Open Access

Aberrant trophoblastic differentiation in human cancer: An emerging novel therapeutic target (Review)

  • Authors:
    • Chen Chang
    • Yi-Lin Chen
    • Yi-Wen Wang
    • Hui-Wen Chen
    • Che-Wei Hsu
    • Kun-Che Lin
    • Yin-Chien Ou
    • Tsunglin Liu
    • Wan-Li Chen
    • Chien-An Chu
    • Chung-Liang Ho
    • Chung-Ta Lee
    • Nan-Haw Chow
  • View Affiliations

  • Published online on: January 15, 2024     https://doi.org/10.3892/or.2024.8701
  • Article Number: 43
  • Copyright: © Chang 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

Various types of human cancer may develop aberrant trophoblastic differentiation, including histological changes and altered expression of β‑human chorionic gonadotropin (β‑hCG). Aberrant trophoblastic differentiation in epithelial cancer is usually associated with poor differentiation, tumor metastasis, unfavorable prognosis and treatment resistance. Since β‑hCG‑targeting vaccines have failed in an early phase II trial, it is crucial to obtain a better understanding of the molecular pathogenesis of trophoblastic differentiation in human cancer. The present review summarizes the clinical and translational research on this topic with the aim of accelerating the development of an effective targeted therapy. Ectopic expression of β‑hCG promotes proliferation, migration, invasion, vasculogenesis and epithelial‑mesenchymal transition (EMT) in vitro, and enhances metastatic and tumorigenic capabilities in vivo. Signaling cascades modulated by β‑hCG include the TGF‑β receptor pathway, EMT‑related pathways, the c‑MET receptor tyrosine kinase and mitogen‑activated protein kinase/ERK pathways, and the SMAD2/4 pathway. Taken together, these findings indicated that TGF‑β receptors, c‑MET and ERK1/2 are potential therapeutic targets. Nevertheless, further investigation on the molecular basis of aberrant trophoblastic differentiation is mandatory to improve the design of precision therapy for this aggressive type of human cancer.

Introduction

Epithelial carcinoma occasionally contains elements reminiscent of trophoblastic differentiation, such as syncytiotrophoblasts and areas resembling choriocarcinoma (1). Immunohistochemical studies have revealed the expression of β-human chorionic gonadotropin (β-hCG) and other trophoblastic hormones in syncytiotrophoblast-like and cytotrophoblast-like cells (1,2). Moreover, non-giant cancer cells in humans may express hCG in primary carcinoma of the bladder, upper urinary tract, prostate, colon, lung, testis and gynecological malignancies. Thus, the expression of β-hCG suggests aberrant differentiation of cancer cells toward a gestational trophoblastic phenotype (16). Ectopic expression of β-hCG is usually associated with poor clinicopathological indicators and unfavorable clinical outcomes (3,4,710). In previous analyses using β-hCG as a biomarker, carcinoma with trophoblastic differentiation was revealed to be significantly associated with poor differentiation (2,4,1015), advanced tumor stage (2,4,9,10,13), early hematogenous spread (7,10), chemoresistance (16,17), radioresistance (3,8,10,18) and shorter disease-specific overall survival (4,8). The most well-known example of trophoblastic differentiation in human cancer is observed in urothelial carcinoma (UC) with trophoblastic differentiation (UCTD) (4).

In vitro studies have indicated that overexpression of β-hCG promotes the migration and invasion of ovarian cancer cells, and facilitates their metastasis into peritoneal xenografts (19). Moreover, upregulation of β-hCG can promote the transition of cancer cells from an epithelial phenotype with relevant biomarkers to a loosely adherent, motile phenotype with mesenchymal markers [epithelial-mesenchymal transition (EMT)], and can reduce their adhesive ability (19,20). In a mouse model of human colorectal cancer (CRC), β-hCG-overexpressing cells were shown to exhibit increased invasiveness, migratory ability and metastatic potential (20).

EMT is the dynamic transition of cells from an epithelial to a mesenchymal phenotype. This transition has been identified as the main driver of tumor progression and metastasis (21). During normal placentation, three main trophoblast populations exist: Cytotrophoblast stem cells and their differentiated derivatives, syncytiotrophoblasts and extravillous cytotrophoblasts. Syncytiotrophoblasts primarily exhibit the epithelial phenotype, whereas extravillous cytotrophoblasts undergo EMT, initially forming multilayered cell columns and then (in humans) deeply infiltrating the maternal decidual stroma and blood vessels (22). In epithelial carcinoma, EMT initiates the dissociation of cancer cells from primary tumors, and these cells subsequently migrate and disseminate to distant sites. To the best of our knowledge, the molecular mechanisms underlying trophoblastic differentiation in nongestational human cancer remain unclear. In an autopsy study, only tumor cells with trophoblastic differentiation, but not UC cells, were identified at multiple metastatic sites after treatment with gemcitabine and oxaliplatin (7), supporting the potential resistance of UCTD to standard chemotherapy. Thus, identification of the mechanisms underlying β-hCG-mediated EMT may facilitate the development of targeted therapy against tumor progression through aberrant trophoblastic differentiation. The present review summarizes the literature on the molecular pathogenesis of β-hCG in nontrophoblastic human cancer, with the aim of accelerating the development of targeted therapies for trophoblastic differentiation in human cancer.

Histopathology of trophoblastic differentiation and diagnosis

There is a spectrum of trophoblastic differentiation in human cancer, from syncytiotrophoblasts, areas resembling choriocarcinoma, pure choriocarcinoma to other trophoblastic tumors, such as epithelioid trophoblastic tumors (8). The tumor cells exhibit hyperchromatic trophoblastic-like cells with deep eosinophilic cytoplasm that usually appear in the periphery of the tumor nests or infiltrate the interstitium. Tumors may also have scattered multinucleate giant cells and well-defined syncytiotrophoblastic cells or syncytiotrophoblastic cells that wrap around mononuclear tumor cells that resemble cytotrophoblasts (4). In contrast with the inner mass of conventional carcinoma cells, which have pale to eosinophilic cytoplasm, the architecture is similar to the normal anatomical relationship in chorionic villi. Only tumors showing definite immunostaining for β-hCG are diagnosed as trophoblastic differentiation phenotype of nongestational carcinoma (4,8).

Trophoblastic differentiation in UC

As per the current classification criteria outlined by the World Health Organization, UCTD includes UC with giant cells resembling syncytiotrophoblastic giant cells and, rarely, those that are indistinguishable from choriocarcinoma (23). To the best of our knowledge, the incidence of UCTD has not been investigated thoroughly. In our recent cohort study, UCTD was detected in 47 of 859 patients (5.5%) and 65 of 635 patients (10.2%) with bladder and upper urinary tract UC, respectively (4). The incidence rate was significantly lower than that (19–36%) reported in small-scale studies (1,2,24). This inconsistency in incidence rate may be because of our strict inclusion criteria regarding trophoblastic histological features compared with the b-hCG immunostaining data used in earlier studies. Our previous study also demonstrated that β-hCG can be expressed in not only UC with syncytiotrophoblast-like and choriocarcinoma-like features, but also in conventional UC (8). Few studies have investigated the expression of other trophoblastic hormones, such as human placental lactogen (hPL), pregnancy-specific β-1-glycoprotein and placental alkaline phosphatase (24). By contrast, the expression of pituitary hormones with structures similar to that of hCG, such as luteinizing hormone (LH), follicle- stimulating hormone (FSH) and growth hormone, was not found in UCTD (2).

Clinical significance of trophoblastic differentiation in UC

UCTD in the bladder is usually detected as nonpapillary, multiple and large (>3 cm) tumors with muscle-invasion and nodal-metastasis at diagnosis (4). In addition, the risks of recurrence, progression and mortality are significantly higher than conventional UC (1,4). Regarding immunohistochemistry, the nonfocal pattern of b-hCG expression is a key predictor of poor prognosis (4). The aforementioned findings corroborate those of other studies indicating the associations of trophoblastic differentiation, either of syncytiotrophoblasts or b-hCG-positive cells, with higher grades of UC and advanced stages of disease (1,8,24). Notably, elevated levels of b-hCG in the serum may be detected in 20–76% of the total number of patients with advanced-stage disease or metastasis (23,2528). Several studies have reported the potential of b-hCG as a biomarker of radioresistance and chemoresistance (3,7,18); however, contradictory findings have also been reported (23,25,27).

Trophoblastic differentiation in nonurothelial carcinoma and its clinical significance

In nonurothelial carcinoma, trophoblastic differentiation is frequently observed in gestational trophoblastic tumors and ovarian germ cell tumors. In addition, sporadic cases of trophoblastic differentiation have been reported in oral cavity, head and neck, lung, stomach, colorectum, prostate, breast and gynecological tract carcinoma (5,913,16,20,2948).

In a study on squamous cell carcinoma of the oral cavity, β-hCG expression (range, 0.5–5%) was detected in 29 of 45 patients (64%) with oral cancer (11); β-hCG expression had a positive association with tumor differentiation. Furthermore, β-hCG expression has been observed in various histological subtypes of lung cancer, including neuroendocrine carcinoma, squamous cell carcinoma, adenocarcinoma and giant cell carcinoma (12,16,49). Trophoblastic hormone immunoreactivity was previously detected in 31% (28/90) of all lung carcinoma cases, regardless of histological differentiation (12).

Although β-hCG has procarcinogenic activities in other types of carcinoma, its role in breast cancer remains controversial. This hormone reportedly inhibits the proliferation and induces the differentiation of human breast cancer cells in vitro (50). Paradoxically, a recent study revealed enhanced proliferation and poor differentiation of β-hCG-expressing breast cancer cells, which translated into higher colonization and invasion abilities of these cells (51). In breast cancer cells, the BRCA1 mutation reportedly promotes β-hCG-mediated tumorigenesis through TGF-βRII signaling (52).

A previous study showed that β-hCG-producing cells can be found in the normal gastric mucosa, particularly the pylorus (31). β-hCG expression has been detected in 6.0–8.2% of all cases of gastric carcinoma, and the rate is even as high as 53% in some reports (32,33). In general, the presence of β-hCG-positive cells or an elevated level of β-hCG in the serum is associated with poor differentiation, adverse prognosis and advanced tumor stage (9); however, contradictory findings have been reported (34).

In ovarian epithelial cancer, elevated levels of serum β-hCG may be detected in both benign (27.6%) and malignant (67%) neoplasms (13), leading to false-positive pregnancy test results (53). Immunohistochemical expression of β-hCG tends to be higher in intermediate- to high-grade ovarian tumors compared with in low-grade tumors; however, the expression does not vary across the histological subtypes of ovarian cancer. Although a study reported higher expression rates of β-hCG in stage III (Federation of Gynecology and Obstetrics) ovarian mucinous carcinomas than in stage I carcinoma, no prominent association was discovered between β-hCG expression and overall survival (13). Regarding molecular mechanisms, β-hCG overexpression reportedly promotes the transformation and tumorigenesis of human ovarian epithelial cells (54). The association of trophoblastic differentiation with high-grade cancer and disease progression has also been observed in endometrial cancer (5,48).

In summary, β-hCG expression is a well-documented phenomenon in nongestational carcinoma of different organs, and may even be observed in some normal tissues and benign tumors. In most cases, the trophoblastic phenotype is associated with poor differentiation, advanced tumor stage and poor prognosis.

Prognostic impact of trophoblastic differentiation in human cancer

In terms of prognostic implications, patients with carcinoma showing aberrant trophoblastic differentiation have been reported to have unfavorable clinical outcomes (35,710,16). Specifically, the phenotype has been associated with early hematogenous spread (7,10,15), higher risk of chemoresistance (16,17) or radioresistance (3,8,10,18), and shorter disease-specific overall survival. Using β-hCG as a biomarker, our recent study revealed a higher risk of recurrence (P=0.005), progression (P<0.0001) and patient death (P<0.0001) for UCTD than for traditional, high-grade UC of the bladder (4). Notably, patients with UCTD and with circumferential, infiltrative or diffuse patterns of β-hCG expression have been reported to have poorer disease-specific overall survival than those with focal β-hCG expression.

In addition to expression in primary tumors, elevated β-hCG serum levels have been observed in sporadic cases of carcinoma with trophoblastic differentiation in UC (1,7,14), squamous cell anal cancer (30), gastric cancer (9), non-small cell carcinoma of the lung (36), pulmonary pleomorphic carcinoma (46), endometrial adenocarcinoma (5,48), lymphoepithelioma-like carcinoma and squamous cell carcinoma of the cervix (55). However, the actual incidence of abnormal laboratory results for this entity has not been thoroughly examined. In our experience, most UCTDs showing a focal pattern of β-hCG expression have normal serum levels (unpublished data). Nevertheless, the levels of serum β-hCG appear to change with treatment (57,30,48) and are associated with clinical outcome (5,9,16). Specifically, a serum β-HCG level ≥4 IU/l prior to chemotherapy has been reported to be a significant prognostic factor for patients with advanced gastric cancer (hazard ratio 1.7; 95% confidence interval 2.8–1.1) (9). Serum β-hCG elevation (≥5 mIU/ml) in patients with non-small cell lung cancer showing trophoblastic differentiation is also significantly associated with chemoresistance (16). Taken together, these findings indicated that serum β-hCG measurement may have potential as a marker of clinical response or prognosis, and thus should be applied as a potential biomarker during follow-up after surgery.

hCG: Gene, protein and structure

The α-subunit of hCG is encoded by a single gene (CGA) on chromosome 6q21.1–23 (56), whereas the β-subunit is encoded by six nonallelic genes (CGB1, CGB2, CGB3, CGB5, CGB7 and CGB8) on chromosome 19q13.3 (57). CGB4, which is adjacent to the aforementioned CGB cluster, encodes the β-subunit of LH. CGB6 is an allelic variant of CGB7, whereas CGB9 is an allelic variant of CGB3. To the best of our knowledge, the functions of CGB1 and CGB2 remain unknown; these genes may be pseudogenes (57). The expression of CGB genes is upregulated to some extent in the first trimester of pregnancy, which suggests a role in implantation (58). A protein encoded by type I genes (CGB6 and CGB7) contains an alanine residue at position 117, whereas β-hCG, which is encoded by type II genes (CGB3, CGB9, CGB5 and CGB8), contains an aspartic acid residue at this position. The effect of this heterogeneity on the function and immunoreactivity of β-hCG remains unknown. Type I genes are expressed primarily in benign nontrophoblastic tissues, whereas type II genes are expressed in trophoblastic and malignant tissues (59).

The hormone hCG comprises 237 amino acids and belongs to the glycoprotein hormone family, which includes LH, thyroid-stimulating hormone (TSH) and FSH. The protein members of the aforementioned family are heterodimers comprising α- and β-subunits. The α-subunit contains 92 amino acids and is common across all family members. By contrast, the β-subunit of hCG exhibits varying degrees of homology with other family members (LH, 80–85%; FSH, 36%; TSH, 46%) (6062). The homology between hCG and LH indicates their common biological function; both bind to the same receptor, the LH/hCG receptor (63). By contrast, FSH and TSH bind to structurally similar but distinct receptors. The β-subunit of hCG contains 145 amino acids, whereas that of LH contains 121 amino acids; this difference originates from a 24-amino-acid-long extension in hCG, known as the C-terminal peptide (61,64).

The α-subunit of hCG comprises two N-linked oligosaccharides (linked to the N atom of asparagine), whereas its β-subunit comprises two N-linked oligosaccharides and four O-linked oligosaccharides (linked to the O atom of serine) (65). The O-linked and N-linked oligosaccharides contain saccharides ranging from trisaccharides to pentasaccharides, and exhibit monoantennary to triantennary structures (66). Posttranslationally modified hCG variants are complex and have three dimeric isoforms: Regular hCG, hyperglycosylated hCG (hCG-H) and sulfated hCG (hCG-S) (62,67). hCG-H is a glycoprotein with excessive branching and complex hCG oligosaccharide side chains. Although hCG and hCG-H share an amino acid sequence, they are distinct glycoproteins with completely different oligosaccharide structures (66). The levels of hCG-H are high in early pregnancy (68) and show a higher trend in malignant diseases than in normal pregnancy. Various aberrant glycosylations have been detected in tumor-derived hCG (69), which has led to the generation of a diverse molecular weight spectra. In one study, the average molecular weight of hCG was determined to be 37,500 Da through matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (70); the molecular weights of α-hCG and β-hCG were 14,000 and 23,500 Da, respectively.

hCG-S originates from the pituitary gland and appears to mimic the activities of LH during the menstrual cycle; LH stimulates ovarian granulosa and theca cells to produce relevant hormones, and facilitates follicular growth, luteinization and ovulation (62,67,71). Both hCG and hCG-S act through the LH/hCG receptor, whereas hCG-H acts through the TGF-β receptor as an autocrine and paracrine factor (62).

Trophoblasts represent a key source of hCG. Syncytiotrophoblasts secrete hCG, whereas cytotrophoblasts secrete hCG-H. Cole et al (72) reported the following five common forms of hCG: hCG, hCG-H, hCG-S, free β-hCG and free β-hCG-H (72). They classified these proteins into group 1 (proteins produced by placental syncytiotrophoblasts and pituitary gonadotropes) and group 2 (proteins produced by placental cytotrophoblasts and human cancer cells). Group 1 proteins are hormones that act through the hCG/LH receptor, and are central to the menstrual cycle and pregnancy, whereas group 2 proteins perform autocrine functions by antagonizing the TGF-β receptor and are critical in advanced malignancies (67,72).

Structurally, hCG can be classified as intact hCG, α-hCG, β-hCG, partially degraded or nicked hCG and β-hCG, and a β-core fragment (73); the International Federation of Clinical Chemistry has approved this classification.

Regulation of hCG expression

The expression and production of hCG may be regulated by various hormones [corticosteroids, progesterone and gonadotropin-releasing hormone (GnRH)], growth factors (epidermal growth factor, placental growth hormone, leukemia inhibitory factor and vascular endothelial growth factor), cytokines (interleukin-6 and tumor necrosis factor-α), ligands of peroxisome proliferator-activated receptors (PPARs), and homeobox genes (62,7477).

Levels of α-hCG and β-hCG expression vary in normal placenta, with the α/β ratio ranging from 1.7 in the first trimester, to 12 once the pregnancy has reached full term (78). Therefore, the regulatory mechanisms of α-hCG and β-hCG expression may be different. Knowledge regarding the pathways involved in the upregulation of hCG genes remains limited. Cyclic adenosine monophosphate (cAMP) appears to regulate the transcription of the α- and β-subunits of hCG in the placenta and choriocarcinoma (79,80) by regulating the 5′ cAMP response element (CRE) enhancers of the promoters of both genes through different mechanisms (81,82).

α-hCG

The promoter of the α-subunit gene contains the following five regulatory elements: A trophoblast-specific element (TSE), an α-activating element (αACT), a tandem or duplicated CRE, a junctional regulatory element (JRE) and a CCAAT box. These elements have been categorized into the following three domains: The upstream regulatory domain (URE; TSE and αACT), tandem CRE and the downstream regulatory domain (JRE and CCAAT box) (8388). Tissue-specific expression of α-hCG has been demonstrated in trophoblasts on the basis of limited or specific tissue distribution of the binding proteins and regulation of the aforementioned elements (88).

The exact locations of αACT and TSE overlap slightly; the sequence of αACT between −161 and −142 overlaps that of TSE between −182 and −159 (89). Furthermore, two regulatory sequences can be noted within this region: A downstream domain located between −172 and −151 and an upstream domain located between −177 and −156 (86). This structure indicates that these regions are activated by at least two types of binding proteins that may be specifically expressed by either pituitary or placental cells (90).

The tandem CRE is responsible for the binding of CRE-binding proteins (CREBs), which belong to the basic region leucine zipper family of proteins. In villous trophoblasts, activating transcription factor (ATF)-1 is more extensively involved in the binding of CRE and, to some extent, CREB-1, than the other members of the ATF/CREB family, such as ATF-2, ATF-3, ATF-4 and CREB-2 (81). In humans, CREB binding may be dependent on URE binding (89). URE-1/αACT binds to members of the ubiquitous GATA family of DNA-binding proteins (91), whereas URE-2/TSE binds to TSE-binding proteins (TSEBs) (92).

β-hCG

The expression of CGB among patients is heterogeneous, and the magnitude of expression has varied across studies, possibly because they focused on various pregnancy trimesters (93,94). CGB expression also varies across tumors and normal tissues. For example, the expression levels of type II genes (CGB3, CGB9, CGB5 and CGB8) are higher in bladder tumors than in the normal urothelium (95).

Similar to the α-hCG promoter, the β-hCG promoter comprises a tandem CRE (two repeated CREs), where c-Jun suppresses β-hCG expression (96). At least two additional TSE elements have been reported to cluster in the regulatory region of CGB5. The genes encoding the α- and β-subunits of hCG may be coordinately regulated by TSEBs (92). Other transcription factors involved in CGB expression include ETS protooncogene 2 (ETS2), activating protein 2 (AP2), promoter selective transcription factor (SP)1, SP3, octamer-binding transcription factor (OCT)3/OCT4, PPARγ, P53 and metastasis-associated protein 3 (MTA3) (97103).

In human choriocarcinoma and murine cells, ETS2 enhances the transcription of CGB5 through activation of the rat sarcoma virus/mitogen-activated protein kinase pathway, and the primary effects of cAMP on the β-hCG promoter are mediated through the proximal ETS2 enhancer (97). AP2, SP1 and SP3 are the key regulators of basal CGB transcription in placental trophoblast cells (103,104). AP2 and SP1 play distinct roles in the regulation of basal activity and cAMP-responsiveness of the β-hCG promoter (105), whereas SP3 suppresses its basal transcription by inhibiting SP1 (105). The expression levels of CGB3-CGB9 have been reported to be considerably higher in ovarian cancer cells than in healthy ovarian cells (103). CGB1 and CGB2 transcripts have been detected in 20% of all ovarian cancer tissue sample, but not in control samples. This may have resulted from demethylation of CGB promoter regions, an increased level of transcription factor AP2 (TFAP2)-α, and a decreased level of SP3 in ovarian tumors (103). OCT3 and OCT4 are essential for maintaining embryonic cells in an undifferentiated state; these transcription factors may silence hCG expression in choriocarcinoma cells (99).

The treatment of trophoblasts with PPARγ or retinoid X receptor (RXR)α ligands increases the levels of CGB transcript, hCG and β-hCG (106). PPARγ/RXRα heterodimers directly bind to the regulatory region of CGB5 (106). Activation of PPARγ enhances the transcript levels of α-hCG and β-hCG in villous trophoblasts, but reduces the level of hCG in invasive extravillous trophoblasts (107). Shalom-Barak et al (108) and Peng et al (109) suggested that PPARγ is distinctly expressed in various trophoblast subsets and during trophoblast stem cell differentiation. p53 selectively induces the expression of CGB7; a p53-responsive element has been identified in the promoter of CGB7 (101). MTA3, a chromatin-remodeling protein, acts directly on the CGB5 promoter and suppresses CGB5 expression in trophoblasts (102). The deregulation of MTA3 has previously been associated with pre-eclampsia (102).

In addition to transcription factors, various epigenetic mechanisms (methylation) are involved in the regulation of CGB expression in the placenta and cancer cells (110,111). Allelic polymorphism for methylation sensitivity in the promoter of CGB5 have been shown to be associated with a high risk of miscarriage (112). The deregulation of epigenetic mechanisms with altered CGB expression has also been associated with an increased risk of early pregnancy loss (104).

Biological effects of β-hCG upregulation in human cancer

Proliferation

To explore the biological effects of trophoblastic differentiation in human cancer, in vitro experiments have been performed using several human cancer models. Ectopic expression of β-hCG appears to enhance the in vitro proliferation of ovarian (54) and stomach (34) cancer cells, but not that of CRC cells (20,35). Similar effects were noted on the in vivo proliferation of primary ovarian cancer cells (54), but not that of CRC (20,35) or bladder (113) cancer cells. Table I summarizes the various biological effects of β-hCG upregulation in human cancer.

Table I.

Biological effects of β-hCG overexpression in human cancer.

Table I.

Biological effects of β-hCG overexpression in human cancer.

First author, yearCancerExperi-mental modelMaterialsActivated pathwayPoor prognosisHigh expressionInvasionMigrationProliferationMetastasisEMTMorphology changeTumorigenesisAntiapoptosis ChemoresistanceAngiogenesisRecurrence marker(Refs.)
Sengodan, 2017Breast cancerIn vitroMCF7 cells, MDAMB-231 cells, SKBR3 cells, T47D cellsTGFβRII signalingN/AVVVN/AN/AVN/AVN/AN/AN/AN/A(52)
Sengodan, 2017 In vivoBRCA1 conditional knockout mouse modelMutated/defective BRCA1 activates β-hCG expressionN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(52)
Kawamata, 2018Colorectal carcinomaIn vitroCaco-2 cells, LoVo cells, HCA7 cells, WiDr cells, T84 cellsEMT via TGF-β signaling pathwayN/AN/AVVXN/AVVN/AN/AN/AN/AN/A(20)
Li, 2018 HCT-116 cells, HT-29 cells, SW480 cellsN/AN/AN/AVVXN/AN/AN/AN/AN/AN/AN/AN/A(35)
Lundin, 2001 In vivoPrimary tumorN/AVVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(40)
Kawamata, 2018 Primary tumorEMT via TGFN/Aβ signaling pathwayVVVN/AN/AVN/AN/AN/AN/AN/AN/AN/A(20)
Kawamata, 2018 LoVo-GFP cells, LoVo-hCGβ cellsEMT via TGF-β signaling pathwayN/AN/AN/AN/AN/AN/AN/AN/AVN/AN/AVN/A(20)
Li, 2018 Primary tumorN/AVN/AVVXVN/AN/AN/AN/AN/AN/AN/A(35)
Konishi, 2018 Primary tumorN/AVN/AN/AN/AN/AN/AVN/AN/AN/AN/AN/AN/A(38)
Białas, 2020Esophageal carcinomaIn vivoPrimary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(118)
Li, 2013GlioblastomaIn vitroU87MG cellsERK1/2N/A pathwayN/AVVN/AN/AN/AN/AN/AN/AN/AN/AV(114)
Białas, 2020Head and neck cancerIn vivoPrimary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(118)
Li, 2013Lung cancerIn vivoPrimary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(114)
Noda, 1990Non-small cell lung cancerIn vivoPrimary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(39)
Arano, 1994 Primary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(41)
Okutur, 2010 Primary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(42)
Seder, 2017 Preoperative serumN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AV(43)
Wong, 2015 Primary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(44)
Khobta, 2012 Primary tumor, preoperative plasmaN/AN/AVVN/AN/AVN/AN/AN/AN/AN/AN/AN/A(36)
Khattri, 2011 Primary tumor, preoperative serumN/AN/AVVN/AN/AVN/AN/AN/AN/AN/AN/AN/A(37)
Szturmowicz, 1999 Primary tumor, preoperative serumN/AVVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(16)
Vicier, 2013 Preoperative serumN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(45)
Dinis de Sousa, 2021 Primary tumor, preoperative serumN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(46)
Liu, 2017Ovarian cancerIn vitroES-2 cells, SKOV3 cellsN/AN/AN/AVVN/AN/AVVN/AN/AN/AN/AN/A(19)
Sengodan, 2017 OVCAR8 cellsBRCA1 regulation on β-hCGN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(52)
Guo, 2011 T29 cells, T29-hCG cells, T80 cells, T80-hCG cellsN/AN/AN/AN/AN/AVN/AN/AN/AN/AVN/AN/AN/A(54)
Śliwa, 2019 In vivoPrimary tumorDemethylation regulating gene expressionN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(103)
Liu, 2017 Primary tumorN/AN/AVN/AN/AN/AN/AVVN/AN/AN/AN/AN/A(19)
Liu, 2017 ES-2 cells, SKOV3 cellsN/AN/AN/AN/AN/AN/AVN/AVVN/AN/AN/AN/A(19)
Guo, 2011 T29 cells, T29-hCG, T80 cells and T80-hCG cells were injected into an athymic nude mouse modelN/AN/AN/AN/AN/AVN/AN/AN/AVVN/AN/AN/A(54)
Guo, 2011 Primary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(54)
Sengodan, 2017Prostate cancerIn vitroDU145 cellsBRCA1 regulation on β-hCGN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(52)
Wu. 2006 DU145 cells, PC3 cellsN/AN/AN/AVVN/AN/AN/AVN/AN/AN/AN/AN/A(115)
Li, 2013 DU145 cellsERK1/2 pathwayN/AN/AVVN/AN/AN/AN/AN/AN/AN/AN/AN/A(116)
Szturmowicz, 1995Small cell lung cancerIn vivoPreoperative serumN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AVN/AN/A(138)
Butler, 2000Urothelial carcinomaIn vitroT24 cells, SCaBER cells, J82 cells, 5637 cells, RT112 cellsN/AN/AN/AN/AN/AN/AN/AN/AN/AVVN/AN/AN/A(113)
Białas, 2020 In vivoPrimary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(118)
Cheng, 2021 Primary tumorN/AVVVN/AN/AVN/AN/AN/AN/AN/AN/AV(4)
Hoshi, 2018 Postoperative serumN/AVVN/AN/AN/AN/AN/AN/AN/AN/AVN/AN/A(10)
Rajabi, 2013 Preoperative serum, urineN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(53)
Armah, 2007 Primary tumorN/AVVN/AN/AN/AVN/AN/AN/AN/AVN/AN/A(120)
Shimada, 2006 Primary tumor, preoperative serumN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(14)
Regalado, 2004 Primary tumorN/AN/AVVN/AN/AVN/AN/AN/AN/AVN/AN/A(117)
Ramakumar, 1998 Primary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(47)
Oyasu, 1994 Primary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(139)
Białas, 2020Uterine corpus endometrial carcinomaIn vivoPrimary tumorN/AN/AVN/AN/AN/AN/AN/AN/AN/AN/AN/AN/AN/A(118)

[i] N/A, not available; V, reported in the literature; X, no significant differences; β-hCG, β-human chorionic gonadotropin; EMT, epithelial-mesenchymal transition.

Apoptosis

Overexpression of β-hCG inhibits the in vitro apoptosis of ovarian and bladder cancer cells (113), as well as that of cells obtained from the xenografts of nude mice (54). In vitro β-hCG treatment of bladder UC exerts antiapoptotic effects (113).

Migration

β-hCG reportedly stimulates the in vitro migration of BRCA1-mutant breast cancer (52), CRC (20,35), glioblastoma (114), ovarian cancer (19) and prostate cancer cells (115,116).

Invasion

β-hCG promotes in vitro cell invasion in BRCA1-mutant breast cancer (52), CRC (20,35), ovarian cancer (19), prostate cancer (115,116) and glioblastoma (114). β-hCG expression has also been strongly associated with tumor invasion in primary CRC (20,35), non-small cell lung cancer (36,37) and bladder cancer (4,117).

EMT

Ectopic expression of β-hCG in cancer cells appears to induce in vitro EMT in BRCA1-mutant breast cancer (52), CRC (20) and ovarian cancer (19). β-hCG has been reported to be essential for in vivo modulation of EMT in mouse tumor xenograft models of CRC (20), CRC metastasis (20,38), large cell carcinoma (39) and lung squamous cell carcinoma (118).

Angiogenesis

Xenografts of CRC with aberrant β-hCG expression have been demonstrated to harbor a higher density of microvessels than control tumors in mice (20). The fifth subunit of β-hCG, CGB5, may also promote tumor growth and vasculogenic mimicry by activating the LH receptor signal transduction pathway (119).

Metastasis

Overexpression of β-hCG is strongly associated with increased metastatic potential in CRC (20,35), non-small cell lung cancer (36,37), ovarian cancer (19) and bladder cancer (4,117,120).

Tumorigenesis

Overexpression of β-hCG has been reported to enhance tumorigenesis in mouse tumor xenograft models of BRCA1-mutant breast cancer (52), CRC (20), ovarian cancer (19,54) and bladder cancer (113).

Taken together, these findings indicated that the molecular mechanisms underlying the effects of β-hCG on increased tumorigenesis include promotion of cell proliferation, differentiation, vasculogenesis, EMT and metastasis.

The hCG-mediated signaling pathways

TGF-β signaling pathway

In an early study conducted using CRC as a model, LoVo and HCA-7 cells were transfected with β-hCG (20). Ectopic expression of β-hCG was shown to upregulate zinc finger protein SNAI1 (SNAIL), zinc finger protein SNAI2, twist-related protein 1 (TWIST) and phosphorylated-SMAD2, but to downregulate epithelial cadherin in hCGβ-transfected HCA-7 cells compared with in control cells. The phosphorylation of SMAD2, which was activated through stimulation of TGF-β receptors during EMT, was also promoted. Together, these results indicated that β-hCG may induce EMT through the TGF-β signaling pathway. Fig. 1 depicts the β-hCG-mediated signaling pathways discussed in the present review.

EMT-related signaling pathway

A recent cell line and mouse model experiment revealed that β-hCG promotes the proliferation of gastric cancer cells in vitro through activation of the c-Met (121).

ERK1/2 signaling pathway

In human glioblastoma, β-hCG has been shown to upregulate the expression of phosphorylated-ERK1/2 in U87MG cells in a dose- and time-dependent manner (114). In addition, the ERK/matrix metalloproteinase 2 (MMP2) signaling pathway is involved in the β-hCG-mediated metastasis of epithelial ovarian cancer cells (122). This observation was supported by an in vitro experiment performed using the DU145 prostate cancer cell line (116).

Expression of non-hCG biomarkers associated with trophoblastic differentiation

In addition to hCG, the other trophoblastic markers that are used for diagnostic purposes include hPL, melanoma cell adhesion molecule (MCAM; also called CD146), p63, mucin (MUC)-4, human leukocyte antigen (HLA)-G, cytokeratin 18, inhibin A, GATA-binding protein 3 (GATA3), HSD3B1 and spalt-like transcription factor 4 (SALL4) (123128).

The expression of these markers varies across trophoblast types, namely, cytotrophoblasts, syncytiotrophoblasts and intermediate trophoblasts (ITs); the latter two types may be derived from cytotrophoblasts. ITs may be further subtyped into villous ITs (villi are anchored to the basal plate through trophoblastic columns), implantation site ITs and chorionic-type (chorion laeve) ITs (129). Each subtype of IT has a different immunohistochemical profile and malignant counterparts. For example, hCG, HSD3B1, hPL, inhibin, MCAM, SALL4, HLA-G, MUC4 and p63 are expressed in the malignant villous ITs; hPL, MUC-4, HSD3B1, HLA-G and CD146 are expressed in the malignant implantation site ITs, with limited expression of hCG and inhibin; while tumor cells of chorionic-type IT origin diffusely express HSD3B1, HLA-G, p63, cyclin E, inhibin A and GATA3, with occasional expression of CD146 and hPL (130).

In general, SALL4 is specifically expressed in mononuclear cytotrophoblasts, in contrast to hCG, which is expressed mainly in syncytiotrophoblasts. HSD3B1 is regarded as a pantrophoblastic marker. HLA-G is useful for all three subtypes of IT. CD146 and hPL are highly specific for implantation site ITs, whereas p63 and PLAP are highly specific for chorionic ITs (129,131).

Notably, some of the aforementioned markers have been detected in nontrophoblastic tumors. For example, one small study (n=16) revealed that hCG was expressed in 93% of patients with UCTD (8). In this study, not only the trophoblastic component but also the UC component expressed hCG, at a rate as high as 85% (8); HSD3B1 was also expressed in the trophoblastic component of all but one case (8). By contrast, SALL4 expression was variable, with a 50% staining rate in trophoblasts and a 43% staining rate in the UC component of hCG-positive cases (8).

Although supplementary potential markers for trophoblastic differentiation have been identified, the application of some of these markers remains debatable, partly because of the difficulty associated with determining the trophoblastic lineage of candidate cells (132). The proposed characteristics of primary first-trimester trophoblasts include the expression of a specific set of protein markers (cytokeratin 7, GATA3 and TFAP2-γ), the HLA class I expression profile, the methylation of ELF5 and the expression of microRNAs (miRNAs) from the chromosome 19 miRNA cluster (132).

Options for targeted therapy

As a target for cancer vaccines, hCG has been explored for decades (133). An early investigation revealed the potential of a monoclonal antibody against β-hCG (6H1) in the inhibition of tumor growth in vitro and in vivo (134). The CDX-1307 vaccine (also called B11-hCG-β) was developed to target β-hCG-expressing bladder cancer cells (135). This vaccine comprises a B11 monoclonal antibody against the mannose receptor of antigen-presenting cells fused to β-hCG. In a phase I clinical trial involving patients with cancer, CDX-1307 was found to be well tolerated. It induced substantial β-hCG-specific cellular and humoral immune responses when co-administered with GM-CSF and the Toll-like receptor agonists Resiquimod (R848) and poly-ICLC (135). However, enrollment for the early phase II trial was slow and the study was terminated prematurely (https://clinicaltrials.gov/ct2/show/record/NCT01094496) (136).

Combination therapies can be used as alternatives. Through its direct and collaborative effects with Toll-like receptor ligands and accessory cell-secreted cytokines, hCG was shown to mediate chemoresistance in gonadotropin-sensitive tumors in a mouse study (137). The coadministration of curcumin and an anti-hCG vaccine (β-hCG conjugated to tetanus toxoid) to mice carrying syngeneic tumors resulted in considerably improved animal survival (137).

On the basis of the aforementioned molecular alterations, inhibitors of type I and II TGF-β receptors appear to successfully reverse the biological effects and overexpression of SNAIL and TWIST induced by β-hCG in human CRC (20). Moreover, an ERK1/2 inhibitor could reduce the expression of MMP2, invasion of human glioblastoma cells (114) and motility of prostate cancer cells in vitro (116). Taken together, these results indicated that both ERK1/2 and MMP2 are potential targets in precision therapy for β-hCG-related cancer progression.

Future perspectives

The understanding of the molecular basis of β-hCG-mediated tumorigenesis in human cancer remains incomplete. Information regarding the mechanisms underlying trophoblastic differentiation in human cancer may facilitate the development of personalized therapy for patients with cancer. With the advancement of research, targeting the constituent(s) of β-hCG-mediated EMT and angiogenesis may improve current therapeutic regimens for patients with epithelial cancer with trophoblastic differentiation.

Acknowledgements

Not applicable.

Funding

This manuscript was supported by research grants [grant nos. MOST 108-2320-B-006-050-MY3, MOST 110-2314-B-006-083 and MOST 111-2320-B-006-025] from the Ministry of Science and Technology, Taiwan, and grants [grant nos. NCKUH-11204052 and NCKUH-11208006] from the National Cheng Kung University Hospital, Taiwan.

Availability of data and materials

Not applicable.

Authors' contributions

CC, YLC, YWW, TL, HWC, CWH. KCL, YCO, CAC, CLH, CTL and NHC performed literature research. CC, YLC, CTL and NHC wrote the original draft. CC, CTL, WLC and NHC revised the article. YLC generated the original figure and table. CTL and NHC performed visualization. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All authors declare that they have no competing interests.

References

1 

Campo E, Algaba F, Palacin A, Germa R, Sole-Balcells FJ and Cardesa A: Placental proteins in high-grade urothelial neoplasms. An immunohistochemical study of human chorionic gonadotropin, human placental lactogen, and pregnancy-specific beta-1-glycoprotein. Cancer. 63:2497–2504. 1989. View Article : Google Scholar : PubMed/NCBI

2 

Dirnhofer S, Koessler P, Ensinger C, Feichtinger H, Madersbacher S and Berger P: Production of trophoblastic hormones by transitional cell carcinoma of the bladder: Association to tumor stage and grade. Hum Pathol. 29:377–382. 1998. View Article : Google Scholar : PubMed/NCBI

3 

Moutzouris G, Yannopoulos D, Barbatis C, Zaharof A and Theodorou C: Is beta-human chorionic gonadotrophin production by transitional cell carcinoma of the bladder a marker of aggressive disease and resistance to radiotherapy? Br J Urol. 72:907–909. 1993. View Article : Google Scholar : PubMed/NCBI

4 

Cheng HL, Chou LP, Tsai HW, Lee CT, Wang YW, Chung-Liang H, Ou JH, Tsai YS and Chow NH: Urothelial carcinoma with trophoblastic differentiation: Reappraisal of the clinical implication and immunohistochemically features. Urol Oncol. 39:732.e17–732.e23. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Bradley CS, Benjamin I, Wheeler JE and Rubin SC: Endometrial adenocarcinoma with trophoblastic differentiation. Gynecol Oncol. 69:74–77. 1998. View Article : Google Scholar : PubMed/NCBI

6 

Coleman RL, Lindberg G, Muller CY, Miller DS and Hameed A: Ectopic production and localization of beta-human chorionic gonadotropin in lymphoepithelioma-like carcinoma of the cervix: A case report. Int J Gynecol Pathol. 19:179–182. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Uchida M, Kawai K, Kurobe M, Ikeda A, Kandori S, Endo T, Miyagawa T, Kojima T, Tsutsumi M and Nishiyama H: Metastases of urothelial carcinoma with trophoblastic differentiation that responded to combination chemotherapy with gemcitabine and oxaliplatin: A case report. Hinyokika Kiyo. 64:55–61. 2018.(In Japanese). PubMed/NCBI

8 

Przybycin CG, McKenney JK, Nguyen JK, Shah RB, Umar SA, Harik L, Shih IM and Cox RM: Urothelial carcinomas with trophoblastic differentiation, including choriocarcinoma: Clinicopathologic series of 16 cases. Am J Surg Pathol. 44:1322–1330. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Webb A, Scott-Mackie P, Cunningham D, Norman A, Andreyev J, O'Brien M and Bensted J: The prognostic value of serum and immunohistochemical tumour markers in advanced gastric cancer. Eur J Cancer. 32A:63–68. 1996. View Article : Google Scholar : PubMed/NCBI

10 

Hoshi S, Numahata K, Morozumi K, Katumata Y, Kuromoto A, Takai Y, Hoshi K, Bilim V and Sasagawa I: Bladder cancer metastasis producing beta-human chorionic gonadotropin, squamous cell carcinoma antigen, granulocyte-colony stimulating factor, and parathyroid hormone-related protein. IJU Case Rep. 2:47–50. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Bhalang K, Kafrawy AH and Miles DA: Immunohistochemical study of the expression of human chorionic gonadotropin-beta in oral squamous cell carcinoma. Cancer. 85:757–762. 1999. View Article : Google Scholar : PubMed/NCBI

12 

Dirnhofer S, Freund M, Rogatsch H, Krabichler S and Berger P: Selective expression of trophoblastic hormones by lung carcinoma: Neurendocrine tumors exclusively produce human chorionic gonadotropin alpha-subunit (hCGalpha). Hum Pathol. 31:966–972. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Lenhard M, Tsvilina A, Schumacher L, Kupka M, Ditsch N, Mayr D, Friese K and Jeschke U: Human chorionic gonadotropin and its relation to grade, stage and patient survival in ovarian cancer. BMC Cancer. 12:22012. View Article : Google Scholar : PubMed/NCBI

14 

Shimada K, Nakamura M, Ishida E and Konishi N: Urothelial carcinoma with plasmacytoid variants producing both human chorionic gonadotropin and carbohydrate antigen 19-9. Urology. 68:891.e7–e10. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Wang Z, Wang J, Zhang W, Wang D, Wang X and Liang X: Case report: Urothelial carcinoma of the renal pelvis with trophoblastic differentiation: A rare case report and review of literature. Pathol Oncol Res. 29:16108562023. View Article : Google Scholar : PubMed/NCBI

16 

Szturmowicz M, Slodkowska J, Zych J, Rudzinski P, Sakowicz A and Rowinska-Zakrzewska E: Frequency and clinical significance of beta-subunit human chorionic gonadotropin expression in non-small cell lung cancer patients. Tumour Biol. 20:99–104. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Cook AM, Huddart RA, Jay G, Norman A, Dearnaley DP and Horwich A: The utility of tumour markers in assessing the response to chemotherapy in advanced bladder cancer. Br J Cancer. 82:1952–1957. 2000.PubMed/NCBI

18 

Martin JE, Jenkins BJ, Zuk RJ, Oliver RT and Baithun SI: Human chorionic gonadotrophin expression and histological findings as predictors of response to radiotherapy in carcinoma of the bladder. Virchows Arch A Pathol Anat Histopathol. 414:273–277. 1989. View Article : Google Scholar : PubMed/NCBI

19 

Liu N, Peng SM, Zhan GX, Yu J, Wu WM, Gao H, Li XF and Guo XQ: Human chorionic gonadotropin β regulates epithelial-mesenchymal transition and metastasis in human ovarian cancer. Oncol Rep. 38:1464–1472. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Kawamata F, Nishihara H, Homma S, Kato Y, Tsuda M, Konishi Y, Wang L, Kohsaka S, Liu C, Yoshida T, et al: Chorionic gonadotropin-β modulates epithelial-mesenchymal transition in colorectal carcinoma metastasis. Am J Pathol. 188:204–215. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Nieto MA, Huang RYJ, Jackson RA and Thiery JP: EMT: 2016. Cell. 166:21–45. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Vićovac L and Aplin JD: Epithelial-mesenchymal transition during trophoblast differentiation. Acta Anat (Basel). 156:202–216. 1996. View Article : Google Scholar : PubMed/NCBI

23 

Comperat EM, Netto GJ and Tsuzuki T: Invasive urothelial carcinoma. In: WHO Classification of of Tumours: Urinary and Male Genital Tumours. Vol 8. 5th edition. IARC; Lyon: pp. 150–165. 2022

24 

Moldavsy M, Sazbon A, Kuchersky N and Turani H: Screening for transitional cell carcinoma of the bladder with trophoblastic differentiation in Upper Galilee. Harefuah. 134:260–263. 3363351998.(In Hebrew). PubMed/NCBI

25 

Dexeus F, Logothetis C, Hossan E and Samuels ML: Carcinoembryonic antigen and beta-human chorionic gonadotropin as serum markers for advanced urothelial malignancies. J Urol. 136:403–407. 1986. View Article : Google Scholar : PubMed/NCBI

26 

Iles RK, Jenkins BJ, Oliver RT, Blandy JP and Chard T: Beta human chorionic gonadotrophin in serum and urine. A marker for metastatic urothelial cancer. Br J Urol. 64:241–244. 1989. View Article : Google Scholar : PubMed/NCBI

27 

Douglas J, Sharp A, Chau C, Head J, Drake T, Wheater M, Geldart T, Mead G and Crabb SJ: Serum total hCGβ level is an independent prognostic factor in transitional cell carcinoma of the urothelial tract. Br J Cancer. 110:1759–1766. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Iles RK: Ectopic hCGbeta expression by epithelial cancer: Malignant behaviour, metastasis and inhibition of tumor cell apoptosis. Mol Cell Endocrinol. 260–262. 264–270. 2007.PubMed/NCBI

29 

Gehring C, Siepmann T, Heidegger H and Jeschke U: The controversial role of human chorionic gonadotropin in the development of breast cancer and other types of tumors. Breast. 26:135–140. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Pokharel K, Gilbar PJ, Mansfield SK, Nair LM and So A: Elevated beta human chorionic gonadotropin in a non-pregnant female diagnosed with anal squamous cell carcinoma. J Oncol Pharm Pract. 26:1266–1269. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Manabe T, Adachi M and Hirao K: Human chorionic gonadotropin in normal, inflammatory, and carcinomatous gastric tissue. Gastroenterology. 89:1319–1325. 1985. View Article : Google Scholar : PubMed/NCBI

32 

Ito H and Tahara E: Human chorionic gonadotropin in human gastric carcinoma. A retrospective immunohistochemical study. Acta Pathol Jpn. 33:287–296. 1983.PubMed/NCBI

33 

Yakeishi Y, Mori M and Enjoji M: Distribution of beta-human chorionic gonadotropin-positive cells in noncancerous gastric mucosa and in malignant gastric tumors. Cancer. 66:695–701. 1990. View Article : Google Scholar : PubMed/NCBI

34 

Murhekar KM, Anuratha JN, Majhi U and Rajkumar T: Expression of human chorionic gonadotropin beta in gastric carcinoma: A retrospective immunohistochemical study. Indian J Med Paediatr Oncol. 30:99–102. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Li J, Yin M, Song W, Cui F, Wang W, Wang S and Zhu H: B subunit of human chorionic gonadotropin promotes tumor invasion and predicts poor prognosis of early-stage colorectal cancer. Cell Physiol Biochem. 45:237–249. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Khobta N, Tomasini P, Garcia ME, Garcia S and Barlesi F: β-Human chorionic gonadotropin (HCG) dosage and lung cancer: A pitfall when screening patients for clinical trials. Bull Cancer. 99:1065–1068. 2012.(In French). View Article : Google Scholar : PubMed/NCBI

37 

Khattri S, Vivekanandarajah A, Varma S and Kong F: Secretion of beta-human chorionic gonadotropin by non-small cell lung cancer: A case report. J Med Case Rep. 5:192011. View Article : Google Scholar : PubMed/NCBI

38 

Konishi Y, Kawamata F, Nishihara H, Homma S, Kato Y, Tsuda M, Kohsaka S, Einama T, Liu C, Yoshida T, et al: Tumor budding and human chorionic gonadotropin-β expression correlate with unfavorable patient outcome in colorectal carcinoma. Med Oncol. 35:1042018. View Article : Google Scholar : PubMed/NCBI

39 

Noda Y, Simodaira M, Ito H, Gonda H, Okada N, Suzuki M and Kaneko M: Two cases of human chorionic gonadotropin-producing large cell carcinoma of the lung accompanied with gynecomastia. Nihon Kyobu Shikkan Gakkai Zasshi. 28:781–785. 1990.(In Japanese). PubMed/NCBI

40 

Lundin M, Nordling S, Lundin J, Alfthan H, Stenman UH and Haglund C: Tissue expression of human chorionic gonadotropin beta predicts outcome in colorectal cancer: A comparison with serum expression. Int J Cancer. 95:18–22. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Arano Y, Shimizu J, Murakami S, Hayashi Y, Kobayashi K, Sekido N, Morita K, Mochiki Y, Tomita S and Watanabe Y: A female case of adenocarcinoma of the lung producing human chorionic gonadotropin. Kyobu Geka. 47:485–487. 1994.(In Japanese). PubMed/NCBI

42 

Okutur K, Hasbal B, Aydin K, Bozkurt M, Namal E, Oz B, Kaynak K and Demir G: Pleomorphic carcinoma of the lung with high serum beta-human chorionic gonadotropin level and gynecomastia. J Korean Med Sci. 25:1805–1808. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Seder CW, Arndt AT, Jordano L, Basu S, Fhied CL, Sayidine S, Chmielewski GW, Gallo K, Liptay MJ and Borgia JA: Serum biomarkers may prognosticate recurrence in node-negative, non-small cell lung cancers less than 4 centimeters. Ann Thorac Surg. 104:1637–1643. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Wong YP, Tan GC, Aziz S, Pongprakyun S and Ismail F: Beta-human chorionic gonadotropin-secreting lung adenocarcinoma. Malays J Med Sci. 22:76–80. 2015.PubMed/NCBI

45 

Vicier C, Tabouret E, Tallet A, Gonçalves A, Chetaille B, Viens P and Madroszyk A: BetaHCG secretion by a pulmonary adenocarcinoma. World J Surg Oncol. 11:2282013. View Article : Google Scholar : PubMed/NCBI

46 

Dinis de Sousa M, Barata M, Miranda AR, Sequeira P, Oliveira A, Xavier L and Mansinho H: Beta-HCG secretion by a pulmonary pleomorphic carcinoma: A case report. Respir Med Case Rep. 34:1015282021.PubMed/NCBI

47 

Ramakumar S, Cheville JC and Zincke H: Urothelial carcinoma of the bladder with choriocarcinomatous differentiation A report of two cases and review of the literature. Urol Oncol. 4:39–42. 1998. View Article : Google Scholar : PubMed/NCBI

48 

Pesce C, Merino MJ, Chambers JT and Nogales F: Endometrial carcinoma with trophoblastic differentiation. An aggressive form of uterine cancer. Cancer. 68:1799–1802. 1991. View Article : Google Scholar : PubMed/NCBI

49 

Attanoos RL, Papagiannis A, Suttinont P, Goddard H, Papotti M and Gibbs AR: Pulmonary giant cell carcinoma: Pathological entity or morphological phenotype? Histopathology. 32:225–231. 1998. View Article : Google Scholar : PubMed/NCBI

50 

Liao XH, Wang Y, Wang N, Yan TB, Xing WJ, Zheng L, Zhao DW, Li YQ, Liu LY, Sun XG, et al: Human chorionic gonadotropin decreases human breast cancer cell proliferation and promotes differentiation. IUBMB Life. 66:352–360. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Dando I, Carmona-Carmona CA and Zampieri N: Human chorionic gonadotropin-mediated induction of breast cancer cell proliferation and differentiation. Cells. 10:2642021. View Article : Google Scholar : PubMed/NCBI

52 

Sengodan SK, Nadhan R, Nair RS, Hemalatha SK, Somasundaram V, Sushama RR, Rajan A, Latha NR, Varghese GR, Thankappan RK, et al: BRCA1 regulation on β-hCG: A mechanism for tumorigenicity in BRCA1 defective breast cancer. Oncogenesis. 6:e3762017. View Article : Google Scholar : PubMed/NCBI

53 

Rajabi B, Khoury J, Brewer C and Goodman OB Jr: Urothelial bladder carcinoma with choriocarcinomatous differentiation presenting with a false-positive pregnancy test. Am J Med Sci. 346:256–258. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Guo X, Liu G, Schauer IG, Yang G, Mercado-Uribe I, Yang F, Zhang S, He Y and Liu J: Overexpression of the β subunit of human chorionic gonadotropin promotes the transformation of human ovarian epithelial cells and ovarian tumorigenesis. Am J Pathol. 179:1385–1393. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Mustafa A, Bozdag Z, Tepe NB and Ozcan HC: An unexpected reason for elevated human chorionic gonadotropin in a young woman. Cervical squamous carcinoma. Saudi Med J. 37:905–907. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Fiddes JC and Goodman HM: The gene encoding the common alpha subunit of the four human glycoprotein hormones. J Mol Appl Genet. 1:3–18. 1981.PubMed/NCBI

57 

Boorstein WR, Vamvakopoulos NC and Fiddes JC: Human chorionic gonadotropin beta-subunit is encoded by at least eight genes arranged in tandem and inverted pairs. Nature. 300:419–422. 1982. View Article : Google Scholar : PubMed/NCBI

58 

Rull K and Laan M: Expression of beta-subunit of HCG genes during normal and failed pregnancy. Hum Reprod. 20:3360–3368. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Bellet D, Lazar V, Bièche I, Paradis V, Giovangrandi Y, Paterlini P, Lidereau R, Bedossa P, Bidart JM and Vidaud M: Malignant transformation of nontrophoblastic cells is associated with the expression of chorionic gonadotropin beta genes normally transcribed in trophoblastic cells. Cancer Res. 57:516–523. 1997.PubMed/NCBI

60 

Lapthorn AJ, Harris DC, Littlejohn A, Lustbader JW, Canfield RE, Machin KJ, Morgan FJ and Isaacs NW: Crystal structure of human chorionic gonadotropin. Nature. 369:455–461. 1994. View Article : Google Scholar : PubMed/NCBI

61 

Stenman UH, Tiitinen A, Alfthan H and Valmu L: The classification, functions and clinical use of different isoforms of HCG. Hum Reprod Update. 12:769–784. 2006. View Article : Google Scholar : PubMed/NCBI

62 

Nwabuobi C, Arlier S, Schatz F, Guzeloglu-Kayisli O, Lockwood CJ and Kayisli UA: hCG: Biological functions and clinical applications. Int J Mol Sci. 18:20372017. View Article : Google Scholar : PubMed/NCBI

63 

Schmitt EJ, Barros CM, Fields PA, Fields MJ, Diaz T, Kluge JM and Thatcher WW: A cellular and endocrine characterization of the original and induced corpus luteum after administration of a gonadotropin-releasing hormone agonist or human chorionic gonadotropin on day five of the estrous cycle. J Anim Sci. 74:1915–1929. 1996. View Article : Google Scholar : PubMed/NCBI

64 

Pierce JG and Parsons TF: Glycoprotein hormones: Structure and function. Annu Rev Biochem. 50:465–495. 1981. View Article : Google Scholar : PubMed/NCBI

65 

Morgan FJ, Birken S and Canfield RE: The amino acid sequence of human chorionic gonadotropin. The alpha subunit and beta subunit. J Biol Chem. 250:5247– 5258. 1975. View Article : Google Scholar : PubMed/NCBI

66 

Elliott MM, Kardana A, Lustbader JW and Cole LA: Carbohydrate and peptide structure of the alpha- and beta-subunits of human chorionic gonadotropin from normal and aberrant pregnancy and choriocarcinoma. Endocrine. 7:15–32. 1997. View Article : Google Scholar : PubMed/NCBI

67 

Cole LA: Biological functions of hCG and hCG-related molecules. Reprod Biol Endocrinol. 8:1022010. View Article : Google Scholar : PubMed/NCBI

68 

Kovalevskaya G, Birken S, Kakuma T, Ozaki N, Sauer M, Lindheim S, Cohen M, Kelly A, Schlatterer J and O'Connor JF: Differential expression of human chorionic gonadotropin (hCG) glycosylation isoforms in failing and continuing pregnancies: Preliminary characterization of the hyperglycosylated hCG epitope. J Endocrinol. 172:497–506. 2002. View Article : Google Scholar : PubMed/NCBI

69 

Kobata A and Takeuchi M: Structure, pathology and function of the N-linked sugar chains of human chorionic gonadotropin. Biochim Biophys Acta. 1455:315–326. 1999. View Article : Google Scholar : PubMed/NCBI

70 

Birken S, Berger P, Bidart JM, Weber M, Bristow A, Norman R, Sturgeon C and Stenman UH: Preparation and characterization of new WHO reference reagents for human chorionic gonadotropin and metabolites. Clin Chem. 49:144–154. 2003. View Article : Google Scholar : PubMed/NCBI

71 

Birken S, Maydelman Y, Gawinowicz MA, Pound A, Liu Y and Hartree AS: Isolation and characterization of human pituitary chorionic gonadotropin. Endocrinology. 137:1402–1411. 1996. View Article : Google Scholar : PubMed/NCBI

72 

Cole LA: hCG, the wonder of today's science. Reprod Biol Endocrinol. 10:242012. View Article : Google Scholar : PubMed/NCBI

73 

Stenman UH, Bidart JM, Birken S, Mann K, Nisula B and O'Connor J: Standardization of protein immunoprocedures. Choriogonadotropin (CG). Scand J Clin Lab Invest Suppl. 216:42–78. 1993. View Article : Google Scholar : PubMed/NCBI

74 

Khodr G and Siler-Khodr TM: The effect of luteinizing hormone-releasing factor on human chorionic gonadotropin secretion. Fertil Steril. 30:301–304. 1978. View Article : Google Scholar : PubMed/NCBI

75 

Szilágyi A, Benz R and Rossmanith WG: The human first-term placenta in vitro: Regulation of hCG secretion by GnRH and its antagonist. Gynecol Endocrinol. 6:293–300. 1992. View Article : Google Scholar : PubMed/NCBI

76 

Wilson EA and Jawad MJ: Stimulation of human chorionic gonadotropin secretion by glucocorticoids. Am J Obstet Gynecol. 142:344–349. 1982. View Article : Google Scholar : PubMed/NCBI

77 

Murthi P, Kalionis B, Cocquebert M, Rajaraman G, Chui A, Keogh RJ, Evain-Brion D and Fournier T: Homeobox genes and down-stream transcription factor PPARγ in normal and pathological human placental development. Placenta. 34:299–309. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Boothby M, Kukowska J and Boime I: Imbalanced synthesis of human choriogonadotropin alpha and beta subunits reflects the steady state levels of the corresponding mRNAs. J Biol Chem. 258:9250–9253. 1983. View Article : Google Scholar : PubMed/NCBI

79 

Ringler GE, Kao LC, Miller WL and Strauss JF III: Effects of 8-bromo-cAMP on expression of endocrine functions by cultured human trophoblast cells. Regulation of specific mRNAs. Mol Cell Endocrinol. 61:13–21. 1989. View Article : Google Scholar : PubMed/NCBI

80 

Burnside J, Nagelberg SB, Lippman SS and Weintraub BD: Differential regulation of hCG alpha and beta subunit mRNAs in JEG-3 choriocarcinoma cells by 8-bromo-cAMP. J Biol Chem. 260:12705–12709. 1985. View Article : Google Scholar : PubMed/NCBI

81 

Knöfler M, Saleh L, Strohmer H, Husslein P and Wolschek MF: Cyclic AMP- and differentiation-dependent regulation of the proximal alphaHCG gene promoter in term villous trophoblasts. Mol Hum Reprod. 5:573–580. 1999. View Article : Google Scholar : PubMed/NCBI

82 

Fenstermaker RA, Milsted A, Virgin JB, Miller WL and Nilson JH: The transcriptional response of the human chorionic gonadotropin beta-subunit gene to cAMP is cycloheximide sensitive and is mediated by cis-acting sequences different from that found in the alpha-subunit gene. Mol Endocrinol. 3:1070–1076. 1989. View Article : Google Scholar : PubMed/NCBI

83 

Deutsch PJ, Jameson JL and Habener JF: Cyclic AMP responsiveness of human gonadotropin-alpha gene transcription is directed by a repeated 18-base pair enhancer. Alpha-promoter receptivity to the enhancer confers cell-preferential expression. J Biol Chem. 262:12169–12174. 1987. View Article : Google Scholar : PubMed/NCBI

84 

Delegeane AM, Ferland LH and Mellon PL: Tissue-specific enhancer of the human glycoprotein hormone alpha-subunit gene: Dependence on cyclic AMP-inducible elements. Mol Cell Biol. 7:3994–4002. 1987. View Article : Google Scholar : PubMed/NCBI

85 

Bokar JA, Keri RA, Farmerie TA, Fenstermaker RA, Andersen B, Hamernik DL, Yun J, Wagner T and Nilson JH: Expression of the glycoprotein hormone alpha-subunit gene in the placenta requires a functional cyclic AMP response element, whereas a different cis-acting element mediates pituitary-specific expression. Mol Cell Biol. 9:5113–5122. 1989. View Article : Google Scholar : PubMed/NCBI

86 

Jameson JL, Albanese C and Habener JF: Distinct adjacent protein-binding domains in the glycoprotein hormone alpha gene interact independently with a cAMP-responsive enhancer. J Biol Chem. 264:16190–16196. 1989. View Article : Google Scholar : PubMed/NCBI

87 

Jameson JL and Hollenberg AN: Regulation of chorionic gonadotropin gene expression. Endocr Rev. 14:203–221. 1993. View Article : Google Scholar : PubMed/NCBI

88 

Budworth PR, Quinn PG and Nilson JH: Multiple characteristics of a pentameric regulatory array endow the human alpha-subunit glycoprotein hormone promoter with trophoblast specificity and maximal activity. Mol Endocrinol. 11:1669–1680. 1997. View Article : Google Scholar : PubMed/NCBI

89 

Pittman RH, Clay CM, Farmerie TA and Nilson JH: Functional analysis of the placenta-specific enhancer of the human glycoprotein hormone alpha subunit gene. Emergence of a new element. J Biol Chem. 269:19360–19368. 1994. View Article : Google Scholar : PubMed/NCBI

90 

Cole LA: Human chorionic gonadotropin and associated molecules. Expert Rev Mol Diagn. 9:51–73. 2009. View Article : Google Scholar : PubMed/NCBI

91 

Steger DJ, Hecht JH and Mellon PL: GATA-binding proteins regulate the human gonadotropin alpha-subunit gene in the placenta and pituitary gland. Mol Cell Biol. 14:5592–5602. 1994. View Article : Google Scholar : PubMed/NCBI

92 

Steger DJ, Büscher M, Hecht JH and Mellon PL: Coordinate control of the alpha- and beta-subunit genes of human chorionic gonadotropin by trophoblast-specific element-binding protein. Mol Endocrinol. 7:1579–1588. 1993. View Article : Google Scholar : PubMed/NCBI

93 

Talmadge K, Boorstein WR, Vamvakopoulos NC, Gething MJ and Fiddes JC: Only three of the seven human chorionic gonadotropin beta subunit genes can be expressed in the placenta. Nucleic Acids Res. 12:8415–8436. 1984. View Article : Google Scholar : PubMed/NCBI

94 

Bo M and Boime I: Identification of the transcriptionally active genes of the chorionic gonadotropin beta gene cluster in vivo. J Biol Chem. 267:3179–3184. 1992. View Article : Google Scholar : PubMed/NCBI

95 

Hotakainen K, Lintula S, Jarvinen R, Paju A, Stenman J, Rintala E and Stenman UH: Overexpression of human chorionic gonadotropin beta genes 3, 5 and 8 in tumor tissue and urinary cells of bladder cancer patients. Tumour Biol. 28:52–56. 2007. View Article : Google Scholar : PubMed/NCBI

96 

Pestell RG, Hollenberg AN, Albanese C and Jameson JL: c-Jun represses transcription of the human chorionic gonadotropin alpha and beta genes through distinct types of CREs. J Biol Chem. 269:31090–31096. 1994. View Article : Google Scholar : PubMed/NCBI

97 

Ghosh D, Ezashi T, Ostrowski MC and Roberts RM: A central role for Ets-2 in the transcriptional regulation and cyclic adenosine 5′-monophosphate responsiveness of the human chorionic gonadotropin-beta subunit gene. Mol Endocrinol. 17:11–26. 2003. View Article : Google Scholar : PubMed/NCBI

98 

Knöfler M, Saleh L, Bauer S, Galos B, Rotheneder H, Husslein P and Helmer H: Transcriptional regulation of the human chorionic gonadotropin beta gene during villous trophoblast differentiation. Endocrinology. 145:1685–1694. 2004. View Article : Google Scholar : PubMed/NCBI

99 

Liu L and Roberts RM: Silencing of the gene for the beta subunit of human chorionic gonadotropin by the embryonic transcription factor Oct-3/4. J Biol Chem. 271:16683–16689. 1996. View Article : Google Scholar : PubMed/NCBI

100 

Fournier T, Guibourdenche J, Handschuh K, Tsatsaris V, Rauwel B, Davrinche C and Evain-Brion D: PPARγ and human trophoblast differentiation. J Reprod Immunol. 90:41–49. 2011. View Article : Google Scholar : PubMed/NCBI

101 

Sohr S and Engeland K: The tumor suppressor p53 induces expression of the pregnancy-supporting human chorionic gonadotropin (hCG) CGB7 gene. Cell Cycle. 10:3758–3767. 2011. View Article : Google Scholar : PubMed/NCBI

102 

Chen Y, Miyazaki J, Nishizawa H, Kurahashi H, Leach R and Wang K: MTA3 regulates CGB5 and Snail genes in trophoblast. Biochem Biophys Res Commun. 433:379–384. 2013. View Article : Google Scholar : PubMed/NCBI

103 

Śliwa A, Kubiczak M, Szczerba A, Walkowiak G, Nowak-Markwitz E, Burczyńska B, Butler S, Iles R, Białas P and Jankowska A: Regulation of human chorionic gonadotropin beta subunit expression in ovarian cancer. BMC Cancer. 19:7462019. View Article : Google Scholar : PubMed/NCBI

104 

Głodek A, Kubiczak MJ, Walkowiak GP, Nowak-Markwitz E and Jankowska A: Methylation status of human chorionic gonadotropin beta subunit promoter and TFAP2A expression as factors regulating CGB gene expression in placenta. Fertil Steril. 102:1175–1182.e8. 2014. View Article : Google Scholar : PubMed/NCBI

105 

Johnson W and Jameson JL: AP-2 (activating protein 2) and Sp1 (selective promoter factor 1) regulatory elements play distinct roles in the control of basal activity and cyclic adenosine 3′,5′-monophosphate responsiveness of the human chorionic gonadotropin-beta promoter. Mol Endocrinol. 13:1963–1975. 1999. View Article : Google Scholar : PubMed/NCBI

106 

Tarrade A, Schoonjans K, Guibourdenche J, Bidart JM, Vidaud M, Auwerx J, Rochette-Egly C and Evain-Brion D: PPAR gamma/RXR alpha heterodimers are involved in human CG beta synthesis and human trophoblast differentiation. Endocrinology. 142:4504–4514. 2001. View Article : Google Scholar : PubMed/NCBI

107 

Handschuh K, Guibourdenche J, Cocquebert M, Tsatsaris V, Vidaud M, Evain-Brion D and Fournier T: Expression and regulation by PPARgamma of hCG alpha- and beta-subunits: Comparison between villous and invasive extravillous trophoblastic cells. Placenta. 30:1016–1022. 2009. View Article : Google Scholar : PubMed/NCBI

108 

Shalom-Barak T, Zhang X, Chu T, Timothy Schaiff W, Reddy JK, Xu J, Sadovsky Y and Barak Y: Placental PPARγ regulates spatiotemporally diverse genes and a unique metabolic network. Dev Biol. 372:143–155. 2012. View Article : Google Scholar : PubMed/NCBI

109 

Peng L, Yang H, Ye Y, Ma Z, Kuhn C, Rahmeh M, Mahner S, Makrigiannakis A, Jeschke U and von Schönfeldt V: Role of peroxisome proliferator-activated receptors (PPARs) in trophoblast functions. Int J Mol Sci. 22:4332021. View Article : Google Scholar : PubMed/NCBI

110 

Campain JA, Gutkin DW and Cox GS: Differential DNA methylation of the chorionic gonadotropin beta-subunit multigene family. Mol Endocrinol. 7:1331–1346. 1993. View Article : Google Scholar : PubMed/NCBI

111 

Grigoriu A, Ferreira JC, Choufani S, Baczyk D, Kingdom J and Weksberg R: Cell specific patterns of methylation in the human placenta. Epigenetics. 6:368–379. 2011. View Article : Google Scholar : PubMed/NCBI

112 

Uusküla L, Rull K, Nagirnaja L and Laan M: Methylation allelic polymorphism (MAP) in chorionic gonadotropin beta5 (CGB5) and its association with pregnancy success. J Clin Endocrinol Metab. 96:E199–E207. 2011. View Article : Google Scholar : PubMed/NCBI

113 

Butler SA, Ikram MS, Mathieu S and Iles RK: The increase in bladder carcinoma cell population induced by the free beta subunit of human chorionic gonadotrophin is a result of an anti-apoptosis effect and not cell proliferation. Br J Cancer. 82:1553–1556. 2000.PubMed/NCBI

114 

Li Z, Du L, Li C and Wu W: Human chorionic gonadotropin β induces cell motility via ERK1/2 and MMP-2 activation in human glioblastoma U87MG cells. J Neurooncol. 111:237–244. 2013. View Article : Google Scholar : PubMed/NCBI

115 

Wu W and Walker AM: Human chorionic gonadotropin beta (HCGbeta) down-regulates E-cadherin and promotes human prostate carcinoma cell migration and invasion. Cancer. 106:68–78. 2006. View Article : Google Scholar : PubMed/NCBI

116 

Li Z, Li C, Du L, Zhou Y and Wu W: Human chorionic gonadotropin β induces migration and invasion via activating ERK1/2 and MMP-2 in human prostate cancer DU145 cells. PLoS One. 8:e545922013. View Article : Google Scholar : PubMed/NCBI

117 

Regalado JJ: Mixed micropapillary and trophoblastic carcinoma of bladder: Report of a first case with new immunohistochemical evidence of urothelial origin. Hum Pathol. 35:382–384. 2004. View Article : Google Scholar : PubMed/NCBI

118 

Białas P, Śliwa A, Szczerba A and Jankowska A: The study of the expression of CGB1 and CGB2 in human cancer tissues. Genes (Basel). 11:10822020. View Article : Google Scholar : PubMed/NCBI

119 

Gao S, Fan C, Huang H, Zhu C, Su M and Zhang Y: Effects of HCG on human epithelial ovarian cancer vasculogenic mimicry formation in vivo. Oncol Lett. 12:459–466. 2016. View Article : Google Scholar : PubMed/NCBI

120 

Armah HB and Parwani AV: Sarcomatoid urothelial carcinoma with choriocarcinomatous features: First report of an unusual case. Urology. 70:812.e11–e14. 2007. View Article : Google Scholar : PubMed/NCBI

121 

Zhao R, Zhang T, Xi W, Sun X, Zhou L, Guo Y, Zhao C and Bao Y: Human chorionic gonadotropin promotes cell proliferation through the activation of c-Met in gastric cancer cells. Oncol Lett. 16:4271–4278. 2018.PubMed/NCBI

122 

Wu W, Gao H, Li X, Peng S, Yu J, Liu N, Zhan G, Zhu Y, Wang K and Guo X: β-hCG promotes epithelial ovarian cancer metastasis through ERK/MMP2 signaling pathway. Cell Cycle. 18:46–59. 2019. View Article : Google Scholar : PubMed/NCBI

123 

Singer G, Kurman RJ, McMaster MT and Shih IM: HLA-G immunoreactivity is specific for intermediate trophoblast in gestational trophoblastic disease and can serve as a useful marker in differential diagnosis. Am J Surg Pathol. 26:914–920. 2002. View Article : Google Scholar : PubMed/NCBI

124 

Shih IM: The role of CD146 (Mel-CAM) in biology and pathology. J Pathol. 189:4–11. 1999. View Article : Google Scholar : PubMed/NCBI

125 

Banet N, Gown AM, Shih IM, Kay Li Q, Roden RB, Nucci MR, Cheng L, Przybycin CG, Nasseri-Nik N, Wu LS, et al: GATA-3 expression in trophoblastic tissues: An immunohistochemical study of 445 cases, including diagnostic utility. Am J Surg Pathol. 39:101–108. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Mao TL, Kurman RJ, Jeng YM, Huang W and Shih IM: HSD3B1 as a novel trophoblast-associated marker that assists in the differential diagnosis of trophoblastic tumors and tumorlike lesions. Am J Surg Pathol. 32:236–242. 2008. View Article : Google Scholar : PubMed/NCBI

127 

Stichelbout M, Devisme L, Franquet-Ansart H, Massardier J, Vinatier D, Renaud F and Kerdraon O: SALL4 expression in gestational trophoblastic tumors: A useful tool to distinguish choriocarcinoma from placental site trophoblastic tumor and epithelioid trophoblastic tumor. Hum Pathol. 54:121–126. 2016. View Article : Google Scholar : PubMed/NCBI

128 

Kaur B and Sebire NJ: Gestational trophoblastic tumours and non-neoplastic trophoblastic lesions: morphology and immunocytochemistry to refine the diagnosis. Diagn Histopathol. 25:53–65. 2019. View Article : Google Scholar

129 

Heller DS: Update on the pathology of gestational trophoblastic disease. APMIS. 126:647–654. 2018. View Article : Google Scholar : PubMed/NCBI

130 

Cheung AN, Hui P and Shih I: Gestational trophoblastic disease. WHO classification of tumours: Female genital tumours. Vol 4. 5th edition. IARC; Lyon: pp. 310–333. 2020

131 

Shih IM: Trophogram, an immunohistochemistry-based algorithmic approach, in the differential diagnosis of trophoblastic tumors and tumorlike lesions. Ann Diagn Pathol. 11:228–234. 2007. View Article : Google Scholar : PubMed/NCBI

132 

Lee CQE, Gardner L, Turco M, Zhao N, Murray MJ, Coleman N, Rossant J, Hemberger M and Moffett A: What is trophoblast? A combination of criteria define human first-trimester trophoblast. Stem Cell Reports. 6:257–272. 2016. View Article : Google Scholar : PubMed/NCBI

133 

Triozzi PL and Stevens VC: Human chorionic gonadotropin as a target for cancer vaccines. Oncol Rep. 6:7–17. 1999.PubMed/NCBI

134 

Yu N, Xu W, Jiang Z, Cao Q, Chu Y and Xiong S: Inhibition of tumor growth in vitro and in vivo by a monoclonal antibody against human chorionic gonadotropin beta. Immunol Lett. 114:94–102. 2007. View Article : Google Scholar : PubMed/NCBI

135 

Morse MA, Bradley DA, Keler T, Laliberte RJ, Green JA, Davis TA and Inman BA: CDX-1307: A novel vaccine under study as treatment for muscle-invasive bladder cancer. Expert Rev Vaccines. 10:733–742. 2011. View Article : Google Scholar : PubMed/NCBI

136 

Rayn KN, Hale GR, Grave GP and Agarwal PK: New therapies in nonmuscle invasive bladder cancer treatment. Indian J Urol. 34:11–19. 2018. View Article : Google Scholar : PubMed/NCBI

137 

Sahoo S, Singh P, Kalha B, Singh O and Pal R: Gonadotropin-mediated chemoresistance: Delineation of molecular pathways and targets. BMC Cancer. 15:9312015. View Article : Google Scholar : PubMed/NCBI

138 

Szturmowicz M, Wiatr E, Sakowicz A, Slodkowska J, Roszkowski K, Filipecki S and Rowinska-Zakrzewska ER: The role of human chorionic gonadotropin beta subunit elevation in small-cell lung cancer patients. J Cancer Res Clin Oncol. 121:309–312. 1995. View Article : Google Scholar : PubMed/NCBI

139 

Oyasu R, Nan L, Smith DP and Kawamata H: Human chorionic gonadotropin beta-subunit synthesis by undifferentiated urothelial carcinoma with syncytiotrophoblastic differentiation. Arch Pathol Lab Med. 118:715–717. 1994.PubMed/NCBI

Related Articles

Journal Cover

March-2024
Volume 51 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
Chang C, Chen Y, Wang Y, Chen H, Hsu C, Lin K, Ou Y, Liu T, Chen W, Chu C, Chu C, et al: Aberrant trophoblastic differentiation in human cancer: An emerging novel therapeutic target (Review). Oncol Rep 51: 43, 2024
APA
Chang, C., Chen, Y., Wang, Y., Chen, H., Hsu, C., Lin, K. ... Chow, N. (2024). Aberrant trophoblastic differentiation in human cancer: An emerging novel therapeutic target (Review). Oncology Reports, 51, 43. https://doi.org/10.3892/or.2024.8701
MLA
Chang, C., Chen, Y., Wang, Y., Chen, H., Hsu, C., Lin, K., Ou, Y., Liu, T., Chen, W., Chu, C., Ho, C., Lee, C., Chow, N."Aberrant trophoblastic differentiation in human cancer: An emerging novel therapeutic target (Review)". Oncology Reports 51.3 (2024): 43.
Chicago
Chang, C., Chen, Y., Wang, Y., Chen, H., Hsu, C., Lin, K., Ou, Y., Liu, T., Chen, W., Chu, C., Ho, C., Lee, C., Chow, N."Aberrant trophoblastic differentiation in human cancer: An emerging novel therapeutic target (Review)". Oncology Reports 51, no. 3 (2024): 43. https://doi.org/10.3892/or.2024.8701