521. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Herraiz M, Barbesino G, Faquin W, Chan-Smutko G, Patel D, Shannon KM, Daniels GH and Chung DC: Prevalence of thyroid cancer in familial adenomatous polyposis syndrome and the role of screening ultrasound examinations. Clin Gastroenterol Hepatol. 5:367–373. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Uchino S, Ishikawa H, Miyauchi A, Hirokawa M, Noguchi S, Ushiama M, Yoshida T, Michikura M, Sugano K and Sakai T: Age- and gender-specific risk of thyroid cancer in patients with familial adenomatous polyposis. J Clin Endocrinol Metab. 101:4611–4617. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Casellas-Cabrera N, Díaz-Algorri Y, Carlo-Chévere VJ, González-Pons M, Rodríguez-Mañón N, Pérez-Mayoral J, Bertrán-Rodríguez C, Soto-Salgado M, Giardiello FM, Rodríguez-Quilichini S and Cruz-Correa M: Risk of thyroid cancer among Caribbean Hispanic patients with familial adenomatous polyposis. Fam Cancer. 15:267–274. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Kurihara K, Shimizu S, Chong J, Hishima T, Funata N, Kashiwagi H, Nagai H, Miyaki M and Fukayama M: Nuclear localization of immunoreactive beta-catenin is specific to familial adenomatous polyposis in papillary thyroid carcinoma. Jpn J Cancer Res. 91:1100–1102. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Tomoda C, Miyauchi A, Uruno T, Takamura Y, Ito Y, Miya A, Kobayashi K, Matsuzuka F, Kuma S, Kuma K and Kakudo K: Cribriform-morular variant of papillary thyroid carcinoma: clue to early detection of familial adenomatous polyposis-associated colon cancer. World J Surg. 28:886–889. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Ito Y, Miyauchi A, Ishikawa H, Hirokawa M, Kudo T, Tomoda C and Miya A: Our experience of treatment of cribriform morular variant of papillary thyroid carcinoma; difference in clinicopathological features of FAP-associated and sporadic patients. Endocr J. 58:685–689. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Cetta F, Moltoni L, Barellini L, Monti M and Gotti G: Familial adenomatous polyposis-associated papillary thyroid carcinoma shows an indolent course and usually, but not always, belongs to the cribriform-morular variant of papillary thyroid carcinoma. Acta Cytol. 56:107–108. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Nieminen TT, Walker CJ, Olkinuora A, Genutis LK, O'Malley M, Wakely PE, LaGuardia L, Koskenvuo L, Arola J, Lepistö AH, et al: Thyroid carcinomas that occur in familial adenomatous polyposis patients recurrently harbor somatic variants in APC, BRAF, and KTM2D. Thyroid. 30:380–388. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Park J, Kim JW, Park H, Park SY, Kim TH, Kim SW, Oh YL and Chung JH: Multifocality in a patient with cribriform-morular variant of papillary thyroid carcinoma is an important clue for the diagnosis of familial adenomatous polyposis. Thyroid. 29:1606–1614. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Yamashita T, Hosoda Y, Kameyama K, Aiba M, Ito K and Fujimoto Y: Peculiar nuclear clearing composed of microfilaments in papillary carcinoma of the thyroid. Cancer. 70:2923–2928. 1992. View Article : Google Scholar : PubMed/NCBI

28 

Dalal KM, Moraitis D, Iwamoto C, Shaha AR, Patel SG and Ghossein RA: Clinical curiosity: Cribriform-morular variant of papillary thyroid carcinoma. Head Neck. 28:471–476. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Hizawa K, Iida M, Yao T, Aoyagi K, Oohata Y, Mibu R, Yamasaki K, Hirata T and Fujishima M: Association between thyroid cancer of cribriform variant and familial adenomatous polyposis. J Clin Pathol. 49:611–613. 1996. View Article : Google Scholar : PubMed/NCBI

30 

Chong Y, Shin JH, Oh YL, Han BK and Ko EY: Cribriform-morular variant of papillary thyroid carcinoma: Ultrasonographic and clinical characteristics. Thyroid. 23:45–49. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Guilmette J and Nosé V: Hereditary and familial thyroid tumours. Histopathology. 72:70–81. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Rehan S and Aye K: In patients with a positive family history of familial adenomatous polyposis can the condition be diagnosed from the presence of congenital hypertrophy of the retinal pigment epithelium detected via an eye examination: A systematic review. Clin Exp Ophthalmol. 48:98–116. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Xu M, Zheng Y, Zuo Z, Zhou Q, Deng Q, Wang J and Wang D: De novo familial adenomatous polyposis associated thyroid cancer with a c.2929delG frameshift deletion mutation in APC: A case report and literature review. World J Surg Oncol. 21:732023. View Article : Google Scholar : PubMed/NCBI

34 

Dinarvand P, Davaro EP, Doan JV, Ising ME, Evans NR, Phillips NJ, Lai J and Guzman MA: Familial adenomatous polyposis syndrome: An update and review of extraintestinal manifestations. Arch Pathol Lab Med. 143:1382–1398. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Truta B, Allen BA, Conrad PG, Kim YS, Berk T, Gallinger S, Bapat B, Terdiman JP and Sleisenger MH: Genotype and phenotype of patients with both familial adenomatous polyposis and thyroid carcinoma. Fam Cancer. 2:95–99. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Fenton PA, Clarke SE, Owen W, Hibbert J and Hodgson SV: Cribriform variant papillary thyroid cancer: A characteristic of familial adenomatous polyposis. Thyroid. 11:193–197. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Lee S, Hong SW, Shin SJ, Kim YM, Rhee Y, Jeon BI, Moon WC, Oh MR and Lim SK: Papillary thyroid carcinoma associated with familial adenomatous polyposis: Molecular analysis of pathogenesis in a family and review of the literature. Endocr J. 51:317–323. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Kashiwagi H, Konishi F, Kanazawa K and Miyaki M: Sisters with familial adenomatous polyposis affected with thyroid carcinoma, desmoid tumour and duodenal polyposis. Br J Surg. 83:2281996. View Article : Google Scholar : PubMed/NCBI

39 

Cetta F, Montalto G, Gori M, Curia MC, Cama A and Olschwang S: Germline mutations of the APC gene in patients with familial adenomatous polyposis-associated thyroid carcinoma: Results from a European cooperative study. J Clin Endocrinol Metab. 85:286–292. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Soravia C, Sugg SL, Berk T, Mitri A, Cheng H, Gallinger S, Cohen Z, Asa SL and Bapat BV: Familial adenomatous polyposis-associated thyroid cancer: A clinical, pathological, and molecular genetics study. Am J Pathol. 154:127–135. 1999. View Article : Google Scholar : PubMed/NCBI

41 

Silva S, Lage P, Cabral F, Alves R, Catarino A, Félix A and André S: Bilateral breast fibromatosis after silicone prosthetics in a patient with classic familial adenomatous polyposis: A case report. Oncol Lett. 16:1449–1454. 2018.PubMed/NCBI

42 

Kameyama K, Mukai M, Takami H and Ito K: Cribriform-morular variant of papillary thyroid carcinoma: Ultrastructural study and somatic/germline mutation analysis of the APC gene. Ultrastruct Pathol. 28:97–102. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Nesland JM: Ultrastructural and molecular analysis of cribriform-morular variant of papillary thyroid carcinoma. Ultrastruct Pathol. 28:532004. View Article : Google Scholar : PubMed/NCBI

44 

Cameselle-Teijeiro J, Menasce LP, Yap BK, Colaco RJ, Castro P, Celestino R, Ruíz-Ponte C, Soares P and Sobrinho-Simões M: Cribriform-morular variant of papillary thyroid carcinoma: Molecular characterization of a case with neuroendocrine differentiation and aggressive behavior. Am J Clin Pathol. 131:134–142. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Jung CK, Choi YJ, Lee KY, Bae JS, Kim HJ, Yoon SK, Son YI, Chung JH and Oh YL: The cytological, clinical, and pathological features of the cribriform-morular variant of papillary thyroid carcinoma and mutation analysis of CTNNB1 and BRAF genes. Thyroid. 19:905–913. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Nakazawa T, Celestino R, Machado JC, Cameselle-Teijeiro JM, Vinagre J, Eloy C, Benserai F, Lameche S, Soares P and Sobrinho-Simões M: Cribriform-morular variant of papillary thyroid carcinoma displaying poorly differentiated features. Int J Surg Pathol. 21:379–389. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Oh EJ, Lee S, Bae JS, Kim Y, Jeon S and Jung CK: TERT Promoter mutation in an aggressive cribriform morular variant of papillary thyroid carcinoma. Endocr Pathol. 28:49–53. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Sahu A, Shahin M, Jain P, Sultania M and Ayyanar P: Cribriform morular thyroid carcinoma: A rare case and associated uncommon features. Int J Surg Pathol. 106689692312065722023.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

49 

Baloch ZW, Segal JP and Livolsi VA: Unique growth pattern in papillary carcinoma of the thyroid gland mimicking adenoid cystic carcinoma. Endocr Pathol. 22:200–205. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Mogoş V, Mogoş S, Sfarti C, Băcăuanu R, Huţanu O, Cotea E, Ciobanu D, Tudorache C and Târcoveanu E: Familial syndromic papillary thyroid carcinoma report of two cases. Rev Med Chir Soc Med Nat Iasi. 116:1048–1054. 2012.PubMed/NCBI

51 

Tsuji H, Yasuoka H, Nakamura Y, Hirokawa M, Hiroshima T, Sakamaki Y, Miyauchi A and Tsujimoto M: Aggressive cribriform-morular variant of papillary thyroid carcinoma: Report of an unusual case with pulmonary metastasis displaying poorly differentiated features. Pathol Int. 68:700–705. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Corean J, Furtado LV, Kadri S, Segal JP and Emerson LL: Cribriform-morular variant of papillary thyroid carcinoma with poorly differentiated features: A case report with immunohistochemical and molecular genetic analysis. Int J Surg Pathol. 27:294–304. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Cameselle-Teijeiro JM, Mete O, Asa SL and LiVolsi V: Inherited follicular epithelial-derived thyroid carcinomas: From molecular biology to histological correlates. Endocr Pathol. 32:77–101. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Mohindra S, Sakr H, Sturgis C and Chute DJ: LEF-1 is a sensitive marker of cribriform morular variant of papillary thyroid carcinoma. Head Neck Pathol. 12:455–462. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Alikhan M, Koshy A, Hyjek E, Stenson K, Cohen RN and Yeo KT: Discrepant serum and urine β-hCG results due to production of β-hCG by a cribriform-morular variant of thyroid papillary carcinoma. Clin Chim Acta. 438:181–185. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Lyu S, Zhong G, Chen H, Li J and Li M: The first case of cribriform-morular thyroid carcinoma and FAP with APC gene mutation in China: A case report and brief review. Case Rep Gastrointest Med. 2023:62224322023.PubMed/NCBI

57 

Chang HY, Lim MY and Bundele MM: Cribriform morular thyroid carcinoma: A recently reclassified entity. ANZ J Surg. 93:2257–2259. 2023. View Article : Google Scholar : PubMed/NCBI

58 

Cetta F, Curia MC, Montalto G, Gori M, Cama A, Battista P and Barbarisi A: Thyroid carcinoma usually occurs in patients with familial adenomatous polyposis in the absence of biallelic inactivation of the adenomatous polyposis coli gene. J Clin Endocrinol Metab. 86:427–432. 2001. View Article : Google Scholar : PubMed/NCBI

59 

Cameselle-Teijeiro J, Ruiz-Ponte C, Loidi L, Suarez-Peñaranda J, Baltar J and Sobrinho-Simoes M: Somatic but not germline mutation of the APC gene in a case of cribriform-morular variant of papillary thyroid carcinoma. Am J Clin Pathol. 115:486–493. 2001. View Article : Google Scholar : PubMed/NCBI

60 

Kumamoto K, Ishida H, Ohsawa T, Ishibashi K, Ushiama M, Yoshida T and Iwama T: Germline and somatic mutations of the APC gene in papillary thyroid carcinoma associated with familial adenomatous polyposis: Analysis of three cases and a review of the literature. Oncol Lett. 10:2239–2243. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Manzoni M, Roversi G, Di Bella C, Pincelli AI, Cimino V, Perotti M, Garancini M and Pagni F: Solid cell nests of the thyroid gland: Morphological, immunohistochemical and genetic features. Histopathology. 68:866–874. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Houghton O, Connolly LE and McCluggage WG: Morules in endometrioid proliferations of the uterus and ovary consistently express the intestinal transcription factor CDX2. Histopathology. 53:156–165. 2008. View Article : Google Scholar : PubMed/NCBI

63 

McCluggage WG and Van de Vijver K: SATB2 is consistently expressed in squamous morules associated with endometrioid proliferative lesions and in the stroma of atypical polypoid adenomyoma. Int J Gynecol Pathol. 38:397–403. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Travaglino A, Raffone A, Russo D, Guadagno E, Pignatiello S, Moretta P, Zullo F, Del Basso De Caro M, Insabato L and Mascolo M: Does endometrial morular metaplasia represent odontogenic differentiation? Virchows Arch. 479:607–616. 2021. View Article : Google Scholar : PubMed/NCBI

65 

Gu Y, Zhang S, Liang X, Zhao H, Li X and Lu J: Clinical and pathological characteristics and prognosis of lung adenocarcinoma with high-grade fetal features: A retrospective analysis. Int J Surg Pathol. 32:667–678. 2024. View Article : Google Scholar : PubMed/NCBI

66 

Isobe T, Seki M, Yoshida K, Sekiguchi M, Shiozawa Y, Shiraishi Y, Kimura S, Yoshida M, Inoue Y, Yokoyama A, et al: Integrated molecular characterization of the lethal pediatric cancer pancreatoblastoma. Cancer Res. 78:865–876. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Xu J, Park KJ, Rehrauer WM and Weisman PS: Mesonephric-like adenocarcinoma of the ovary with squamoid morular metaplasia, aberrant β-catenin expression, and concurrent FGFR2 and CTNNB1 mutations: A case report. Virchows Arch. 484:147–150. 2024. View Article : Google Scholar : PubMed/NCBI

68 

Lee HE, Chandan VS, Lee CT and Wu TT: Squamoid morules in the pseudoinvasive foci of colonic polyp morphologically mimic invasive carcinoma. Hum Pathol. 68:54–60. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Cameselle-Teijeiro J, Alberte-Lista L, Chiarelli S, Buriticá C, Gonçalves L, González-Cámpora R and Nogales FF: CD10 is a characteristic marker of tumours forming morules with biotin-rich, optically clear nuclei that occur in different organs. Histopathology. 52:389–392. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Sherwood RI, Maehr R, Mazzoni EO and Melton DA: Wnt signaling specifies and patterns intestinal endoderm. Mech Dev. 128:387–400. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Cameselle-Teijeiro J, Febles-Pérez C and Sobrinho-Simões M: Papillary and mucoepidermoid carcinoma of the thyroid with anaplastic transformation: A case report with histologic and immunohistochemical findings that support a provocative histogenetic hypothesis. Pathol Res Pract. 191:1214–1221. 1995. View Article : Google Scholar : PubMed/NCBI

72 

Davies TF, Latif R, Sachidanandam R and Ma R: The transient human thyroid progenitor cell: Examining the thyroid continuum from stem cell to follicular cell. Thyroid. 31:1151–1159. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Laforga JB, Pedro T and Gasent JM: Pulmonary metastasis of cribriform-morular variant of thyroid carcinoma mimicking primary adenocarcinoma of the lung: A potential pitfall. Diagn Cytopathol. 48:78–81. 2020. View Article : Google Scholar : PubMed/NCBI

74 

Ito Y, Ishikawa H, Kihara M, Hirokawa M, Kiyota N, Kasahara T and Miyauchi A: Control of lung metastases and colon polyposis with lenvatinib therapy in a patient with cribriform-morular variant of papillary thyroid carcinoma and an APC gene mutation: A case study. Thyroid. 29:1511–1517. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Septer S, Slowik V, Morgan R, Dai H and Attard T: Thyroid cancer complicating familial adenomatous polyposis: Mutation spectrum of at-risk individuals. Hered Cancer Clin Pract. 11:132013. View Article : Google Scholar : PubMed/NCBI

76 

Domingues GAB, Kizys MML, Janovsky CCPS, de Barros Maciel RM, Dias-da-Silva MR, Martins JRM, Camacho CP and Cunha LL: The impact of the genetic background in a patient with papillary thyroid cancer and familial adenomatous polyposis. Arch Endocrinol Metab. 66:112–117. 2022. View Article : Google Scholar : PubMed/NCBI

77 

Krausova M and Korinek V: Wnt signaling in adult intestinal stem cells and cancer. Cell Signal. 26:570–579. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Mah AT, Yan KS and Kuo CJ: Wnt pathway regulation of intestinal stem cells. J Physiol. 594:4837–4847. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Miyaki M, Iijima T, Ishii R, Hishima T, Mori T, Yoshinaga K, Takami H, Kuroki T and Iwama T: Molecular evidence for multicentric development of thyroid carcinomas in patients with familial adenomatous polyposis. Am J Pathol. 157:1825–1827. 2000. View Article : Google Scholar : PubMed/NCBI

80 

Cameselle-Teijeiro JM, Peteiro-González D, Carreira M, Abdulkader I, Reyes-Santías R, Celestino R, Romero Rojas A, Ruíz-Ponte C, Soares P, Casanueva F and Sobrinho-Simões M: Molecular alterations in the cribriform-morular variant of papillary thyroid carcinoma. Virchows Arch. 469 (1 Supp):S722016.

81 

Hussain I, Deb P, Chini A, Obaid M, Bhan A, Ansari KI, Mishra BP, Bobzean SA, Udden SMN, Alluri PG, et al: HOXA5 expression is elevated in breast cancer and is transcriptionally regulated by estradiol. Front Genet. 11:5924362020. View Article : Google Scholar : PubMed/NCBI

82 

Aydemirli MD, van der Tuin K, Hes FJ, van den Ouweland AMW, van Wezel T, Kapiteijn E and Morreau H: A unique case of two somatic APC mutations in an early onset cribriform-morular variant of papillary thyroid carcinoma and overview of the literature. Fam Cancer. 19:15–21. 2020. View Article : Google Scholar : PubMed/NCBI

83 

Xu B, Yoshimoto K, Miyauchi A, Kuma S, Mizusawa N, Hirokawa M and Sano T: Cribriform-morular variant of papillary thyroid carcinoma: A pathological and molecular genetic study with evidence of frequent somatic mutations in exon 3 of the beta-catenin gene. J Pathol. 199:58–67. 2003. View Article : Google Scholar : PubMed/NCBI

84 

Giannelli SM, McPhaul L, Nakamoto J and Gianoukakis AG: Familial adenomatous polyposis-associated, cribriform morular variant of papillary thyroid carcinoma harboring a K-RAS mutation: Case presentation and review of molecular mechanisms. Thyroid. 24:1184–1189. 2014. View Article : Google Scholar : PubMed/NCBI

85 

Kwon MJ, Rho YS, Jeong JC, Shin HS, Lee JS, Cho SJ and Nam ES: Cribriform-morular variant of papillary thyroid carcinoma: A study of 3 cases featuring the PIK3CA mutation. Hum Pathol. 46:1180–1188. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Melo M, da Rocha AG, Vinagre J, Batista R, Peixoto J, Tavares C, Celestino R, Almeida A, Salgado C, Eloy C, et al: TERT promoter mutations are a major indicator of poor outcome in differentiated thyroid carcinomas. J Clin Endocrinol Metab. 99:E754–E765. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Denaro N, Romanò R, Alfieri S, Dolci A, Licitra L, Nuzzolese I, Ghidini M, Bareggi C, Bertaglia V, Solinas C and Garrone O: The tumor microenvironment and the estrogen loop in thyroid cancer. Cancers (Basel). 15:24582023. View Article : Google Scholar : PubMed/NCBI

88 

Cancer Genome Atlas Research Network, . Integrated genomic characterization of papillary thyroid carcinoma. Cell. 159:676–690. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Wan H, Li J, Chen X, Sellers ZM and Dong H: Divergent roles of estrogen receptor subtypes in regulating estrogen-modulated colonic ion transports and epithelial repair. J Biol Chem. 299:1050682023. View Article : Google Scholar : PubMed/NCBI

90 

Li M, Chai HF, Peng F, Meng YT, Zhang LZ, Zhang L, Zou H, Liang QL, Li MM, Mao KG, et al: Estrogen receptor β upregulated by lncRNA-H19 to promote cancer stem-like properties in papillary thyroid carcinoma. Cell Death Dis. 9:11202018. View Article : Google Scholar : PubMed/NCBI

91 

Guo X, Yue L, Li M, Dai A, Sun J, Fang L, Zhao H and Sun Q: Nuclear receptor estrogen-related receptor gamma suppresses colorectal cancer aggressiveness by regulating Wnt/β-catenin signaling. Carcinogenesis. 43:865–873. 2022. View Article : Google Scholar : PubMed/NCBI

92 

Singh TD, Lee JE, Son KH, Lee BR, Kim SK, Gulwani D, Sarangthem V and Jeon YH: An inverse agonist of estrogen-related receptor gamma, GSK5182, enhances Na+/I symporter function in radioiodine-refractory papillary thyroid cancer cells. Cells. 12:4702023. View Article : Google Scholar : PubMed/NCBI

93 

Akaishi J, Kondo T, Sugino K, Ogimi Y, Masaki C, Hames KY, Yabuta T, Tomoda C, Suzuki A, Matsuzu K, et al: Cribriform-morular variant of papillary thyroid carcinoma: Clinical and pathological features of 30 cases. World J Surg. 42:3616–3623. 2018. View Article : Google Scholar : PubMed/NCBI

94 

Rossi ED, Revelli L, Martini M, Taddei A, Pintus C, Panunzi C and Fadda G: Cribriform-morular variant of papillary thyroid carcinoma in an 8-year-old girl: A case report with immunohistochemical and molecular testing. Int J Surg Pathol. 20:629–632. 2012. View Article : Google Scholar : PubMed/NCBI

95 

Hirokawa M, Matsuda K, Kudo T, Higuchi M, Suzuki A, Takada N, Nakashima M and Miyauchi A: Cribriform-morular variant of papillary thyroid carcinoma shows high Ki-67 labeling indices, despite its excellent prognosis. Pathobiology. 86:248–253. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Fujimoto T, Hirokawa M, Ota H, Yabuta T, Fukushima M, Kobayashi K, Amino N and Miyauchi A: Characteristic sonographic features of cribriform papillary thyroid carcinoma for differentiation from other thyroid nodules. J Med Ultrason (2001). 42:83–87. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger M, et al: 2015 American thyroid association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: The American thyroid association guidelines task force on thyroid nodules and differentiated thyroid cancer. Thyroid. 26:1–133. 2016. View Article : Google Scholar : PubMed/NCBI

98 

Weber M, Kersting D, Riemann B, Brandenburg T, Führer-Sakel D, Grünwald F, Kreissl MC, Dralle H, Weber F, Schmid KW, et al: Enhancing radioiodine incorporation into radioiodine-refractory thyroid cancer with MAPK inhibition (ERRITI): A single-center prospective two-arm study. Clin Cancer Res. 28:4194–4202. 2022. View Article : Google Scholar : PubMed/NCBI

99 

Van Nostrand D, Veytsman I, Kulkarni K, Heimlich L and Burman KD: Redifferentiation of differentiated thyroid cancer: Clinical insights from a narrative review of literature. Thyroid. 33:674–681. 2023. View Article : Google Scholar : PubMed/NCBI

100 

Montes de Jesus FM, Espeli V, Paone G and Giovanella L: Add-on radioiodine during long-term BRAF/MEK inhibition in patients with RAI-refractory thyroid cancers: a reasonable option? Endocrine. 81:450–454. 2023. View Article : Google Scholar : PubMed/NCBI

101 

Carmona Matos DM, Jang S, Hijaz B, Chang AW, Lloyd RV, Chen H and Jaskula-Sztul R: Characterization of somatostatin receptors (SSTRs) expression and antiproliferative effect of somatostatin analogues in aggressive thyroid cancers. Surgery. 165:64–68. 2019. View Article : Google Scholar : PubMed/NCBI

102 

Loibl S, André F, Bachelot T, Barrios CH, Bergh J, Burstein HJ, Cardoso MJ, Carey LA, Dawood S, Del Mastro L, et al: Early breast cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol. 35:159–182. 2024. View Article : Google Scholar : PubMed/NCBI

103 

Shete N, Calabrese J and Tonetti DA: Revisiting estrogen for the treatment of endocrine-resistant breast cancer: Novel therapeutic approaches. Cancers (Basel). 15:36472023. View Article : Google Scholar : PubMed/NCBI

104 

Generali D, Berardi R, Caruso M, Cazzaniga M, Garrone O, Minchella I, Paris I, Pinto C and De Placido S: Aromatase inhibitors: The journey from the state of the art to clinical open questions. Front Oncol. 13:12491602023. View Article : Google Scholar : PubMed/NCBI

105 

Li B, Liang J, Lu F, Zeng G, Zhang J, Ma Y, Liu P, Wang Q, Zhou Q and Chen L: Discovery of novel inhibitor for WNT/β-catenin pathway by tankyrase 1/2 structure-based virtual screening. Molecules. 25:16802020. View Article : Google Scholar : PubMed/NCBI

106 

Li B, Orton D, Neitzel LR, Astudillo L, Shen C, Long J, Chen X, Kirkbride KC, Doundoulakis T, Guerra ML, et al: Differential abundance of CK1α provides selectivity for pharmacological CK1α activators to target WNT-dependent tumors. Sci Signal. 10:eaak99162017. View Article : Google Scholar : PubMed/NCBI

107 

Fang L, Zhu Q, Neuenschwander M, Specker E, Wulf-Goldenberg A, Weis WI, von Kries JP and Birchmeier W: A small-molecule antagonist of the β-catenin/TCF4 interaction blocks the self-renewal of cancer stem cells and suppresses tumorigenesis. Cancer Res. 76:891–901. 2016. View Article : Google Scholar : PubMed/NCBI

108 

Wiese M, Walther N, Diederichs C, Schill F, Monecke S, Salinas G, Sturm D, Pfister SM, Dressel R, Johnsen SA and Kramm CM: The β-catenin/CBP-antagonist ICG-001 inhibits pediatric glioma tumorigenicity in a Wnt-independent manner. Oncotarget. 8:27300–27313. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Chatterjee A, Paul S, Bisht B, Bhattacharya S, Sivasubramaniam S and Paul MK: Advances in targeting the WNT/β-catenin signaling pathway in cancer. Drug Discov Today. 27:82–101. 2022. View Article : Google Scholar : PubMed/NCBI

110 

Tabernero J, Van Cutsem E, Garralda E, Tai D, De Braud F, Geva R, van Bussel MTJ, Fiorella Dotti K, Elez E, de Miguel MJ, et al: A phase Ib/II study of WNT974 + encorafenib + cetuximab in patients with BRAF V600E-mutant KRAS wild-type metastatic colorectal cancer. Oncologist. 28:230–238. 2023. View Article : Google Scholar : PubMed/NCBI

111 

Burock S, Daum S, Keilholz U, Neumann K, Walther W and Stein U: Phase II trial to investigate the safety and efficacy of orally applied niclosamide in patients with metachronous or sychronous metastases of a colorectal cancer progressing after therapy: The NIKOLO trial. BMC Cancer. 18:2972018. View Article : Google Scholar : PubMed/NCBI

112 

Mraz KA, Hodan R, Rodgers-Fouche L, Arora S, Balaguer F, Guillem JG, Jeter JM, Kanth P, Li D, Liska D, et al: Current chemoprevention approaches in lynch syndrome and familial adenomatous polyposis: A global clinical practice survey. Front Oncol. 13:11418102023. View Article : Google Scholar : PubMed/NCBI

113 

Peixoto RD, Oliveira LJC, Passarini TM, Andrade AC, Diniz PH, Prolla G, Amorim LC, Gil M, Lino F, Garicochea B, et al: Vitamin D and colorectal cancer-A practical review of the literature. Cancer Treat Res Commun. 32:1006162022. View Article : Google Scholar : PubMed/NCBI

114 

Filetti S, Durante C, Hartl DM, Leboulleux S, Locati LD, Newbold K, Papotti MG and Berruti A; ESMO Guidelines Committee. Electronic address, : simpleclinicalguidelines@esmo.org: ESMO clinical practice guideline update on the use of systemic therapy in advanced thyroid cancer. Ann Oncol. 33:674–684. 2022. View Article : Google Scholar : PubMed/NCBI

115 

Gouda MA and Subbiah V: Precision oncology with selective RET inhibitor selpercatinib in RET-rearranged cancers. Ther Adv Med Oncol. 15:175883592311770152023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2024
Volume 52 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cameselle‑García S, Abdulkader‑Nallib I, Sánchez‑Ares M and Cameselle‑Teijeiro JM: Cribriform morular thyroid carcinoma: Clinicopathological and molecular basis for both a preventive and therapeutic approach for a rare tumor (Review). Oncol Rep 52: 119, 2024.
APA
Cameselle‑García, S., Abdulkader‑Nallib, I., Sánchez‑Ares, M., & Cameselle‑Teijeiro, J.M. (2024). Cribriform morular thyroid carcinoma: Clinicopathological and molecular basis for both a preventive and therapeutic approach for a rare tumor (Review). Oncology Reports, 52, 119. https://doi.org/10.3892/or.2024.8778
MLA
Cameselle‑García, S., Abdulkader‑Nallib, I., Sánchez‑Ares, M., Cameselle‑Teijeiro, J. M."Cribriform morular thyroid carcinoma: Clinicopathological and molecular basis for both a preventive and therapeutic approach for a rare tumor (Review)". Oncology Reports 52.3 (2024): 119.
Chicago
Cameselle‑García, S., Abdulkader‑Nallib, I., Sánchez‑Ares, M., Cameselle‑Teijeiro, J. M."Cribriform morular thyroid carcinoma: Clinicopathological and molecular basis for both a preventive and therapeutic approach for a rare tumor (Review)". Oncology Reports 52, no. 3 (2024): 119. https://doi.org/10.3892/or.2024.8778