Next Article in Journal
Comprehensive 3DCRT Hypofractionated Radiotherapy Schedule for Localized Prostate Adenocarcinoma in the Era of IMRT: Dosimetric and Endoscopic Analysis
Next Article in Special Issue
Unlocking Overexpressed Membrane Proteins to Guide Breast Cancer Precision Medicine
Previous Article in Journal
Responses to Medical Treatment in 192 Patients with Pancreatic Neuroendocrine Neoplasms Referred to the Copenhagen Neuroendocrine Tumour Centre in 2000–2020
Previous Article in Special Issue
Interleukin-6 as a Predictive Factor of Pathological Response to FLOT Regimen Systemic Treatment in Locally Advanced Gastroesophageal Junction or Gastric Cancer Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cancer Screening: Present Recommendations, the Development of Multi-Cancer Early Development Tests, and the Prospect of Universal Cancer Screening

by
Laurenția Nicoleta Galeș
1,2,
Mihai-Andrei Păun
3,*,
Rodica Maricela Anghel
1,3 and
Oana Gabriela Trifănescu
1,3
1
Department of Oncology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
2
Department of Medical Oncology II, Prof. Dr. Al. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
3
Department of Radiotherapy II, Prof. Dr. Al. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(6), 1191; https://doi.org/10.3390/cancers16061191
Submission received: 15 February 2024 / Revised: 8 March 2024 / Accepted: 15 March 2024 / Published: 18 March 2024
(This article belongs to the Special Issue Biomarkers for the Early Detection and Treatment of Cancers)

Abstract

:

Simple Summary

This article is intended to serve as a roadmap for scientists, steering them toward fruitful avenues of investigation. Moreover, the review makes a compelling argument for the implementation of universal cancer screening and multi-cancer early detection screening. It synthesizes evidence supporting the efficacy of such screenings, emphasizing their potential to detect various cancers at early, more treatable stages. By presenting a well-reasoned case, the review contributes to the discourse on the importance of widespread screening, urging healthcare providers and policymakers to consider its adoption for improved cancer detection and outcomes.

Abstract

Cancer continues to pose a considerable challenge to global health. In the search for innovative strategies to combat this complex enemy, the concept of universal cancer screening has emerged as a promising avenue for early detection and prevention. In contrast to targeted approaches that focus on specific populations or high-risk individuals, universal screening seeks to cast a wide net to detect incipient malignancies in different demographic groups. This paradigm shift in cancer care underscores the importance of comprehensive screening programs that go beyond conventional boundaries. As our understanding of the complex molecular and genetic basis of cancer deepens, the need to develop comprehensive screening methods becomes increasingly apparent. In this article, we look at the rationale and potential benefits of universal cancer screening.

1. Introduction

In 2020, the World Health Organization (WHO) statistics showed that cancer exacts an alarming toll, one in six deaths per year worldwide, with an alarming increase in overall mortality globally [1,2,3].
Cancer mortality can be prevented through cancer screening programs, which have a considerable impact on reducing cancer-specific mortality by selecting pre-symptomatic cases and driving more extensive diagnostic procedures and finally prompt treatment. For instance, breast cancer mortality in the United States declined by 53.2% between 1975 and 2014, with similar trends observed in Europe, which is attributed to both screening and improved treatments [4,5,6]. There is a need to avoid late detection, which is when cancer is diagnosed when it has already progressed beyond any potential curative options, or metastasized, where it is most often fatal, which in an estimated 15% of cases could have been avoided by early disease detection.
Among the countries with the highest per capita healthcare spending, cancer screening recommendations are relatively consistent, including breast, cervical, colorectal, prostate, skin, and lung cancer screening. However, differences in healthcare utilization exist between countries (and even within them), with a particular impact on cancer screening recommendations [4,7]. As such, there is a lack of universal consensus on which cancers should be screened for in the general population, with important malignancies left unchecked. For instance, cancers of the lung, pancreas, esophagus, stomach, and ovary are most often diagnosed in the metastatic setting, with a lack of population-based methods for screening, which is the case for up to 60% of cancer-related deaths where nationwide strategies for screening do not exist [8,9,10].
Current screening methods have increased survival rates, but they also have some drawbacks, including the risk of overdiagnosis, false-positive results, exposure to radiation, invasiveness leading to a decrease in patience compliance, and lack of support from the general public to undergo these investigations [10,11,12,13,14,15,16].
Furthermore, the current perspective on cancer screening might be conceptually inexact, as detection relies on “one organ at a time” intervention (Figure 1). Thus, most screening strategies are ineffective due to the high cost of a method designed only for one malignancy, thus excluding the potential diagnosis of other cancers that the patient might be suffering from. In contrast, universal cancer screening is a concept that predicts a future where detection is directed at multiple organs and multiple tumor types simultaneously with a single, potentially non-invasive method. So far, myriad multi-cancer early detection (MCED) methods have been proposed, gathering interest from clinicians to improve cancer detection and outcomes [7,17,18].
In this article, our goal is to provide insights on new developments in MCED biomarker research by sharing the latest results and directions that bring us closer to universal cancer screening, a concept that envisions testing persons from the general population to detect early cancers, no matter which type or site, in their pre-symptomatic phase, with a single, cost-effective, minimally invasive test.

2. Status Quo of Cancer Screening

Before we present the updates in screening biomarker research, we want to briefly present the current materials and methods for cancer screening. It is important to note that there is still a long way to go until consensus is reached over the targeted population (risk categories, age, frequency of testing) and that these tests have different categories of recommendation (Table 1).

2.1. Colorectal Cancer

Colorectal cancer benefits from some of the best screening tools available, including stool tests and colonoscopies.
Stool-based tests include the guaiac fecal occult bleeding test (g-FOBT), fecal immunochemical test (FIT), and stool DNA test (DNA-FIT). The g-FOBT and FIT tests detect blood in stool, relying on the premise that cancers will cause ruptures of small vessels, leading to clinically insignificant bleeding [20,21]. Although these tests have high sensitivity and specificity, it is worth noting that they have lower sensitivity for pre-malignant lesions such as adenomas (especially smaller ones), which do not invade the blood vessels in the submucosa [22,23].
Detection (and also resection) of precancerous lesions is only possible through colonoscopy, which is considered the gold standard for colorectal cancer screening due to its high sensitivity and specificity, relative safety, and demonstrating reduction of overall colorectal cancer mortality [24]. Possible limitations to this test include a certain degree of invasiveness and dependence on the training level and experience of the gastroenterologist, leading to discrepancies in the outcomes shown by different studies [25].
There are no validated serum biomarkers used in colorectal cancer screening. Serum CEA and CA19-9 markers are not used in screening due to low sensitivity and specificity, but are validated for prognosis and post-resection follow-up [16,26].
The United States Preventive Services Taskforce (USPSTF) highly recommends colorectal cancer screening for patients aged 50–75 (grade A). For other age groups, the USPSTF suggests screening for patients aged 45–49 (grade B) and also 76–85 (grade C—based on life expectancy and patient risk). Screening modalities include colonoscopy every 10 years, g-FOBT or FIT yearly, with stool DNA-FIT every 1–3 years, and flexible sigmoidoscopy every 5 years (or flexible sigmoidoscopy every 10 years plus FIT every year) [27]. The American Cancer Society (ACS) also recommends screening for adults aged 45–75, selective screening for those 76–85, and no screening for those over 85. For patients under the age of 45, high-risk features may cause earlier intervention. Patients with relatives diagnosed with colorectal cancer should be screened 10 years before the diagnosis age of the relative. Patients with abdominal or pelvic irradiation history should begin screening 5 years post-intervention or starting at age 30. Patients suffering from inflammatory bowel disease also require colonoscopies at most 8 years after diagnosis, repeated every 1–3 years. Most importantly, patients with removed polyps during colonoscopy should repeat the test after 3 years maximum, possibly earlier if malignant or high-risk features are found in pathology [28]. A comparison of colon cancer screening recommendations is offered in Table 2, with relevant information about the eligible population and type of testing. In the relevant literature column, we cited the source we found for the information from published guidelines, statements, or relevant articles showing the outcomes of the screening methods that are in place.

2.2. Breast Cancer

Breast cancer screening depends mostly on imaging techniques such as mammography, ultrasonography, magnetic resonance imaging, and digital breast tomosynthesis, offering a view of the breast and surrounding tissue, including the axillary region. Unfortunately, there is a lack of consensus on stratifying risk groups and offering which method to which type of patient, with a lack of homogeneity observed at both the international and national level [47]. Furthermore, the use of mammography is a subject of dispute among clinicians, due to the risk of irradiation and overdiagnosis (i.e., interpreting benign lesions as malignant, leading to overtreatment, burdening the patient psychologically and financially) [13,48]. Serum biomarkers such as CA15-3 are not suitable for early detection of breast cancer, since the level rarely increases in patients with early or localized breast cancer, although methods have been proposed to improve the sensitivity in early disease detection [49]. Currently, CA15-3 is endorsed for surveillance of resected treated breast cancer or for monitoring progression on treatment for metastatic disease [50].
The USPSTF recommends that all women aged 50–74 years receive every two years screening mammography (grade B), with a suggestion for women aged 40–49 years to receive individualized screening or no mammography [8,51]. However, the ACS recommends that women aged 45–54 receive annual screening mammography and women over the age of 55 annual or biennial mammography until their life expectancy is below 10 years. Women at high risk of breast cancer (such as carriers of BRCA1/2 mutations, women with personal or family history of breast cancer or tumor syndromes such as Li–Fraumeni or Cowden) or with chest radiation therapy administered between 10–30 years of age are recommended to receive breast MRI and yearly mammogram screening starting at age 30 [52]. In Table 3, we summarize the breast cancer screening recommendations in different high-income countries. In the relevant literature column, we cited the source we found for the information from published guidelines, statements, or relevant articles showing the outcomes of the screening methods that are in place.

2.3. Prostate Cancer

The recommendations for prostate cancer management include digital rectal examination and serum prostate-specific antigen (PSA) determination for screening. However, evidence so far does not demonstrate a survival improvement for PSA screening. A systematic review of five randomized controlled trials, including a total of 721,718 men enrolled, concluded that testing PSA levels does not affect overall mortality, with a significant risk of overdiagnosis and overtreatment [77]. It is worth remembering that PSA can be detected in the bloodstream after the cancer cells have ruptured the basement layer of the prostate, indicating that the test is conceptually not designed for early tumors or premalignant lesions [78,79].
PSA testing is no longer recommended for prostate cancer screening. Men aged 55–69 have a C grade recommendation according to the USPSTF, meaning that testing is suggested only if recommended by their physician due to increased risk. For men older than 70, the USPSTF recommends against screening. These changes have led to fewer false-positive diagnoses, but also more metastatic prostate cancer diagnoses in the US Caucasian population. Similarly, the ACS recommends discussing PSA testing with men aged 50 and above with an average risk, aged 45 and above who are African American, those with a first-degree relative below the age of 65 diagnosed with prostate cancer, or those aged 40 with multiple first-degree relatives with early PC diagnoses [8,80,81].

2.4. Lung Cancer

Low-dose computed tomography (LDCT) is being offered in the United States for high-risk populations (individuals between the ages of 55 and 80 who have a smoking history equivalent to 30 packs of cigarettes per year, who are currently smoking, or have quit within the last 15 years) to screen for lung cancer. LDCT has good results, showing a 20% reduction in lung cancer mortality compared to the conventional chest X-ray, and also higher sensitivity [82]. It is worth noting that in Europe, LDCT does not benefit from the same level of adoption, with several trials failing to show benefit [83,84]. Also, LDCT can give false-positive results, leading to unnecessary diagnostic and therapeutic procedures, as well as false-negative results, as LDCT may not detect all cancers. Radiation exposure and the high cost of this procedure are also limitations for the large-scale adoption of LDCT [82,84,85].
Current recommendations from the USPSTF include annual low-dose computer tomography (LDCT) for adults aged 50–80 with a >20 pack-year history of smoking, and who have either quit within the last 15 years or who are current smokers (grade B recommendation) [86]. LDCT can be discontinued once a patient has not smoked for at least 15 years or if there would not be a benefit on life expectancy any longer. The 2023 ACS Recommendations for Lung Screening follow the USPSTF recommendations, with no limit to the cessation period (all former smokers eligible for LDCT) [87].

2.5. Cervical Cancer

Screening for cervical cancer can be performed through a Pap smear followed by cytologic examination of the gynecological tract cells and HPV detection (the causal agent in the overwhelming majority of cases). Pap smears are largely regarded as an example of screening success, due to the impressive reduction in cervical cancer mortality after implementing screening strategies using this method [88], because of the high diagnostic accuracy for premalignant lesions [89].
Cervical cytology is another screening method that benefits from a grade A recommendation from the USPSTF, for women aged 21–29, given every three years, with the choice to continue until 65 years or combine cervical cytology with high-risk human papilloma virus (hrHPV) testing every five years from ages 30–65. The American College of Obstetrics and Gynecology (ACOG) and the Society of Gynecological Oncology (SGO) have both endorsed these recommendations. ACS also adds primary HPV testing as screening every 5 years from age 25–65, with co-testing or cytology (according to USPSTF recommendations) offered only when HPV testing is not available [8,90,91]. Further information on cervical cancer screening recommendations across high-income countries is offered below in Table 4.

2.6. Oral Cancer Screening

Visual oral examination (VOE) is the recommended test for oral cancer and oral potentially malignant disorders (OPMDs), which implies systematic visual inspection and palpation of the oral cavity under a bright light source, to detect suspect lesions of the oral mucosa [110,111]. Although most national organizations, including the USPSTF, do not recommend population-based screening through VOE, an RCT showed a reduction in mortality of 81%, and a reduction of 38% in oral cancer incidence [112] and further studies have demonstrated high sensitivity and specificity for this type of screening [110,113]. What is particular about this form of screening is the non-invasiveness, its low cost, and the importance of primary care doctors and dentists, which make the test more accessible and also lowers the burden of manpower needed for population-based screening.

2.7. Screening for Other Cancers

Other types of cancers (such as ovarian [114], pancreatic [115], or testicular [116]) may benefit from current methods of screening. However, the current methods have yet to prove a reduction in mortality or reduction in the financial burden, thus lacking a consensus on a standardized valid test. This underlines the need for new validated methods to improve the current management of early detections.

3. Fantastic Biomarkers and Where to Find Them

Cancer presence can be discovered in its pre-symptomatic phase through careful evaluation of bodily fluids, where trace amounts of catabolites, nucleic acids, and peptides might reflect the existence of complex mechanisms involved in tumorigenesis and host response directed towards the malignancy. Direct vascular invasion permits cancer cells to be found in blood, but other processes such as necrosis, apoptosis, micro-vesicle budding, and phagocytosis (which occur earlier), or the formation of auto-antibodies, contribute with a host of byproducts that can serve as markers of early-stage disease. As such, bodily fluids can function as a strategic key point of early cancer detection [117,118,119].

3.1. Blood Tumor Markers

3.1.1. Circulating Tumor Cells

Circulating tumor cells (CTCs) occur as part of the natural evolution of malignancies towards metastasis. Although this process usually involves lymphovascular invasion, CTCs have been nonetheless found before imaging detection of a primary tumor, indicating a potential value in cancer screening [120,121,122,123].
Huang et al. showed an increase in mean CTC for patients with early (stage I/II) lung, colorectal, gastric, liver, and esophageal cancer, providing a glimpse of establishing a new biomarker, by using EpCAM+/vimentin+ specific immunomagnetic beads for CTC isolation [124,125]. Other possible strategies include the analysis of circulating ensembles of tumor-associated cells (C-ETACs), defined as clusters of pan-cytokeratin and EpCAM-positive cells (at least three). This method has so far outperformed conventional CTC assays, with 89.8% sensitivity and 97% specificity [126,127].

3.1.2. Plasma Cell-Free Nucleic Acids

As described earlier, conventional protein tumor markers such as carcinoembryonic antigen (CEA), cancer antigens 15-3 (CA15-3) or 19-9 (CA19-9), are routinely used in clinical practice, but they suffer from low sensitivity, especially at early stages, and are not routinely used for screening [128]. Novel tests such as liquid biopsies hold promise for accurate screening of multiple tumor types and represent an exciting avenue for the research and development of new biomarkers [129,130,131].
Circulating biomarkers, including microRNAs (miRNAs), cell-free DNA (cfDNA), and circulating tumor DNA (ctDNA), represent active targets as biomarkers of cancer detections and MCED can be achieved through genome sequencing of plasma, as shown by several groups. Malignancy can be detected and tumor sites, including ovary, liver, stomach, pancreas, esophagus, colorectum, lung or breast, can be predicted using nucleic acid assays [17,132].
One of the most discussed tests available is the Galleri (GRAIL) test, which is a targeted methylation assay combined with a machine learning classifier for the detection and discrimination of more than 50 cancers using cfDNA. The test obtained 54.9% sensitivity and 99.3% specificity, with 93% accuracy for tissue-of-origin (TOO) prediction [133,134,135]. Further MCED tests are showing promising results, and we are eagerly awaiting clinical trials to demonstrate their screening value. A review conducted by Brito-Rocha et al. provides further insights into the development of biomarkers that can be evaluated through liquid biopsies, including the clinical trials where these methods were used. The article provides a comprehensive summary of the state of the art in liquid biopsy for MCED research and further encourages the need for thorough clinical research of the new screening tools for them to be used in current practice [18]. As a tribute to their work, we updated Table 3 from their manuscript with the current state of the art in clinical trials for the validation of MCED tests (Table 5 and Table 6).

3.1.3. Extracellular Vesicle-Based Tests for MCED

Extracellular vesicles (EVs), such as exosomes, microvesicles, and apoptotic bodies, are lipid membrane vesicles that get secreted by a large number of cells, including cancer cells. They play major roles in cell–cell-mediated communication, including molecule transportation. Whether they are constituted by nucleic acids, proteins, or metabolites, these molecules can be of interest as cancer biomarkers [136,137,138,139].
Goldvaser et al. studied the use of exosome human telomerase reverse transcriptase (hTERT) mRNA as a minimally invasive pan-cancer biomarker [140]. Their study showed a 62% sensitivity and 100% specificity for detecting 15 cancer types, both solid and hematologic.
The VeritaTM test using EV-protein profiling combined with machine learning is capable of detecting pancreatic, ovarian, and bladder cancer in early, curative stages (stage I and II), with varying degrees of sensitivity pancreatic cancer (95.5%), ovarian cancer (74.4%), and bladder cancer (43.8%) [141]. Most encouragingly, targeting the CD9+ EVs and aptamer recognition of CD63/EpCAM/MUC1 (epithelial markers), carcinomas were detected with 100% sensitivity and specificity, indicating that specific targeting of tumor type-specific proteins can be used for both cancer screening and identification of the tumor’s origin [142,143].

3.2. Non-Invasive Early Cancer Detection Using Indirect Media—Saliva, Tears, Urine, Breath

Blood is not the only compartment where biomarkers can be identified. Other media, for instance, urine, saliva, or breath, may contain materials derived from circulation. These molecules can be interrogated through non-invasive testing for the oncogenic pathways they uncover [17].
Although the glomerular filter impedes the passing of larger molecules, extracellular vesicles containing tumor-specific RNA signatures can be detected through a nanowire device embedded in a microfluidic system, as described by Takao Yasui et al. [144].
Furthermore, saliva is an easily available bodily fluid that may emerge as a haven for MCED development, as innovations in the assay of circulatory biomarkers have been exciting thus far. The review by Kaczor-Urbanowicz et al. delves deeper into the recent progress made in salivaomics and the impact on cancer diagnosis [145].
Tear samples, although difficult to collect, maybe an interesting avenue for MCED biomarker discovery, as a study by Daily et al. has identified several protein markers in tears of early-stage breast cancer [146].
Last but not least, exhaled breath can become a viable site for biomarker identification, with devices showing promising results in detecting multiple cancers, including respiratory, gastrointestinal, or gynecological cancers. We recommend the systematic review done by Krilaviciute et al. for further insights on exhaled breath biomarker discovery [147].

3.3. Tumor Exfoliation

Anatomic corridors such as the gastrointestinal (GI) tract or the female reproductive tracts are lined by epithelial cells, which have a physiological tendency to exfoliate into a common efflux route. Similar to CTCs, the excretory medium for these exfoliates can be interrogated for clinically useful biomarkers, as exfoliation occurs from both precursor lesions and earliest-stage cancers, even before any substantial invasion can occur [17].
The stool can be a valuable sample for colorectal cancer screening, since even precursor lesions can be accurately detected using multi-target DNA tests, achieving detection rates comparable to those of colonoscopy [148,149,150]. Attempts at improving diagnostic accuracy have been made by targeting molecular debris in stool, including abnormal DNA (such as mutant KRAS, actin, FIT, aberrantly methylated BMP, and NDRG4 promoter regions), using the Cologuard® stool DNA test. Unfortunately, the test had lower sensitivity towards adenomas, thus limiting its preventive role, and lower specificity compared to FIT (87–90% compared to 95–96%) [17,150]. The mt-sRNA ColoSense® test showed improvements in sensitivity (94%), but remains insufficient for advanced adenomas (46%) [151]. However, molecular tumor fingerprinting of early cancers using stool samples remains an attractive venue in biomarker research. Also, compared to the standard fecal occult blood test, early data suggest that stool testing can be expanded to include the previously unscreened upper GI tract as well. However, techniques need to be optimized and rigorously tested in the clinic to be adequately assessed to have a pan-GI screening test [17]. In our opinion, the diagnostic yield of these tests is dependent on accurately predicting the mutations occurring earliest in tumorigenesis, which may influence the transition between premalignant lesions towards neoplasia [152].
Detecting gynecological neoplasms with tampons is a concept that has been proven by analyzing methylated DNA markers extracted from vaginal tampons. Also, molecular analysis may allow the detection of both endometrial and ovarian cancer using a conventional cervical Papanicolaou test, further expanding the clinical utility of this routine test. Further clinical trials are needed to demonstrate the practicality of these tests, as they might turn a single cancer detection test into an MCED examination [153,154].
Table 5. Clinical trials conducted/ongoing for validation of multi-cancer early detection tests (updated and redesigned from Table 3 of Brito-Rocha et al. [18]).
Table 5. Clinical trials conducted/ongoing for validation of multi-cancer early detection tests (updated and redesigned from Table 3 of Brito-Rocha et al. [18]).
Trial IDTrial NameMCED TestSponsorStatus (as of February 2024)Est. CompletionEnrolledPublications
NCT02889978CCGA, GRAIL-001GalleriGRAIL, LLCActive, not recruitingMarch 202415,254[135,155]
NCT03085888STRIVE, GRAIL-002GalleriGRAIL, LLCActive, not recruitingMay 202599,481
NCT03934866SUMMITGalleri (combined with LDCT)University College London and GrailActive, not recruitingAugust 203013,035[156]
NCT04241796PATHFINDER, GRAIL-007GalleriGRAIL, LLCCompletedNA6662[157,158,159]
NCT05155605PATHFINDER2GalleriGRAIL, LLCRecruitingJanuary 202835,000
NCT05205967REFLECTIONGalleriGRAIL, LLCRecruitingAugust 202617,000
NCT05611632,ISRCTN91431511NHS-Galleri, GRAIL-009GalleriGRAIL, Bio UK Ltd.Active, not recruitingFebruary 2026140,000[160,161,162]
NCT05235009LEV87AGAGomeElyptaRecruitingMarch 20259170[163]
NCT05295017LEV93AGAGomeElyptaRecruitingMarch 20251256
NCT05780957Multi-Cancer Early Detection (MCED) of FirefightersGAGomeElyptaRecruitingJanuary 20312000 (estimated)
NCT05227534PREVENTOverCBurning Rock DxRecruitingJanuary 202912,500-
NCT04972201PROMISEOverCChinese Academy of Medical Sciences and
Burning Rock Dx
CompletedNA2305[164]
NCT04822792PRESCIENTOverCChinese Academy of Medical Sciences and
Burning Rock Dx
CompletedNA11,879-
NCT04820868THUNDEROverCShanghai Zhongshan Hospital and
Burning Rock Dx
CompletedNA2508[165,166]
NCT04817306PREDICTOverCShanghai Zhongshan Hospital and
Burning Rock Dx
CompletedNA14,206-
NCT03517332-DEEPGENQuantgene Inc.CompletedNA675[167]
NCT03967652-Na-noseAnhui Medical University-NANA[168,169]
NCT05366881CAMPERR-Adela, Inc.Active, recruitingDecember 20267000[170]
NCT05254834Vallania-Freenome Holdings Inc.Active, recruitingJune 20255400-
NCT05227261K-DETEKSPOT-MASGene SolutionsActive, recruiting-10,000[171,172]
NCT05159544FuSion-Singlera Genomics Inc.--60,000[173]
NCT04405557PREDICT-Geneplus-Beijing Co.CompletedNA757Article under review
NCT02662621EXODIAG-Centre Georges Francois LeclercCompletedNA80[174]
NCT04197414--Yonsei UniversityActive, recruitingDecember 20293000-
NCT03951428--LifeStory Health Inc.CompletedNA30-
NCT03869814--Bluestar Genomics Inc.CompletedNA660-
NCT02612350--Pathway GenomicsCompletedNA1106-
NCT06011694The Jinling CohortMERCURY testNanjing Shihejiyin Technology, Inc.RecruitingMay 202715,000 (estimated)[175]
NCT05633342CADENCEMiRXES MCSTMiRXES Pte Ltd.RecruitingMay 202515,000 (estimated)-
NCT04825834DELFI-L101DELFIDelfi Diagnostics Inc.RecruitingJune 20262660 (estimated)[176]
NCT05306288DELFI-L201 (CASCADE-LUNG)Delfi Diagnostics Inc.Active, not recruitingMarch 202515,000 (estimated)-
NCT05492617DETECTRigshospitalet, DenmarkCompletedNA3628[177]
NCT05435066CORE-HHHarbringer HxHarbringer HealthRecruitingJuly 202510,000 (estimated)[178]
NCT05117840SHIELDLUNAR-2Guardant Health, Inc.RecruitingJanuary 20269000 (estimated)[179,180]
NCT04136002ECLIPSEJanuary 202440,000[181]
Table 6. Technical description of MCED Tests currently on clinical trials conducted/ongoing for validation of multi-cancer early detection tests (updated and redesigned from Table 3 of Brito-Rocha et al. [18]).
Table 6. Technical description of MCED Tests currently on clinical trials conducted/ongoing for validation of multi-cancer early detection tests (updated and redesigned from Table 3 of Brito-Rocha et al. [18]).
MCED TestSponsorPublicationsSampleMethod of DetectionTumor Types
GalleriGRAIL, LLC[135,155]plasmaTargeted methylation assay (cfDNA), combined with a machine learning classifier for detecting (54.9% sensitivity, 99.3% specificity) and discriminating TOO (93% accuracy) [134,135].<40 different tumor types: GI, hematological, head and neck, lung, ovary, etc.
GAGomeElypta[163]plasma and urineFree glycosaminoglycan profiles (GAGome)14 different cancers: gliomas, genito-urinary, etc.
OverCBurning Rock Dx[164]plasmaTargeted methylation sequencing assay (ELSA-seq) combined with machine learning for cancer detection (80.6% sensitivity, 98.3% specificity) and TOO discrimination (81.0% accuracy)Lung, colorectal, liver, ovarian, pancreatic, esophageal [182]
DEEPGENQuantgene Inc.[167]PlasmaNGS combined with machine learning for cancer detection (57% sensitivity, 95% specificity)Lung, breast, colorectal, prostate, bladder, pancreatic, and liver
Na-noseAnhui Medical University[168,169]Exhaled breathNanoparticle sensors for Volatile Organic Compounds found in exhaled breathHead and neck, lung, breast, colorectal, etc.
-Adela, Inc.[170]PlasmacfMeDIP-seq [183] combined with machine learningKidney, pancreas, lung, gliomas, etc.
SPOT-MASGene Solutions[171,172]PlasmaNGS sequencing of ctDNA to detect genome-wide hypomethylation, targeted hypermethylated regions, genome-wide copy number variation, length profile of ctDNAGastric, lung, breast, etc.
-Singlera Genomics Inc.[173]PlasmaDNA methylationlung, esophagus, stomach, liver, pancreatic and colorectal
-Geneplus-Beijing Co.Article under reviewPlasmacfDNA analysisHepatic, pancreatic, ovarian, breast, colorectal and gastric cancer.
-Centre Georges Francois Leclerc[174]Blood and urineAnalysis of HSP70-exosomesBreast, lung, ovarian
-Yonsei University-Blood and urinectDNA analysisUrological malignancies
-LifeStory Health Inc.-Menstrual blood Breast, endometrial, lung
-Bluestar Genomics Inc.-PlasmacfDNA analysisPancreatic
-Pathway Genomics-PlasmacfDNA analysisPancancer
MERCURY testNanjing Shihejiyin Technology, Inc.[175]plasmacfDNA analysisLiver, lung, colorectal
MiRXES MCSTMiRXES Pte Ltd.-plasmamiRNA expression in tandem with DNA methylationlung, breast, colorectal, liver, stomach, esophageal, ovarian, pancreatic, and prostate
DELFIDelfi Diagnostics Inc.[176]plasmacfDNA analysisLung, liver cancer
Harbringer HxHarbringer Health[178]plasmacfDNA analysisVariety of solid and hematological cancers
LUNAR-2Guardant Health, Inc.[179,180]plasmactDNA analysisColon, lung cancer

4. Expanding the Horizons towards Universal Cancer Screening

4.1. Improved Imaging Techniques

Conceptually, computed tomography (CT) scans with or without contrast enhancement of the whole body can be considered MCED. However, CT scans have low sensitivity and specificity, and pose a significant threat of side effects, due to irradiation and contrast-induced nephropathy, which severely limit the repeated use for one person [184,185].
Next-generation approaches towards imaging involve molecular, nanoparticle, and fluorescent constructs, which have sparked clinical interest for their potential to capture a precise in vivo snapshot of the person’s oncological status, providing details for both screening and treatment monitoring. Further research is warranted to optimize the technical details and to provide information about the examination performance and clinical outcomes [17,186,187,188].

4.2. Volatile Organic Compounds

Byproducts of metabolic processes involving cellular transformation and tumorigenesis can be found in trace amounts in urine and expired breath. These catabolites and the patterns of volatile organic compounds (VOCs) can be studied, with numerous studies indicating potential relationships between VOCs and different cancers. It has been suggested that in breast cancer patients, six specific VOCs could be used for a screening test. Other studies have highlighted the importance of VOCs in cancers such as prostate, kidney, or lung [7,133,169,189,190,191,192,193,194,195,196,197].
So far, volatilome research has been impeded by technical issues and a high financial burden, requiring complex technology for the identification of specific patterns. However, a biosensor has recently been proposed, Caenorhabditis elegans, a nematode with a refined olfactory sense (with over 1200 olfactory receptor-like genes, encoding 1.5 times as many different types of olfactory receptors as a dog, capable of detecting subtle odorant concentration changes in its habitat). With the observation that C. elegans is attracted to urine from cancer patients, but avoids urine from healthy persons, a team led by Hirotsu et al. perfected an innovative, highly sensitive cancer screening test called N-Nose, which has been available in Japan since 2020. It is capable of detecting the presence of cancer for 15 malignancies, with a sensitivity of 87.5% and specificity of 90.2%. Unfortunately, N-Nose only correlates with the presence/absence of cancer, without the capability of pinpointing the cancer types. However, efforts are being made to further understand the chemotaxis mechanism of C. elegans and methods of perfecting this potentially powerful screening tool. It is important to mention that the N-Nose behavioral assay has been thoroughly evaluated in clinical studies with large cohorts of patients, showing reliable results. Also, the results have been replicated at other prestigious institutions in the US and Italy [7,128,198,199,200,201,202,203].

4.3. Always on PATROL for Early Lung Cancer

A particular assay that has drawn the authors’ attention has been the diagnostic platform PATROL (point-of-care aerosolizable nanosensors with tumor-responsive oligonucleotide barcodes), which is currently tested for the early diagnosis of lung cancer, but with the potential to become an MCED test for airway cancers. The diagnostic platform incorporates three modules, as follows. (1) Artificially synthesized low-plex activity-based nanomers (ABNs), consisting of protease substrates (for dysregulated proteases produced by tumoral cells on the lung epithelia) conjugated to eight-armed PEG nanoscaffolds with single-stranded modified DNA 20-mers. These molecules are nebulized through a (2) portable inhalation unit, and then delivered to the lung, where the DNA barcodes are liberated upon substrate cleavage by dysregulated proteases associated with early lung adenocarcinoma, shed into the systemic circulation, and then excreted by the kidneys in the urine. The urine is then sampled (2 h after inhalation), and the DNA barcodes are analyzed through a (3) multiplex lateral flow assay, quantified, and classified to provide diagnosis. The results of this test have so far been promising with high sensitivity and specificity for stage I/II lung adenocarcinoma [204]. This particular assay sparked our interest not only for the good results shown so far and the cost-effectiveness promised by the authors but also for the concept of activity-based diagnostics (ABD), in which the tumoral metabolic processes are leveraged to provide the result via the excretion of a synthesized biomarker. We believe that variants of this concept can be proposed and used for other cancers by mastering the triad this assay proposes: (1) a highly sensible, conjugable complex consisting of a readable DNA barcode and a cleavable enzyme substrate that can be targeted by dysregulated tumoral enzymes; (2) an accessible, preferably non-invasive entry route for this complex, which cannot be degraded along this path, towards the organ(s) in which cancer is screened; and (3) an excretion route where the DNA can be available for analyzing and interpretation of the results.

5. Discussion on the Prospect of Universal Cancer Screening

5.1. “One Test to Rule Them All” vs. “One Organ at a Time”

The universal cancer screening approach is a paradigm-changing approach for the early detection of cancers through the non-invasive interrogation of multiple markers and media on the possible existence of malignancy. With the potential of multi-organ detection, we can imagine tests that can include all cancers in a single efficient modality of testing, which would be potentially both cost- and life-saving. Perhaps, in the era of treating cancer using “precision medicine” tailored to the most intimate characteristics of tumor cell biology, a “one test to rule them all” approach toward cancer screening would be a match made in heaven, as simulations have already modeled, promising reductions of up to a quarter in all cancer-related deaths in patients aged 50–79 years in the United States and a reduction of almost 80% for late-stage (III, IV) disease [7,17,205].

5.2. Aggregate Prevalence

The value of a screening intervention is given by the combination of a low estimated number of persons that need to be screened to detect one cancer (NNS), and a high positive predictive value (PPV—the probability that a screening test indicates the presence of a tumor). Given that prevalence varies across tumor locations and subtypes, a single test that incorporates multiple targets can decrease the NNS. Furthermore, aggregating multiple cancers with different prevalence rates in a universal test would yield higher PPVs than single-organ screening. This argument has been comprehensively supported by Ahlquist et. al, offering an insightful rationale for the need for MCED [17].

5.3. Manpower Considerations

It is difficult to estimate if MCED screening would represent a burden or a relief for medical healthcare workers. One of the pivotal requirements of a successful, efficient screening program is a central, well-funded authority, responsible for setting recommendations and organizing the framework of how these recommendations can be applied (see Table 2, Table 3 and Table 4). It is most encouraging to see cancer screening programs offering eligible population tests that do not require healthcare workers (such as FIT) at home [35]. These tests are generally accompanied by greater compliance from the general public because of their non-invasiveness, privacy, and simple nature (see Table 1). Collecting samples such as stool, urine, or saliva can be done by the patient and sent through the mail to a specialized laboratory. Though most of the MCED screening tests in clinical trials (see Table 5 and Table 6) require blood samples, this should not be an impediment, as these tests can be offered simultaneously with other regular health checkups that require this minimally invasive medical procedure. In conclusion, we believe that MCED would not burden already-pressured health services.

5.4. Cost-Effectiveness

Most cancers are diagnosed in late stages, needing expensive long-term systemic treatment, which leads to an increase in the economic burden of cancers. In 2020 alone, the US healthcare system covered 157.7 billion USD, and 16.1 million cancer patients had out-of-pocket expenses, with a quarter of them declaring they had difficulties paying the bills. Mitigation of the economic, medical, and psychological issues related to late-stage diagnosis is an overarching drive of universal cancer screening. Such expenses warrant investments in MCED, as tests such as Galleri (~945 USD), or N-Nose (~109 USD) can be cheaper than PET-CT, for instance (~1100 USD). Large-scale implementation of these tests can lower the price even further, and the biggest benefit would be offering prompt treatment, with lower financial burden for both the patient and healthcare providers [7,206]. The reductions in the financial burden have been estimated by Hackshaw et al. and Tafazzoli et al., showing that hypothetically MCED could reduce the cost per cancer treatment by up to 5421 USD per cancer patient [19,207]. With permission, we decided to present the estimated data from Hackshaw et al.’s article in Table 7.

5.5. Bioinformatics and Artificial Intelligence (AI)

Biomarker research has evolved in close relation with bioinformatic development. Since the development of the Cancer Genome Atlas (TCGA) project, large databases containing valuable data on the multisystem biology features of multiple tumor types have led to the development of data mining methods to propose valuable biomarkers [18,208,209,210,211,212,213,214]. Examples include the study of molecular aberrations of the SST gene for multiple GI cancers [215], the difference in HSP90α protein across 9 different tumors [216], or the identification of CpG markers to detect 27 cancer types [217].
Furthermore, the existence of large databases allows machine learning algorithms to be developed and trained to improve the detection capacity of current biomarkers. Several neural network-based algorithms have been developed to improve detection sensitivity and specificity, as well as tumor-of-origin (TOO) discrimination [18]. The undeniable benefit of artificial intelligence becomes evident when we observe that the vast majority of MCED tests with ongoing trials have applied deep learning algorithms to improve their diagnostic yield (Table 6). As such, it is important to recognize AI as a key element of biomarker discovery that boosts the output and outcomes of MCED tests.

6. Conclusions

The dream of universal cancer screening remains elusive at the moment. However, translational studies coupled with clinical research offer us an optimistic view for the future, as we move closer to significant improvements in oncological outcomes through early diagnosis and curative treatment. We are hopeful that the tools presented in this article will soon be incorporated into current guidelines for screening the general population and will help improve the survival rate of cancer patients.

Author Contributions

Conceptualization, L.N.G. and M.-A.P.; methodology, L.N.G. and R.M.A.; software, M.-A.P.; writing—original draft preparation, M.-A.P.; writing—review and editing, L.N.G. and O.G.T.; supervision, L.N.G. and O.G.T.; project administration, R.M.A. and L.N.G.; tables and figures—M.-A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Dyba, T.; Randi, G.; Bray, F.; Martos, C.; Giusti, F.; Nicholson, N.; Gavin, A.; Flego, M.; Neamtiu, L.; Dimitrova, N.; et al. The European Cancer Burden in 2020: Incidence and Mortality Estimates for 40 Countries and 25 Major Cancers. Eur J Cancer 2021, 157, 308–347. [Google Scholar] [CrossRef]
  3. Torre, L.A.; Siegel, R.L.; Ward, E.M.; Jemal, A. Global Cancer Incidence and Mortality Rates and Trends—An Update. Cancer Epidemiol. Biomark. Prev. 2016, 25, 16–27. [Google Scholar] [CrossRef]
  4. Basu, P.; Ponti, A.; Anttila, A.; Ronco, G.; Senore, C.; Vale, D.B.; Segnan, N.; Tomatis, M.; Soerjomataram, I.; Primic Žakelj, M.; et al. Status of Implementation and Organization of Cancer Screening in The European Union Member States—Summary Results from the Second European Screening Report. Int. J. Cancer 2018, 142, 44–56. [Google Scholar] [CrossRef] [PubMed]
  5. Berry, D.A.; Cronin, K.A.; Plevritis, S.K.; Fryback, D.G.; Clarke, L.; Zelen, M.; Mandelblatt, J.S.; Yakovlev, A.Y.; Habbema, J.D.F.; Feuer, E.J. Effect of Screening and Adjuvant Therapy on Mortality from Breast Cancer. N. Engl. J. Med. 2005, 353, 1784–1792. [Google Scholar] [CrossRef] [PubMed]
  6. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  7. Di Luccio, E.; Morishita, M.; Hirotsu, T.C. Elegans as a Powerful Tool for Cancer Screening. Biomedicines 2022, 10, 2371. [Google Scholar] [CrossRef] [PubMed]
  8. Barsouk, A.; Saginala, K.; Aluru, J.S.; Rawla, P.; Barsouk, A. US Cancer Screening Recommendations: Developments and the Impact of COVID-19. Med. Sci. 2022, 10, 16. [Google Scholar] [CrossRef] [PubMed]
  9. Armaroli, P.; Villain, P.; Suonio, E.; Almonte, M.; Anttila, A.; Atkin, W.S.; Dean, P.B.; de Koning, H.J.; Dillner, L.; Herrero, R.; et al. European Code against Cancer, 4th Edition: Cancer Screening. Cancer Epidemiol. 2015, 39, S139–S152. [Google Scholar] [CrossRef] [PubMed]
  10. US Preventive Services Task Force Screening for Prostate Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2018, 319, 1901–1913. [CrossRef]
  11. Wadsworth, L.P.; Wessman, I.; Björnsson, A.S.; Jonsdottir, G.; Kristinsson, S.Y. The Half-Painted Picture: Reviewing the Mental Health Impacts of Cancer Screening. Medicine 2022, 101, e30479. [Google Scholar] [CrossRef]
  12. Wu, G.X.; Raz, D.J. Lung Cancer Screening. Cancer Treat. Res. 2016, 170, 1–23. [Google Scholar] [CrossRef]
  13. Independent UK Panel on Breast Cancer Screening. The Benefits and Harms of Breast Cancer Screening: An Independent Review. Lancet 2012, 380, 1778–1786. [Google Scholar] [CrossRef]
  14. Barry, M.J.; Simmons, L.H. Prevention of Prostate Cancer Morbidity and Mortality: Primary Prevention and Early Detection. Med. Clin. North. Am. 2017, 101, 787–806. [Google Scholar] [CrossRef] [PubMed]
  15. Geneve, N.; Kairys, D.; Bean, B.; Provost, T.; Mathew, R.; Taheri, N. Colorectal Cancer Screening. Prim. Care 2019, 46, 135–148. [Google Scholar] [CrossRef] [PubMed]
  16. Duffy, M.J. Carcinoembryonic Antigen as a Marker for Colorectal Cancer: Is It Clinically Useful? Clin. Chem. 2001, 47, 624–630. [Google Scholar] [CrossRef] [PubMed]
  17. Ahlquist, D.A. Universal Cancer Screening: Revolutionary, Rational, and Realizable. NPJ Precis. Onc. 2018, 2, 23. [Google Scholar] [CrossRef] [PubMed]
  18. Brito-Rocha, T.; Constâncio, V.; Henrique, R.; Jerónimo, C. Shifting the Cancer Screening Paradigm: The Rising Potential of Blood-Based Multi-Cancer Early Detection Tests. Cells 2023, 12, 935. [Google Scholar] [CrossRef] [PubMed]
  19. Hackshaw, A.; Cohen, S.S.; Reichert, H.; Kansal, A.R.; Chung, K.C.; Ofman, J.J. Estimating the Population Health Impact of a Multi-Cancer Early Detection Genomic Blood Test to Complement Existing Screening in the US and UK. Br. J. Cancer 2021, 125, 1432–1442. [Google Scholar] [CrossRef] [PubMed]
  20. Moleyar-Narayana, P.; Ranganathan, S. Cancer Screening. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  21. Issa, I.A.; Noureddine, M. Colorectal Cancer Screening: An Updated Review of the Available Options. World J. Gastroenterol. 2017, 23, 5086–5096. [Google Scholar] [CrossRef] [PubMed]
  22. Bond, J.H. Fecal Occult Blood Test Screening for Colorectal Cancer. Gastrointest. Endosc. Clin. N. Am. 2002, 12, 11–21. [Google Scholar] [CrossRef] [PubMed]
  23. Meklin, J.; Syrjänen, K.; Eskelinen, M. Colorectal Cancer Screening with Traditional and New-Generation Fecal Immunochemical Tests: A Critical Review of Fecal Occult Blood Tests. Anticancer. Res. 2020, 40, 575–581. [Google Scholar] [CrossRef] [PubMed]
  24. Singh, H.; Nugent, Z.; Demers, A.A.; Kliewer, E.V.; Mahmud, S.M.; Bernstein, C.N. The Reduction in Colorectal Cancer Mortality after Colonoscopy Varies by Site of the Cancer. Gastroenterology 2010, 139, 1128–1137. [Google Scholar] [CrossRef] [PubMed]
  25. Kaminski, M.F.; Regula, J.; Kraszewska, E.; Polkowski, M.; Wojciechowska, U.; Didkowska, J.; Zwierko, M.; Rupinski, M.; Nowacki, M.P.; Butruk, E. Quality Indicators for Colonoscopy and the Risk of Interval Cancer. N. Engl. J. Med. 2010, 362, 1795–1803. [Google Scholar] [CrossRef] [PubMed]
  26. Lakemeyer, L.; Sander, S.; Wittau, M.; Henne-Bruns, D.; Kornmann, M.; Lemke, J. Diagnostic and Prognostic Value of CEA and CA19-9 in Colorectal Cancer. Diseases 2021, 9, 21. [Google Scholar] [CrossRef]
  27. US Preventive Services Task Force; Davidson, K.W.; Barry, M.J.; Mangione, C.M.; Cabana, M.; Caughey, A.B.; Davis, E.M.; Donahue, K.E.; Doubeni, C.A.; Krist, A.H.; et al. Screening for Colorectal Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2021, 325, 1965–1977. [Google Scholar] [CrossRef]
  28. Wolf, A.M.D.; Fontham, E.T.H.; Church, T.R.; Flowers, C.R.; Guerra, C.E.; LaMonte, S.J.; Etzioni, R.; McKenna, M.T.; Oeffinger, K.C.; Shih, Y.-C.T.; et al. Colorectal Cancer Screening for Average-Risk Adults: 2018 Guideline Update from the American Cancer Society. CA Cancer J. Clin. 2018, 68, 250–281. [Google Scholar] [CrossRef]
  29. Ebell, M.H.; Thai, T.N.; Royalty, K.J. Cancer Screening Recommendations: An International Comparison of High Income Countries. Public Health Rev. 2018, 39, 7. [Google Scholar] [CrossRef]
  30. Schneider, R.; Syrogiannouli, L.; Bissig, S.; Scharf, T.; Bulliard, J.-L.; Ducros, C.; Del Giovane, C.; Tal, K.; Zwahlen, M.; Selby, K.; et al. Ten-Year Changes in Colorectal Cancer Screening in Switzerland: The Swiss Health Interview Survey 2007, 2012 and 2017. Prev. Med. Rep. 2022, 27, 101815. [Google Scholar] [CrossRef] [PubMed]
  31. Bretthauer, M.; Løberg, M.; Wieszczy, P.; Kalager, M.; Emilsson, L.; Garborg, K.; Rupinski, M.; Dekker, E.; Spaander, M.; Bugajski, M.; et al. Effect of Colonoscopy Screening on Risks of Colorectal Cancer and Related Death. N. Engl. J. Med. 2022, 387, 1547–1556. [Google Scholar] [CrossRef] [PubMed]
  32. Hoffmeister, M. Interim Evaluation of the Colorectal Cancer Screening Programme in the Netherlands. Lancet Gastroenterol. Hepatol. 2022, 7, 8–9. [Google Scholar] [CrossRef] [PubMed]
  33. Brinkmann, M.; Diedrich, L.; Krauth, C.; Robra, B.-P.; Stahmeyer, J.T.; Dreier, M. General Populations’ Preferences for Colorectal Cancer Screening: Rationale and Protocol for the Discrete Choice Experiment in the SIGMO Study. BMJ Open 2021, 11, e042399. [Google Scholar] [CrossRef] [PubMed]
  34. Blom, J.; Saraste, D.; Törnberg, S.; Jonsson, H. Routine Fecal Occult Blood Screening and Colorectal Cancer Mortality in Sweden. JAMA Netw. Open 2024, 7, e240516. [Google Scholar] [CrossRef] [PubMed]
  35. O’Donoghue, D.; Sheahan, K.; MacMathuna, P.; Stephens, R.B.; Fenlon, H.; Morrin, M.; Mooney, J.; Fahy, L.E.; Mooney, T.; Smith, A. A National Bowel Cancer Screening Programme Using FIT: Achievements and Challenges. Cancer Prev. Res. 2019, 12, 89–94. [Google Scholar] [CrossRef] [PubMed]
  36. Gartlehner, G.; Schernhammer, E.; Lax, S.F.; Preusser, M.; Bachler, H.; Titzer, H.; Kletecka-Pulker, M.; Turnher, H.; Siebert, U. Screening for Colorectal Cancer. Wien. Klin. Wochenschr. 2023, 135, 447–455. [Google Scholar] [CrossRef] [PubMed]
  37. Olesen, T.B.; Jensen, H.; Møller, H.; Jensen, J.W.; Andersen, B.; Rasmussen, M. Nationwide Participation in FIT-Based Colorectal Cancer Screening in Denmark during the COVID-19 Pandemic: An Observational Study. eLife 2023, 12, e81808. [Google Scholar] [CrossRef] [PubMed]
  38. Tran, T.N.; Hoeck, S.; De Schutter, H.; Janssens, S.; Peeters, M.; Van Hal, G. The Impact of a Six-Year Existing Screening Programme Using the Faecal Immunochemical Test in Flanders (Belgium) on Colorectal Cancer Incidence, Mortality and Survival: A Population-Based Study. Int. J. Environ. Res. Public Health 2023, 20, 1654. [Google Scholar] [CrossRef]
  39. Canadian Task Force on Preventive Health Care. Recommendations on Screening for Colorectal Cancer in Primary Care. CMAJ 2016, 188, 340–348. [Google Scholar] [CrossRef]
  40. Clinical Practice Guidelines for the Prevention, Early Detection, and Management of Colorectal Cancer: Population Screening. Available online: https://app.magicapp.org/#/guideline/j1Q1Xj (accessed on 4 March 2024).
  41. Seitz, J.-F.; Lapalus, D.; Arlotto, S.; Gentile, S.; Ettori, F.; Rinaldi, Y.; Grandval, P.; Delasalle, P. Colorectal Cancer Screening by Fecal Immunochemical Test or Colonoscopy in France: How Many People Are Actually Covered? Focus on the Provence-Alpes-Côte d’Azur Region. Eur. J. Gastroenterol. Hepatol. 2022, 34, 405–410. [Google Scholar] [CrossRef]
  42. Saito, Y.; Oka, S.; Kawamura, T.; Shimoda, R.; Sekiguchi, M.; Tamai, N.; Hotta, K.; Matsuda, T.; Misawa, M.; Tanaka, S.; et al. Colonoscopy Screening and Surveillance Guidelines. Dig. Endosc. 2021, 33, 486–519. [Google Scholar] [CrossRef]
  43. Bowel Cancer-UK National Screening Committee (UK NSC)-GOV.UK. Available online: https://view-health-screening-recommendations.service.gov.uk/bowel-cancer/ (accessed on 4 March 2024).
  44. Clinical Practice Guidelines for Bowel Screening in New Zealand—Health New Zealand|Te Whatu Ora. Available online: https://www.tewhatuora.govt.nz/publications/clinical-practice-guidelines-for-bowel-screening-in-new-zealand/ (accessed on 4 March 2024).
  45. Bucchi, L.; Mancini, S.; Baldacchini, F.; Ravaioli, A.; Giuliani, O.; Vattiato, R.; Zamagni, F.; Giorgi Rossi, P.; Campari, C.; Canuti, D.; et al. How a Faecal Immunochemical Test Screening Programme Changes Annual Colorectal Cancer Incidence Rates: An Italian Intention-to-Screen Study. Br. J. Cancer 2022, 127, 541–548. [Google Scholar] [CrossRef]
  46. Nouni-García, R.; Lara-López, Á.; Carratalá-Munuera, C.; Gil-Guillén, V.F.; López-Pineda, A.; Orozco-Beltrán, D.; Quesada, J.A. Factors Associated with Colorectal Cancer Screening in Spain: Results of the 2017 National Health Survey. Int. J. Environ. Res. Public Health 2022, 19, 5460. [Google Scholar] [CrossRef]
  47. Budh, D.P.; Sapra, A. Breast Cancer Screening. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  48. Autier, P.; Boniol, M. Mammography Screening: A Major Issue in Medicine. Eur. J. Cancer 2018, 90, 34–62. [Google Scholar] [CrossRef]
  49. Choi, J.W.; Moon, B.-I.; Lee, J.W.; Kim, H.J.; Jin, Y.; Kim, H.-J. Use of CA15-3 for Screening Breast Cancer: An Antibody-Lectin Sandwich Assay for Detecting Glycosylation of CA15-3 in Sera. Oncol. Rep. 2018, 40, 145–154. [Google Scholar] [CrossRef]
  50. Duffy, M.J.; Evoy, D.; McDermott, E.W. CA 15-3: Uses and Limitation as a Biomarker for Breast Cancer. Clin. Chim. Acta 2010, 411, 1869–1874. [Google Scholar] [CrossRef]
  51. Davisson, L. USPSTF Breast Cancer Screening Guidelines. W. V. Med. J. 2016, 112, 29–31. [Google Scholar]
  52. Oeffinger, K.C.; Fontham, E.T.H.; Etzioni, R.; Herzig, A.; Michaelson, J.S.; Shih, Y.-C.T.; Walter, L.C.; Church, T.R.; Flowers, C.R.; LaMonte, S.J.; et al. Breast Cancer Screening for Women at Average Risk: 2015 Guideline Update from the American Cancer Society. JAMA 2015, 314, 1599–1614. [Google Scholar] [CrossRef]
  53. Draft Recommendation: Breast Cancer: Screening|United States Preventive Services Taskforce. Available online: https://www.uspreventiveservicestaskforce.org/uspstf/draft-recommendation/breast-cancer-screening-adults#bcei-recommendation-title-area (accessed on 4 March 2024).
  54. Saslow, D.; Boetes, C.; Burke, W.; Harms, S.; Leach, M.O.; Lehman, C.D.; Morris, E.; Pisano, E.; Schnall, M.; Sener, S.; et al. American Cancer Society Guidelines for Breast Screening with MRI as an Adjunct to Mammography. CA Cancer J. Clin. 2007, 57, 75–89. [Google Scholar] [CrossRef] [PubMed]
  55. Breast Cancer Risk Assessment and Screening in Average-Risk Women. Available online: https://www.acog.org/clinical/clinical-guidance/practice-bulletin/articles/2017/07/breast-cancer-risk-assessment-and-screening-in-average-risk-women (accessed on 4 March 2024).
  56. Monticciolo, D.L.; Malak, S.F.; Friedewald, S.M.; Eby, P.R.; Newell, M.S.; Moy, L.; Destounis, S.; Leung, J.W.T.; Hendrick, R.E.; Smetherman, D. Breast Cancer Screening Recommendations Inclusive of All Women at Average Risk: Update from the ACR and Society of Breast Imaging. J. Am. Coll. Radiol. 2021, 18, 1280–1288. [Google Scholar] [CrossRef] [PubMed]
  57. Monticciolo, D.L.; Newell, M.S.; Moy, L.; Lee, C.S.; Destounis, S.V. Breast Cancer Screening for Women at Higher-Than-Average Risk: Updated Recommendations from the ACR. J. Am. Coll. Radiol. 2023, 20, 902–914. [Google Scholar] [CrossRef] [PubMed]
  58. Herrmann, C.; Vounatsou, P.; Thürlimann, B.; Probst-Hensch, N.; Rothermundt, C.; Ess, S. Impact of Mammography Screening Programmes on Breast Cancer Mortality in Switzerland, a Country with Different Regional Screening Policies. BMJ Open 2018, 8, e017806. [Google Scholar] [CrossRef] [PubMed]
  59. Bringedal, B.; Fretheim, A.; Nilsen, S.; Isaksson Rø, K. Do You Recommend Cancer Screening to Your Patients? A Cross-Sectional Study of Norwegian Doctors. BMJ Open 2019, 9, e029739. [Google Scholar] [CrossRef] [PubMed]
  60. Kregting, L.M.; Olthof, E.M.G.; Breekveldt, E.C.H.; Aitken, C.A.; Heijnsdijk, E.A.M.; Toes-Zoutendijk, E.; De Koning, H.J.; Van Ravesteyn, N.T. Concurrent Participation in Breast, Cervical, and Colorectal Cancer Screening in the Netherlands. Eur. J. Cancer 2022, 175, 180–186. [Google Scholar] [CrossRef] [PubMed]
  61. Heinig, M.; Schäfer, W.; Langner, I.; Zeeb, H.; Haug, U. German Mammography Screening Program: Adherence, Characteristics of (Non-)Participants and Utilization of Non-Screening Mammography—A Longitudinal Analysis. BMC Public Health 2023, 23, 1678. [Google Scholar] [CrossRef] [PubMed]
  62. Lagerlund, M.; Åkesson, A.; Zackrisson, S. Population-Based Mammography Screening Attendance in Sweden 2017–2018: A Cross-Sectional Register Study to Assess the Impact of Sociodemographic Factors. Breast 2021, 59, 16–26. [Google Scholar] [CrossRef]
  63. O’Sullivan, K.; O’Donovan, A. Factors Associated with Breast Cancer Mammography Screening and Breast Self-Examination in Irish Women: Results from the Irish Longitudinal Study on Ageing (TILDA). Acta Oncol. 2022, 61, 1301–1308. [Google Scholar] [CrossRef]
  64. Schiller-Fruehwirth, I.; Jahn, B.; Einzinger, P.; Zauner, G.; Urach, C.; Siebert, U. The Long-Term Effectiveness and Cost Effectiveness of Organized versus Opportunistic Screening for Breast Cancer in Austria. Value Health 2017, 20, 1048–1057. [Google Scholar] [CrossRef]
  65. Lynge, E.; Bak, M.; von Euler-Chelpin, M.; Kroman, N.; Lernevall, A.; Mogensen, N.B.; Schwartz, W.; Wronecki, A.J.; Vejborg, I. Outcome of Breast Cancer Screening in Denmark. BMC Cancer 2017, 17, 897. [Google Scholar] [CrossRef]
  66. De Troeyer, K.; Silversmit, G.; Rosskamp, M.; Truyen, I.; Van Herck, K.; Goossens, M.M.; Martens, P.; Kellen, E.; Hendrickx, E.; Rummens, E.; et al. The Effect of the Flemish Breast Cancer Screening Program on Breast Cancer-Specific Mortality: A Case-Referent Study. Cancer Epidemiol. 2023, 82, 102320. [Google Scholar] [CrossRef]
  67. Klarenbach, S.; Sims-Jones, N.; Lewin, G.; Singh, H.; Thériault, G.; Tonelli, M.; Doull, M.; Courage, S.; Garcia, A.J.; Thombs, B.D. Recommendations on Screening for Breast Cancer in Women Aged 40–74 Years Who Are Not at Increased Risk for Breast Cancer. CMAJ 2018, 190, E1441–E1451. [Google Scholar] [CrossRef]
  68. Australian Institute of Health and Welfare. BreastScreen Australia Monitoring Report. 2022. Available online: https://www.breastcheck.ie/_fileupload/documents/BC-PR-PM-12-Rev0-BreastCheck-Programme-Report_2018_and_2019.pdf (accessed on 4 March 2024).
  69. Nickson, C.; Velentzis, L.; Brennan, P.; Mann, G.B.; Houssami, N. Improving Breast Cancer Screening in Australia: A Public Health Perspective. Public Health Res. Pract. 2019, 29, 1–7. [Google Scholar] [CrossRef] [PubMed]
  70. Seradour, B. The French breast cancer screening program in 2022: Ensuring progress. Bull Cancer 2022, 109, 756–759. [Google Scholar] [CrossRef] [PubMed]
  71. Uematsu, T.; Izumori, A.; Moon, W.K. Overcoming the Limitations of Screening Mammography in Japan and Korea: A Paradigm Shift to Personalized Breast Cancer Screening Based on Ultrasonography. Ultrasonography 2023, 42, 508–517. [Google Scholar] [CrossRef] [PubMed]
  72. Njor, S.H.; Heinävaara, S.; Stefansdóttir, H.; Nygård, M.; Guðmundsdóttir, E.M.; Bhargava, S.; Leivonen, A.; Campbell, S.; Søborg, B.; Hofvind, S.; et al. Differences in Mammography Screening Attendance among Non-Western Immigrants in Denmark, Finland, Iceland and Norway. Prev. Med. Rep. 2023, 36, 102516. [Google Scholar] [CrossRef] [PubMed]
  73. Taylor-Phillips, S.; Seedat, F.; Kijauskaite, G.; Marshall, J.; Halligan, S.; Hyde, C.; Given-Wilson, R.; Wilkinson, L.; Denniston, A.K.; Glocker, B.; et al. UK National Screening Committee’s Approach to Reviewing Evidence on Artificial Intelligence in Breast Cancer Screening. Lancet Digit. Health 2022, 4, e558–e565. [Google Scholar] [CrossRef]
  74. Taylor, R.; Gregory, M.; Sexton, K.; Wharton, J.; Sharma, N.; Amoyal, G.; Morrell, S. Breast Cancer Mortality and Screening Mammography in New Zealand: Incidence-Based and Aggregate Analyses. J. Med. Screen 2019, 26, 35–43. [Google Scholar] [CrossRef]
  75. Pelullo, C.P.; Cantore, F.; Lisciotto, A.; Di Giuseppe, G.; Pavia, M. Organized Breast and Cervical Cancer Screening: Attendance and Determinants in Southern Italy. Cancers 2021, 13, 1578. [Google Scholar] [CrossRef]
  76. Portero de la Cruz, S.; Béjar, L.M.; Cebrino, J. Temporal Evolution and Associated Factors of Adherence to Mammography Screening among Women in Spain: Results from Two National Health Surveys (2017–2020). Healthcare 2023, 11, 2934. [Google Scholar] [CrossRef] [PubMed]
  77. Ilic, D.; Djulbegovic, M.; Jung, J.H.; Hwang, E.C.; Zhou, Q.; Cleves, A.; Agoritsas, T.; Dahm, P. Prostate Cancer Screening with Prostate-Specific Antigen (PSA) Test: A Systematic Review and Meta-Analysis. BMJ 2018, 362, k3519. [Google Scholar] [CrossRef] [PubMed]
  78. Balk, S.P.; Ko, Y.-J.; Bubley, G.J. Biology of Prostate-Specific Antigen. JCO 2003, 21, 383–391. [Google Scholar] [CrossRef]
  79. David, M.K.; Leslie, S.W. Prostate Specific Antigen. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  80. London, J.W.; Fazio-Eynullayeva, E.; Palchuk, M.B.; Sankey, P.; McNair, C. Effects of the COVID-19 Pandemic on Cancer-Related Patient Encounters. JCO Clin. Cancer Inform. 2020, 657–665. [Google Scholar] [CrossRef] [PubMed]
  81. Jeong, S.H.; Raman, J.D. Impact of the Evolving United States Preventative Services Task Force Policy Statements on Incidence and Distribution of Prostate Cancer over 15 Years in a Statewide Cancer Registry. Prostate Int. 2021, 9, 12–17. [Google Scholar] [CrossRef] [PubMed]
  82. National Lung Screening Trial Research Team; Church, T.R.; Black, W.C.; Aberle, D.R.; Berg, C.D.; Clingan, K.L.; Duan, F.; Fagerstrom, R.M.; Gareen, I.F.; Gierada, D.S.; et al. Results of Initial Low-Dose Computed Tomographic Screening for Lung Cancer. N. Engl. J. Med. 2013, 368, 1980–1991. [Google Scholar] [CrossRef] [PubMed]
  83. Silva, M.; Pastorino, U.; Sverzellati, N. Lung Cancer Screening with Low-Dose CT in Europe: Strength and Weakness of Diverse Independent Screening Trials. Clin. Radiol. 2017, 72, 389–400. [Google Scholar] [CrossRef] [PubMed]
  84. Amicizia, D.; Piazza, M.F.; Marchini, F.; Astengo, M.; Grammatico, F.; Battaglini, A.; Schenone, I.; Sticchi, C.; Lavieri, R.; Di Silverio, B.; et al. Systematic Review of Lung Cancer Screening: Advancements and Strategies for Implementation. Healthcare 2023, 11, 2085. [Google Scholar] [CrossRef]
  85. Bartlett, E.C.; Silva, M.; Callister, M.E.; Devaraj, A. False-Negative Results in Lung Cancer Screening-Evidence and Controversies. J. Thorac. Oncol. 2021, 16, 912–921. [Google Scholar] [CrossRef] [PubMed]
  86. Colson, Y.L.; Shepard, J.-A.O.; Lennes, I.T. New USPSTF Guidelines for Lung Cancer Screening: Better but Not Enough. JAMA Surg. 2021, 156, 513–514. [Google Scholar] [CrossRef] [PubMed]
  87. Wolf, A.M.D.; Oeffinger, K.C.; Shih, T.Y.-C.; Walter, L.C.; Church, T.R.; Fontham, E.T.H.; Elkin, E.B.; Etzioni, R.D.; Guerra, C.E.; Perkins, R.B.; et al. Screening for Lung Cancer: 2023 Guideline Update from the American Cancer Society. CA Cancer J. Clin. 2024, 74, 50–81. [Google Scholar] [CrossRef]
  88. Jansen, E.E.L.; Zielonke, N.; Gini, A.; Anttila, A.; Segnan, N.; Vokó, Z.; Ivanuš, U.; McKee, M.; de Koning, H.J.; de Kok, I.M.C.M.; et al. Effect of Organised Cervical Cancer Screening on Cervical Cancer Mortality in Europe: A Systematic Review. Eur. J. Cancer 2020, 127, 207–223. [Google Scholar] [CrossRef]
  89. Melnikow, J.; Henderson, J.T.; Burda, B.U.; Senger, C.A.; Durbin, S.; Soulsby, M.A. Screening for Cervical Cancer with High-Risk Human Papillomavirus Testing: A Systematic Evidence Review for the U.S. Preventive Services Task Force; U.S. In Preventive Services Task Force Evidence Syntheses, Formerly Systematic Evidence Reviews; Agency for Healthcare Research and Quality: Rockville, MD, USA, 2018. [Google Scholar]
  90. Fontham, E.T.H.; Wolf, A.M.D.; Church, T.R.; Etzioni, R.; Flowers, C.R.; Herzig, A.; Guerra, C.E.; Oeffinger, K.C.; Shih, Y.-C.T.; Walter, L.C.; et al. Cervical Cancer Screening for Individuals at Average Risk: 2020 Guideline Update from the American Cancer Society. CA Cancer J. Clin. 2020, 70, 321–346. [Google Scholar] [CrossRef]
  91. US Preventive Services Task Force; Curry, S.J.; Krist, A.H.; Owens, D.K.; Barry, M.J.; Caughey, A.B.; Davidson, K.W.; Doubeni, C.A.; Epling, J.W.; Kemper, A.R.; et al. Screening for Cervical Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2018, 320, 674–686. [Google Scholar] [CrossRef] [PubMed]
  92. Updated Cervical Cancer Screening Guidelines. Available online: https://www.acog.org/clinical/clinical-guidance/practice-advisory/articles/2021/04/updated-cervical-cancer-screening-guidelines (accessed on 5 March 2024).
  93. Tirri, B.F.; Petignat, P.; Jacot-Guillarmod, M.; Mueller, M.D.; Fehr, M.; Kind, A.B. Recommandations pour la prévention du cancer du col de l’utérus. Avis, D’expert 2018, 50, 1–5. [Google Scholar]
  94. Bjørge, T.; Engesæter, B.; Skare, G.B.; Tropé, A. CervicalScreen Norway—A Screening Programme in Transition. Nor. J. Epidemiol. 2022, 30, 55–60. [Google Scholar] [CrossRef]
  95. Großmann, L.M.; Napierala, H.; Herrmann, W.J. Differences in Breast and Cervical Cancer Screening between West and East Germany: A Secondary Analysis of a German Nationwide Health Survey. BMC Public Health 2023, 23, 1931. [Google Scholar] [CrossRef] [PubMed]
  96. Neis, F.; Holleczek, B.; Henes, M.; Juhasz-Böss, I.; Wallwiener, D.; Neis, K.J. Proposal for a Descriptive and Differentiated Presentation of the Longitudinal Impact of the New Organized Cancer Screening Guideline and HPV Vaccination in Germany. Arch. Gynecol. Obstet. 2022, 307, 1125–1136. [Google Scholar] [CrossRef]
  97. Wang, W.; Arcà, E.; Sinha, A.; Hartl, K.; Houwing, N.; Kothari, S. Cervical Cancer Screening Guidelines and Screening Practices in 11 Countries: A Systematic Literature Review. Prev. Med. Rep. 2022, 28, 101813. [Google Scholar] [CrossRef]
  98. Cervical Cancer Screening|Irish Cancer Society. Available online: https://www.cancer.ie/cancer-information-and-support/cancer-types/cervical-cancer/cervical-cancer-screening (accessed on 5 March 2024).
  99. Brzoska, P.; Wahidie, D.; Yilmaz-Aslan, Y. An Intersectional Perspective on the Utilization of Cervical Cancer Screening among Migrants. A Cross-Sectional Analysis of Survey Data from Austria. Cancers 2021, 13, 6082. [Google Scholar] [CrossRef]
  100. Bonde, J.; Schroll, J.B.; Kj, B. Differentieret implementering af HPV-baseret screening i dansk livmoderhalskræftscreening. Ugeskr. Læger 2022, 184, V04210327. [Google Scholar]
  101. Olesen, T.B.; Jensen, H.; Møller, H.; Jensen, J.W.; Waldstrøm, M.; Andersen, B. Participation in the Nationwide Cervical Cancer Screening Programme in Denmark during the COVID-19 Pandemic: An Observational Study. eLife 2023, 12, e81522. [Google Scholar] [CrossRef]
  102. Dombrowski, C.; Bourgain, C.; Ma, Y.; Meiwald, A.; Pinsent, A.; Weynand, B.; Turner, K.M.; Huntington, S.; Adams, E.J.; Bogers, J.; et al. An Economic Evaluation of Two Cervical Screening Algorithms in Belgium: HR-HPV Primary Compared to HR-HPV and Liquid-Based Cytology (LBC) Co-Testing. Eur. J. Cancer Prev. 2023, 10, 1097. [Google Scholar] [CrossRef]
  103. Canadian Task Force on Preventive Health Care Recommendations on Screening for Cervical Cancer. CMAJ 2013, 185, 35–45. [CrossRef]
  104. Cervical Cancer Screening in Australia and New Zealand. Available online: https://ranzcog.edu.au/wp-content/uploads/2022/05/Cervical-cancer-screening-in-Australia-and-New-Zealand-C-Gyn-19-Amended-July-2020.pdf (accessed on 5 March 2024).
  105. Hamers, F.F.; Poullié, A.-I.; Arbyn, M. Updated Evidence-Based Recommendations for Cervical Cancer Screening in France. Eur. J. Cancer Prev. 2022, 31, 279–286. [Google Scholar] [CrossRef]
  106. Palmer, M.R.; Saito, E.; Katanoda, K.; Sakamoto, H.; Hocking, J.S.; Brotherton, J.M.L.; Ong, J.J. The Impact of Alternate HPV Vaccination and Cervical Screening Strategies in Japan: A Cost-Effectiveness Analysis. Lancet Reg. Health—West. Pac. 2024, 44, 101018. [Google Scholar] [CrossRef]
  107. Cervical Cancer—UK National Screening Committee (UK NSC)—GOV.UK. Available online: https://view-health-screening-recommendations.service.gov.uk/cervical-cancer/ (accessed on 5 March 2024).
  108. Finland: Human Papillomavirus and Related Cancers, Fact Sheet 2023. Fact Sheet. 2023. Available online: https://hpvcentre.net/statistics/reports/FIN_FS.pdf (accessed on 5 March 2024).
  109. Portero de la Cruz, S.; Cebrino, J. Trends and Determinants in Uptake of Cervical Cancer Screening in Spain: An Analysis of National Surveys from 2017 and 2020. Cancers 2022, 14, 2481. [Google Scholar] [CrossRef] [PubMed]
  110. Warnakulasuriya, S.; Kerr, A.R. Oral Cancer Screening: Past, Present, and Future. J. Dent. Res. 2021, 100, 1313–1320. [Google Scholar] [CrossRef] [PubMed]
  111. Nagao, T.; Warnakulasuriya, S. Screening for Oral Cancer: Future Prospects, Research and Policy Development for Asia. Oral Oncol. 2020, 105, 104632. [Google Scholar] [CrossRef]
  112. Sankaranarayanan, R.; Ramadas, K.; Thara, S.; Muwonge, R.; Thomas, G.; Anju, G.; Mathew, B. Long Term Effect of Visual Screening on Oral Cancer Incidence and Mortality in a Randomized Trial in Kerala, India. Oral Oncol. 2013, 49, 314–321. [Google Scholar] [CrossRef] [PubMed]
  113. Ribeiro, M.F.A.; Oliveira, M.C.M.; Leite, A.C.; Bruzinga, F.F.B.; Mendes, P.A.; Grossmann, S.d.M.C.; de Araújo, V.E.; Souto, G.R. Assessment of Screening Programs as a Strategy for Early Detection of Oral Cancer: A Systematic Review. Oral Oncol. 2022, 130, 105936. [Google Scholar] [CrossRef]
  114. Liberto, J.M.; Chen, S.-Y.; Shih, I.-M.; Wang, T.-H.; Wang, T.-L.; Pisanic, T.R. Current and Emerging Methods for Ovarian Cancer Screening and Diagnostics: A Comprehensive Review. Cancers 2022, 14, 2885. [Google Scholar] [CrossRef] [PubMed]
  115. Waleleng, B.J.; Adiwinata, R.; Wenas, N.T.; Haroen, H.; Rotty, L.; Gosal, F.; Rotty, L.; Winarta, J.; Waleleng, A.; Simadibrata, M. Screening of Pancreatic Cancer: Target Population, Optimal Timing and How? Ann. Med. Surg. 2022, 84, 104814. [Google Scholar] [CrossRef] [PubMed]
  116. Rovito, M.J.; Manjelievskaia, J.; Leone, J.E.; Lutz, M.J.; Nangia, A. From ‘D’ to ‘I’: A Critique of the Current United States Preventive Services Task Force Recommendation for Testicular Cancer Screening. Prev. Med. Rep. 2016, 3, 361–366. [Google Scholar] [CrossRef] [PubMed]
  117. D’Souza-Schorey, C.; Clancy, J.W. Tumor-Derived Microvesicles: Shedding Light on Novel Microenvironment Modulators and Prospective Cancer Biomarkers. Genes. Dev. 2012, 26, 1287–1299. [Google Scholar] [CrossRef] [PubMed]
  118. Adams, D.L.; Martin, S.S.; Alpaugh, R.K.; Charpentier, M.; Tsai, S.; Bergan, R.C.; Ogden, I.M.; Catalona, W.; Chumsri, S.; Tang, C.-M.; et al. Circulating Giant Macrophages as a Potential Biomarker of Solid Tumors. Proc. Natl. Acad. Sci. USA 2014, 111, 3514–3519. [Google Scholar] [CrossRef]
  119. Macdonald, I.K.; Parsy-Kowalska, C.B.; Chapman, C.J. Autoantibodies: Opportunities for Early Cancer Detection. Trends Cancer 2017, 3, 198–213. [Google Scholar] [CrossRef]
  120. Vasseur, A.; Kiavue, N.; Bidard, F.; Pierga, J.; Cabel, L. Clinical Utility of Circulating Tumor Cells: An Update. Mol. Oncol. 2021, 15, 1647–1666. [Google Scholar] [CrossRef]
  121. Flores, B.C.T.; Correia, M.P.; Rodríguez, J.G.; Henrique, R.; Jerónimo, C. Bridging the Gaps between Circulating Tumor Cells and DNA Methylation in Prostate Cancer. Cancers 2021, 13, 4209. [Google Scholar] [CrossRef] [PubMed]
  122. Crook, T.; Leonard, R.; Mokbel, K.; Thompson, A.; Michell, M.; Page, R.; Vaid, A.; Mehrotra, R.; Ranade, A.; Limaye, S.; et al. Accurate Screening for Early-Stage Breast Cancer by Detection and Profiling of Circulating Tumor Cells. Cancers 2022, 14, 3341. [Google Scholar] [CrossRef]
  123. Ilie, M.; Hofman, V.; Long-Mira, E.; Selva, E.; Vignaud, J.-M.; Padovani, B.; Mouroux, J.; Marquette, C.-H.; Hofman, P. “Sentinel” Circulating Tumor Cells Allow Early Diagnosis of Lung Cancer in Patients with Chronic Obstructive Pulmonary Disease. PLoS ONE 2014, 9, e111597. [Google Scholar] [CrossRef]
  124. Huang, C.; Ding, S.; Huang, C.; Pan, F.; Liu, X.; Zhang, H.; Zhou, J.; Liang, X.; Wang, X.; Song, P. Distribution and Clinical Analysis of EpCAM+/Vimentin+ Circulating Tumor Cells in High-Risk Population and Cancer Patients. Front. Oncol. 2021, 11, 642971. [Google Scholar] [CrossRef]
  125. Hillig, T.; Horn, P.; Nygaard, A.-B.; Haugaard, A.S.; Nejlund, S.; Brandslund, I.; Sölétormos, G. In Vitro Detection of Circulating Tumor Cells Compared by the CytoTrack and CellSearch Methods. Tumour Biol. 2015, 36, 4597–4601. [Google Scholar] [CrossRef]
  126. Akolkar, D.; Patil, D.; Crook, T.; Limaye, S.; Page, R.; Datta, V.; Patil, R.; Sims, C.; Ranade, A.; Fulmali, P.; et al. Circulating Ensembles of Tumor-Associated Cells: A Redoubtable New Systemic Hallmark of Cancer. Int. J. Cancer 2020, 146, 3485–3494. [Google Scholar] [CrossRef] [PubMed]
  127. Gaya, A.; Crook, T.; Plowman, N.; Ranade, A.; Limaye, S.; Bhatt, A.; Page, R.; Patil, R.; Fulmali, P.; Datta, V.; et al. Evaluation of Circulating Tumor Cell Clusters for Pan-cancer Noninvasive Diagnostic Triaging. Cancer Cytopathol. 2021, 129, 226–238. [Google Scholar] [CrossRef]
  128. Inaba, S.; Shimozono, N.; Yabuki, H.; Enomoto, M.; Morishita, M.; Hirotsu, T.; di Luccio, E. Accuracy Evaluation of the C. Elegans Cancer Test (N-NOSE) Using a New Combined Method. Cancer Treat. Res. Commun. 2021, 27, 100370. [Google Scholar] [CrossRef] [PubMed]
  129. Markou, A.; Tzanikou, E.; Lianidou, E. The Potential of Liquid Biopsy in the Management of Cancer Patients. Semin. Cancer Biol. 2022, 84, 69–79. [Google Scholar] [CrossRef] [PubMed]
  130. Poulet, G.; Massias, J.; Taly, V. Liquid Biopsy: General Concepts. Acta Cytol. 2019, 63, 449–455. [Google Scholar] [CrossRef] [PubMed]
  131. Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid Biopsy Enters the Clinic—Implementation Issues and Future Challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef]
  132. Cohen, S.; Mehrabi, S.; Yao, X.; Millingen, S.; Aikhionbare, F.O. Reactive Oxygen Species and Serous Epithelial Ovarian Adenocarcinoma. Cancer Res. J. 2016, 4, 106–114. [Google Scholar] [CrossRef]
  133. Pons-Belda, O.D.; Fernandez-Uriarte, A.; Diamandis, E.P. Multi Cancer Early Detection by Using Circulating Tumor DNA-The Galleri Test. Reply to Klein et al. The Promise of Multicancer Early Detection. Comment on “Pons-Belda et al. Can Circulating Tumor DNA Support a Successful Screening Test for Early Cancer Detection? The Grail Paradigm. Diagnostics 2021, 11, 2171”. Diagnostics 2022, 12, 1244. [Google Scholar] [CrossRef]
  134. Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Swanton, C.; Seiden, M.V.; Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Smith, D.; Richards, D.; et al. Sensitive and Specific Multi-Cancer Detection and Localization Using Methylation Signatures in Cell-Free DNA. Ann. Oncol. 2020, 31, 745–759. [Google Scholar] [CrossRef]
  135. Klein, E.A.; Richards, D.; Cohn, A.; Tummala, M.; Lapham, R.; Cosgrove, D.; Chung, G.; Clement, J.; Gao, J.; Hunkapiller, N.; et al. Clinical Validation of a Targeted Methylation-Based Multi-Cancer Early Detection Test Using an Independent Validation Set. Ann. Oncol. 2021, 32, 1167–1177. [Google Scholar] [CrossRef]
  136. Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020, 182, 1044–1061.e18. [Google Scholar] [CrossRef] [PubMed]
  137. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed]
  138. Urabe, F.; Kosaka, N.; Ito, K.; Kimura, T.; Egawa, S.; Ochiya, T. Extracellular Vesicles as Biomarkers and Therapeutic Targets for Cancer. Am. J. Physiol.-Cell Physiol. 2020, 318, C29–C39. [Google Scholar] [CrossRef] [PubMed]
  139. Yu, W.; Hurley, J.; Roberts, D.; Chakrabortty, S.K.; Enderle, D.; Noerholm, M.; Breakefield, X.O.; Skog, J.K. Exosome-Based Liquid Biopsies in Cancer: Opportunities and Challenges. Ann. Oncol. 2021, 32, 466–477. [Google Scholar] [CrossRef] [PubMed]
  140. Goldvaser, H.; Gutkin, A.; Beery, E.; Edel, Y.; Nordenberg, J.; Wolach, O.; Rabizadeh, E.; Uziel, O.; Lahav, M. Characterisation of Blood-Derived Exosomal hTERT mRNA Secretion in Cancer Patients: A Potential Pan-Cancer Marker. Br. J. Cancer 2017, 117, 353–357. [Google Scholar] [CrossRef] [PubMed]
  141. Hinestrosa, J.P.; Kurzrock, R.; Lewis, J.M.; Schork, N.J.; Schroeder, G.; Kamat, A.M.; Lowy, A.M.; Eskander, R.N.; Perrera, O.; Searson, D.; et al. Early-Stage Multi-Cancer Detection Using an Extracellular Vesicle Protein-Based Blood Test. Commun. Med. 2022, 2, 29. [Google Scholar] [CrossRef] [PubMed]
  142. Zhang, J.; Shi, J.; Zhang, H.; Zhu, Y.; Liu, W.; Zhang, K.; Zhang, Z. Localized Fluorescent Imaging of Multiple Proteins on Individual Extracellular Vesicles Using Rolling Circle Amplification for Cancer Diagnosis. J. Extracell. Vesicles 2020, 10, 12025. [Google Scholar] [CrossRef]
  143. Liu, C.; Zhao, J.; Tian, F.; Cai, L.; Zhang, W.; Feng, Q.; Chang, J.; Wan, F.; Yang, Y.; Dai, B.; et al. Low-Cost Thermophoretic Profiling of Extracellular-Vesicle Surface Proteins for the Early Detection and Classification of Cancers. Nat. Biomed. Eng. 2019, 3, 183–193. [Google Scholar] [CrossRef]
  144. Yasui, T.; Yanagida, T.; Ito, S.; Konakade, Y.; Takeshita, D.; Naganawa, T.; Nagashima, K.; Shimada, T.; Kaji, N.; Nakamura, Y.; et al. Unveiling Massive Numbers of Cancer-Related Urinary-microRNA Candidates via Nanowires. Sci. Adv. 2017, 3, e1701133. [Google Scholar] [CrossRef]
  145. Kaczor-Urbanowicz, K.E.; Martín Carreras-Presas, C.; Kaczor, T.; Tu, M.; Wei, F.; Garcia-Godoy, F.; Wong, D.T.W. Emerging Technologies for Salivaomics in Cancer Detection. J. Cell Mol. Med. 2017, 21, 640–647. [Google Scholar] [CrossRef]
  146. Daily, A.; Ravishankar, P.; Harms, S.; Klimberg, V.S. Using Tears as a Non-Invasive Source for Early Detection of Breast Cancer. PLoS ONE 2022, 17, e0267676. [Google Scholar] [CrossRef] [PubMed]
  147. Krilaviciute, A.; Heiss, J.A.; Leja, M.; Kupcinskas, J.; Haick, H.; Brenner, H. Detection of Cancer through Exhaled Breath: A Systematic Review. Oncotarget 2015, 6, 38643–38657. [Google Scholar] [CrossRef] [PubMed]
  148. Lidgard, G.P.; Domanico, M.J.; Bruinsma, J.J.; Light, J.; Gagrat, Z.D.; Oldham–Haltom, R.L.; Fourrier, K.D.; Allawi, H.; Yab, T.C.; Taylor, W.R.; et al. Clinical Performance of an Automated Stool DNA Assay for Detection of Colorectal Neoplasia. Clin. Gastroenterol. Hepatol. 2013, 11, 1313–1318. [Google Scholar] [CrossRef] [PubMed]
  149. Redwood, D.G.; Asay, E.D.; Blake, I.D.; Sacco, P.E.; Christensen, C.M.; Sacco, F.D.; Tiesinga, J.J.; Devens, M.E.; Alberts, S.R.; Mahoney, D.W.; et al. Stool DNA Testing for Screening Detection of Colorectal Neoplasia in Alaska Native People. Mayo Clin. Proc. 2016, 91, 61–70. [Google Scholar] [CrossRef] [PubMed]
  150. Imperiale, T.F.; Ransohoff, D.F.; Itzkowitz, S.H.; Levin, T.R.; Lavin, P.; Lidgard, G.P.; Ahlquist, D.A.; Berger, B.M. Multitarget Stool DNA Testing for Colorectal-Cancer Screening. N. Engl. J. Med. 2014, 370, 1287–1297. [Google Scholar] [CrossRef] [PubMed]
  151. Barnell, E.K.; Wurtzler, E.M.; La Rocca, J.; Fitzgerald, T.; Petrone, J.; Hao, Y.; Kang, Y.; Holmes, F.L.; Lieberman, D.A. Multitarget Stool RNA Test for Colorectal Cancer Screening. JAMA 2023, 330, 1760–1768. [Google Scholar] [CrossRef] [PubMed]
  152. Currais, P.; Rosa, I.; Claro, I. Colorectal Cancer Carcinogenesis: From Bench to Bedside. World J. Gastrointest. Oncol. 2022, 14, 654–663. [Google Scholar] [CrossRef] [PubMed]
  153. 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]
  154. 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]
  155. Bryce, A.H.; Thiel, D.D.; Seiden, M.V.; Richards, D.; Luan, Y.; Coignet, M.; Zhang, Q.; Zhang, N.; Hubbell, E.; Kurtzman, K.N.; et al. Performance of a Cell-Free DNA-Based Multi-Cancer Detection Test in Individuals Presenting with Symptoms Suspicious for Cancers. JCO Precis. Oncol. 2023, 7, e2200679. [Google Scholar] [CrossRef]
  156. Bhamani, A.; Horst, C.; Bojang, F.; Quaife, S.L.; Dickson, J.L.; Tisi, S.; Hall, H.; Verghese, P.; Creamer, A.; Prendecki, R.; et al. The SUMMIT Study: Utilising a Written ‘Next Steps’ Information Booklet to Prepare Participants for Potential Lung Cancer Screening Results and Follow-Up. Lung Cancer 2023, 176, 75–81. [Google Scholar] [CrossRef]
  157. Nadauld, L.D.; McDonnell, C.H.; Beer, T.M.; Liu, M.C.; Klein, E.A.; Hudnut, A.; Whittington, R.A.; Taylor, B.; Oxnard, G.R.; Lipson, J.; et al. The PATHFINDER Study: Assessment of the Implementation of an Investigational Multi-Cancer Early Detection Test into Clinical Practice. Cancers 2021, 13, 3501. [Google Scholar] [CrossRef]
  158. Schrag, D.; Beer, T.M.; McDonnell, C.H.; Nadauld, L.; Dilaveri, C.A.; Reid, R.; Marinac, C.R.; Chung, K.C.; Lopatin, M.; Fung, E.T.; et al. Blood-Based Tests for Multicancer Early Detection (PATHFINDER): A Prospective Cohort Study. Lancet 2023, 402, 1251–1260. [Google Scholar] [CrossRef]
  159. Westgate, C.; Salles, G.; Shih, S.; Lopatin, M.; Kurtzman, K.N.; Kappes, K.; Venn, O.; Parekh, S.; Venstrom, J.M.; Younes, A.; et al. Multi-Cancer Early Detection Test Is Sensitive and Accurate in Detecting Shared DNA Methylation Signal in a Variety of Lymphoid and Plasma Cell Neoplasms. Blood 2023, 142, 3646. [Google Scholar] [CrossRef]
  160. Neal, R.D.; Johnson, P.; Clarke, C.A.; Hamilton, S.A.; Zhang, N.; Kumar, H.; Swanton, C.; Sasieni, P. Cell-Free DNA–Based Multi-Cancer Early Detection Test in an Asymptomatic Screening Population (NHS-Galleri): Design of a Pragmatic, Prospective Randomised Controlled Trial. Cancers 2022, 14, 4818. [Google Scholar] [CrossRef]
  161. Sasieni, P.; Smittenaar, R.; Hubbell, E.; Broggio, J.; Neal, R.D.; Swanton, C. Modelled Mortality Benefits of Multi-Cancer Early Detection Screening in England. Br. J. Cancer 2023, 129, 72–80. [Google Scholar] [CrossRef]
  162. Brentnall, A.R.; Mathews, C.; Beare, S.; Ching, J.; Sleeth, M.; Sasieni, P. Dynamic Data-Enabled Stratified Sampling for Trial Invitations with Application in NHS-Galleri. Clin. Trials 2023, 20, 425–433. [Google Scholar] [CrossRef]
  163. Bratulic, S.; Limeta, A.; Dabestani, S.; Birgisson, H.; Enblad, G.; Stålberg, K.; Hesselager, G.; Häggman, M.; Höglund, M.; Simonson, O.E.; et al. Noninvasive Detection of Any-Stage Cancer Using Free Glycosaminoglycans. Proc. Natl. Acad. Sci. USA 2022, 119, e2115328119. [Google Scholar] [CrossRef] [PubMed]
  164. Gao, Q.; Wang, C.; Yang, X.; Fang, S.; Zhang, Y.; Wang, G.; Liu, F.; Wen, X.; Zhao, J.; Zhou, G.; et al. 909P A Multi-Cancer Early Detection Model Based on Liquid Biopsy of Multi-Omics Biomarkers: A Proof of Concept Study (PROMISE Study). Ann. Oncol. 2022, 33, S963–S964. [Google Scholar] [CrossRef]
  165. Gao, Q.; Lin, Y.P.; Li, B.S.; Wang, G.Q.; Dong, L.Q.; Shen, B.Y.; Lou, W.H.; Wu, W.C.; Ge, D.; Zhu, Q.L.; et al. Unintrusive Multi-Cancer Detection by Circulating Cell-Free DNA Methylation Sequencing (THUNDER): Development and Independent Validation Studies. Ann. Oncol. 2023, 34, 486–495. [Google Scholar] [CrossRef] [PubMed]
  166. Gao, Q.; Li, B.; Cai, S.; Xu, J.; Wang, C.; Fang, S.; Qiu, F.; Su, J.; Xu, F.; Wen, X.; et al. LBA3 Early Detection and Localization of Multiple Cancers Using a Blood-Based Methylation Assay (ELSA-Seq). Ann. Oncol. 2020, 31, S1358. [Google Scholar] [CrossRef]
  167. Ris, F.; Hellan, M.; Douissard, J.; Nieva, J.J.; Triponez, F.; Woo, Y.; Geller, D.; Buchs, N.C.; Buehler, L.; Moenig, S.; et al. Blood-Based Multi-Cancer Detection Using a Novel Variant Calling Assay (DEEPGENTM): Early Clinical Results. Cancers 2021, 13, 4104. [Google Scholar] [CrossRef]
  168. Hakim, M.; Billan, S.; Tisch, U.; Peng, G.; Dvrokind, I.; Marom, O.; Abdah-Bortnyak, R.; Kuten, A.; Haick, H. Diagnosis of Head-and-Neck Cancer from Exhaled Breath. Br. J. Cancer 2011, 104, 1649–1655. [Google Scholar] [CrossRef]
  169. Peng, G.; Hakim, M.; Broza, Y.Y.; Billan, S.; Abdah-Bortnyak, R.; Kuten, A.; Tisch, U.; Haick, H. Detection of Lung, Breast, Colorectal, and Prostate Cancers from Exhaled Breath Using a Single Array of Nanosensors. Br. J. Cancer 2010, 103, 542–551. [Google Scholar] [CrossRef]
  170. Park, B.; Shen, S.Y.; Min, J.; Fleshner, N.; Knox, J.; May, T.; Ailles, L.; Newton, Y.; Zhang, J.; Singhania, R.; et al. Abstract 1030: Development of a Genome-Wide Methylome Enrichment Platform for Multi-Cancer Early Detection (MCED). Cancer Res. 2023, 83, 1030. [Google Scholar] [CrossRef]
  171. Nguyen, H.T.-H.; Tran, L.S. Clinical Validation of a ctDNA-Based Assay for Multi-Cancer Detection: An Interim Report from a Vietnamese Longitudinal Prospective Cohort Study of 2795 Participants. JCO Glob. Oncol. 2023, 9, 135. [Google Scholar] [CrossRef]
  172. Tran, L.S.; Nguyen, L.H.D.; Nguyen, T.H.H. LBA2 Analytical and Clinical Validation of a ctDNA-Based Assay for Multi-Cancer Detection. Ann. Oncol. 2023, 34, S1467. [Google Scholar] [CrossRef]
  173. Suo, C.; Zhao, R.; Jiang, Y.; Zhang, Y.; He, Q.; Su, Z.; Liu, R.; Jin, L.; Chen, X. Abstract 4194: The FuSion Project of Pan-Cancer Early Screening in Chinese– An Integrative Study by Fudan University and Singlera. Cancer Res. 2023, 83, 4194. [Google Scholar] [CrossRef]
  174. Chanteloup, G.; Cordonnier, M.; Isambert, N.; Bertaut, A.; Marcion, G.; Garrido, C.; Gobbo, J. Membrane-Bound Exosomal HSP70 as a Biomarker for Detection and Monitoring of Malignant Solid Tumours: A Pilot Study. Pilot. Feasibility Stud. 2020, 6, 35. [Google Scholar] [CrossRef] [PubMed]
  175. Bao, H.; Wang, Z.; Ma, X.; Guo, W.; Zhang, X.; Tang, W.; Chen, X.; Wang, X.; Chen, Y.; Mo, S.; et al. Letter to the Editor: An Ultra-Sensitive Assay Using Cell-Free DNA Fragmentomics for Multi-Cancer Early Detection. Mol. Cancer 2022, 21, 129. [Google Scholar] [CrossRef] [PubMed]
  176. Mazzone, P.J.; Wong, K.-K.; Tsay, J.-C.J.; Pass, H.I.; Vachani, A.; Ryan, A.; Carey, J.; Jakubowski, D.; Wu, T.; Zong, Y.; et al. Abstract 5766: Prospective Evaluation of Cell-Free DNA Fragmentomes for Lung Cancer Detection. Cancer Res. 2023, 83, 5766. [Google Scholar] [CrossRef]
  177. Pommergaard, H.-C. Early Detection of de Novo Cancer in Liver Transplant Recipients—A ScandiaTransplant Collaboration. Available online: https://clinicaltrials.gov/study/NCT05492617 (accessed on 3 March 2024).
  178. Harbinger Health Cancer ORigin Epigenetics-Harbinger Health—Collection of Blood and Tissue Samples From Cancer and Non-Cancer Subjects for Validation of a Novel Blood-Based Multi-Cancer Screening Test. Available online: https://clinicaltrials.gov/study/NCT05435066 (accessed on 3 March 2024).
  179. Nguyen, H.; Raymond, V.M.; Vento-Gaudens, E.; Marino, E.; Lang, K.; Eagle, C. Screening for High Frequency Malignant Disease (SHIELD). JCO 2022, 40, TPS1602. [Google Scholar] [CrossRef]
  180. Raymond, V.; Nguyen, H.; Cotton, L.; Vento-Gaudens, E.; Eagle, C. PP01.20 Trial in Progress: Screening for High Frequency Malignant Disease (SHIELD). J. Thorac. Oncol. 2023, 18, e19. [Google Scholar] [CrossRef]
  181. Raymond, V.M.; Higashi, L.; Marino, E.; Lang, K. Evaluation of the ctDNA LUNAR-2 Test in an Average Patient Screening Episode (ECLIPSE). JCO 2021, 39, TPS142. [Google Scholar] [CrossRef]
  182. Hackshaw, A.; Clarke, C.A.; Hartman, A.-R. New Genomic Technologies for Multi-Cancer Early Detection: Rethinking the Scope of Cancer Screening. Cancer Cell 2022, 40, 109–113. [Google Scholar] [CrossRef] [PubMed]
  183. Shen, S.Y.; Burgener, J.M.; Bratman, S.V.; De Carvalho, D.D. Preparation of cfMeDIP-Seq Libraries for Methylome Profiling of Plasma Cell-Free DNA. Nat. Protoc. 2019, 14, 2749–2780. [Google Scholar] [CrossRef] [PubMed]
  184. Twombly, R. Full-Body CT Screening: Preventing or Producing Cancer? JNCI J. Natl. Cancer Inst. 2004, 96, 1650–1651. [Google Scholar] [CrossRef] [PubMed]
  185. Welch, H.G.; Skinner, J.S.; Schroeck, F.R.; Zhou, W.; Black, W.C. Regional Variation of Computed Tomographic Imaging in the United States and the Risk of Nephrectomy. JAMA Intern. Med. 2018, 178, 221–227. [Google Scholar] [CrossRef]
  186. Tipirneni, K.E.; Rosenthal, E.L.; Moore, L.S.; Haskins, A.D.; Udayakumar, N.; Jani, A.H.; Carroll, W.R.; Morlandt, A.B.; Bogyo, M.; Rao, J.; et al. Fluorescence Imaging for Cancer Screening and Surveillance. Mol. Imaging Biol. 2017, 19, 645–655. [Google Scholar] [CrossRef] [PubMed]
  187. Yu, E.Y.; Bishop, M.; Zheng, B.; Ferguson, R.M.; Khandhar, A.P.; Kemp, S.J.; Krishnan, K.M.; Goodwill, P.W.; Conolly, S.M. Magnetic Particle Imaging: A Novel in Vivo Imaging Platform for Cancer Detection. Nano Lett. 2017, 17, 1648–1654. [Google Scholar] [CrossRef]
  188. Cheng, Z.; Yan, X.; Sun, X.; Shen, B.; Gambhir, S.S. Tumor Molecular Imaging with Nanoparticles. Engineering 2016, 2, 132–140. [Google Scholar] [CrossRef]
  189. Shumilov, E.; Flach, J.; Pabst, T.; Fiedler, M.; Angelillo-Scherrer, A.; Trümper, L.; Joncourt, R.; Kohlmann, A.; Bacher, U. Genetic Alterations Crossing the Borders of Distinct Hematopoetic Lineages and Solid Tumors: Diagnostic Challenges in the Era of High-Throughput Sequencing in Hemato-Oncology. Crit. Rev. Oncol. Hematol. 2018, 126, 64–79. [Google Scholar] [CrossRef] [PubMed]
  190. Tivey, A.; Church, M.; Rothwell, D.; Dive, C.; Cook, N. Circulating Tumour DNA—Looking beyond the Blood. Nat. Rev. Clin. Oncol. 2022, 19, 600–612. [Google Scholar] [CrossRef] [PubMed]
  191. Wen, Q.; Boshier, P.; Myridakis, A.; Belluomo, I.; Hanna, G.B. Urinary Volatile Organic Compound Analysis for the Diagnosis of Cancer: A Systematic Literature Review and Quality Assessment. Metabolites 2020, 11, 17. [Google Scholar] [CrossRef]
  192. Saalberg, Y.; Wolff, M. VOC Breath Biomarkers in Lung Cancer. Clin. Chim. Acta 2016, 459, 5–9. [Google Scholar] [CrossRef]
  193. Feil, C.; Staib, F.; Berger, M.R.; Stein, T.; Schmidtmann, I.; Forster, A.; Schimanski, C.C. Sniffer Dogs Can Identify Lung Cancer Patients from Breath and Urine Samples. BMC Cancer 2021, 21, 917. [Google Scholar] [CrossRef]
  194. Mallafré-Muro, C.; Llambrich, M.; Cumeras, R.; Pardo, A.; Brezmes, J.; Marco, S.; Gumà, J. Comprehensive Volatilome and Metabolome Signatures of Colorectal Cancer in Urine: A Systematic Review and Meta-Analysis. Cancers 2021, 13, 2534. [Google Scholar] [CrossRef]
  195. Lima, A.R.; Pinto, J.; Carvalho-Maia, C.; Jerónimo, C.; Henrique, R.; Bastos, M.d.L.; Carvalho, M.; Guedes de Pinho, P. A Panel of Urinary Volatile Biomarkers for Differential Diagnosis of Prostate Cancer from Other Urological Cancers. Cancers 2020, 12, 2017. [Google Scholar] [CrossRef]
  196. Monteiro, M.; Moreira, N.; Pinto, J.; Pires-Luís, A.S.; Henrique, R.; Jerónimo, C.; Bastos, M. de L.; Gil, A.M.; Carvalho, M.; Guedes de Pinho, P. GC-MS Metabolomics-based Approach for the Identification of a Potential VOC-biomarker Panel in the Urine of Renal Cell Carcinoma Patients. J. Cell Mol. Med. 2017, 21, 2092–2105. [Google Scholar] [CrossRef]
  197. Giró Benet, J.; Seo, M.; Khine, M.; Gumà Padró, J.; Pardo Martnez, A.; Kurdahi, F. Breast Cancer Detection by Analyzing the Volatile Organic Compound (VOC) Signature in Human Urine. Sci. Rep. 2022, 12, 14873. [Google Scholar] [CrossRef]
  198. Iliff, A.J.; Xu, X.Z.S.C. Elegans: A Sensible Model for Sensory Biology. J. Neurogenet. 2020, 34, 347–350. [Google Scholar] [CrossRef] [PubMed]
  199. Girard, L.R.; Fiedler, T.J.; Harris, T.W.; Carvalho, F.; Antoshechkin, I.; Han, M.; Sternberg, P.W.; Stein, L.D.; Chalfie, M. WormBook: The Online Review of Caenorhabditis Elegans Biology. Nucleic Acids Res. 2007, 35, D472–D475. [Google Scholar] [CrossRef]
  200. Bargmann, C.I. Comparative Chemosensation from Receptors to Ecology. Nature 2006, 444, 295–301. [Google Scholar] [CrossRef] [PubMed]
  201. Suzuki, M.; Hattori, Y.; Saito, T.; Harada, Y. Pond Assay for the Sensory Systems of Caenorhabditis Elegans: A Novel Anesthesia-Free Method Enabling Detection of Responses to Extremely Low Chemical Concentrations. Biology 2022, 11, 335. [Google Scholar] [CrossRef]
  202. Hirotsu, T.; Sonoda, H.; Uozumi, T.; Shinden, Y.; Mimori, K.; Maehara, Y.; Ueda, N.; Hamakawa, M. A Highly Accurate Inclusive Cancer Screening Test Using Caenorhabditis Elegans Scent Detection. PLoS ONE 2015, 10, e0118699. [Google Scholar] [CrossRef]
  203. Bargmann, C.I.; Hartwieg, E.; Horvitz, H.R. Odorant-Selective Genes and Neurons Mediate Olfaction in C. Elegans. Cell 1993, 74, 515–527. [Google Scholar] [CrossRef]
  204. Zhong, Q.; Tan, E.K.W.; Martin-Alonso, C.; Parisi, T.; Hao, L.; Kirkpatrick, J.D.; Fadel, T.; Fleming, H.E.; Jacks, T.; Bhatia, S.N. Inhalable Point-of-Care Urinary Diagnostic Platform. Sci. Adv. 2024, 10, eadj9591. [Google Scholar] [CrossRef] [PubMed]
  205. Hubbell, E.; Clarke, C.A.; Aravanis, A.M.; Berg, C.D. Modeled Reductions in Late-Stage Cancer with a Multi-Cancer Early Detection Test. Cancer Epidemiol. Biomark. Prev. 2021, 30, 460–468. [Google Scholar] [CrossRef]
  206. Brill, J.V. Screening for Cancer: The Economic, Medical, and Psychosocial Issues. Am. J. Manag. Care 2020, 26 (Suppl. S14), S300–S306. [Google Scholar]
  207. Tafazzoli, A.; Ramsey, S.D.; Shaul, A.; Chavan, A.; Ye, W.; Kansal, A.R.; Ofman, J.; Fendrick, A.M. The Potential Value-Based Price of a Multi-Cancer Early Detection Genomic Blood Test to Complement Current Single Cancer Screening in the USA. Pharm. Econ. 2022, 40, 1107–1117. [Google Scholar] [CrossRef]
  208. Tomczak, K.; Czerwińska, P.; Wiznerowicz, M. The Cancer Genome Atlas (TCGA): An Immeasurable Source of Knowledge. Contemp. Oncol. 2015, 19, A68–A77. [Google Scholar] [CrossRef] [PubMed]
  209. Goldman, M.; Craft, B.; Swatloski, T.; Ellrott, K.; Cline, M.; Diekhans, M.; Ma, S.; Wilks, C.; Stuart, J.; Haussler, D.; et al. The UCSC Cancer Genomics Browser: Update 2013. Nucleic Acids Res. 2013, 41, D949–D954. [Google Scholar] [CrossRef]
  210. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed]
  211. Chandrashekar, D.S.; Bashel, B.; Balasubramanya, S.A.H.; Creighton, C.J.; Ponce-Rodriguez, I.; Chakravarthi, B.V.S.K.; Varambally, S. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia 2017, 19, 649–658. [Google Scholar] [CrossRef] [PubMed]
  212. Cancer Genome Atlas Research Network; Weinstein, J.N.; Collisson, E.A.; Mills, G.B.; Shaw, K.R.M.; Ozenberger, B.A.; Ellrott, K.; Shmulevich, I.; Sander, C.; Stuart, J.M. The Cancer Genome Atlas Pan-Cancer Analysis Project. Nat. Genet. 2013, 45, 1113–1120. [Google Scholar] [CrossRef] [PubMed]
  213. GTEx Consortium Human Genomics. The Genotype-Tissue Expression (GTEx) Pilot Analysis: Multitissue Gene Regulation in Humans. Science 2015, 348, 648–660. [Google Scholar] [CrossRef] [PubMed]
  214. Barrett, T.; Troup, D.B.; Wilhite, S.E.; Ledoux, P.; Rudnev, D.; Evangelista, C.; Kim, I.F.; Soboleva, A.; Tomashevsky, M.; Edgar, R. NCBI GEO: Mining Tens of Millions of Expression Profiles—Database and Tools Update. Nucleic Acids Res. 2007, 35, D760–D765. [Google Scholar] [CrossRef] [PubMed]
  215. Manoochehri, M.; Wu, Y.; Giese, N.A.; Strobel, O.; Kutschmann, S.; Haller, F.; Hoheisel, J.D.; Moskalev, E.A.; Hackert, T.; Bauer, A.S. SST Gene Hypermethylation Acts as a Pan-Cancer Marker for Pancreatic Ductal Adenocarcinoma and Multiple Other Tumors: Toward Its Use for Blood-Based Diagnosis. Mol. Oncol. 2020, 14, 1252–1267. [Google Scholar] [CrossRef]
  216. Chen, W.; Li, G.; Peng, J.; Dai, W.; Su, Q.; He, Y. Transcriptomic Analysis Reveals That Heat Shock Protein 90α Is a Potential Diagnostic and Prognostic Biomarker for Cancer. Eur. J. Cancer Prev. 2020, 29, 357–364. [Google Scholar] [CrossRef]
  217. Liu, B.; Liu, Y.; Pan, X.; Li, M.; Yang, S.; Li, S.C. DNA Methylation Markers for Pan-Cancer Prediction by Deep Learning. Genes 2019, 10, 778. [Google Scholar] [CrossRef]
Figure 1. Comparison between the present concept of screening versus multi-cancer early detection screening. Inspired by Ahlquist et. al, and redesigned [17].
Figure 1. Comparison between the present concept of screening versus multi-cancer early detection screening. Inspired by Ahlquist et. al, and redesigned [17].
Cancers 16 01191 g001
Table 1. Differences in screening recommendations and adherence for the United States and the United Kingdom, with data regarding adherence, sensitivity, and specificity (data from Supplementary Table S1 from Hackshaw et al. [19]).
Table 1. Differences in screening recommendations and adherence for the United States and the United Kingdom, with data regarding adherence, sensitivity, and specificity (data from Supplementary Table S1 from Hackshaw et al. [19]).
CancerScreening ProcedureScreening Interval% Eligible AdultsAdherenceSensitivitySpecificity
USUKUSUKUSUKUSUKUSUKUSUK
LungLow-dose computed tomographyNo RecommendationAnnuallyNA7%NA14.4%NA84.9%NA87.2%NA
ColorectalCologuardFITEvery 3 yearsEvery 2 years93%68%67.7%57.7%92%79%87%94%
BreastBilateral mammographyAnnuallyEvery 3 years90% of women77% of women77.6%70.5%87%87%89%97.2%
CervicalCytological examination and hrHPV testingEvery 5 yearsEvery 5 years63% of women60% of women95%76.2%95%95%85%85%
Table 2. Comparison between colon cancer screening recommendations for the average-risk population in different high-income countries (inspired by Ebell et al. [29], and updated).
Table 2. Comparison between colon cancer screening recommendations for the average-risk population in different high-income countries (inspired by Ebell et al. [29], and updated).
CountryOrganizationTypeYearTestEligible PopulationRelevant Literature
United StatesUSPSTFNational Guideline Committee2021Shared decision-making between clinician and patient for one of the following strategies:
-
g-FOBT/FIT every year.
-
Stool DNA-FIT every 1–3 years.
-
CT colonography every 5 years.
-
Flexible sigmoidoscopy every 5 years.
-
Flexible sigmoidoscopy every 10 years and annual FIT.
-
Colonoscopy screening every 10 years.
All adults aged 45–75 years; selectively screen adults aged 76–85 years[27]
ACSCancer Society2018
-
g-FOBT/FIT every year.
-
Stool DNA-FIT every 3 years.
-
Colonoscopy every 10 years
-
CT colonography every 5 years.
-
Flexible sigmoidoscopy every 5 years.
All adults aged 45–75; selectively screen adults aged 76–85 years[28]
LuxembourgMinistry of HealthNational Guideline Committee2016FIT every 2 years. Colonoscopy if positive test.All adults between 55–74 years old invited to participateNA
SwitzerlandLeague Against CancerCancer Society2013Colonoscopy every 10 years—g-FOBT every two years.All adults aged 50–69[30]
NorwayCancer Registry of NorwayCancer Society2022FIT every 2 years. Colonoscopy if positive test.All adults aged 55–74[31]
NetherlandsNIPHENational Guideline Committee2019FIT every 2 years.All adults aged 55–75[32]
GermanyFederal Joint CommitteeNational Guideline Committee2019Women, 50–54: annual FIT.
Women >55: FIT every 2 years or colonoscopy twice at an interval of 10 years.
All adults from 55 years onwards[33]
Men, 50–54: annual FIT or colonoscopy twice at an interval of 10 years.
Men, >55: FIT once every 2 years or colonoscopy twice at intervals of 10 years.
SwedenNational Board of Health and WelfareNational Guideline Committee FIT once every 2 years. Colonoscopy if positive test.All adults aged 60–74[34]
IrelandNational Screening ServiceNational Guideline Committee2013Bowel Screen Programme: FIT once every 2 years. Colonoscopy if positive test.All adults aged 59–69[35]
AustriaAustrian Cancer CareCancer Society2023Colonoscopy every 10 years, or FIT every 2 years.All adults 45–75 years[36]
DenmarkNational Board of HealthNational Guideline Committee2014FIT once every 2 years. Colonoscopy if positive test.All adults aged 50–74[37]
BelgiumFoundation Against CancerCancer Society2013FIT once every 2 years. Colonoscopy if positive test.All adults aged 50–74[38]
CanadaCTFPHCNational Guideline Committee201650–59: g-FOBT/FIT every 2 years or flexible sigmoidoscopy every 10 years (weak recommendation).
60–74 years: g-FOBT/FIT every 2 years or flexible sigmoidoscopy every 10 years (strong recommendation).
All adults aged 50–74 years[39]
AustraliaAustralian Government Department of HealthNational Guideline Committee2023FIT once every 2 years. Colonoscopy if positive test.All adults aged 50–74 years[40]
FranceNational Cancer InstituteNational Guideline Committee2015FIT once every 2 years. Colonoscopy if positive test.All adults aged 50–74 years[41]
JapanNational Cancer CenterNational Guideline Committee2016g-FOBT once per year.All adults aged 40 and over[42]
IcelandIcelandic Cancer SocietyCancer Society2015FIT once every 2 years.
Colonoscopy if positive test.
All adults aged 60–69NA
UKUK National Screening CommitteeNational Guideline Committee2020FIT once every 2 years.
Colonoscopy if positive test.
All adults aged 50–74[43]
FinlandCancer Society of FinlandCancer Society2021FIT once every 2 years.
Colonoscopy if positive test.
Adults aged 56–74 years oldNA
New ZealandMinistry of HealthCancer Society2023Time to Screen program offering FIT once every 2 years. Colonoscopy if the test is positiveAdults aged 60–74[44]
ItalyNational Screening ObservatoryNational Guideline Committee2015FIT once every 2 years. Colonoscopy if the test is positive.Adults aged 50–69 years[45]
SpainCancer Strategy of National Health SystemNational Guideline Committee2009FIT once every 2 years.Adults aged 50–69[46]
Abbreviations: CTFPHC = Canadian Task Force on Preventive Health Care; NA = Not available; NIPHE = National Institute for Public Health and the Environment, USPSTF = United Stated Preventive Services Task Force.
Table 3. Comparison between breast cancer screening recommendations in different high-income countries (inspired by Ebell et al. [29], and updated).
Table 3. Comparison between breast cancer screening recommendations in different high-income countries (inspired by Ebell et al. [29], and updated).
CountryOrganizationTypeYearPrimary GroupFurther DiscussionRelevant Literature
United StatesUSPSTFNational Guideline Committee2023 *Women aged 40–75: biennial screening mammographyInsufficient evidence for mammography screening in women >75[53]
Insufficient evidence for breast US, or MRI in women identified with dense breasts on an otherwise negative screening mammogram
ACSCancer Society2015, 2007Women with average risk of breast cancer:
ages 40–45—option to start yearly mammography
ages 45–54—yearly mammography;
ages ≥55—biennial/annual screening;
continue mammography until life expectancy ≥10 years
Annual breast MRI screening in higher-risk patients: BRCA mutation, a first-degree relative of BRCA carrier (untested), the lifetime risk of ≥20–25% (as defined by BRCAPRO/other models), radiation to the chest between 10–30 years, Li–Fraumeni/Cowden/Bannayan–Riley–-Ruvalcaba syndrome and first-degree relatives[52,54]
ACOGSpecialty Society2017, reaffirmed 2021Offer mammography starting at 40 years. Initiate at ages 40–49 years after counseling. Recommend by no later than age 50 if the patient has not already been initiated. Continue until age 75. Beyond 75, consider life expectancy.Annual or biennial screening. Clinical breast examination may be offered for women aged 25–39 years and annually for women 40 years and older.[55]
ACRSpecialty Society2023, 2021Risk assessment by age 25. Annual mammography starting at the age of 40. Screening should continue past age 74 unless severe comorbidities limit life expectancy.Women with genetically based increased risk, or with a calculated lifetime risk of ≥20% are recommended to undergo breast MRI surveillance starting at ages 25–30 and annual mammography starting between ages 25–40, depending on the risk.[56,57]
LuxembourgMinistry of HealthNational Guideline CommitteeNAProgramme Mammographie targets women aged 50–70, inviting them for mammography every 23 months.
SwitzerlandSwiss Cancer LeagueCancer Society2016Mammography once every 2 years for women aged 50–70 (74 in some cantons) [58]
NorwayCancer Registry of NorwayCancer Society2010Mammography once every 2 years for women 50–69 years of age [59]
NetherlandsNIPHENational Guideline Committee2017Mammography once every 2 years for women 50–74 years of age [60]
GermanyFederal Joint CommitteeNational Guideline Committee2015Mammography once every 2 years for women 50–69 years of age [61]
SwedenNational Board of Health and WelfareNational Guideline Committee2013Mammography once every 2 years for women 40–74 years of age [62]
IrelandNational Screening ServiceNational Guideline Committee2015BreastCheck program offers mammography once every 2 years for all women aged 50–69 years. [63]
AustriaAustrian Cancer Aid SocietyCancer Society2014Mammography once every 2 years for women aged 45–69 years [64]
DenmarkNational Board of HealthNational Guideline Committee2014Mammography once every 2 years for women aged 50–69 years [65]
BelgiumFoundation Against CancerCancer Society2017Mammography once every 2 years for women aged 50–69 years [66]
CanadaCTFPHCNational Guideline Committee2011Mammography once every 2–3 years for participants aged 50–74For women aged 40–49 years, recommendation against mammography (shared decision-making with healthcare provider)[67]
AustraliaAustralian Government Department of HealthNational Guideline Committee2015BreastScreen program offers mammography every 2 years for women aged 50–74Program available for women from age 40, with mammography every 2 years[68,69]
FranceNational Cancer InstituteNational Guideline Committee2015, 2016Mammography once every 2 years for participants aged 50–74. Screening also includes manual breast examYearly MRI, mammography, and US for patients from the age of 30 onwards in high-risk cases (genetic factor, family history, personal history of radiation treatments)[70]
JapanNational Cancer CenterNational Guideline Committee2021Mammography once every two years to women aged 40–69 ± manual breast examination [71]
IcelandIcelandic Cancer SocietyCancer SocietyNAMammography once every 2 years for women aged 40–69 years. Invitation for women aged 70–74 to have mammography every three years [72]
UKUK National Screening CommitteeNational Guideline Committee2012Mammography once every 3 years form women aged 50–70 years [73]
FinlandCancer Society of FinlandCancer Society2010Mammography once every 2 years for women aged 50–69 years [72]
New ZealandMinistry of HealthCancer Society2014BreastScreen Aotearoa program offers mammography every two years for women aged 45–69 [74]
ItalyNational Screening ObservatoryNational Guideline Committee2015Mammography once every 2 years for women aged 50–69 years (45–69 in Campania region) [75]
SpainCancer Strategy of National Health SystemNational Guideline Committee2009Mammography once every 2 years for women aged 50–69 years [76]
* Currently a draft recommendation at the time of writing (March 2024). Abbreviations: ACOG = American College of Obstetrics and Gynecology, ACR = American College of Radiology; ACS = American Cancer Society; CTFPHC = Canadian Task Force on Preventive Health Care; NA = Not Available; NIPHE = National Institute for Public Health and the Environment, USPSTF = United Stated Preventive Services Task Force.
Table 4. Comparison between cervical cancer screening recommendations in different high-income countries (inspired by Ebell et al. [29], and updated).
Table 4. Comparison between cervical cancer screening recommendations in different high-income countries (inspired by Ebell et al. [29], and updated).
CountryOrganizationTypeYearScreening MethodEligible PopulationRelevant Literature
United StatesUSPSTFNational Guideline Committee2018Women aged 21–29: cytology alone every 3 years
Women aged 30–65 years: cytology alone every 3 years, with HR-HPV testing alone every 5 years, or co-testing every 5 years
Women between years 21–65[91]
ACSCancer Society2020Screen with HR-HPV testing every 5 years
If not available, co-testing every 5 years, or cytology every 3 years
Women between years 25–65[90]
ACOGSpecialty Society2021Endorses USPSTF recommendationsWomen between years 21–65[92]
LuxembourgMinistry of HealthNational Guideline CommitteeNANANANA
SwitzerlandLeague Against CancerCancer Society2018Women aged 21–29: cytological screening
Women aged 30–70 years: cytological/HR-HPV screening every 3 years
Women between years 21–70[93]
NorwayCancer Registry of NorwayCancer Society2022Cytology screening from the age of 25, HR-HPV testing from the age of 34Women aged 25–69 years[94]
NetherlandsNIPHENational Guideline Committee2021HR-HPV screening every five years between 30–60 yearsWomen aged 30–60[60]
GermanyFederal Joint CommitteeNational Guideline Committee2020Women aged 21–35: annual genital examination, medical history, cytology
Women aged >35 are offered HR-HPV and cytology co-testing
Women aged 20[95,96]
SwedenNational Board of Health and WelfareNational Guideline Committee2015Women aged 23–29: Cytology-based screening once every three years
Women aged 30–49: HR-HPV testing every three years with a cytology co-test at age 41
Women aged 50–64: HR-HPV test every seven years
Women aged 23–64[97]
IrelandNational Screening ServiceNational Guideline Committee2020CervicalCheck program—women aged 25–29 years: cervical screening every 3 years
Women aged 30–65 years: cervical screening every 5 years
Women between 25–65 years of age[98]
AustriaAustrian Cancer CareCancer Society2014Cytology every 3 yearsWomen between 19–69 years of age[99]
DenmarkNational Board of HealthNational Guideline Committee2022Women aged 23–49 years: co-testing every 3 years
Women aged 50–64 years: co-testing every 5 years
Women between 23–64 years of age[100,101]
BelgiumFoundation Against CancerCancer Society2022Cytology tests every 3 years
Women aged 30–64 years—HR-HPV test every five years
Women aged between 25–64 years[102]
CanadaCTFPHCNational Guideline Committee2013Cytology every 3 yearsWomen aged between 25–69 years[103]
AustraliaAustralian Government Department of HealthNational Guideline Committee2022HR-HPV testing every 5 years with cytology for positive testsWomen aged 25–74 years of age[104]
FranceNational Cancer InstituteNational Guideline Committee2022Women 25–30 years: cytology testing every 3 years
Women 30–65 years HR-HPV testing every 5 years
Women aged 25–65 years[105]
JapanNational Cancer CenterNational Guideline Committee2020Women aged 20–69 years: cytology testing once every 2 yearsWomen aged 20–69[106]
IcelandIcelandic Cancer SocietyCancer Society2020Women aged 23–29 years: HR-HPV every three years
Women aged 30–59 years: HR-HPV every five years
Women aged 60–64 years: HR-HPV test—if a test is negative, women are discharged from screening
Women aged 23–64NA
UKUK National Screening CommitteeNational Guideline Committee2019HR-HPV test once every 3 yearsWomen aged 25–64[107]
FinlandCancer Society of FinlandCancer Society2017Cytology once every five years (can be substituted with HR-HPV)Women aged 25–65[108]
New ZealandMinistry of HealthCancer Society2023HR-HPV once every five years (once every 3 years if immunodeficient) screening extended to women aged 70–74 years if unscreened or under-screenedWomen aged 25–69[104]
ItalyNational Screening ObservatoryNational Guideline Committee Women aged 25–30 years: Cytology test once every three years
Women aged 30–65 years: HPV-based screening every five years
Women aged 25–65 years[97]
SpainCancer Strategy of National Health SystemNational Guideline Committee2019Women aged 25–34 years: cytology co-testing once every 3 years
Women aged 35–65 years: HR-HPV testing every five years
Women aged 25–65 years[109]
Abbreviations: ACOG = American College of Obstetricians and Gynecologists; ACS = American Cancer Society; CTFPHC = Canadian Task Force on Preventive Health Care; NA = Not Available; NIPHE = National Institute for Public Health and the Environment.
Table 7. Estimates of screening outcomes for the UK, per year, from Hackshaw et al. [19] with permission from the authors.
Table 7. Estimates of screening outcomes for the UK, per year, from Hackshaw et al. [19] with permission from the authors.
Current Screening OnlyIncremental MCED Test
AnalysisTotal PositivesTrue Positive Cancers (Diagnostic Yield per 1000)True Positive/False Positive RationCancer Detection RateDiagnostic cost per Confirmed Cancer DiagnosisTotal PositivesDiagnostic Yield per 1000True Positive/False Positive RationCancer Detection RateDiagnostic Cost per Confirmed Cancer Diagnosis
100% MCED uptake481,87624,888 (3.43)1:1812%10,452244,15392,817 (4.26)1:1.643%2175
20% decrease cancer incidence in screening population476,89919,910 (2.74)1:239%12,921246,22094,884 (4.35)1:1.644%2134
Difference−4977−4978--246920672067--−41
Prostate cancer added635,79727,754 (2.61%)1:2213%12,917243,58692,250 (4.23%)1:1.643%2187
Difference153,9212866 −2465−567−567--12
50% MCED uptake-----122,07646,409 (4.23)1:1.622%2175
Difference00--0−122,077−46,408--0
25% MCED uptake-----61,03823,204 (4.23)1:1.611%2175
Difference00--0−183,115−69,613--0
30% decline in standard-of-care screening, only receiving MCED test-----76,34630,945 (4.72)1:1.514%1989
Difference-----−167,807−61,872--−186
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Galeș, L.N.; Păun, M.-A.; Anghel, R.M.; Trifănescu, O.G. Cancer Screening: Present Recommendations, the Development of Multi-Cancer Early Development Tests, and the Prospect of Universal Cancer Screening. Cancers 2024, 16, 1191. https://doi.org/10.3390/cancers16061191

AMA Style

Galeș LN, Păun M-A, Anghel RM, Trifănescu OG. Cancer Screening: Present Recommendations, the Development of Multi-Cancer Early Development Tests, and the Prospect of Universal Cancer Screening. Cancers. 2024; 16(6):1191. https://doi.org/10.3390/cancers16061191

Chicago/Turabian Style

Galeș, Laurenția Nicoleta, Mihai-Andrei Păun, Rodica Maricela Anghel, and Oana Gabriela Trifănescu. 2024. "Cancer Screening: Present Recommendations, the Development of Multi-Cancer Early Development Tests, and the Prospect of Universal Cancer Screening" Cancers 16, no. 6: 1191. https://doi.org/10.3390/cancers16061191

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