Open Access

Combination of circulating tumor cells, lncRNAs and DNA methylation for the diagnosis of endometrial carcinoma

  • Authors:
    • Hongmei Ding
    • Juan Wang
    • Xiaoyu Zhao
    • Shi Xiu
    • Honghong Cai
    • Jingjing Ma
    • Li Fu
    • Jinhua Zhou
    • Fangrong Shen
    • Hong Zhang
    • Youguo Chen
    • Bingyan Li
    • Jing Yan
  • View Affiliations

  • Published online on: September 11, 2024     https://doi.org/10.3892/ol.2024.14678
  • Article Number: 545
  • Copyright: © Ding 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

Endometrial carcinoma (EC) is one of the most common gynecological malignant neoplasms, the prognosis of which is strongly related to the time of diagnosis, with an earlier diagnosis leading to a better prognosis. Therefore, effective diagnostic indicators and methods are needed to ensure early detection. The present study explored the following in EC: Circulating tumor cells (CTCs); the long noncoding RNAs (lncRNAs) RP4‑616B8.5, RP11‑389G6.3 and carboxy‑terminal domain (CTD)‑2377D24.6; and the methylation of cysteine dioxygenase type 1 (CDO1) and CUGBP Elav‑like family member 4 (CELF4). In total, 85 patients, including 71 with EC, and 14 without EC (NO‑EC) but with uterine fibroids or polyps, were included in the present study. In total, 46 patients with EC and 8 NO‑EC patients underwent CTC detection. In the evaluation of the EC vs. NO‑EC groups, the results showed that the CTC‑positive rate of the EC group was 80.43% and that the area under the curve (AUC) value of CTCs was 0.8872 (P=0.0098). A total of 35 patients with EC and 14 NO‑EC patients underwent detection of the RP4‑616B8.5, RP11‑389G6.3 and CTD‑2377D24.6 lncRNAs. When the levels of the three lncRNAs RP4‑616B8.5, RP11‑389G6.3 and CTD‑2377D24.6 were compared between the EC and NO‑EC groups, they were higher in the EC group; the P‑values were 0.0002, 0.0001 and <0.0001, respectively, and the AUC values were 0.8184, 0.8347 and 0.8265, respectively. In addition, a total of 35 patients with EC and 8 NO‑EC patients underwent CDO1 and CELF4 DNA methylation analysis. The positive rates of the methylated genes CDO1 and CELF4 were 20% (7/35) and 5.71% (2/35), and the P‑values of the comparisons between the EC and NO‑EC groups were 0.1748 and 0.5004, respectively; the AUC values were 0.6000 and 0.5286. Furthermore, the combination of CTCs, and lncRNAs RP4‑616B8.5, RP11‑389G6.3 and CTD‑2377D24.6 exhibited high performance in the detection of EC (AUC=0.9375).

Introduction

Endometrial carcinoma (EC) is one of the most common gynecological malignancies in developed countries (1). Notably, the incidence of EC has been increasing in a number of countries, including the United States, as well as in Europe and East Asia, which may be due to a greater exposure to environmental risk factors, such as obesity, increasing age (≥55 years) and a shift in female reproductive patterns (24). Moreover, the incidence of EC is estimated to increase by 55% from 2010 to 2030 (5).

Current clinical data have indicated that the prognosis of EC is closely related to the time of diagnosis, with an earlier diagnosis associated with a better prognosis [International Federation of Gynecology and Obstetrics (FIGO) stage I–II]; for example, the 5-year survival rate has been reported to decrease from 85% for stage I disease to 25% for stage IV disease (68). Currently, EC is diagnosed by a combination of transvaginal ultrasound (TVUS) and endometrial biopsy; however, there is marked heterogeneity in the accuracy of TVUS for detecting malignancies, as its sensitivity ranges from 0.5 to 0.8 for gynecologic oncological diseases (9). On the other hand, endometrial biopsy is invasive and uncomfortable for the patient, and pathological assessment sometimes cannot be carried out due to the failure of the sampling owing to the pain of the sampling process or problems of cervical stenosis (10). Furthermore, the role of test results in guiding personalized treatment plans requires more research support. There are still unanswered questions regarding EC, including a number in the domains of treatment toxicity, diagnostic procedures and adjuvant therapy (11,12). Therefore, reliable detection of EC is necessary to ensure adequate treatment and reduce EC-associated mortality.

With recent advancements in technology, researchers have focused on developing robust and sensitive detection methods, such as circulating tumor cell (CTC) detection, as well as methods involving genomics, epigenomics and transcriptomics (13,14). CTCs disseminate into the bloodstream from either the primary tumor site or metastatic sites (15). Consequently, the number of CTCs is higher in patients with various types of cancer, such as lung cancer and breast cancer, than in healthy volunteers (16,17). In a study of the detection of EC CTCs, aminopeptidase N (CD13) was identified as an alternative prognostic marker for both cervical and endometrial cancer, as its expression was detected in patients with EC before surgery and after recurrence (18).

Long noncoding RNAs (lncRNAs) have an important role in the epigenetic regulatory network, and can regulate gene expression and post-transcriptional processes by influencing the structures of protomers, chromatin and transcription factors (19). A number of studies have reported that some lncRNAs affect various hallmarks of human cancer, such as replicative immortality, antagonism of cell death and evasion of immunosurveillance (20,21); therefore, lncRNAs, such as RP4-616B8.5, RP11-389G6.3, carboxy-terminal domain (CTD)-2377D24.6, AC138904.1 and AC099329.2, are used as biomarkers in numerous cancer diagnoses (22,23). Ding et al (24) reported that the combination of lncRNAs RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 had good performance (P<0.0001) in EC diagnosis. Xin et al (25) reported that low RP11-395G23.3 expression was significantly associated with advanced histological grade and lymphovascular space invasion in patients with EC, and that RP11-395G23.3 may be a target for the diagnosis and treatment of EC.

Cytosine methylation of DNA within CpG dinucleotides is the most well-researched epigenetic alteration in humans (26). Hypermethylation of the CpG islands of gene promoters can silence genes, and this is the basis of the clinical use of a number of biomarkers (27,28). DNA methylation is a highly stable molecular feature that can be detected in tumor tissues and cells (29,30). During malignant transformation, EC cells acquire two main types of aberrant DNA methylation patterns: Local DNA hypermethylation and global DNA hypomethylation (31). Qi et al (32) reported that hypermethylated cysteine dioxygenase type 1 (CDO1) and CUGBP Elav-like family member 4 (CELF4) could serve as triage strategy biomarkers in the non-invasive examination of endometrial malignant lesions, and the sensitivity and specificity of CDO1/CELF4 dual-gene methylation assay for endometrial atypical hyperplasia and endometrial cancer reached 84.9 and 86.6%, respectively. CDO1 and zinc finger protein 454 hypermethylation has also been verified in histological samples from patients with EC and atypical hyperplasia (AH) compared with those from patients with benign and normal endometria (P<0.001) (33).

To increase the efficacy of EC screening, a combination of major biomarkers, namely, CTCs, lncRNAs (RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6) and DNA methylation (CDO1 and CELF4), was evaluated in the present study to construct a better diagnostic model for EC.

Materials and methods

Specimens

A total of 85 patients, including 71 with EC, and 14 without EC (NO-EC) but with uterine fibroids or polyps, were enrolled from The First Affiliated Hospital of Soochow University (Suzhou, China) between March 2023 and March 2024. All enrolled patients were female; aged 32–84 years (mean age, 57.75 years); and had provided written informed consent before participation in the present study, with permission given for sample collection and analysis. The present study was approved by the Ethics Committee of The First Affiliated Hospital of Soochow University (approval no. 2021.351). The diagnostic criteria for EC were based on the 2014 World Health Organization Classification of Tumours of the Female Reproductive Organs. The samples were collected before any anticancer drug treatment. The specific clinical information of the subjects is shown in Table I and the groups are shown in Fig. 1.

Table I.

Patient information.

Table I.

Patient information.

Patients with ECa (n=71)NO-ECa patients (n=14)EC vs. NO-EC



CharacteristicEndometrioid adenocarcinoma (n=55)Serous adenocarcinoma (n=9)Other EC (n=7)Uterine fibroids (n=10)Polyps (n=4)P-value
Mean ± SD age, years57.80±10.2059.11±8.82457.8±9.82253.70±8.00164.00±10.000.6448
Hypertension
  Yes33/55 (60.00%)2/9 (22.22%)3/7 (42.86%)2/10 (20.00%)1/4 (25.00%)0.0281
  No22/55 (40.00%)7/9 (77.78%)4/7 (57.14%)8/10 (80.00%)3/4 (75.00%)
Diabetes
  Yes13/55 (23.64%)2/9 (22.22%)1/7 (14.29%)0/10 (0.00%)0/4 (0.00%)0.0494
  No42/55 (76.36%)7/9 (77.78%)6/7 (85.71%)10/10 (100.00%)4/4 (100.00%)
Fatty liver
  Yes19/55 (34.55%)3/9 (33.33%)0/7 (0.00%)1/10 (10.00%)2/4 (50.00%)0.4791
  No36/55 (65.45%)6/9 (66.67%)7/7 (100.00%)9/10 (90.00%)2/4 (50.00%)
LDLa
  High (>3.4 mmol/l)19/55 (34.55%)5/9 (55.56%)5/7 (71.43%)1/10 (10.00%)2/4 (50.00%)0.1746
  Normal (≤3.4 mmol/l)36/55 (65.45%)4/9 (44.44%)2/7 (28.57%)9/10 (90.00%)2/4 (50.00%)
HDLa
  Low (<1.0 mmol/l)16/55 (29.09%)1/9 (11.11%)2/7 (28.57%)1/10 (10.00%)0/4 (0.00%)0.1165
  Normal (≥1.0 mmol/l)39/55 (70.91%)8/9 (88.89%)5/7 (71.43%)9/10 (90.00%)4/4 (100.00%)
TAGa
  High (>1.7 mmol/l)20/55 (36.36%)5/9 (55.56%)5/7 (71.43%)3/10 (30.00%)1/4 (25.00%)0.8599
  Normal (≤1.7 mmol/l)35/55 (63.64%)4/9 (44.44%)2/7 (28.57%)7/10 (70.00%)3/4 (75.00%)
Cholesterol
  High (>5.2 mmol/l)19/55 (34.55%)5/9 (55.56%)3/7 (42.86%)2/10 (20.00%)1/4 (25.00%)0.2399
  Normal (≤5.2 mmol/l)36/55 (65.45%)4/9 (44.44%)4/7 (57.14%)8/10 (80.00%)3/4 (75.00%)
HE4a
  High (>70 pmol/l, before menopause;   >140 pmol/l, post-menopause)18/55 (32.73%)1/9 (11.11%)2/7 (28.57%)0/10 (00.00%)0/4 (0.00%)0.0188
  Normal (≤70 pmol/l, before menopause; ≤140 pmol/l, post-menopause)37/55 (67.27%)8/9 (88.89%)5/7 (71.43%)10/10 (100.00%)4/4 (100.00%)
Glucose
  Normal (3.9-6.1 mmol/l)16/55 (29.09%)1/9 (11.11%)1/7 (14.29%)1/10 (10.00%)0/4 (0.00%)0.1165
  High (>6.1 mmol/l)39/55 (70.91%)8/9 (88.89%)6/7 (85.71%)9/10 (90.00%)4/4 (100.00%)
Number of pregnancies
  03/55 (5.45%)0/9 (0.00%)0/7 (0.00%)0/10 (0.00%)0/4 (0.00%)0.4257
  112/55 (21.82%)2/9 (22.22%)0/7 (0.00%)3/10 (30.00%)1/4 (25.00%)
  214/55 (25.45%)3/9 (33.33%)4/7 (57.14%)4/10 (40.00%)2/4 (50.00%)
  312/55 (21.82%)3/9 (33.33%)1/7 (14.29%)1/10 (10.00%)0/4 (0.00%)
  ≥414/55 (25.45%)1/9 (11.11%)2/7 (28.57%)2/10 (20.00%)1/4 (25.00%)
Stage
  Stage I48/55 (87.27%)5/9 (55.56%)3/7 (42.86%)
  Stage II4/55 (7.27%)1/9 (11.11%)3/7 (42.86%)
  Stage III3/55 (5.45%)1/9 (11.11%)1/7 (14.29%)
  Stage IV0/55 (0.00%)2/9 (22.22%)0/7 (0.00%)
Muscular layer infiltration depth
  <1/244/55 (80.00%)6/9 (66.67%)4/7 (57.14%)
  ≥1/211/55 (20.00%)3/9 (33.33%)3/7 (42.86%)
Tumor size, cm
  <216/55 (29.09%)3/9 (33.33%)2/7 (28.57%)
  ≥239/55 (70.91%)6/9 (66.67%)5/7 (71.43%)
HPVa
  Positive6/31 (19.35%)4/8 (50.00%)2/6 (33.33%) 0/3 (0.00%)0.3695
  Negative25/31 (80.65%)4/8 (50.00%)4/6 (66.67%) 3/3 (100.00%)
CEAa
  High (>5 ng/ml, no smoking; >10 ng/ml, smoking)1/39 (2.57%)0/7 (0.00%)0/3 (0.00%)0/10 (0.00%)1/3 (33.33%)0.3131
  Normal (0–5 ng/ml, no smoking; 0–10 ng/ml, smoking)38/39 (97.43%)7/7 (100.00%)3/3 (100.00%)10/10 (100.00%)2/3 (66.67%)
CA19-9a
  High (>37 U/ml)9/51 (17.65%)1/8 (12.50%)0/6 (0.00%)1/10 (10.00%)0/3 (0.00%)0.4734
  Normal (0–37 U/ml)42/51 (82.35%)7/8 (87.50%)6/6 (100.00%)9/10 (90.00%)3/3 (100.00%)
CA125a
  High (>35 U/ml)9/53 (16.98%)2/9 (22.22%)0/6 (0.00%)2/10 (20.00%)0/2 (0.00%)0.9667
  Normal (0–35 U/ml)44/53 (83.02%)7/9 (77.78%)6/6 (100.00%)8/10 (80.00%)2/2 (100.00%)
ERa
  Positive44/50 (88.00%)7/7 (100.00%)4/4 (100.00%)
  Negative6/50 (12.00%)0/7 (0.00%)0/4 (0.00%)
PRa
  Positive42/54 (77.78%)6/6 (100.00%)5/5 (100.00%)
  Negative12/54 (22.22%)0/6 (0.00%)0/5 (0.00%)
Ki67a
  Positive55/55 (100.00%)7/7 (100.00%)5/5 (100.00%)
  Negative0/55 (0.00%)0/7 (0.00%)0/5 (0.00%)
MSH2a
  Positive28/28 (100.00%)4/4 (100.00%)3/3 (100.00%)
  Negative0/28 (0.00%)0/4 (0.00%)0/3 (0.00%)
CTCa
  Positive31/36 (86.11%)4/8 (50.00%)2/2 (100.00%)0/8 (0.00%) 0.0098
  Negative5/36 (13.89%)4/8 (50.00%)0/2 (0.00%)8/8 (100.00%)
CDO1 DNA methylationa
  Positive7/29 (24.14%)0/5 (0.00%)0/1 (0.00%)0/8 (0.00%) 0.1748
  Negative22/29 (75.86%)5/5 (100.00%)1/1 (100.00%)8/8 (100.00%)
CELF4 DNA methylationa
  Positive2/29 (6.90%)0/5 (0.00%)0/1 (0.00%)0/8 (0.00%) 0.5004
  Negative27/29 (93.10%)5/5 (100.00%)1/1 (100.00%)8/8 (100.00%)

a Detected in some patients. CDO1, cysteine dioxygenase type 1; CA, cancer antigen; CEA, carcinoembryonic antigen; CELF4, CUGBP Elav-like family member 4; CTC, circulating tumor cell; EC, endometrial carcinoma; ER, estrogen receptor; HDL, high-density lipoprotein; HE4, human epididymis protein 4; LDL, low-density lipoprotein; NO-EC, without EC; PR, progesterone receptor.

CTC enrichment and detection

A total of 46 patients with EC and 8 NO-EC patients underwent CTC detection. Peripheral blood (PB) samples (4 ml/patient) were collected before surgery or treatment, stored in EDTA tubes (Becton, Dickinson and Company) and CTCs were detected within 6 h using the CytoBot® 2000 system (Holosensor Medical Technology Ltd.). Before CTC detection, PB mononuclear cells (PBMCs) were isolated from the PB. Briefly, 4 ml density gradient separation solution (Shenzhen DAKEWE Bio-engineering Co., Ltd.) and a diluted blood sample (4 ml PB with an equal volume of phosphate buffer, pH 7.0; Biological Industries) were added sequentially to a sterile 15-ml centrifuge tube and centrifuged at 700 × g for 20 min at room temperature. The PBMCs were then carefully pipetted into a new 15-ml centrifuge tube, washed twice with 5–10 ml PBS (pH 7.2) and centrifuged at 500 × g for 5 min at 25°C.

CTCs were detected using the CytoBot 2000 system, a novel CTC platform based on advanced technology, including microfluidics and immunoenrichment. Briefly, CTC capture chips were manufactured using a metal mesh with pores measuring 15 µm in diameter, and gold-covered polymers and the purified anti-human CD326 (Ep-CAM) capture antibody (cat. no. 324202; BioLegend, Inc.) were seeded onto the surface to form a capture chip with unique functionality. In the present study, the PBMCs were resuspended in PBS (pH 7.2) to a volume of 300 µl and were loaded onto the capture chip. CTCs were captured and stained by the CytoBot® 2000 system using the preset procedures and pre-prepared reagents from the CTCs detection kit (Holosensor, Inc.).

The immunofluorescence staining was carried out using the CytoBot 2000 system and the CTCs detection kit (Holosensor, Inc.), and the indicators used were Alexa Fluor® 488 anti-pancytokeratin (CK), PE anti-human CD45 antibodies and DAPI. The cell types were determined under a fluorescence microscope [RX50M; Sunny Optical Technology (Group) Company Limited]. The evaluation criteria of CTCs was CK+CD45DAPI+, and the threshold for CTC positivity was a CK+CD45DAPI+ CTC number of ≥2.

Tissue sample collection, RNA isolation and reverse transcription-quantitative PCR (RT-qPCR) analysis

In total, 35 patients with EC and 14 NO-EC patients underwent RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 lncRNA detection. Tumor tissues, paracancerous tissues (at a 1-cm distance from tumor tissues), uterine fibroid and polyp tissues were obtained during surgery before treatment. Total RNA was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and RNA was subsequently reverse transcribed into cDNA with a PrimeScript RT reagent kit (Takara Biotechnology, Ltd.) according to the manufacturer's protocol. The expression levels of RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 lncRNAs were measured by qPCR using the Hiff qPCR SYBR Green Master Mix (Shanghai Yeasen Biotechnology Co., Ltd.) and the QuantStudio 6 system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The qPCR conditions were as follows: Pre-denaturation at 95°C for 10 min; followed by 40 cycles of denaturation at 95°C for 10 sec, annealing at 60°C for 10 sec and extension at 70°C for 30 sec; and a hold at 95°C for 15 sec, 60°C for 1 min. The positive standard was a Cq value of ≤40. The expression levels were normalized to the levels of GAPDH mRNA and were calculated using the ΔCq method (34). The primers used for analysis are listed in Table II.

Table II.

Primer sequences used for reverse transcription-quantitative PCR.

Table II.

Primer sequences used for reverse transcription-quantitative PCR.

PrimerSequence, 5′-3′
CTD-2377D24.6-F TTCCGGTGTCCAGATGTTCA
CTD-2377D24.6-R AAGGTGAGTTGGGGAGGATG
RP4-616B8.5-F ATGAGTGTGGCAGCCTATGT
RP4-616B8.5-R AACTCCTGACCTCGTGATCC
RP11-389G6.3-F GGCCTTGAGAGATAGAGGGG
RP11-389G6.3-R ATACGTCCTTCCCATCCTGC
GAPDH-F GCACAGTCAAGGCTGAGAATG
GAPDH-R ATGGTGGTGAAGACGCCAGTA

[i] F, forward; R, reverse.

CDO1 and CELF4 DNA methylation analysis

In total, 35 patients with EC and 8 NO-EC patients were included in this analysis. For clinical testing, cervical epithelial cells and endocervix cells were collected using a cervical brush or cervical epidermal cell sampler. In this experiment, the cervical epidermal cell samples were scraped from subjects with endometrial cell collectors (SAP-I) and were placed in sample preservation solution (cat. no. AM7020; Thermo Fisher Scientific, Inc.). Genomic DNA was extracted using the TIANamp Genomic DNA Kit (cat. no. DP304; Tiangen Biotech Co., Ltd.) and 20 µl DNA eluent was obtained.

A custom-developed bisulfite conversion kit (methylation detection sample pretreatment kit; Holosensor Medical Technology Ltd.) was used to convert the extracted DNA into bisulfite and obtain the transformed bis-DNA. Finally, CDO1 and CELF4 amplification was performed on an ABI 7500 device (Applied Biosystems; Thermo Fisher Scientific, Inc.). The reaction mixture consisted of the PCR solution and primer probes, and the transformed bis-DNA samples were added to the mixture. The reaction conditions were as follows: Pre-denaturation at 96°C for 5 min, followed by 45 cycles of denaturation at 94°C for 15 sec and annealing at 60°C for 35 sec, and a hold at 25°C for 10 min. The positive standard was a Cq value of ≤38. The primers used for the analysis are listed in Table III.

Table III.

Primer sequences used for DNA methylation detection.

Table III.

Primer sequences used for DNA methylation detection.

PrimerSequence, 5′-3′
CDO1 F ATCAACGTTTATATTTTTAAGTTATCG
CDO1 R GACTTAGACCCTCTACTAATCCG
CDO1 FP FAM-CATTCTATTTCGGGCGCGGAGATGCGG-BHQ1
CELF4 F ATCTCCATGTATATAAAGATGGITACG
CELF4 R GATATAAGAACTATAACTTAATCCG
CELF4 FP ROX-ATACCTATAACGGGTTCGGTAGTAGTT-BHQ2

[i] CDO1, cysteine dioxygenase type 1; CELF4, CUGBP Elav-like family member 4; F, forward; R, reverse; FP, fluorescent probe; FAM, carboxyfluorescein; BHQ, Black Hole Quencher; ROX, Rhodamine X.

Statistical analysis

Statistical analyses, including receiver operating characteristic (ROC) curve analysis, paired Student's t-test and unpaired Student's t-test, were performed using GraphPad Prism 10.1.2 software (Dotmatics). P<0.05 was considered to indicate a statistically significant difference.

Results

Diagnostic value of CTC detection

CTC detection, a classic screening method for tumors, has been applied effectively in numerous types of cancer (3537). In the present study, CTC detection was used to evaluate patients with EC and NO-EC patients. The results of CTC enrichment and detection are shown in Fig. 2 and Table I. The classic staining characteristics of the CTCs were CK+CD45DAPI+ (Fig. 2A).

A total of 54 subjects, including 46 patients with EC and 8 NO-EC patients, underwent CTC detection. The total CTC positivity rates for all patients with EC, those with stage I EC, those with stage II EC and those with stage III EC were 80.43% (37/46), 82.05% (32/39), 50% (2/4) and 100% (3/3), respectively (Fig. 2B). In the present study, no patients with stage IV EC underwent CTC detection. Among the 8 NO-EC patients, the CTC positivity rate was 0% (0/8), and the threshold for CTC-positive patients was a CK+CD45DAPI+ CTC number of ≥2. The number of CTCs was significantly increased in patients with EC compared with in NO-EC patients (Fig. 2C). In addition, CTCs performed well in distinguishing between the EC and NO-EC groups, with an area under the curve (AUC) value of 0.8872 (Fig. 2D). These findings indicated that CTCs had a good effect on EC diagnosis.

Diagnostic value of lncRNA detection in EC

Ding et al (24) measured the lncRNAs RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 in clinical samples, and reported that they had good diagnostic performance regarding histological subtype (P=0.0001), advanced clinical stage (P=0.011) and clinical grade (P<0.0001) in patients with EC. The present study evaluated the lncRNAs RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 in patients with EC and NO-EC patients by RT-qPCR analysis; however, the results obtained were different from the results of the previous study (24). The expression levels of the RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 lncRNAs were not significantly different between tumor (n=35) and paracancerous (n=35) tissues according to the results of RT-qPCR (P=0.2730, 0.0517 and 0.5180, respectively; Fig. 3A, E and I) and ROC curve analyses (AUC=0.5380, 0.5747 and 0.5192, respectively; Fig. 3C, G and K). However, the performance of RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 in distinguishing the EC group (n=35) from the NO-EC group (n=14) was good (Fig. 3B, F and J), with AUC values of 0.8184, 0.8347 and 0.8265, respectively (Fig. 3D, H and L).

Diagnostic value of CDO1 and CELF4 DNA methylation

DNA methylation detection has been widely used in cancer screening studies (3840). Huang et al (41) reported that a panel comprising any two of the three hypermethylated genes, BHLHE22, CDO1 and CELF4, reached a sensitivity of 91.8% and specificity of 95.5%. In view of the good performance that has previously been reported, the present study performed a CDO1 and CELF4 DNA methylation analysis. CDO1 and CELF4 were detected in 35 EC samples and 8 NO-EC samples (Fig. 1; Table I). The positive rates of CDO1 and CELF4 methylation were 20% (7/35) and 5.71% (2/35) in EC, respectively, and these values were lower than those reported in other studies (32,33,4042). CDO1 and CELF4 DNA methylation did not significantly differ between the EC (n=35) and NO-EC (n=8) groups (P=0.1748 and 0.5004, respectively; Table I). In addition, the AUC values were only 0.6000 and 0.5286 for CDO1 and CELF4 methylation, respectively (Fig. 4A and B).

To better understand the diagnostic performance of these biomarkers, CTCs and lncRNAs (RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6) were combined, and the AUC value reached 0.9375 (Fig. 4C), thus indicating that CTCs and these lncRNAs had good performance in distinguishing the EC and NO-EC groups. When all three groups of tumor markers were combined, the AUC value was also 0.9375 (Fig. 4D). These results revealed that the diagnostic performance was not markedly improved after adding methylated genes.

Associations of microsatellite instability-high (MSI-H) status or tumor mutational burden (TMB) with tumor invasiveness and tumor volume

MSI-H and TMB are predictive biomarkers for immune checkpoint inhibitors (43). MSI is an indicator of DNA instability and represents a novel cascade in the carcinogenesis of EC in which MSI mutates hMSH6 (C8), increases gene instability, and leads to the accumulation of mutations in other cancer-related genes (44). The present study investigated the effects of MSI-H status and TMB on EC invasion and tumor volume. TMB was detected in 14 patients with EC, and its association with the depth of muscle infiltration (<1/2 or ≥1/2) or tumor volume (<2 cm or ≥2 cm) was evaluated. The results revealed that high TMB was not significantly correlated with muscle infiltration (P=0.4637; Fig. 5A) or tumor volume (P=0.4637; Fig. 5B). Moreover, MSI-H status was detected in 16 patients with EC, and muscle infiltration (P>0.9999; Fig. 5C) and tumor volume (P=0.1676; Fig. 5D) were not significantly correlated with MSI-H status. These findings indicated that MSI-H status and TMB were not significantly associated with tumor invasion and tumor volume in EC.

Discussion

In the bloodstream of patients with solid tumors, the ratio of CTCs to white blood cells has been reported to be 1:106−1:107, thus these cells are considered quite rare. Even so, the prognostic role of CTCs has been clearly demonstrated in numerous types of cancer (45). Magbanua et al (46) reported that the CTC trajectory pattern over the course of treatment was a good predictor of progression-free survival (PFS) and overall survival (OS). CTC counts of ≥2 and 5 per 7.5 ml have also been shown to be associated with reduced PFS and OS in patients with non-small cell lung cancer (47). Chen et al (48) reported that the CTC test accurately identified patients who were at a high risk for prostate cancer, allowing for the early intervention and effective treatment of patients. There are a number of types of sorting methods for CTCs, including the dielectrophoretic DLD method (49), the DEPArray™ system (50), emerging microfluidic technologies (51), dielectrophoretic enrichment (52) and the negative-selection enrichment method (48,53,54).

In the present study, CTCs were detected using the CytoBot® 2000 system, which works based on microscale meshes with a nanofunctionalized coating that enables the efficient capture of CTCs (55). The results revealed that the positive rate for CTCs in patients with EC was 80.43%. The AUC value between EC and NO-EC groups was 0.8872, indicating that CTC detection had a good screening performance on EC.

lncRNAs are a class of RNA transcripts that are >200 nucleotides long (56). It has been reported that some lncRNAs have specific effects on tumor screening. For example, risk scores have been obtained for lncRNAs RP4-792G4.2 and RP11-325122.2 in glioblastoma, and their scores can be used for risk assessment (57). Additionally, RP11-54H7.4 is a possible prognostic target for tongue squamous cell carcinoma (58). Furthermore, the performance of the three-lncRNA signature comprising RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 has been reported to be higher in EC than in paracancerous tissue (24). On the basis of existing studies, the present study explored whether combining more indicators could improve the performance of a diagnostic model for EC.

In the present study, the levels RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 were measured in tumor tissues (n=35) and matched paracancerous tissues (n=35). However, these three indicators did not significantly distinguish tumor tissue from paracancerous tissue (P=0.2730, 0.0517 and 0.5180). The present study further evaluated whether these three indicators were effective in distinguishing between the EC (n=35) and NO-EC (n=14) groups. Notably, the performance of these indicators in differentiating the EC group (n=35) from the NO-EC group (n=14) was good (P=0.0002, 0.0001 and P<0.0001, respectively). Therefore, the lncRNAs RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6 may be suitable for distinguishing between the EC and NO-EC groups.

Qi et al (32) reported that the CDO1/CELF4 dual-gene methylation assay had high sensitivity and specificity for AH and EC. Similarly, Krasnyi et al (42) reported that CDO1 and CDH13 gene methylation could predict early EC treatment outcomes. In the present study, methylated CDO1 and CELF4 were used to distinguish the EC group from the NO-EC group; however, the AUC values were only 0.6000 and 0.5286, respectively. Notably, when methylated CDO1 and CELF4 were added into the CTCs and lncRNAs panel, these two indicators could not improve the screening performance. The reason for this result may be only two methylated genes (CDO1 and CELF4) were assessed. In addition, due to limited cell samples, the present study could not simultaneously conduct a number of molecular biology experiments. In the future, more gene indicators could be added and next generation sequencing may be used to improve EC screening performance.

MSI-H status and TMB are cancer-related conditions (43,44). The present study investigated whether these two indicators were related to the depth of muscle infiltration or EC tumor volume; however, the results revealed no significant correlation.

In conclusion, in the differentiation between EC and NO-EC groups, the performance of the combined model comprising CTCs and three lncRNAs (RP4-616B8.5, RP11-389G6.3 and CTD-2377D24.6) was promising.

Acknowledgements

Not applicable.

Funding

The present study was funded by the Suzhou Science and Technology Plan Project (grant no. SKY2021035).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

HD, YC and JY contributed to the study design, data analysis and writing of the manuscript. BL, JW and XZ contributed to the study design and writing of the manuscript. SX and HC contributed to CTC detection and statistics. JY and XZ provided the CTC detection instruments and chips. JM and LF contributed to experimental system verification and DNA methylation detection. JZ contributed to patient clinical information arrangement and data analysis. FS and HZ contributed to sample collection, lncRNA qPCR detection and statistics. HD provided funding. HD and JY confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of The First Affiliated Hospital of Soochow University (approval no. 2021.351). The enrolled patients provided written informed consent before participation in this study, with permission for sample collection and analysis.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Li W, Xu Y, Zeng X, Tan J, Wang Y, Wu H, Li M and Yi C: Etiological relationship between lipid metabolism and endometrial carcinoma. Lipids Health Dis. 22:1162023. View Article : Google Scholar : PubMed/NCBI

2 

Kokts-Porietis RL, Elmrayed S, Brenner DR and Friedenreich CM: Obesity and mortality among endometrial cancer survivors: A systematic review and meta-analysis. Obes Rev. 22:e133372021. View Article : Google Scholar : PubMed/NCBI

3 

Wang X, Glubb DM and O'Mara TA: Dietary factors and endometrial cancer risk: A mendelian randomization study. Nutrients. 15:6032023. View Article : Google Scholar : PubMed/NCBI

4 

Makker V, MacKay H, Ray-Coquard I, Levine DA, Westin SN, Aoki D and Oaknin A: Endometrial cancer. Nat Rev Dis Primers. 7:882021. View Article : Google Scholar : PubMed/NCBI

5 

Sheikh MA, Althouse AD, Freese KE, Soisson S, Edwards RP, Welburn S, Sukumvanich P, Comerci J, Kelley J, LaPorte RE and Linkov F: USA endometrial cancer projections to 2030: Should we be concerned? Future Oncol. 10:2561–2568. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Van Wijk F, Huikeshoven F, Abdulkadir L, Ewing P and Burger C: Stage III and IV endometrial cancer: A 20-year review of patients. Int J Gynecol Cancer. 16:1648–1655. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Vermij L, Jobsen JJ, León-Castillo A, Brinkhuis M, Roothaan S, Powell ME, de Boer SM, Khaw P, Mileshkin LR, Fyles A, et al: Prognostic refinement of NSMP high-risk endometrial cancers using oestrogen receptor immunohistochemistry. Br J Cancer. 128:1360–1368. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Crosbie EJ, Kitson SJ, McAlpine JN, Mukhopadhyay A, Powell ME and Singh N: Endometrial cancer. Lancet. 399:1412–1428. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Tian Y and Luo H: Diagnostic accuracy of transvaginal ultrasound examination for local staging of cervical cancer: A systematic review and meta-analysis. Med Ultrason. 24:348–355. 2022.PubMed/NCBI

10 

Elmstrøm-Christensen LB and Lauszus FF: Diagnostic delay of gynaecological cancer in women with postmenopausal bleeding. Dan Med J. 69:A092107442022.PubMed/NCBI

11 

Oaknin A, Bosse TJ, Creutzberg CL, Giornelli G, Harter P, Joly F, Lorusso D, Marth C, Makker V, Mirza MR, et al: Endometrial cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol. 33:860–877. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Tronconi F, Nero C, Giudice E, Salutari V, Musacchio L, Ricci C, Carbone MV, Ghizzoni V, Perri MT, Camarda F, et al: Advanced and recurrent endometrial cancer: State of the art and future perspectives. Crit Rev Oncol Hematol. 180:1038512022. View Article : Google Scholar : PubMed/NCBI

13 

Vermij L, Smit V, Nout R and Bosse T: Incorporation of molecular characteristics into endometrial cancer management. Histopathology. 76:52–63. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Kasius JC, Pijnenborg JMA, Lindemann K, Forsse D, van Zwol J, Kristensen GB, Krakstad C, Werner HMJ and Amant F: Risk stratification of endometrial cancer patients: FIGO stage, biomarkers and molecular classification. Cancers (Basel). 13:58482021. View Article : Google Scholar : PubMed/NCBI

15 

Kiss I, Kolostova K, Pawlak I and Bobek V: Circulating tumor cells in gynaecological malignancies. J BUON. 25:40–50. 2020.PubMed/NCBI

16 

Hu X, Zang X and Lv Y: Detection of circulating tumor cells: Advances and critical concerns. Oncol Lett. 21:4222021. View Article : Google Scholar : PubMed/NCBI

17 

Castro-Giner F and Aceto N: Tracking cancer progression: From circulating tumor cells to metastasis. Genome Med. 12:312020. View Article : Google Scholar : PubMed/NCBI

18 

Law KS, Huang CE and Chen SW: Detection of circulating tumor cell-related markers in gynecologic cancer using microfluidic devices: A pilot study. Int J Mol Sci. 24:23002023. View Article : Google Scholar : PubMed/NCBI

19 

Lin W, Zhou Q, Wang CQ, Zhu L, Bi C, Zhang S, Wang X and Jin H: LncRNAs regulate metabolism in cancer. Int J Biol Sci. 16:1194–1206. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Park EG, Pyo SJ, Cui Y, Yoon SH and Nam JW: Tumor immune microenvironment lncRNAs. Brief Bioinform. 23:bbab5042022. View Article : Google Scholar : PubMed/NCBI

21 

Tan YT, Lin JF, Li T, Li JJ, Xu RH and Ju HQ: LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun (Lond). 41:109–120. 2021. View Article : Google Scholar : PubMed/NCBI

22 

Xing C, Sun SG, Yue ZQ and Bai F: Role of lncRNA LUCAT1 in cancer. Biomed Pharmacother. 134:1111582021. View Article : Google Scholar : PubMed/NCBI

23 

Zhang G, Sun J and Zhang X: A novel cuproptosis-related LncRNA signature to predict prognosis in hepatocellular carcinoma. Sci Rep. 12:113252022. View Article : Google Scholar : PubMed/NCBI

24 

Ding H, Jiang F, Deng L, Wang J, Wang P, Ji M, Li J, Shi W, Pei Y, Li J, et al: Prediction of clinical outcome in endometrial carcinoma based on a 3-lncRNA signature. Front Cell Dev Biol. 9:8144562021. View Article : Google Scholar : PubMed/NCBI

25 

Xin W, Gao X, Zhao S, Zhao P, Yu H, Wu Q and Hua K: LncRNA RP11-395G23.3 suppresses the endometrial cancer progression via regulating microRNA-205-5p/PTEN axis. Am J Transl Res. 12:4422–4433. 2020.PubMed/NCBI

26 

Esteller M: Aberrant DNA methylation as a cancer-inducing mechanism. Annu Rev Pharmacol Toxicol. 45:629–656. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Nishiyama A and Nakanishi M: Navigating the DNA methylation landscape of cancer. Trends Genet. 37:1012–1027. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Wang Q, Xiong F, Wu G, Liu W, Chen J, Wang B and Chen Y: Gene body methylation in cancer: Molecular mechanisms and clinical applications. Clin Epigenetics. 14:1542022. View Article : Google Scholar : PubMed/NCBI

29 

Papanicolau-Sengos A and Aldape K: DNA methylation profiling: An emerging paradigm for cancer diagnosis. Annu Rev Pathol. 17:295–321. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Jamshidi A, Liu MC, Klein EA, Venn O, Hubbell E, Beausang JF, Gross S, Melton C, Fields AP, Liu Q, et al: Evaluation of cell-free DNA approaches for multi-cancer early detection. Cancer Cell. 40:1537–1549.e12. 2022. View Article : Google Scholar : PubMed/NCBI

31 

Caplakova V, Babusikova E, Blahovcova E, Balharek T, Zelieskova M and Hatok J: DNA methylation machinery in the endometrium and endometrial cancer. Anticancer Res. 36:4407–4420. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Qi B, Sun Y, Lv Y, Hu P, Ma Y, Gao W, Li S, Zhang X, Jin X, Liou Y, et al: Hypermethylated CDO1 and CELF4 in cytological specimens as triage strategy biomarkers in endometrial malignant lesions. Front Oncol. 13:12893662023. View Article : Google Scholar : PubMed/NCBI

33 

Wang L, Dong L, Xu J, Guo L, Wang Y, Wan K, Jing W, Zhao L, Feng X, Zhang K, et al: Hypermethylated CDO1 and ZNF454 in cytological specimens as screening biomarkers for endometrial cancer. Front Oncol. 12:7146632022. View Article : Google Scholar : PubMed/NCBI

34 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Lawrence R, Watters M, Davies CR, Pantel K and Lu YJ: Circulating tumour cells for early detection of clinically relevant cancer. Nat Rev Clin Oncol. 20:487–500. 2023. View Article : Google Scholar : PubMed/NCBI

36 

Yao H, Wen L, Li Z and Xia C: Analysis of diagnostic value of CTC and CTDNA in early lung cancer. Cell Mol Biol (Noisy-le-grand). 69:57–62. 2023. View Article : Google Scholar

37 

Francini S, Duraes M, Rathat G, Macioce V, Mollevi C, Pages L, Ferrer C, Cayrefourcq L and Alix-Panabières C: Circulating tumor cell detection by liquid biopsy during early-stage endometrial cancer surgery: A pilot study. Biomolecules. 13:4282023. View Article : Google Scholar : PubMed/NCBI

38 

Li Y, Fan Z, Meng Y, Liu S and Zhan H: Blood-based DNA methylation signatures in cancer: A systematic review. Biochim Biophys Acta Mol Basis Dis. 1869:1665832023. View Article : Google Scholar : PubMed/NCBI

39 

Harada H, Hosoda K, Moriya H, Mieno H, Ema A, Ushiku H, Washio M, Nishizawa N, Ishii S, Yokota K, et al: Cancer-specific promoter DNA methylation of cysteine dioxygenase type 1 (CDO1) gene as an important prognostic biomarker of gastric cancer. PLoS One. 14:e02148722019. View Article : Google Scholar : PubMed/NCBI

40 

Kong LH, Xiao XP, Wan R, Chao XP, Chen XJ, Wang J, Wu HW and Li L: The role of DNA methylation in the screening of endometrial cancer in postmenopausal women. Zhonghua Yi Xue Za Zhi. 103:907–912. 2023.(In Chinese). PubMed/NCBI

41 

Huang RL, Su PH, Liao YP, Wu TI, Hsu YT, Lin WY, Wang HC, Weng YC, Ou YC, Huang TH and Lai HC: Integrated epigenomics analysis reveals a DNA methylation panel for endometrial cancer detection using cervical scrapings. Clin Cancer Res. 23:263–272. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Krasnyi AM, Gadzhieva LT, Kokoeva DN, Kosenko MG, Yarotskaya EL, Pavlovich SV, Ashrafyan LA and Sukhikh GT: Analysis of CDO1, PITX2, and CDH13 gene methylation in early endometrial cancer for prediction of medical treatment outcomes. Int J Mol Sci. 25:48922024. View Article : Google Scholar : PubMed/NCBI

43 

Salem ME, Bodor JN, Puccini A, Xiu J, Goldberg RM, Grothey A, Korn WM, Shields AF, Worrilow WM, Kim ES, et al: Relationship between MLH1, PMS2, MSH2 and MSH6 gene-specific alterations and tumor mutational burden in 1057 microsatellite instability-high solid tumors. Int J Cancer. 147:2948–2956. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Kawaguchi M, Banno K, Yanokura M, Kobayashi Y, Kishimi A, Ogawa S, Kisu I, Nomura H, Hirasawa A, Susumu N and Aoki D: Analysis of candidate target genes for mononucleotide repeat mutation in microsatellite instability-high (MSI-H) endometrial cancer. Int J Oncol. 35:977–982. 2009.PubMed/NCBI

45 

Vasseur A, Kiavue N, Bidard FC, Pierga JY and Cabel L: Clinical utility of circulating tumor cells: An update. Mol Oncol. 15:1647–1666. 2021. View Article : Google Scholar : PubMed/NCBI

46 

Magbanua MJM, Hendrix LH, Hyslop T, Barry WT, Winer EP, Hudis C, Toppmeyer D, Carey LA, Partridge AH, Pierga JY, et al: Serial analysis of circulating tumor cells in metastatic breast cancer receiving first-line chemotherapy. J Natl Cancer Inst. 113:443–452. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Krebs MG, Sloane R, Priest L, Lancashire L, Hou JM, Greystoke A, Ward TH, Ferraldeschi R, Hughes A, Clack G, et al: Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol. 29:1556–1563. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Chen J, Xie T, Yang J, Lin X, Huang L, Su S and Deng J: Feasibility study of expressing epcam +/vimentin + CTC in prostate cancer diagnosis. J Cancer Res Clin Oncol. 149:8699–8709. 2023. View Article : Google Scholar : PubMed/NCBI

49 

Rahmati M and Chen X: Separation of circulating tumor cells from blood using dielectrophoretic DLD manipulation. Biomed Microdevices. 23:492021. View Article : Google Scholar : PubMed/NCBI

50 

Di Trapani M, Manaresi N and Medoro G: DEPArray system: An automatic image-based sorter for isolation of pure circulating tumor cells. Cytometry A. 93:1260–1266. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Wei X, Chen K, Guo S, Liu W and Zhao XZ: Emerging microfluidic technologies for the detection of circulating tumor cells and fetal nucleated red blood cells. ACS Appl Bio Mater. 4:1140–1155. 2021. View Article : Google Scholar : PubMed/NCBI

52 

S Iliescu F, Sim WJ, Heidari H, P Poenar D, Miao J, Taylor HK and Iliescu C: Highlighting the uniqueness in dielectrophoretic enrichment of circulating tumor cells. Electrophoresis. 40:1457–1477. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Burr R, Edd JF, Chirn B, Mishra A, Haber DA, Toner M and Maheswaran S: Negative-selection enrichment of circulating tumor cells from peripheral blood using the microfluidic CTC-iChip. Methods Mol Biol. 2471:309–321. 2022. View Article : Google Scholar : PubMed/NCBI

54 

Andree KC, van Dalum G and Terstappen LW: Challenges in circulating tumor cell detection by the CellSearch system. Mol Oncol. 10:395–407. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Wang J, Dallmann R, Lu R, Yan J and Charmet J: Flow rate-independent multiscale liquid biopsy for precision oncology. ACS Sens. 8:1200–1210. 2023. View Article : Google Scholar : PubMed/NCBI

56 

Chi Y, Wang D, Wang J, Yu W and Yang J: Long non-coding RNA in the pathogenesis of cancers. Cells. 8:10152019. View Article : Google Scholar : PubMed/NCBI

57 

Paul Y, Thomas S, Patil V, Kumar N, Mondal B, Hegde AS, Arivazhagan A, Santosh V, Mahalingam K and Somasundaram K: Genetic landscape of long noncoding RNA (lncRNAs) in glioblastoma: Identification of complex lncRNA regulatory networks and clinically relevant lncRNAs in glioblastoma. Oncotarget. 9:29548–29564. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Zhang M, Chen Z, Zhang S, Wu L, Jie Y, Liao Y, Huang Y, Chen J and Shi B: Analysis of differentially expressed long non-coding RNAs and the associated TF-mRNA network in tongue squamous cell carcinoma. Front Oncol. 10:14212020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 28 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ding H, Wang J, Zhao X, Xiu S, Cai H, Ma J, Fu L, Zhou J, Shen F, Zhang H, Zhang H, et al: Combination of circulating tumor cells, lncRNAs and DNA methylation for the diagnosis of endometrial carcinoma. Oncol Lett 28: 545, 2024.
APA
Ding, H., Wang, J., Zhao, X., Xiu, S., Cai, H., Ma, J. ... Yan, J. (2024). Combination of circulating tumor cells, lncRNAs and DNA methylation for the diagnosis of endometrial carcinoma. Oncology Letters, 28, 545. https://doi.org/10.3892/ol.2024.14678
MLA
Ding, H., Wang, J., Zhao, X., Xiu, S., Cai, H., Ma, J., Fu, L., Zhou, J., Shen, F., Zhang, H., Chen, Y., Li, B., Yan, J."Combination of circulating tumor cells, lncRNAs and DNA methylation for the diagnosis of endometrial carcinoma". Oncology Letters 28.5 (2024): 545.
Chicago
Ding, H., Wang, J., Zhao, X., Xiu, S., Cai, H., Ma, J., Fu, L., Zhou, J., Shen, F., Zhang, H., Chen, Y., Li, B., Yan, J."Combination of circulating tumor cells, lncRNAs and DNA methylation for the diagnosis of endometrial carcinoma". Oncology Letters 28, no. 5 (2024): 545. https://doi.org/10.3892/ol.2024.14678