Next Article in Journal
Reply to Mazzei et al. Some Concerns from a Radiological Point of View. Comment on “Huang et al. Outcomes of Conversion Surgery for Metastatic Gastric Cancer Compared with In-Front Surgery plus Palliative Chemotherapy or In-Front Surgery Alone. J. Pers. Med. 2022, 12, 555”
Previous Article in Journal
Atlas of PD-L1 for Pathologists: Indications, Scores, Diagnostic Platforms and Reporting Systems
 
 
jpm-logo
Article Menu

Article Menu

Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Protocol

An Integrated Approach for the Early Detection of Endometrial and Ovarian Cancers (Screenwide Study): Rationale, Study Design and Pilot Study

by
Paula Peremiquel-Trillas
1,2,3,†,
Sonia Paytubi
1,†,
Beatriz Pelegrina
1,†,
Jon Frias-Gomez
1,2,3,
Álvaro Carmona
1,
José Manuel Martínez
4,
Javier de Francisco
5,
Yolanda Benavente
1,3,
Marc Barahona
4,
Ferran Briansó
6,7,
Júlia Canet-Hermida
8,9,
Víctor Caño
5,
August Vidal
9,10,
Alba Zanca
10,
Núria Baixeras
10,
Axel Rodríguez
10,
Sergi Fernández-Gonzalez
4,
Núria Dueñas
8,9,
Laura Càrdenas
11,
Álvaro Aytés
12,
Ilaria Bianchi
13,
Miquel Àngel Pavón
1,3,
Jaume Reventós
14,
Gabriel Capellà
8,9,
David Gómez
1,
Mireia Diaz
1,3,
Jordi Ponce
4,
Joan Brunet
8,9,15,
Xavier Matias-Guiu
9,10,
Francesc Xavier Bosch
1,3,16,
Silvia de Sanjosé
3,17,†,
Laia Alemany
1,3,†,
Marta Pineda
8,9,†,
Fátima Marin
8,9,† and
Laura Costas
1,3,*
add Show full author list remove Hide full author list
1
Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, l’Hospitalet de Llobregat, 08908 Barcelona, Spain
2
Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain
3
Consortium for Biomedical Research in Epidemiology and Public Health-CIBERESP, Carlos III Institute of Health, 28029 Madrid, Spain
4
Department of Gynecology, IDIBELL, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08908 Barcelona, Spain
5
Department of Anesthesiology, IDIBELL, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, 08908 Barcelona, Spain
6
Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, 08028 Barcelona, Spain
7
Roche Diagnostics, Sant Cugat del Vallès, 08174 Barcelona, Spain
8
Hereditary Cancer Group, ONCOBELL Program, Catalan Institute of Oncology, IDIBELL, L’Hospitalet, 08908 Barcelona, Spain
9
Consortium for Biomedical Research in Cancer–CIBERONC, Carlos III Institute of Health, 28029 Madrid, Spain
10
Department of Pathology, IDIBELL, Hospitalet de Llobregat, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
11
Department of Gynecology and Obstetrics, Hospital Universitari Dr. Josep Trueta, 17007 Girona, Spain
12
Program against Cancer Therapeutic Resistance (ProCURE), IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain
13
ASSIR Delta, Serveis d’Atenció Primària Delta del Llobregat, Direcció d’Atenció Primària Costa de Ponent, Gerència Territorial Metropolitana Sud, Institut Català de la Salut, 08006 Barcelona, Spain
14
Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
15
Medical Oncology Department, Catalan Institute of Oncology, Doctor Josep Trueta Girona University Hospital, 17007 Girona, Spain
16
Universitat Oberta de Catalunya, 08018 Barcelona, Spain
17
Consultant, National Cancer Institute, Bethesda, MA 20814, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Pers. Med. 2022, 12(7), 1074; https://doi.org/10.3390/jpm12071074
Submission received: 30 May 2022 / Revised: 23 June 2022 / Accepted: 27 June 2022 / Published: 29 June 2022
(This article belongs to the Section Epidemiology)

Abstract

:
Screenwide is a case-control study (2017–2021) including women with incident endometrial and ovarian cancers (EC and OC), BRCA1/2 and MMR pathogenic variant carriers, and age-matched controls from three centers in Spain. Participants completed a personal interview on their sociodemographic factors, occupational exposure, medication, lifestyle, and medical history. We collected biological specimens, including blood samples, self-collected vaginal specimens, cervical pap-brush samples, uterine specimens, and, when available, tumor samples. The planned analyses included evaluation of the potential risk factors for EC/OC; evaluation of molecular biomarkers in minimally invasive samples; evaluation of the cost-effectiveness of molecular tests; and the generation of predictive scores to integrate different epidemiologic, clinical, and molecular factors. Overall, 182 EC, 69 OC, 98 BRCA pathogenic variant carriers, 104 MMR pathogenic variant carriers, and 385 controls were enrolled. The overall participation rate was 85.7%. The pilot study using 61 samples from nine EC cases and four controls showed that genetic variants at the variant allele fraction > 5% found in tumors (n = 61 variants across the nine tumors) were detected in paired endometrial aspirates, clinician-collected cervical samples, and vaginal self-samples with detection rates of 90% (55/61), 79% (48/61), and 72% (44/61) by duplex sequencing, respectively. Among the controls, only one somatic mutation was detected in a cervical sample. We enrolled more than 800 women to evaluate new early detection strategies. The preliminary data suggest that our methodological approach could be useful for the early detection of gynecological cancers.

Graphical Abstract

1. Introduction

Endometrial cancer (EC) is the most common gynecological tumor in very high human development index regions, and is the second most common globally [1]. Ovarian cancer (OC) is the most lethal gynecological cancer due to diagnosis at an advanced stage [1]. Worldwide, the burden of both tumors is expected to increase in the following years [2,3]. A genetic susceptibility component exists for both tumors: Lynch syndrome (MMR pathogenic variant carriers) is strongly associated with an increased risk of EC, to a lesser extent of OC, while BRCA pathogenic variant carriers have a higher risk of developing OC than the general population [4,5]. Estrogens play a relevant role in EC etiology. Nulliparity, infertility, and age at last birth have been repeatedly associated with EC [6,7,8]. Obesity is also an established risk factor of EC; among women, EC is the most consistently associated with a high body mass index (BMI) [9]. Obesity may increase the risk of EC by a variety of mechanisms, including the conversion of androgens to estrogens via the aromatase activity [10]. OC is also associated with hormonal factors, such as nulliparity, early-onset menarche, late-onset menopause, and the use of hormonal treatments [4,11]. Other factors that interact with sex hormones could potentially modify the risk of EC and OC. Night shift work has been associated with higher concentrations of sex hormones [12], but the role night shift plays in EC and OC is still unknown, with few and discordant results [13,14,15,16,17,18]. Identifying modifiable risk factors for EC and OC is relevant from a public health perspective.
EC arises from malignant transformation of the endometrium via the development of precancerous lesions such as atypical hyperplasia/endometrioid intraepithelial neoplasia (AH/EIN), endometrial glandular dysplasia (EmGD), or serous endometrial intraepithelial carcinoma (SEIC) [19,20]. However, the current paradigm in the genesis of high-grade serous carcinoma, the most common ovarian cancer, consists of a tubal origin of precancerous lesions, such as serous tubal intraepithelial carcinoma (STIC) [21,22]. Theoretically, precancer cells should be molecularly different from the normal endometrium, show a monoclonal growth pattern, and share some but not all features of a malignant endometrium [23]. Around 90% of EC serous cancers show somatic pathogenic variants in TP53 [24], and these variants have been identified in 43% and 72% of EmGD and SEIC, respectively [25]. Complex AH/EIN shows genetic changes frequently observed in type I endometrial cancer, including microsatellite instability and pathogenic variants in PTEN, PIK3CA, KRAS, and CTNNB1 [26,27]. Mathematical models suggest that it takes decades for a TP53 pathogenic variant to develop into a STIC, followed by a shorter span of few years for progression to OC [22].
Currently, no convincing approaches provide the necessary accuracy to be introduced as EC or OC screening tests for the general population. Available strategies to detect EC and OC rely on the presence of symptoms. EC presents abnormal bleeding in up to 90% of cases at diagnosis, allowing for early detection of the disease. Contrary, the broad range of OC symptoms leads to delayed diagnosis in many of the cases, leading to an unfavorable prognosis. Therefore, considerable efforts have been made to implement general population screening to diagnose OC early on, using tools like the transvaginal ultrasound and biomarkers such as serum cancer antigen (CA125), with no significant mortality reductions [28]. Among high-risk populations, such as women with hereditary susceptibility, annual surveillance is recommended until prophylactic surgery, although screening methods are associated with significant pain and distress and include an annual endometrial biopsy. New molecular tests may help refine current diagnostic algorithms among EC symptomatic women by improving the performance and failure rate of histological diagnosis, which currently limits the success of endometrial aspirate-based diagnosis. Accurate non-invasive tests would be especially helpful in screening settings and in high-risk populations, as these suboptimal methods are still used to intensively screen asymptomatic women with a family history of cancer.
The anatomical continuity of the uterine cavity with the cervix represents a unique opportunity to detect signs of disease in the upper genital tract using material from routine cervical pap brush samples (cervical cytology) and other non-invasive sampling methods [29]. Thus, cervical cytology is repeatedly recognized as a potential source of information about gynecological tumors, and molecular approaches (genomic, epigenomic, and proteomic) have been evaluated to detect EC and OC with considerable sensitivity and specificity [30,31,32]. Other minimally invasive methods to detect EC/OC using molecular approaches have been evaluated, such as blood samples, tampons, and vaginal self-samples, with promising results [33,34,35]. These recent findings offer an exciting perspective on the early detection of EC/OC. However, some aspects still need to be assessed to accelerate the implementation of novel technologies in a routine screening or clinical setting. Health economics models, such as cost-effectiveness analyses and budget impact analyses, can help identify the most efficient preventive approaches and inform decision-makers about what screening and early detection strategies may be included in the healthcare plans.
In this context, the Screenwide study was launched in 2017 to evaluate epidemiologic, serological, and genomic factors in the diagnosis of EC and OC for comprehensive primary and secondary prevention of gynecologic cancer in women. Specific objectives include evaluating modifiable risk factors using data from personal questionnaires and from blood biomarkers; evaluating genomic biomarkers in cervical pap samples and vaginal self-collected samples in women with EC/OC and high-risk populations compared with women without cancer; estimating the cost-effectiveness of the introduction of molecular tests at the population level; and generating scores to integrate different epidemiologic, clinical, and molecular factors, to predict the individualized risk of cancer. We show here the enrolment protocols and the methodology used to evaluate genomic biomarkers in minimally invasive samples, and we present the results from the pilot study using 61 samples from nine EC cases and four controls.

2. Materials and Methods

Screenwide is a prospective case-control study. Enrolment started in 2017 and finished in 2021. Inclusion criteria included having an intact uterus and, for cases, having an incident diagnosis of EC/OC. Consecutive cases were enrolled during the study period. Gynecologic benign conditions included endometriosis, fibroids, benign cysts, prolapse, and polyps. Hospital controls without gynecologic conditions were enrolled at the anesthetic visits for surgery for conditions, such as ophthalmic or traumatology diseases. High-risk populations without EC/OC were informed about the study at the hereditary cancer clinics and were enrolled during their annual gynecologic check-ups. Exclusion criteria included pregnancy, puerperium (8 weeks), prior treatment with chemotherapy and radiotherapy during the previous 6 months, and communication difficulties that precluded signing informed consent and answering the questionnaire, such as not understanding Spanish or having an intellectual disability. Participants were enrolled in the Bellvitge University Hospital, Josep Trueta University Hospital Catalan Institute of Oncology, and the Sexual and Reproductive Health Care (ASSIR) Delta.

2.1. Epidemiological and Clinical Questionnaires

Epidemiological and clinical questionnaires were designed to gather and cover previous existing knowledge on EC and OC. The data collection and entry were regularly monitored for quality control purposes. A structured epidemiological questionnaire was administered by trained personnel in personal interviews. The questionnaire included basic epidemiologic information such as demographic factors, tobacco consumption, lifetime occupational history (including working night shifts), coffee consumption, physical activity, family history of cancer, anthropometric factors, reproductive factors and exogenous hormone use, sun exposure, sleeping habits, and chronotype information.
Clinical data were extracted from the electronic medical records using a predefined form. The information collected included endometrial thickness measured by transvaginal ultrasound, symptomatology (such as abdominal pain, postmenopausal bleeding, or metrorrhagia), tumor stage and grade, tumor type, presence of lymph node invasion, CA-125 levels, treatments received and their duration, and follow-up data on potential relapses.

2.2. Biological Samples and Histopathological Examination

Sample collection included blood samples, vaginal samples, cervical pap brush samples, endometrial aspirates, and, when available, tumor samples (Table 1). Gynecological samples were performed in the following order: (1) vaginal sampling, (2) cervical pap brush samples, and (3) endometrial cytologies/aspirates. First, 30 ml of peripheral blood was drawn from participants using an EDTA BD Vacutainer® K2E and SSTTM II Advance BD Vacutainer®. The samples® (Becton, Dickinson and Company, Franklin Lakes, NJ, USA). Samples were centrifuged for 15 min at 2500 rpm and the different fractions were aliquoted in whole blood, plasma, cellular fraction for DNA isolation (buffy coat), and serum and were stored at −80 °C. Vaginal samples were collected using the Evalyn brush self-sampling device (Rovers® Medical Devices, The Netherlands) device and were suspended in 5 mL of liquid-based cytology solution (ThinPrep PreservCyt®, Hologic, Bedford, MA, USA). Cervical cytologies were collected by the gynecologist using a Cervex brush (Rovers® Medical Devices, The Netherlands) and were suspended in 20 mL of ThinPrep liquid-based solution, as performed in the regular cervical cancer screening program in Catalonia. Once processed, both samples were aliquoted in three vials and were kept at room temperature, as indicated in the manufacturer’s protocol. Some of the vaginal samples and some cytologies were stored at −80 °C for research purposes. Endometrial aspirates were collected by the gynecologist using a pipelle cannula. Half of the sample was formalin-fixed paraffin-embedded for pathology examination, and the remaining fresh sample was frozen at −80 °C. The tumor samples were collected during surgical treatment. Fresh frozen tissues were stored at −80 °C and were formalin-fixed paraffin-embedded and stored at room temperature.
The cervical cytologies, aspirates, and tumor samples were examined by pathologists at the Bellvitge University Hospital and Josep Trueta University Hospital. Pathologic analysis of the hysterectomy and salpingo-oophorectomy specimens were performed in order to (1) confirm the diagnosis of cancer; (2) select different areas for molecular analysis; and (3) check the presence of pathological abnormalities, such as cervical stenosis. Histological evaluation of all of the uterine tissue samples was performed and classified according to the WHO criteria and was staged and graded according to the FIGO classification.

2.3. Genomic Biomarkers in Minimally Invasive Sampling Methods—Pilot Study

An analysis of the variants in EC and OC from The Cancer Genome Atlas (TCGA) dataset [36] was performed and, using this along with information from previous literature, we constructed a panel of exonic regions and intron-exon boundaries of 49 genes. In particular, we selected exonic regions from 13 genes that achieved >95% sensitivity to detect EC (PTEN, TP53, PIK3CA, and ARID1A, among others), exonic regions from 14 genes that contained frequent point variants (PPP2R1A, RPL22, SETD1B, and RNF43, among others), exonic regions from 12 genes including variants among less common histologies (serous cancers; PIK3R1, LZTR1, AP4E1, ARHGAP35, among others), and exonic regions from 10 genes selected according to previous literature and our TCGA analyses on ovarian cancer (unpublished; PAX2, AKT1, APC, and BRAF, among others). The panel estimated size was 247 kb, and the reference genome used to design the SeqCap EZ probe pool (NimbleGen, Roche) was GRCh38 (hg38) from Homo sapiens. Each gene was classified as a tumor suppressor, oncogene, and ambiguous or not driver according to IntOGen [37]. Preparation of the genomic DNA libraries was performed according to the manufacturer’s recommendations, with certain variations to add unique molecular identifiers (UMIs) for duplex sequencing. Target enrichment for next-generation sequencing using SeqCap EZ probes was performed. The deduplication process was performed using a combination of Picard, fgbio, and bwa tools. VarDictJava and Mutect2, in tumor-only mode, were used for variant calling. Variants detected by both callers and with variant allele frequency (VAF) > 0.5% were considered and filtered by quality and functional impact (all non-synonymous and consensus splicing variants were retained). A minimum VAF of 5% was set in aspirates to filter out the low frequency variants of normal tissue. Additional details are provided in the Supplementary Material file S1.

2.4. Statistical and Cost-Effectiveness Analyses

Self-reported data from the epidemiological questionnaires were analyzed to evaluate potential novel risk factors, such as working night shift, early life BMI, and sun exposure. Logistic regression models were adjusted for potential confounders to estimate the odds ratios (OR) and 95% confidence intervals (CI). An algorithm to calculate a predictive score was developed based on regression models, including epidemiologic, clinic, and molecular variables. The contribution of statistically significant variables was determined on a scale of 1 to 10, according to the methodology of Sullivan et al., 2004 [38].
Cost-effectiveness analyses using Markov models were performed to evaluate the best preventive approaches in order to implement different screening and early detection strategies. Different models were designed to assess the impact of the introduction of genomic biomarkers in minimally invasive sampling methods among symptomatic women, women with inherited susceptibility, and the general population. The different strategies were compared using the incremental cost-effectiveness ratio (ICER), expressed as the ratio of the difference in costs (€) between strategies to the difference in effectiveness (QALY). The analyses were performed from the healthcare system perspective. Deterministic and probabilistic sensitivity analyses were carried out to determine the robustness of the results.

2.5. Ethical Approval

Screenwide followed all the requirements established by the Ethics Committee for Clinical Research and was approved by the Ethics Committee for Clinical Research from the Bellvitge University Hospital (references: PR128/16 and PR348/19). Participation in the study was voluntary, and all eligible subjects signed an informed consent form after receiving information about the study, before participating in any intervention. The Screenwide study followed the national and international directives on ethics and data protection (Declaration of Helsinki and subsequent amendments; EU Reglament 2016/679) and the Spanish laws on data protection (Organic Law 3/2018; Law 14/2007 biomedical research). The study was registered in the National Register of Biobanks/Collections (C.0004389).

3. Results

3.1. Overall Enrollment

In total, 838 subjects were enrolled in the study (Table 1), including 251 incident cancer cases (182 EC and 69 OC), 385 controls (119 asymptomatic women in cervical cancer screening programs, 190 hospital controls with benign gynecological pathology, and 76 women attending hospital for non-gynecological diseases frequency-matched to cases by age), and 202 high-risk participants without cancer (98 BRCA and 104 MMR mutation carriers). Participation rates were calculated using the women accepting participation in the numerator and all subjects, including refusals, in the denominator [39]. The response rates among cases were 89.7% for EC and 86.3% for OC. Among the controls, the response rate was 96.7% for asymptomatic women attending cervical cancer screening programs, 80.5% for patients with benign gynecological pathology, and 76.8% for asymptomatic women attending hospital for non-gynecological diseases. Among high-risk populations (BRCA and MMR mutation carriers), the response rate was 85.2% in both groups. Thirty cases (12%) and twenty-one controls (5%) had a previous history of cancer, other than EC or OC. The calculated power was ≥80% to detect associations with a prevalence of exposure among controls between 0.2 and 0.7, and odds ratios of ≥1.8, with the given sample size of 182 cases and 385 controls, or between 0.3 and 0.6 for 182 cases and 266 controls.
In Table 2, we present the main characteristics of the study population. In summary, EC cases were more likely to have a BMI ≥ 30 than the controls (p < 0.001), and significant differences were also observed in the distribution of menopausal status and hormonal contraception (p < 0.001 and 0.002, respectively, Table 2). Pathologic assessment of cervical pap tests slides was performed. We observed that 26% of slides among EC cases were abnormal [40], while 7% of slides among OC cases were abnormal (contained atypical or malignant cells).

3.2. Pilot Study to Detect Somatic Variants in Minimally Invasive Samples

Patients: A total of 61 samples (13 blood samples, 13 vaginal self-collected samples, 13 cervical clinician-collected samples, 13 endometrial aspirates, and 9 tumor samples) were obtained from 13 postmenopausal women (4 controls and 9 sporadic EC cases). The median age was 74 years for cases (range 54–94 years) and 75 for controls (range 58–92). All tumors were histologically confirmed, five cases had endometrioid histology, two cases were serous carcinoma, one was a neuroendocrine tumor (with an endometrioid component), and one was a carcinosarcoma (Supplementary Figure S1). Four cases were FIGO stage I, four cases were stage III, and one case was stage IV.
Target panel sequencing metrics: The mean raw coverage was 13.477X and final coverage after deduplication and filtering was 868X (Supplementary Table S1). No differences were observed between metrics by type of sample (tumor, aspirate biopsy, pap brush sample, and self-sample, p > 0.05 for all comparisons).
Surgical specimens: Altogether, 61 variants at VAF > 5% were identified in the tumor samples (Figure 1). All of the tumors harbored at least one known or predicted driver variants of EC (Supplementary Table S2). Overall, 17% of variants were identified as known drivers, 52% as predicted drivers, and 31% as passengers. PTEN was the gene most mutated in six out of nine EC surgical specimens, followed by ARID1A (4/9) and CDH4 (4/9). Other known driver variants were also found in other genes such as TP53, PIK3CA, or CTNNB1.
Endometrial aspirates: Overall, a total of 58 variants were identified in the endometrial aspirates from EC cases with VAF > 5%, and 55 of them (55/61, 90%) were previously found in paired tumors from surgical specimens (Table 3 and Table S3). The VAF distributions of the variants in the aspirates were similar to those in the surgical specimens (p > 0.05; Figure 2). No variants were detected in the aspirates from the controls (Table 3).
Minimally invasive samples: No variants were detected in one pap brush sample (endometrioid histology) and two vaginal self-samples (endometrioid and serous histologies) from the cases (Table 3, Supplementary Figure S1). Genetic variants found in tumors at VAF > 5% were detected in paired clinician-collected cervical samples and vaginal self-samples with detection rates of 79% (48/61) and 72% (44/61) through duplex sequencing, respectively (Figure 1). The mean VAF of somatic variants was similar in these minimally invasive samples: 6.4% (range 0.58–40.72%) in pap brush samples and 5.3% (range 0.64–33.19%) in vaginal self-samples, but significantly fewer variants were identified in the aspirates (p < 0.001, Figure 2 and Supplementary Table S4). In all minimally invasive samples from EC cases where somatic variants could be detected, at least one of them was a predicted or known cancer driver (Supplementary Table S4). The mean number of variants in the pap brush samples was 6.8 and 7.2 for stage I and stage > I, respectively, and 6.5 and 7.4, respectively, in the self-samples, although analyses by stage were limited by small sample sizes. Regarding controls, only one variant with VAF > 0.5% was detected in one pap brush sample. This variant (TP53 p.A138V) was not described as a cancer driver, a dominant-negative, nor a loss of function variant. It was not detected in the aspirate or self-sample of the same individual and no (pre)malignant lesions were found in the follow-up after 45 months, suggesting it could correspond to the somatic mutational burden of the normal endometrium [41].

4. Discussion

We have presented the protocol of the Screenwide study, which has achieved optimal recruitment based on our target population. The Screenwide study aims to comprehensively evaluate epidemiologic and molecular factors to better understand the epidemiology of EC and OC. We collected detailed epidemiologic data in order to assess new modifiable risk factors. In addition, we will examine sociodemographic, lifestyle, occupational and environmental exposures, as well as EC/OC risk in collaboration with the Epidemiology of Endometrial Cancer Consortium (E2C2). The study has the ambitious mission to characterize molecular markers for the early detection of EC/OC using minimally invasive samples, including vaginal self-collected samples and cervical pap brush samples. The preliminary data suggest that our methodological approach could be useful to evaluate new early detection strategies for gynecological cancers. Finally, our approach will integrate different epidemiologic, clinical, and molecular factors to generate a predictive score to assess individual risk of EC/OC, with the aim to translate the generated epidemiological knowledge into clinical practice. A cost-effectiveness evaluation is expected to identify scenarios that better contribute to the sustainability of healthcare systems with the introduction of these novel technologies.
EC and OC are expected to rise worldwide in the following decades and, currently, neither screening nor efficient early diagnosis approaches exist. Recent developments on non-invasive sampling methods using genomic, epigenomic, and proteomic approaches offer promising prospects for assessing this issue [29]. Vaginal self-sampling is being implemented in many settings to increase cervical cancer screening coverage and is offered as an alternative screening approach in some regions [42]. The evaluation of EC/OC markers in these specimens could provide women with a noninvasive screening option, thus avoiding pain and discomfort, as well as decreasing medical complications. Our approach described in the pilot study is useful to detect somatic variants in these gynecological minimally invasive samples at a high sensitivity. We have observed a few variants in minimally invasive samples that were not present in the tumor samples. This may be due, in part, to the mutational burden of normal endometrium [41]. Tumor heterogeneity could also contribute to this issue, as it hampers genetic characterization when a single tumor biopsy is analyzed, and genetic analyses of uterine aspirates have been shown to capture this heterogeneity [43]. However, we did not always observe concordant variants in aspirates and minimally invasive samples that were not found in tumors, suggesting that our findings might not be due to this phenomenon. We used duplex sequencing, as it has been described as the most reliable method to detect ultra-low frequency variants, and the theoretical error rate has been estimated to be around 1 × 10−7 [44]. We enriched our sample for non-endometrioid histologies (four out of nine cases, 44% rather than 10–15%) in order to ensure that we would also detect rarer histologies, which are commonly associated with a poorer prognosis. A larger sample size is required to validate these results. In this regard, we are currently sequencing more than 550 additional samples from EC, OC cases, controls, and high-risk populations, which will be split in a training (70% of the sample) and validation dataset (30% of the sample). This sample size provides power (99%) to detect ≥30% difference in the prevalence of variants. The detection of OC would probably need further adjustments of the bioinformatics pipeline, given the expected lower VAF in endometrial aspirates and minimally invasive samples compared with EC samples. The rest of the collected samples will be used to evaluate other molecular techniques (including other genomic approaches, proteomics, and epigenomics), according to the obtained results, and in collaboration with other partners. As this is an exciting and emerging field of research—novel hypotheses and further opportunities for collaboration are expected.
Because of the retrospective nature of case-control studies, we should consider that certain information obtained may be a result of the disease and not the cause of it. The variables collected in the epidemiological questionnaire are mostly self-reported, and participants may have a recall bias, which could lead to misclassification. The inclusion of controls from higher socioeconomic levels may lead to selection bias. This is especially relevant for controls from cancer screening programs. These controls are expected to contribute mostly to technological development and will be potentially excluded from etiologic factors analyses. In order to control for these possible biases, several quantitative analyses of the selection and misclassification biases of the exposure are planned with plausible parameters under a probabilistic approach. The molecular characterization of tumors allows for defining the molecular phenotype and for evaluating the specific risk factors for each molecular subtype. The available sample size allows for an adequate characterization of genomic biomarkers and permits collaboration with different related consortia, although it might be insufficient to evaluate certain associations, especially in the case of OC.

5. Conclusions

We enrolled more than 800 women to evaluate new early detection strategies. The preliminary data suggest that our methodological approach could be useful for the early detection of gynecological cancers. The Screenwide study aims to provide new evidence regarding personalized gynecologic cancer care by offering non-invasive early detection methods for women.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jpm12071074/s1. Table S1. Sequencing metrics; Table S2. Variants observed in tumour samples; Table S3. Variants observed in endometrial aspirates; Table S4. Variants observed in minimally invasive samples; Figure S1. Variant allele frequencies (VAFs) by sample type and participant. File S1: Supplemental Methods [29,45,46,47,48,49,50,51,52,53,54,55].

Author Contributions

Conceptualization, X.M.-G., J.P., J.B., F.X.B., S.d.S., L.A. and L.C. (Laura Costas); methodology, M.P., F.M., S.d.S. and L.C. (Laura Costas); software, B.P., F.M. and F.B.; formal analysis, B.P., F.M., F.B. and J.d.F.; investigation, S.P. and Á.C.; resources, J.C.-H., S.P., J.F.-G., V.C., M.B. and M.À.P.; data curation, B.P., F.M. and F.B.; writing—original draft preparation, P.P.-T., S.P., B.P., F.M. and L.C. (Laura Costas); writing—review and editing, P.P.-T., S.P., B.P., J.F.-G., Á.C., J.M.M., J.d.F., Y.B., M.B., F.B., J.C.-H., A.V., A.Z., N.B., A.R., S.F.-G., N.D., L.C. (Laura Càrdenas), Á.A., I.B., M.À.P., J.R., G.C., D.G., M.D., J.P., J.B., X.M.-G., F.X.B., S.d.S., L.A., M.P., F.M., L.C. (Laura Costas) and Screenwide Team; visualization, P.P.-T., S.P., B.P., F.M. and L.C. (Laura Costas); supervision, S.d.S., L.A. and L.C. (Laura Costas); project administration, L.C. (Laura Costas); funding acquisition, S.d.S., F.X.B., X.M.-G., J.P., Á.A., J.B. and L.C. (Laura Costas). All authors have read and agreed to the published version of the manuscript.

Funding

This work was conducted with the contribution of the Carlos III Health Institute through projects PIE16/0049, PI17/01179, PI19/01835, CM19/00216, FI20/00031, MV20/00029 and MV21/00061, as well as through CIBERESP CB06/02/0073 and CIBERONC CB16/12/00401 and CB16/12/00234, PID2019-111254RB-I00, co-financed by the European Regional Development Fund ERDF, a way to build Europe. It also counts with the support of the Secretariat for Universities and Research of the Department of Business and Knowledge of the Generalitat de Catalunya, Roche Diagnostics, AECC Grupos estables coordinados, grants to support the activities of research groups 2017SGR01085, 2017SGR01718, 2017SGR1282 and 2017SGR00735 and also with funding from the Health Department of the Generalitat de Catalunya (PERIS SLT006/17/76). We thank CERCA Programme/Generalitat de Catalunya for institutional support.

Institutional Review Board Statement

Screenwide follows all the requirements established by the Ethics Committee for Clinical Research and counts with the approval by the Ethics Committee for Clinical Research from the Bellvitge University Hospital (reference: PR128/16 and PR348/19). The Screenwide study followed the national and international directives on ethics and data protection (Declaration of Helsinki and subsequent amendments; EU Reglament 2016/679) and the Spanish laws on data protection (Organic Law3/2018; Law 14/2007 biomedical research). The study is registered in the National Register of Biobanks/Collections (C.0004389).

Informed Consent Statement

Participation in the study is voluntary, and all eligible subjects signed an informed consent form after receiving information from the study and before any intervention.

Data Availability Statement

Data can be found in supplemental material.

Acknowledgments

We thank Maria Eulalia Fernández, Dolores Martí, Dalia Mañá, Gemma Sas, Begoña Carral, Laura Fernández, Vanesa Garcia, Maite Climent, Yolanda Pérez, Maria Trinidad Álvarez Ibañez, Sara Martorell Ubeda, Mònica Salinas, Silvia Iglesias, Ares Solanes, Èlia Grau, Mònica Pardo, Vanesa Florencia, Yolanda Camon, Ana Esteban, Marleny Vergara, Glòria Oliveras, and Gema Carretero for their valuable contributions. We want to acknowledge patients and Biobank HUB-ICO-IDIBELL (PT20/00171) integrated in the ISCIII Biobanks and Biomodels Platform and Xarxa Banc de Tumors de Catalunya (XBTC) for their collaboration.

Conflicts of Interest

The authors received supplies from IDT and Roche Diagnostics at 50% discount. Laura Costas received speaking honoraria from Roche Diagnostics. Ferran Briansó has an employment relationship with Roche Diagnostics. Idibell and Roche Diagnostics signed a contract to collaborate in the development of the bioinformatics pipeline of the project. The authors do not have other conflicts of interest to declare.

References

  1. Ferlay, J.; Ervik, M.; Lam, F.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today; International Agency for Research on Cancer: Lyon, France, 2018. [Google Scholar]
  2. Lortet-Tieulent, J.; Ferlay, J.; Bray, F.; Jemal, A. International Patterns and Trends in Endometrial Cancer Incidence, 1978–2013. JNCI J. Natl. Cancer Inst. 2018, 110, 354–361. [Google Scholar] [CrossRef]
  3. Hao, W.; Zhang, Y.; Li, Z.; Zhang, E.; Gao, S.; Yin, C.; Yue, W. International Trends in Ovarian Cancer Incidence from 1973 to 2012. Arch. Gynecol. Obstet. 2021, 303, 1589–1597. [Google Scholar] [CrossRef]
  4. Lheureux, S.; Gourley, C.; Vergote, I.; Oza, A.M. Epithelial Ovarian Cancer. Lancet 2019, 393, 1240–1253. [Google Scholar] [CrossRef] [Green Version]
  5. Crosbie, E.J.; Kitson, S.J.; McAlpine, J.N.; Mukhopadhyay, A.; Powell, M.E.; Singh, N. Endometrial Cancer. Lancet 2022, 399, 1412–1428. [Google Scholar] [CrossRef]
  6. Setiawan, V.W.; Yang, H.P.; Pike, M.C.; McCann, S.E.; Yu, H.; Xiang, Y.-B.; Wolk, A.; Wentzensen, N.; Weiss, N.S.; Webb, P.M.; et al. Type I and II Endometrial Cancers: Have They Different Risk Factors? J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2013, 31, 2607–2618. [Google Scholar] [CrossRef]
  7. Yang, H.P.; Cook, L.S.; Weiderpass, E.; Adami, H.-O.; Anderson, K.E.; Cai, H.; Cerhan, J.R.; Clendenen, T.V.; Felix, A.S.; Friedenreich, C.M.; et al. Infertility and Incident Endometrial Cancer Risk: A Pooled Analysis from the Epidemiology of Endometrial Cancer Consortium (E2C2). Br. J. Cancer 2015, 112, 925–933. [Google Scholar] [CrossRef] [Green Version]
  8. Setiawan, V.W.; Pike, M.C.; Karageorgi, S.; Deming, S.L.; Anderson, K.; Bernstein, L.; Brinton, L.A.; Cai, H.; Cerhan, J.R.; Cozen, W.; et al. Age at Last Birth in Relation to Risk of Endometrial Cancer: Pooled Analysis in the Epidemiology of Endometrial Cancer Consortium. Am. J. Epidemiol. 2012, 176, 269–278. [Google Scholar] [CrossRef]
  9. Raglan, O.; Kalliala, I.; Markozannes, G.; Cividini, S.; Gunter, M.J.; Nautiyal, J.; Gabra, H.; Paraskevaidis, E.; Martin-Hirsch, P.; Tsilidis, K.K.; et al. Risk Factors for Endometrial Cancer: An Umbrella Review of the Literature. Int. J. Cancer 2019, 145, 1719–1730. [Google Scholar] [CrossRef] [Green Version]
  10. Onstad, M.A.; Schmandt, R.E.; Lu, K.H. Addressing the Role of Obesity in Endometrial Cancer Risk, Prevention, and Treatment. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 4225–4230. [Google Scholar] [CrossRef]
  11. Tanha, K.; Mottaghi, A.; Nojomi, M.; Moradi, M.; Rajabzadeh, R.; Lotfi, S.; Janani, L. Investigation on Factors Associated with Ovarian Cancer: An Umbrella Review of Systematic Review and Meta-Analyses. J. Ovarian Res. 2021, 14, 153. [Google Scholar] [CrossRef]
  12. Papantoniou, K.; Pozo, O.J.; Espinosa, A.; Marcos, J.; Castaño-Vinyals, G.; Basagaña, X.; Juanola Pagès, E.; Mirabent, J.; Martín, J.; Such Faro, P.; et al. Increased and Mistimed Sex Hormone Production in Night Shift Workers. Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol. 2015, 24, 854–863. [Google Scholar] [CrossRef] [Green Version]
  13. Viswanathan, A.N.; Hankinson, S.E.; Schernhammer, E.S. Night Shift Work and the Risk of Endometrial Cancer. Cancer Res. 2007, 67, 10618–10622. [Google Scholar] [CrossRef] [Green Version]
  14. Schwartzbaum, J.; Ahlbom, A.; Feychting, M. Cohort Study of Cancer Risk among Male and Female Shift Workers. Scand. J. Work. Environ. Health 2007, 33, 336–343. [Google Scholar] [CrossRef]
  15. Leung, L.; Grundy, A.; Siemiatycki, J.; Arseneau, J.; Gilbert, L.; Gotlieb, W.H.; Provencher, D.M.; Aronson, K.J.; Koushik, A. Shift Work Patterns, Chronotype, and Epithelial Ovarian Cancer Risk. Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol. 2019, 28, 987–995. [Google Scholar] [CrossRef] [Green Version]
  16. Bhatti, P.; Cushing-Haugen, K.L.; Wicklund, K.G.; Doherty, J.A.; Rossing, M.A. Nightshift Work and Risk of Ovarian Cancer. Occup. Environ. Med. 2013, 70, 231–237. [Google Scholar] [CrossRef] [Green Version]
  17. Poole, E.M.; Schernhammer, E.S.; Tworoger, S.S. Rotating Night Shift Work and Risk of Ovarian Cancer. Cancer Epidemiol. Biomark. Prev. 2011, 20, 934–938. [Google Scholar] [CrossRef] [Green Version]
  18. Dun, A.; Zhao, X.; Jin, X.; Wei, T.; Gao, X.; Wang, Y.; Hou, H. Association Between Night-Shift Work and Cancer Risk: Updated Systematic Review and Meta-Analysis. Front. Oncol. 2020, 10, 1006. [Google Scholar] [CrossRef]
  19. Zheng, W.; Xiang, L.; Fadare, O.; Kong, B. A Proposed Model for Endometrial Serous Carcinogenesis. Am. J. Surg. Pathol. 2011, 35, e1–e14. [Google Scholar] [CrossRef]
  20. Fadare, O.; Zheng, W. Endometrial Glandular Dysplasia (EmGD): Morphologically and Biologically Distinctive Putative Precursor Lesions of Type II Endometrial Cancers. Diagn. Pathol. 2008, 3, 6. [Google Scholar] [CrossRef] [Green Version]
  21. Labidi-Galy, S.I.; Papp, E.; Hallberg, D.; Niknafs, N.; Adleff, V.; Noe, M.; Bhattacharya, R.; Novak, M.; Jones, S.; Phallen, J.; et al. High Grade Serous Ovarian Carcinomas Originate in the Fallopian Tube. Nat. Commun. 2017, 8, 1093. [Google Scholar] [CrossRef]
  22. Shih, I.-M.; Wang, Y.; Wang, T.-L. The Origin of Ovarian Cancer Species and Precancerous Landscape. Am. J. Pathol. 2021, 191, 26–39. [Google Scholar] [CrossRef] [PubMed]
  23. Maksem, J.A.; Robboy, S.J.; Bishop, J.W.; Meiers, I. Endometrial Precancer. In Endometrial Cytology with Tissue Correlations; Essentials in Cytopathology; Springer: Boston, MA, USA, 2009; pp. 189–230. ISBN 978-0-387-89909-1. [Google Scholar]
  24. Cancer Genome Atlas Research Network; Kandoth, C.; Schultz, N.; Cherniack, A.D.; Akbani, R.; Liu, Y.; Shen, H.; Robertson, A.G.; Pashtan, I.; Shen, R.; et al. Integrated Genomic Characterization of Endometrial Carcinoma. Nature 2013, 497, 67–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Jia, L.; Liu, Y.; Yi, X.; Miron, A.; Crum, C.P.; Kong, B.; Zheng, W. Endometrial Glandular Dysplasia with Frequent P53 Gene Mutation: A Genetic Evidence Supporting Its Precancer Nature for Endometrial Serous Carcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2008, 14, 2263–2269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. van der Putten, L.J.M.; van Hoof, R.; Tops, B.B.J.; Snijders, M.P.L.M.; van den Berg-van Erp, S.H.; van der Wurff, A.A.M.; Bulten, J.; Pijnenborg, J.M.A.; Massuger, L.F.A.G. Molecular Profiles of Benign and (Pre)Malignant Endometrial Lesions. Carcinogenesis 2017, 38, 329–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Kurman, R.J. WHO Classification of Tumours of Female Reproductive Organs. In World Health Organization Classification of Tumours, 4th ed.; International Agency for Research on Cancer, World Health Organization, Eds.; International Agency for Research on Cancer: Lyon, France, 2014; ISBN 978-92-832-2435-8. [Google Scholar]
  28. Jacobs, I.J.; Menon, U.; Ryan, A.; Gentry-Maharaj, A.; Burnell, M.; Kalsi, J.K.; Amso, N.N.; Apostolidou, S.; Benjamin, E.; Cruickshank, D.; et al. Ovarian Cancer Screening and Mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): A Randomised Controlled Trial. Lancet Lond. Engl. 2016, 387, 945–956. [Google Scholar] [CrossRef] [Green Version]
  29. Costas, L.; Frias-Gomez, J.; Guardiola, M.; Benavente, Y.; Pineda, M.; Pavón, M.Á.; Martínez, J.M.; Climent, M.; Barahona, M.; Canet, J.; et al. New Perspectives on Screening and Early Detection of Endometrial Cancer. Int. J. Cancer 2019, 145, 3194–3206. [Google Scholar] [CrossRef]
  30. Nebgen, D.R.; Lu, K.H.; Bast, R.C. Novel Approaches to Ovarian Cancer Screening. Curr. Oncol. Rep. 2019, 21, 75. [Google Scholar] [CrossRef]
  31. Wang, Y.; Li, L.; Douville, C.; Cohen, J.D.; Yen, T.-T.; Kinde, I.; Sundfelt, K.; Kjær, S.K.; Hruban, R.H.; Shih, I.-M.; et al. Evaluation of Liquid from the Papanicolaou Test and Other Liquid Biopsies for the Detection of Endometrial and Ovarian Cancers. Sci. Transl. Med. 2018, 10, eaap8793. [Google Scholar] [CrossRef] [Green Version]
  32. Chang, C.C.; Wang, H.C.; Liao, Y.P.; Chen, Y.C.; Weng, Y.C.; Yu, M.H.; Lai, H.C. The Feasibility of Detecting Endometrial and Ovarian Cancer Using DNA Methylation Biomarkers in Cervical Scrapings. J. Gynecol. Oncol. 2018, 29, e17. [Google Scholar] [CrossRef] [Green Version]
  33. Chen, Q.; Zhang, Z.-H.; Wang, S.; Lang, J.-H. Circulating Cell-Free DNA or Circulating Tumor DNA in the Management of Ovarian and Endometrial Cancer. OncoTargets Ther. 2019, 12, 11517–11530. [Google Scholar] [CrossRef] [Green Version]
  34. Reijnen, C.; Putten, L.J.M.; Bulten, J.; Snijders, M.P.L.M.; Küsters-Vandevelde, H.V.N.; Sweegers, S.; Vos, M.C.; Wurff, A.A.M.; Ligtenberg, M.J.L.; Massuger, L.F.A.G.; et al. Mutational Analysis of Cervical Cytology Improves Diagnosis of Endometrial Cancer: A Prospective Multicentre Cohort Study. Int. J. Cancer 2020, 146, 2628–2635. [Google Scholar] [CrossRef] [PubMed]
  35. Bakkum-Gamez, J.N.; Wentzensen, N.; Maurer, M.J.; Hawthorne, K.M.; Voss, J.S.; Kroneman, T.N.; Famuyide, A.O.; Clayton, A.C.; Halling, K.C.; Kerr, S.E.; et al. Detection of Endometrial Cancer via Molecular Analysis of DNA Collected with Vaginal Tampons. Gynecol. Oncol. 2015, 137, 14–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Costas, L.; Palomero, L.; Benavente, Y.; Guardiola, M.; Frias-Gomez, J.; Pavón, M.Á.; Climent, M.; Martinez, J.M.; Barahona, M.; Salinas, M.; et al. Defining a Mutational Signature for Endometrial Cancer Screening and Early Detection. Cancer Epidemiol. 2019, 61, 129–132. [Google Scholar] [CrossRef] [PubMed]
  37. Martínez-Jiménez, F.; Muiños, F.; Sentís, I.; Deu-Pons, J.; Reyes-Salazar, I.; Arnedo-Pac, C.; Mularoni, L.; Pich, O.; Bonet, J.; Kranas, H.; et al. A Compendium of Mutational Cancer Driver Genes. Nat. Rev. Cancer 2020, 20, 555–572. [Google Scholar] [CrossRef]
  38. Sullivan, L.M.; Massaro, J.M.; D’Agostino, R.B. Presentation of Multivariate Data for Clinical Use: The Framingham Study Risk Score Functions. Stat. Med. 2004, 23, 1631–1660. [Google Scholar] [CrossRef]
  39. Slattery, M.L.; Edwards, S.L.; Caan, B.J.; Kerber, R.A.; Potter, J.D. Response Rates among Control Subjects in Case-Control Studies. Ann. Epidemiol. 1995, 5, 245–249. [Google Scholar] [CrossRef]
  40. Frias-Gomez, J.; Tovar, E.; Vidal, A.; Murgui, L.; Ibáñez, R.; Peremiquel-Trillas, P.; Paytubi, S.; Baixeras, N.; Zanca, A.; Ponce, J.; et al. Sensitivity of Cervical Cytology in Endometrial Cancer Detection in a Tertiary Hospital in Spain. Cancer Med. 2021, 10, 6762–6766. [Google Scholar] [CrossRef]
  41. Lac, V.; Nazeran, T.M.; Tessier-Cloutier, B.; Aguirre-Hernandez, R.; Albert, A.; Lum, A.; Khattra, J.; Praetorius, T.; Mason, M.; Chiu, D.; et al. Oncogenic Mutations in Histologically Normal Endometrium: The New Normal? J. Pathol. 2019, 249, 173–181. [Google Scholar] [CrossRef]
  42. Ketelaars, P.J.W.; Bosgraaf, R.P.; Siebers, A.G.; Massuger, L.F.a.G.; van der Linden, J.C.; Wauters, C.a.P.; Rahamat-Langendoen, J.C.; van den Brule, A.J.C.; IntHout, J.; Melchers, W.J.G.; et al. High-Risk Human Papillomavirus Detection in Self-Sampling Compared to Physician-Taken Smear in a Responder Population of the Dutch Cervical Screening: Results of the VERA Study. Prev. Med. 2017, 101, 96–101. [Google Scholar] [CrossRef]
  43. Mota, A.; Colás, E.; García-Sanz, P.; Campoy, I.; Rojo-Sebastián, A.; Gatius, S.; García, Á.; Chiva, L.; Alonso, S.; Gil-Moreno, A.; et al. Genetic Analysis of Uterine Aspirates Improves the Diagnostic Value and Captures the Intra-Tumor Heterogeneity of Endometrial Cancers. Mod. Pathol. 2017, 30, 134–145. [Google Scholar] [CrossRef] [Green Version]
  44. Salk, J.J.; Schmitt, M.W.; Loeb, L.A. Enhancing the Accuracy of Next-Generation Sequencing for Detecting Rare and Subclonal Mutations. Nat. Rev. Genet. 2018, 19, 269–285. [Google Scholar] [CrossRef]
  45. Broad Institute Picard Toolkit; Broad Institute, GitHub Repository, 2019. Available online: https://broadinstitute.github.io/picard/ (accessed on 28 May 2021).
  46. Fulcrum Genomics Fgbio. 2020. Available online: http://fulcrumgenomics.github.io/fgbio/ (accessed on 29 May 2021).
  47. Li, H. Aligning Sequence Reads, Clone Sequences and Assembly Contigs with BWA-MEM. arXiv 2013, arXiv:1303.3997. [Google Scholar]
  48. Smith, T.; Heger, A.; Sudbery, I. UMI-Tools: Modeling Sequencing Errors in Unique Molecular Identifiers to Improve Quantification Accuracy. Genome Res. 2017, 27, 491–499. [Google Scholar] [CrossRef] [Green Version]
  49. Lai, Z.; Markovets, A.; Ahdesmaki, M.; Chapman, B.; Hofmann, O.; McEwen, R.; Johnson, J.; Dougherty, B.; Barrett, J.C.; Dry, J.R. VarDict: A Novel and Versatile Variant Caller for next-Generation Sequencing in Cancer Research. Nucleic Acids Res. 2016, 44, e108. [Google Scholar] [CrossRef] [PubMed]
  50. Benjamin, D.; Sato, T.; Cibulskis, K.; Getz, G.; Stewart, C.; Lichtenstein, L. Calling Somatic SNVs and Indels with Mutect2. Available online: https://www.biorxiv.org/content/10.1101/861054v1 (accessed on 22 June 2022).
  51. Robinson, J.T.; Thorvaldsdóttir, H.; Wenger, A.M.; Zehir, A.; Mesirov, J.P. Variant review with the Integrative Genomics Viewer. Cancer Res. 2017, 77, e31–e34. [Google Scholar] [CrossRef] [Green Version]
  52. Wang, K.; Li, M.; Hakonarson, H. ANNOVAR: Functional Annotation of Genetic Variants from High-Throughput Sequencing Data. Nucleic Acids Res. 2010, 38, e164. [Google Scholar] [CrossRef] [PubMed]
  53. Yang, H.; Wang, K. Genomic Variant Annotation and Prioritization with ANNOVAR and WANNOVAR. Nat. Protoc. 2015, 10, 1556–1566. [Google Scholar] [CrossRef] [PubMed]
  54. Moore, L.; Leongamornlert, D.; Coorens, T.H.H.; Sanders, M.A.; Ellis, P.; Dentro, S.C.; Dawson, K.J.; Butler, T.; Rahbari, R.; Mitchell, T.J.; et al. The Mutational Landscape of Normal Human Endometrial Epithelium. Nature 2020, 580, 640–646. [Google Scholar] [CrossRef] [PubMed]
  55. Tamborero, D.; Rubio-Perez, C.; Deu-Pons, J.; Schroeder, M.P.; Vivancos, A.; Rovira, A.; Tusquets, I.; Albanell, J.; Rodon, J.; Tabernero, J.; et al. Cancer Genome Interpreter Annotates the Biological and Clinical Relevance of Tumor Alterations. Genome Med. 2018, 10, 25. [Google Scholar] [CrossRef]
Figure 1. Venn diagrams showing concordance between number of variants detected in the tumor (A) or in the endometrial aspirate (B) and the minimally-invasive samples.
Figure 1. Venn diagrams showing concordance between number of variants detected in the tumor (A) or in the endometrial aspirate (B) and the minimally-invasive samples.
Jpm 12 01074 g001
Figure 2. Boxplots for variant allele frequencies by sample type.
Figure 2. Boxplots for variant allele frequencies by sample type.
Jpm 12 01074 g002
Table 1. Number of participants, epidemiologic questionnaires, and biologic samples.
Table 1. Number of participants, epidemiologic questionnaires, and biologic samples.
ParticipantsEpidemiologic QuestionnaireBloodVaginal Self-SamplesCervical Pap-Brush SamplesEndometrial AspiratesTumour Samples
Endometrial cancer 1182180174175168161165
Ovarian cancer69696766625652
MMR pathogenic variant carriers 2104103110410290NA
BRCA pathogenic variant carriers 39898098980NA
Healthy women attending CC screening119119331191180NA
Controls with gynaecologic benign conditions190146117176151105NA
Hospital controls (non-gynaecologic)7672764800NA
TOTAL838787468786699412217
NA = not applicable; CC = cervical cancer. 1 Includes two synchronic (endometrial and ovarian) cancer cases. The table also includes 154 EC cases in which a Lynch syndrome tumor screening was performed with immunohistochemistry of mismatch repair system proteins (MMR). Among these EC cases, 119 were MMR proficient (MMRp) and 35 were MMR deficient (MMRd) cases. MMRd patients included four cases of confirmed Lynch syndrome, 14 presumed sporadic (MLH1 methylated), and 17 under evaluation. 2 Women without EC/OC included five women with premalignant lesions (three cases of complex atypical hyperplasia; one case of complex hyperplasia without atypia, and one case of simple atypical hyperplasia), 17 women with a history of colorectal cancer, 3 women with a previous breast cancer diagnosis, and 3 women with other cancer types. 3 Women without EC/OC included 32 women with a history of breast cancer.
Table 2. The main characteristics of the population of the Screenwide study.
Table 2. The main characteristics of the population of the Screenwide study.
High Risk Populations 1Controls 2EC 3OCp-Values 4
n (%)n (%)n (%)n (%)
Participants20238518269
Epidemiologic questionnaire201 (99.5)337 (87.5)180 (98.9)69 (100.0)
Age 0.740/0.124
<60188 (93.1)165 (42.9)51 (28.0)26 (37.7)
60–6912 (5.9)108 (28.1)61 (33.5)22 (31.9)
≥702 (1.0)112 (29.1)70 (38.5)21 (30.4)
Education 5 0.426/0.997
High School or below63 (31.3)240 (71.2)131 (72.8)52 (75.4)
Some college/associate64 (31.8)70 (20.8)29 (16.1)12 (17.4)
College or above74 (36.8)27 (8.0)20 (11.1)5 (7.2)
BMI 5 <0.001/0.500
<18.510 (5.0)4 (1.2)1 (0.6)3 (4.3)
18.5–24.99110 (54.7)99 (29.4)26 (14.4)21 (30.4)
25–29.9946 (22.9)131 (38.9)52 (28.9)24 (34.8)
≥3028 (13.9)89 (26.4)95 (52.8)20 (29.0)
Previous history of cancer (other than EC or OC) 0.346/0.791
Yes56 (27.7)47 (12.2)29 (15.9)8 (11.6)
No146 (57.2)338 (87.8)153 (84.1)61 (88.4)
Menopausal status 5 0.001/0.633
Premenopausal151 (75.1)63 (18.7)7 (3.9)13 (18.8)
Perimenopausal14 (7.0)29 (8.6)12 (6.7)2 (2.9)
Postmenopausal36 (17.9)245 (72.7)161 (89.4)54 (78.3)
Parity 5 0.792/0.122
Nulliparous77 (38.3)36 (10.7)26 (14.4)10 (14.5)
143 (21.4)70 (20.8)28 (15.6)20 (29.0)
≥281 (40.3)229 (68.0)126 (70.0)39 (56.5)
Hormonal contraception5 0.002/0.483
Never53 (26.4)114 (33.8)107 (59.4)34 (49.3)
Ever148 (73.6)221 (65.6)72 (40.0)35 (50.7)
Tobacco consumption 5 0.651/0.794
Never81 (40.3)193 (57.3)126 (70.0)48 (69.6)
Ever120 (59.7)144 (42.7)54 (30.0)21 (30.4)
BMI = body mass index. 1 Includes BRCA and MMR pathogenic variants carriers. 2 Includes women attending cervical cancer screening programs, patients with a benign gynecological pathology, and non-gynecological hospital controls. 3 Includes two synchronic (endometrial and ovarian) cancer cases. 4 p-value for controls and EC, and OC, respectively, excluding women attending cervical cancer screening programs. 5 Data obtained from epidemiological questionnaires, numbers do not always add up due to missing data. Missing data are <5% for all variables.
Table 3. Number of somatic variants found in gynaecological samples compared with variants identified in tumor samples in the pilot study.
Table 3. Number of somatic variants found in gynaecological samples compared with variants identified in tumor samples in the pilot study.
Patient IDCase-Control StatusTumor 1Endometrial Aspirates 2Cervical Pap Brush Samples 3Vaginal Self-Samples 3
Total Nº variants Total Nº variants Total Nº variants Total Nº variants
S110444ControlNA0 0 0
S110449ControlNA0 1 0
S110565ControlNA0 0 0
S110574ControlNA0 0 0
Total Nº variantsTotal Nº variantsNº variants also identified in tumourTotal Nº variantsNº variants also identified in tumourTotal Nº variantsNº variants also identified in tumour
S110036Case4444322
S110081Case25202024211919
S110124Case4440000
S110424Case3433322
S110435Case5555555
S110451Case3331100
S110474Case11121014101610
S110475Case5556455
S110501Case1111111
Total 61585558485044
% (95% CI) 95 (86–98) 590 (80–95) 484 (73–92) 579 (67–87) 490 (79–96) 572 (60–82) 4
Nº = Number of. NA = not applicable. For each type of sample, other than the tumor samples, two columns are specified for the following cases: the total number of variants in that sample and the number of variants that are jointly identified in the tumor. 1 Nº of variants identified in tumor surgical specimens (cases). 2 VAF > 5%. 3 VAF > 0.5%. 4 Nº variants also identified in the tumor/Nº variants in the tumor. 5 Nº variants also identified in the tumor/Nº variants in the corresponding sample.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Peremiquel-Trillas, P.; Paytubi, S.; Pelegrina, B.; Frias-Gomez, J.; Carmona, Á.; Martínez, J.M.; de Francisco, J.; Benavente, Y.; Barahona, M.; Briansó, F.; et al. An Integrated Approach for the Early Detection of Endometrial and Ovarian Cancers (Screenwide Study): Rationale, Study Design and Pilot Study. J. Pers. Med. 2022, 12, 1074. https://doi.org/10.3390/jpm12071074

AMA Style

Peremiquel-Trillas P, Paytubi S, Pelegrina B, Frias-Gomez J, Carmona Á, Martínez JM, de Francisco J, Benavente Y, Barahona M, Briansó F, et al. An Integrated Approach for the Early Detection of Endometrial and Ovarian Cancers (Screenwide Study): Rationale, Study Design and Pilot Study. Journal of Personalized Medicine. 2022; 12(7):1074. https://doi.org/10.3390/jpm12071074

Chicago/Turabian Style

Peremiquel-Trillas, Paula, Sonia Paytubi, Beatriz Pelegrina, Jon Frias-Gomez, Álvaro Carmona, José Manuel Martínez, Javier de Francisco, Yolanda Benavente, Marc Barahona, Ferran Briansó, and et al. 2022. "An Integrated Approach for the Early Detection of Endometrial and Ovarian Cancers (Screenwide Study): Rationale, Study Design and Pilot Study" Journal of Personalized Medicine 12, no. 7: 1074. https://doi.org/10.3390/jpm12071074

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

Article Metrics

Back to TopTop