Fact-checked by Grok 2 weeks ago

Cancer screening

Cancer screening involves the application of diagnostic tests to detect cancer in asymptomatic individuals, with the objective of identifying malignancies at an early, potentially curable stage to reduce morbidity and mortality. The practice targets specific cancer types where evidence demonstrates net benefits, such as breast, cervical, colorectal, and lung cancers, through methods including mammography, Pap smears combined with HPV testing, colonoscopy or stool-based tests, and low-dose computed tomography, respectively. Systematic reviews indicate that effective screening programs can lower cancer-specific mortality—for instance, mammographic screening is associated with approximately a 20% reduction in breast cancer deaths among average-risk women—though absolute risk reductions remain modest, often extending life by months rather than years on average. Major achievements include substantial declines in incidence and mortality for screened cancers following widespread implementation; for example, cervical cancer rates have dropped dramatically due to HPV-based screening preventing progression from precancerous lesions. Current evidence-based guidelines, such as those from the U.S. Preventive Services Task Force, recommend biennial mammography for women aged 40–74, colorectal screening for adults 45–75, and targeted lung screening for high-risk smokers, emphasizing individualized risk assessment to maximize benefits. Controversies center on the balance of benefits against harms, including false-positive results leading to invasive follow-ups, radiation exposure, and particularly overdiagnosis—the detection of indolent tumors that would never progress to cause symptoms or death, potentially subjecting patients to unnecessary treatments with their attendant risks. Peer-reviewed analyses highlight overdiagnosis rates varying by cancer type, such as up to 10–20% in breast screening and higher in prostate-specific antigen testing, which has led to its non-routine recommendation due to insufficient mortality benefits relative to harms like overtreatment of non-lethal cancers. Rigorous evaluation underscores that while screening saves lives for certain high-burden cancers, uncritical expansion without empirical validation can inflate healthcare costs and patient anxiety without proportional gains, necessitating ongoing randomized trial data to refine protocols.

Definition and Principles

Core Concepts

Cancer screening involves the systematic application of tests or examinations to asymptomatic populations to identify occult malignancies or precancerous conditions, enabling earlier intervention that may improve outcomes compared to detection after symptom onset. The primary goal is to reduce cancer-specific mortality by detecting disease during a preclinical phase when curative treatments are feasible, rather than merely advancing the time of diagnosis without altering disease progression. This approach relies on the existence of a detectable latent period in the cancer's natural history, during which the tumor is present but not yet symptomatic or metastatic. Foundational principles for screening programs, codified by Wilson and Jungner in 1968 under World Health Organization auspices, stipulate that the targeted condition must represent a significant public health burden, feature a well-understood natural history with a treatable early stage, and possess a reliable test that is acceptable to the population, sufficiently sensitive to detect most cases, and specific enough to limit false positives. Additional criteria include availability of diagnostic and therapeutic resources, an established policy for managing positives, costs of screening that are economically viable relative to benefits, and implementation as an ongoing process rather than a single event. For cancers, these principles adapt to account for tumor heterogeneity, where screening preferentially identifies slower-growing lesions susceptible to length bias—overrepresentation of indolent tumors that would not have caused harm—and requires validation through randomized trials showing net mortality reduction, as surrogate endpoints like stage shift can mislead due to lead-time bias. Effective screening demands balancing potential benefits, such as decreased incidence of advanced disease, against inherent limitations: tests are probabilistic rather than diagnostic, necessitating confirmatory biopsies that carry risks, and population-level implementation must consider disease prevalence, as benefits diminish in low-incidence groups while harms from overtesting persist. Compliance, equity in access, and ongoing evaluation for evolving evidence are integral, with guidelines from bodies like the U.S. Preventive Services Task Force emphasizing trial-derived mortality data over observational associations.

Historical Development

The concept of systematic cancer screening emerged in the early 20th century, building on advances in cytology and radiology that enabled detection of preclinical disease. Initial efforts focused on accessible sites like the cervix, where exfoliated cells could be examined microscopically. In 1928, George Papanicolaou reported the use of vaginal smears to identify cervical cancer precursors, marking the first practical screening method for a common malignancy. By 1941, clinical studies confirmed the Pap test's efficacy in detecting early-stage lesions, leading to its widespread adoption in the 1940s and 1950s, which correlated with subsequent declines in cervical cancer mortality. Radiological screening advanced concurrently, with mammography originating from Albert Salomon's 1913 examinations of excised breast tissue using X-rays to identify tumors. Practical application for living patients developed in the 1950s and 1960s; by 1962, reports documented detection of occult breast cancers via mammography in over 2,000 screened women. Large-scale trials, such as the Health Insurance Plan study initiated in 1963, provided initial evidence of mortality reduction from biennial mammography in women aged 40-64, spurring national programs like those recommended by the American Cancer Society in 1976. Subsequent decades saw expansion to other cancers, including fecal occult blood testing for colorectal cancer, first evaluated in randomized trials in the 1970s, and prostate-specific antigen (PSA) testing introduced in the 1980s. These developments reflected growing emphasis on randomized controlled trials to validate screening benefits, contrasting earlier intuitive adoption driven by technological feasibility rather than rigorous outcome data. By the late 20th century, screening guidelines from bodies like the U.S. Preventive Services Task Force incorporated trial results, prioritizing tests with demonstrated reductions in cancer-specific mortality.

Evidence Base

Proven Benefits from Trials

Randomized controlled trials (RCTs) provide the strongest evidence for mortality benefits from cancer screening, primarily through reductions in cancer-specific mortality rather than all-cause mortality, which often shows smaller or non-significant effects due to competing risks and long follow-up requirements. Meta-analyses of such trials confirm benefits for colorectal, breast, lung, and cervical cancers under specific conditions, with relative risk reductions typically ranging from 15-30%, though absolute benefits remain modest given low disease incidence in screened populations. These findings derive from high-quality RCTs, such as those evaluating fecal occult blood testing (FOBT) for colorectal cancer and low-dose computed tomography (LDCT) for lung cancer, where screening led to earlier detection of treatable lesions and subsequent mortality declines. For colorectal cancer, eight RCTs of guaiac-based FOBT demonstrated a 16% relative reduction in colorectal cancer mortality (rate ratio 0.84, 95% CI 0.78-0.91) compared to no screening, with benefits persisting over 10-30 years of follow-up; sigmoidoscopy trials similarly showed reductions, though with limited lifetime extension of about 3 months on average. These effects stem from detection of early-stage adenomas and cancers amenable to polypectomy or resection, as evidenced by consistent trial results from the Minnesota and Nottingham studies, among others. Breast cancer screening via mammography has yielded a 25-30% reduction in breast cancer mortality for women aged 50 and older in RCTs, including the Health Insurance Plan trial and Swedish two-county trial, where invitation to screening lowered mortality rates by detecting smaller, node-negative tumors. A meta-analysis of these trials estimated a prevented fraction of mortality of approximately 20%, though benefits diminish for younger or older age groups due to lower incidence or comorbidities. Lung cancer screening with LDCT in high-risk smokers (aged 55-74 with ≥30 pack-years) reduced lung cancer-specific mortality by 20-21% in major RCTs like the National Lung Screening Trial (NLST), which reported 247 fewer lung cancer deaths per 100,000 person-years, and meta-analyses confirming a 17% relative risk reduction across seven trials involving over 84,000 participants. This benefit correlates with reductions in late-stage diagnoses, as screening identifies subsolid nodules for timely intervention. Cervical cancer screening through cytology (Pap smears) or HPV testing has shown mortality reductions in RCTs and trial-based evaluations, with Finnish and other organized programs demonstrating up to 80% decreases in incidence and mortality attributable to detection of pre-invasive lesions, though direct RCT evidence is limited by ethical constraints on unscreened controls. In contrast, prostate cancer screening with PSA testing in RCTs like the European Randomized Study of Screening for Prostate Cancer (ERSPC) showed no significant all-cause or prostate-specific mortality benefit in meta-analyses of five trials, with potential harms outweighing gains. Overall, trial evidence underscores that benefits accrue primarily from reducing advanced-stage disease, with a linear association observed between late-stage incidence drops and mortality reductions across historical RCTs.

Methodological Challenges and Biases

Randomized controlled trials (RCTs) represent the gold standard for assessing cancer screening efficacy, as they mitigate many biases inherent in observational studies by randomizing participants to screened or control groups and measuring outcomes like cancer-specific mortality from the time of randomization. However, even RCTs face challenges, including noncompliance in the screened arm, contamination (where control participants receive screening elsewhere), and insufficient follow-up duration to capture long-term effects, which can underestimate true benefits or mask harms. For instance, lead-time bias—where earlier detection via screening extends apparent survival without altering mortality—is largely avoided in RCTs by using randomization as the baseline, unlike in non-randomized comparisons of screen-detected versus clinically detected cases. Length bias arises because screening preferentially detects slower-growing, less aggressive tumors that are more likely to be asymptomatic and thus have inherently better prognoses, skewing survival estimates upward regardless of intervention. This bias, an extension of which is overdiagnosis (detection of non-progressive lesions that would never cause symptoms or death), persists in RCTs as it reflects the underlying biology of detectable cancers rather than study design flaws. Quantification of overdiagnosis in trials is often imprecise due to inadequate long-term follow-up and failure to account for competing causes of death, with estimates varying widely; for example, systematic reviews highlight methodological inconsistencies in trial designs that inflate or deflate overdiagnosis rates. Observational studies, which form much of the historical evidence base for screening recommendations, are particularly susceptible to self-selection bias, where participants opting for screening differ systematically from non-participants in health behaviors, socioeconomic status, or comorbidities, leading to overestimated mortality reductions. For breast cancer mammography, non-randomized evaluations have reported up to twofold reductions in mortality among screened women, but adjustments for self-selection reveal much smaller effects, often aligning closer to RCT findings of 15-20% reductions in select populations. Such biases underscore the need for caution in extrapolating from cohort or case-control designs, where immortal time bias and healthy screenee effects further confound results. Additional challenges include prognostic selection bias, where screen-detected cases appear less lethal due to detection at earlier stages, independent of screening's causal impact, and the difficulty in distinguishing true shifts in disease natural history from artifacts of detection timing. Trials must extend beyond screening cessation to detect excess diagnoses in screened arms, yet many underpower overdiagnosis assessments or ignore lead-time adjustments, perpetuating uncertainty. Overall, while RCTs provide causal insights, their generalizability is limited by era-specific technologies and populations, and biases like length and overdiagnosis necessitate integrated modeling of preclinical detectable periods to refine estimates.

Harms and Limitations

Overdiagnosis and Overtreatment

Overdiagnosis refers to the detection of cancers through screening that would not have caused symptoms or death during an individual's lifetime, often due to indolent or slow-growing tumors. This phenomenon arises because screening advances the time of diagnosis via lead-time bias, identifying preclinical lesions that may remain harmless if undetected. In randomized controlled trials of cancer screening, overdiagnosis estimates vary widely by cancer type and methodology, ranging from 6% to 67% across programs. For instance, in mammography screening for breast cancer, a Duke Cancer Institute analysis of U.S. data estimated that approximately one in seven detected cases (14%) represents overdiagnosis, particularly of ductal carcinoma in situ (DCIS) lesions that rarely progress. Overdiagnosis directly contributes to overtreatment, as clinicians typically intervene with surgery, radiation, or chemotherapy, exposing patients to risks without altering disease outcomes. In prostate cancer screening using prostate-specific antigen (PSA) tests, the European Randomized Study of Screening for Prostate Cancer (ERSPC) demonstrated a 20% reduction in prostate cancer mortality but highlighted substantial overdiagnosis, with estimates indicating that 50% to 60% of screen-detected cancers may never become clinically significant. Modeling studies calibrated to ERSPC data further quantify overdiagnosis at around 66% in some scenarios, driven by the detection of low-grade, non-lethal tumors. For colorectal cancer screening, overdiagnosis occurs with the identification and removal of polyps or early-stage lesions that would regress or remain asymptomatic; endoscopic resection, while generally safe, carries risks of perforation, bleeding, and subsequent surveillance colonoscopies, amplifying cumulative harms. Overtreatment harms include surgical complications such as incontinence and erectile dysfunction following prostatectomy, mastectomy-related lymphedema in breast cancer, and cardiovascular risks from adjuvant therapies, all inflicted on individuals who would otherwise avoid morbidity. Systematic assessments note that overdiagnosis confounds mortality reductions in trials, as treating pseudodisease does not benefit those with progressive cancers, yet inflates incidence without proportional survival gains. Estimating overdiagnosis remains challenging due to ethical barriers in conducting non-intervention arms and reliance on post-trial incidence trends, which may underestimate rates by ignoring lead-time effects. Critics like H. Gilbert Welch argue that incidental detection via advanced imaging exacerbates overdiagnosis beyond traditional screening, urging risk-benefit discussions to mitigate unnecessary interventions.

False Positives and Psychological Burden

False-positive results in cancer screening arise when tests detect abnormalities suggestive of malignancy that are later confirmed absent upon further evaluation, prompting unnecessary diagnostic workups such as biopsies or imaging. These outcomes impose a psychological burden, including heightened anxiety, cancer-specific worry, and emotional distress, which can persist beyond resolution of the false alarm. In mammography screening, false-positive rates per screening round average 7-12% in organized programs, with cumulative risks reaching 40-60% over 10 annual screens depending on age and breast density. Similar issues occur in prostate-specific antigen (PSA) testing, where up to 70-75% of elevated results prove non-cancerous, often leading to biopsies. Short-term psychological effects are well-documented across screening modalities. Women with false-positive mammograms report significantly elevated anxiety and breast cancer worry immediately following recall, with effects peaking within weeks and often lasting months; one review identified fear, uncertainty, and disrupted daily functioning as common themes. In prostate screening, men experiencing suspicious PSA results followed by benign biopsies exhibit increased general anxiety and prostate cancer-specific fears, comparable to levels in those diagnosed with low-grade disease. Lung cancer screening via low-dose CT also yields short-term negative psychosocial impacts, including greater worry and reduced quality of life at 1 week post-false positive, though many resolve by 1 month. Longer-term consequences remain contentious, with evidence suggesting modest but detectable persistence in cancer-specific domains rather than generalized mental health decrements. A 2010 meta-analysis of mammography false positives found small associations with increased generalized anxiety but stronger links to breast cancer-specific outcomes like intrusive thoughts and avoidance behaviors. Conversely, a 2014 cohort study reported no sustained anxiety or health utility loss beyond 1 year, attributing short-term effects to transient uncertainty. However, a 2023 Danish registry-based analysis linked false-positive mammograms to enduring negative impacts—such as dejection, sleep disturbances, and reduced sense of well-being—12-14 years later, particularly among women requiring invasive follow-up. Systematic reviews highlight that while overall psychological harms are often mild and short-lived, vulnerable subgroups (e.g., those with preexisting anxiety) experience amplified and prolonged effects. Beyond direct emotional toll, false positives can erode trust in screening programs and influence behavior. Multiple studies indicate reduced adherence to subsequent screens, with women facing mammography false positives 10-20% less likely to return for recommended follow-ups, potentially forgoing mortality benefits. This deterrence underscores a trade-off where the psychological burden not only affects individuals but may indirectly amplify net harms by undermining program efficacy. Evidence quality varies, with randomized trial data limited by ethical constraints on measuring harms, leading some reviews to note underreporting in observational studies reliant on self-reported scales.

Direct Procedural Risks

Screening procedures for cancer carry inherent risks stemming from the physical interventions involved, including radiation exposure from imaging techniques and mechanical trauma from invasive methods. These risks, while generally low in absolute terms, can lead to immediate complications such as tissue damage, infection, or secondary malignancies over time. In mammography for breast cancer screening, the primary procedural risk is exposure to ionizing radiation, with each standard two-view exam delivering approximately 4 millisieverts (mSv) to the breast tissue. Cumulative exposure from annual screenings over multiple years has been estimated to induce an additional breast cancer incidence of 20 to 25 cases per 100,000 women screened, though this risk is concentrated in younger women and those with dense breasts due to higher radiosensitivity. Modeling studies project that radiation from digital mammography screening could result in 125 attributable breast cancers and 16 related deaths among 100,000 women screened annually from ages 40 to 74, relative to baseline detection benefits. Additional minor risks include breast compression-induced discomfort or bruising, but severe immediate harms like allergic reactions to contrast (if used) are rare. Colonoscopy, a common direct visualization method for colorectal cancer screening, poses risks of colonic perforation and bleeding due to scope insertion, insufflation, and potential polypectomy during the procedure. Perforation rates in screening contexts range from 0.01% to 0.067%, with an estimated incidence of 3.1 perforations per 10,000 procedures, often requiring surgical intervention. Major bleeding events occur at approximately 14.6 per 10,000 procedures, exacerbated by factors such as older age, therapeutic interventions, or sedation use. Sedation-related cardiopulmonary events, including respiratory depression, add further procedural hazards, though overall severe complication rates remain below 0.2% in asymptomatic screening populations. Low-dose computed tomography (LDCT) for lung cancer screening involves radiation doses of 1.0 to 2.4 mSv per scan, accumulating to an estimated 8 mSv over three annual screens in trial cohorts. This exposure carries a small lifetime attributable cancer risk, modeled at a benefit-to-risk ratio of 16:1 overall, with higher ratios for women due to lower baseline lung cancer incidence. Risks are mitigated by dose-reduction protocols but persist as a stochastic effect, potentially elevating incidence of lung or other radiation-sensitive cancers in heavy smokers. Cervical cancer screening via Pap smear or HPV testing entails minimal procedural risks, primarily limited to transient vaginal spotting, cramping, or rare infection from speculum insertion and cell collection. Serious complications such as significant bleeding or uterine perforation occur in fewer than 0.01% of cases, rendering these harms negligible compared to diagnostic yield. Prostate-specific antigen (PSA) blood testing and digital rectal examination (DRE) for prostate cancer screening involve negligible direct risks, with DRE occasionally causing minor rectal discomfort or rare mucosal trauma but no substantive complication rates documented in large cohorts. Non-invasive stool-based tests for colorectal screening, such as fecal immunochemical testing, avoid procedural risks entirely.

Screening by Cancer Type

Breast Cancer

Breast cancer screening primarily employs mammography to detect non-palpable tumors in asymptomatic women, with the goal of reducing mortality through early intervention. Randomized controlled trials (RCTs), such as the Health Insurance Plan (HIP) study initiated in 1963 and the Swedish Two-County Trial starting in 1977, form the core evidence base. Meta-analyses of these and similar trials indicate a relative risk reduction in breast cancer mortality of 15-20% for women aged 40-74 invited to screening compared to controls, with stronger effects observed in women aged 50-69. Absolute mortality reductions are modest; for instance, biennial screening from ages 50-69 may avert approximately 1-2 deaths per 1,000 women screened over a decade. Despite these benefits, methodological critiques highlight potential biases in early trials, including inadequate randomization and volunteer effects, which Cochrane reviews argue may inflate estimated mortality reductions, particularly for women under 50 where benefits are statistically non-significant or marginal. Overdiagnosis represents a major harm, where indolent cancers unlikely to progress are detected and treated unnecessarily; estimates suggest 15-20% of screen-detected invasive cancers and up to 50% of ductal carcinoma in situ (DCIS) cases are overdiagnosed, leading to mastectomies, radiation, or chemotherapy without survival gain. False-positive results occur in 10-12% of initial mammograms, cumulatively affecting 49% of women over a decade of screening, prompting biopsies, anxiety, and additional imaging. Direct procedural risks from mammography are low, involving minimal radiation exposure equivalent to 2-7 mSv per exam, but cumulative doses raise theoretical concerns for long-term screening. For high-risk women (e.g., BRCA mutation carriers), supplemental MRI detects more cancers but increases overdiagnosis and false positives without proven mortality benefits in RCTs. Current U.S. Preventive Services Task Force (USPSTF) guidelines, updated in 2024, recommend biennial mammography for average-risk women aged 40-74 (Grade B), citing insufficient evidence for those 75 and older or for annual versus biennial intervals. Net benefit diminishes in younger women due to lower incidence, higher false-positive rates from denser breasts, and similar overdiagnosis risks, prompting debates on informed consent emphasizing absolute risks over relative gains.

Cervical Cancer

Cervical cancer screening targets precancerous lesions, primarily cervical intraepithelial neoplasia (CIN), caused by persistent infection with high-risk human papillomavirus (HPV) types, which account for over 99% of cases. The primary methods are cervical cytology (Pap test), which examines cells for abnormalities, and primary HPV testing, which detects oncogenic HPV DNA or RNA. HPV testing demonstrates superior sensitivity for detecting high-grade precancerous lesions compared to cytology alone, enabling risk stratification and longer screening intervals of up to 5 years for HPV-negative women.62218-7/fulltext) Co-testing (HPV plus cytology) or HPV primary with cytology triage is used in many programs to balance sensitivity and specificity. Organized screening programs have substantially reduced cervical cancer incidence and mortality, with observational data from England estimating a 70% prevention of deaths across all ages attributable to screening. Population-based studies and trials, such as the HPV FOCAL randomized controlled trial, confirm that primary HPV screening detects fewer missed precancers over time than cytology, supporting a 60-70% greater protection against invasive cervical cancer.62218-7/fulltext) While randomized trials specifically for cytology are limited, ecological evidence from longstanding programs like those in Nordic countries shows at least 80% reductions in incidence and mortality with regular screening. These benefits stem from identifying and treating regressive or progressive CIN lesions before invasion, with causal links reinforced by the near-elimination of squamous cell carcinomas in screened cohorts. Current U.S. Preventive Services Task Force (USPSTF) guidelines, updated in draft form as of December 2024, recommend cytology every 3 years for women aged 21-29 and, for ages 30-65, primary HPV testing every 5 years, cytology every 3 years, or co-testing every 5 years, with cessation after age 65 for adequately screened women at low risk. The American Cancer Society aligns but starts HPV screening at age 25. Screening before age 21 or after 65 in low-risk individuals lacks evidence of benefit and increases harms. Harms include false positives leading to anxiety and unnecessary colposcopy (affecting 5-10% of screened women), biopsies, and excisional treatments like loop electrosurgical excision procedure (LEEP), which carry risks of bleeding, infection, and future preterm birth (odds ratio 1.7-2.5 for treated CIN). Overtreatment concerns arise from regressing low-grade lesions (CIN1) or even some CIN2 that resolve spontaneously in up to 40-60% of cases, particularly with sensitive HPV testing detecting transient infections. However, overdiagnosis rates remain lower than in breast or prostate screening due to the treatable precancerous nature of lesions and high progression risk for untreated high-grade CIN3 (30-50% to invasion if followed long-term), making net benefits favorable with proper triage protocols.

Colorectal Cancer

Colorectal cancer screening aims to detect adenomatous polyps or early-stage cancers in asymptomatic individuals, primarily through endoscopic or stool-based tests, reducing mortality by enabling polyp removal or early treatment. Randomized controlled trials (RCTs) demonstrate significant efficacy: biennial fecal immunochemical testing (FIT) or guaiac-based fecal occult blood testing (gFOBT) reduces colorectal cancer-specific mortality by 15-33%, as shown in trials like the Minnesota Colon Cancer Control Study. Flexible sigmoidoscopy (FS) screening, performed once or at intervals, lowers incidence and mortality, particularly for distal colorectal cancers, with a 31% mortality reduction in the UK FS trial. Colonoscopy, recommended every 10 years for average-risk adults, detects and removes precancerous lesions throughout the colon; observational data and RCTs like the NordICC trial indicate approximately 15% lower colorectal cancer mortality risk compared to no screening. Major screening methods include stool-based tests (annual FIT or multitarget stool DNA every 3 years), direct visualization via colonoscopy or FS, and CT colonography every 5 years. The U.S. Preventive Services Task Force (USPSTF) recommends screening for adults aged 45-75 years, with a Grade A recommendation for ages 50-75 and Grade B for 45-49, emphasizing that benefits outweigh harms for average-risk individuals. The American Cancer Society similarly advises starting at age 45 for average-risk persons, selecting tests based on patient preferences and local resources. Emerging blood-based tests, approved in recent years, show promise but are considered inferior to established stool or endoscopic options by bodies like the American Society for Gastrointestinal Endoscopy due to lower sensitivity for advanced adenomas. Harms include procedural risks from colonoscopy, such as perforation at rates of 2.3-5.7 per 10,000 procedures and bleeding at similar frequencies, increasing with age and therapeutic interventions. Overdiagnosis arises from removing non-progressive polyps, which constitute a substantial portion of detected lesions; estimates suggest many adenomas identified in screening would not evolve into symptomatic cancers, leading to unnecessary interventions without net clinical benefit. Despite these, meta-analyses confirm net mortality benefits, with RCTs providing robust evidence less prone to biases seen in observational studies for other cancers.

Prostate Cancer

Prostate cancer screening primarily involves measurement of prostate-specific antigen (PSA) levels in the blood, often combined with digital rectal examination (DRE), followed by prostate biopsy if results are suspicious. The PSA test detects elevated levels of this protein produced by prostate cells, which can indicate cancer but also benign conditions like enlargement or infection. Randomized controlled trials, such as the European Randomized Study of Screening for Prostate Cancer (ERSPC) and the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial, provide the primary evidence base, with ERSPC demonstrating a relative reduction in prostate cancer mortality of 20% at 13 years of follow-up in screened men aged 55-69 undergoing PSA testing every 2-4 years. In contrast, the PLCO trial, which included annual PSA and DRE for six years, found no significant mortality reduction after up to 19 years, likely due to high baseline screening contamination in the control group. Adjusted analyses of ERSPC suggest up to a 31% mortality reduction when accounting for noncompliance and contamination. The absolute benefits remain modest, with ERSPC data indicating approximately 1 prostate cancer death prevented per 1,000 men screened over 13 years, though longer follow-up to 16 years shows sustained relative risk reduction without proportional increases in absolute lives saved due to competing causes of death. These gains stem from earlier detection of aggressive tumors amenable to curative treatment, such as radical prostatectomy or radiation, but do not extend to overall mortality reduction, as non-cancer deaths remain unaffected. Empirical data highlight that most prostate cancers are slow-growing and indolent, meaning screening shifts detection toward clinically insignificant disease rather than solely preventing lethal progression. Harms predominate in discussions of PSA screening efficacy, with overdiagnosis rates estimated at 20-50% of detected cases, representing tumors that would not cause symptoms or death within a man's lifetime. In the United States from 1988-2017, modeling suggests 1.5-1.9 million men were overdiagnosed due to PSA screening, leading to unnecessary biopsies and treatments. Overtreatment follows, with 0.9-1.5 million men receiving interventions like surgery or androgen deprivation for indolent cancers, incurring risks of incontinence (up to 20% post-prostatectomy), erectile dysfunction (30-50%), and bowel issues from radiation. Biopsy complications affect 1-2% of procedures, including infections requiring hospitalization (0.5-2.5%), hematuria, hematospermia, and sepsis, with risks elevated in screened populations due to frequent testing. False positives occur in 70-80% of initial abnormal PSA results, prompting anxiety, repeated testing, and invasive follow-up without cancer diagnosis. Major guidelines emphasize shared decision-making over routine screening. The U.S. Preventive Services Task Force (USPSTF) assigns a C recommendation for men aged 55-69, advising individualized discussion of modest benefits against substantial harms, and a D recommendation against screening for those 70 and older. The American Urological Association (AUA) 2023 guidelines endorse PSA screening via shared decision-making for average-risk men 55-69, with earlier initiation (ages 40-45) for high-risk groups like African American men or those with family history, and recommend against routine screening beyond age 75 unless life expectancy exceeds 10 years. The American Cancer Society similarly advocates discussing risks and benefits starting at age 50 for average risk, or 45 for higher risk, with biennial testing preferred over annual to mitigate overdiagnosis. These positions reflect trial data showing net benefit only in select subsets, with causal evidence indicating screening's value hinges on avoiding overtreatment of low-grade disease through tools like active surveillance or multiparametric MRI.

Lung Cancer

Lung cancer screening primarily utilizes low-dose computed tomography (LDCT) scans to detect early-stage tumors in high-risk individuals, particularly long-term smokers. The National Lung Screening Trial (NLST), a randomized controlled trial involving 53,454 participants aged 55-74 with at least a 30 pack-year smoking history, demonstrated that three annual LDCT screenings reduced lung cancer mortality by 20% compared to chest radiography (95% CI, 6.8%-26.7%; 247 vs. 309 deaths per 100,000 person-years). The Dutch-Belgian NELSON trial, screening 15,423 high-risk men and women aged 50-74 with a 15 pack-year history, reported a 24% reduction in lung cancer mortality after volume-based CT screening intervals of 1, 2, and 4 years (hazard ratio 0.76; 95% CI, 0.61-0.94). These trials establish LDCT's efficacy in mortality reduction for selected high-risk groups, though benefits derive from early detection of aggressive cancers rather than indolent ones. Current guidelines from the U.S. Preventive Services Task Force (USPSTF) endorse annual LDCT screening for adults aged 50-80 years with a ≥20 pack-year smoking history who currently smoke or quit within the past 15 years, provided they have no substantial comorbidities limiting life expectancy or ability to undergo curative treatment. The American Cancer Society aligns with similar criteria, emphasizing yearly LDCT for ages 50-80 in this cohort. Screening protocols typically involve nodule assessment using size, volume doubling time, and morphology to guide follow-up, reducing unnecessary interventions; for instance, NELSON's volume-doubling time threshold minimized biopsies for slow-growing nodules. Adherence exceeds 90% in trials, but real-world implementation faces challenges like variable nodule management protocols across centers. Despite mortality benefits, LDCT screening incurs harms, including overdiagnosis of indolent tumors that would not progress to cause death. In NLST, approximately 18% of LDCT-detected lung cancers were potentially overdiagnosed, based on excess incidence without corresponding mortality reduction. Estimates range up to 67% for screen-detected cancers, particularly adenocarcinomas in situ, leading to overtreatment with surgery or radiation that offers no survival gain but introduces morbidity. False-positive rates reached 24.2% in NLST's initial screen, prompting invasive diagnostics like biopsies in 1.1% of participants, with attendant risks of pneumothorax (15% complication rate) and bleeding. Cumulative radiation exposure from annual LDCT (about 1.5 mSv per scan) poses a small long-term cancer risk, estimated at 1-2 additional cases per 1,000 screened over a decade. Psychological distress from false positives persists, with studies noting elevated anxiety levels post-abnormal findings, though most resolve without intervention. Net benefit requires rigorous risk stratification, as extending screening to lower-risk groups dilutes efficacy without proportional harm reduction.

Other Cancers

The United States Preventive Services Task Force (USPSTF) recommends against routine screening for ovarian cancer in asymptomatic women, citing a lack of mortality benefit from randomized trials such as the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial, where multimodal screening with transvaginal ultrasound and CA-125 detected cancers but failed to reduce deaths, while incurring harms including false-positive rates exceeding 10% and leading to unnecessary surgeries with complication rates up to 15%, such as bowel injury or infection. Similar findings emerged from UK Collaborative Trial of Ovarian Cancer Screening, which reported no significant survival advantage after long-term follow-up, with positive predictive values below 50% for invasive cancers prompting oophorectomies in non-cancer cases. For high-risk women with BRCA mutations, risk-reducing salpingo-oophorectomy remains preferred over screening due to superior outcomes in preventing mortality without the diagnostic uncertainties. Pancreatic cancer screening receives an insufficient evidence rating from the USPSTF for the general population, as cohort studies in high-familial-risk groups using CT, MRI, or endoscopic ultrasound detected early lesions but showed no clear impact on survival, with procedural risks including pancreatitis (up to 2-4% for EUS) and radiation exposure from imaging. Guidelines from the American Gastroenterological Association endorse surveillance only for select high-risk individuals, such as those with two or more first-degree relatives affected or germline mutations like BRCA2, initiating MRI or EUS at age 50 or 10 years before the youngest family diagnosis, though evidence derives from non-randomized studies with detection rates of 1-2% yielding resectable cancers but uncertain long-term benefits. For skin cancer, primarily melanoma, the USPSTF issued an I statement in 2023, finding inadequate evidence to assess the balance of benefits and harms from visual skin examinations in asymptomatic adults, as observational data suggest potential early detection but lack randomized trial confirmation of mortality reduction, while false positives prompt biopsies with scarring or infection risks in 1-2% of cases. Screening is selectively advised for fair-skinned individuals with extensive sun exposure or family history, emphasizing self-examination over clinician-led programs due to variable inter-observer reliability. Routine screening for testicular cancer in asymptomatic adolescent or adult males is not recommended by the USPSTF, based on low incidence and evidence that self- or clinician exams do not reduce mortality while increasing unnecessary ultrasounds and orchiectomies from benign findings like epididymitis. Liver cancer screening via ultrasound with or without alpha-fetoprotein is reserved for high-risk groups such as those with cirrhosis or chronic hepatitis B, per American Association for the Study of Liver Diseases guidelines, showing modest survival gains in select cohorts but with false-positive rates over 80% necessitating invasive follow-up. Esophageal and gastric cancer lack general-population screening endorsements, with endoscopy targeted to high-risk populations like those with Barrett's esophagus or endemic regions, where benefits remain unproven in average-risk settings due to procedural complications including perforation (0.1-0.5%). Oral cancer exams are suggested for tobacco or alcohol users, but systematic reviews indicate no mortality benefit in low-risk groups from routine visual inspection. Across these cancers, the absence of large-scale trials demonstrating net benefits underscores prioritization of risk-stratified approaches over broad implementation.

Guidelines and Decision-Making

Organizational Recommendations

The United States Preventive Services Task Force (USPSTF) issues evidence-based recommendations graded A (high certainty of substantial net benefit) to D (recommend against). For breast cancer, it recommends biennial screening mammography for women aged 40 to 74 years (B grade), with insufficient evidence for those 75 and older. Cervical cancer screening is recommended every 3 years with cytology alone for women aged 21 to 29, and every 3 to 5 years with cytology or human papillomavirus testing for those 30 to 65 (A grade), ceasing after hysterectomy or if low risk. Colorectal cancer screening is advised for adults aged 50 to 75 using stool-based or direct visualization tests (A grade), with individualized decisions for ages 45 to 49 and 76 to 85 (C grade). Lung cancer screening with annual low-dose computed tomography is recommended for adults aged 50 to 80 with a 20 pack-year smoking history who currently smoke or quit within 15 years (B grade). Prostate cancer screening with prostate-specific antigen testing receives a C grade for men aged 55 to 69, favoring individualized decisions due to limited net benefit, and D grade against for those 70 and older. The American Cancer Society (ACS) provides guidelines emphasizing early detection for average-risk individuals, updated periodically based on evidence reviews. For breast cancer, women aged 40 to 44 may choose annual mammograms, with annual screening recommended from age 45 and continuing as long as benefits outweigh harms. Cervical cancer screening starts at age 25 with primary HPV testing every 5 years or co-testing every 5 years up to age 65, avoiding routine screening beyond that unless prior inadequate tests. Colorectal cancer screening begins at age 45 using multitarget stool DNA every 3 years, fecal immunochemical test annually, or colonoscopy every 10 years, extending to age 75 with shared decision-making thereafter. Lung cancer screening with low-dose CT is advised annually for ages 50 to 80 with significant smoking history. Prostate cancer screening discussions start at age 50 for average risk, or earlier for higher risk, focusing on informed choice. The National Comprehensive Cancer Network (NCCN) offers guidelines primarily for high-risk populations but includes screening for average risk, drawing from clinical expertise and evidence. Breast cancer screening involves annual mammography starting at age 40, with consideration of MRI for high-risk women. Colorectal screening recommends colonoscopy every 10 years from age 45 for average risk, or stool tests with follow-up. For cervical cancer, it aligns with cytology or HPV-based screening from ages 21 to 65. Lung and prostate guidelines emphasize risk-stratified approaches, including shared decision-making for PSA in men over 50. Internationally, the World Health Organization (WHO) endorses organized screening programs only for breast, cervical, and colorectal cancers where effectiveness is proven and resources allow, cautioning against unproven programs due to potential harms outweighing benefits in low-resource settings. WHO advises against routine screening for other cancers like prostate or lung absent strong evidence of net benefit. Variations among organizations reflect differing emphases on randomized trial data versus observational studies, with USPSTF prioritizing strict net benefit calculations that sometimes lead to more conservative age thresholds compared to ACS.

Individual Risk-Benefit Assessment

Individual risk-benefit assessment for cancer screening requires evaluating a person's specific risk factors, including age, sex, family history, genetic predispositions, lifestyle exposures, and comorbidities, against the potential benefits of early detection—such as reduced cancer-specific mortality—and the harms, including false-positive results, overdiagnosis of indolent lesions, procedural complications, and psychological distress. Risk models, such as the Breast Cancer Risk Assessment Tool (BCRAT) for breast cancer or the PLCOm2012 for lung cancer, quantify lifetime or short-term probabilities of developing invasive disease, enabling tailored recommendations that extend screening intervals for low-risk individuals or intensify them for high-risk ones, thereby optimizing net benefit. For instance, in lung cancer, models incorporating smoking pack-years and duration identify those aged 55-80 with at least a 1.5% six-year risk who derive greater mortality reduction from low-dose CT screening compared to the risks of radiation and unnecessary interventions. Shared decision-making (SDM) is integral to this process, involving clinician-patient discussions of evidence-based outcomes, where benefits like a 20-30% relative reduction in breast cancer mortality from mammography must be weighed against overdiagnosis rates estimated at 15-50% in screened populations, leading to overtreatment without survival gains. SDM interventions have been shown to enhance patient knowledge and align choices with preferences, particularly for prostate cancer screening via PSA testing, where guidelines recommend discussing a potential 20% relative mortality reduction alongside risks of biopsy complications and overdiagnosis of low-grade tumors that may never progress. In colorectal cancer, benefit-harm analyses indicate net positive outcomes over 30 years for most adults starting at age 45-50, but individual factors like frailty or short life expectancy may tip the balance toward harms from colonoscopy, such as perforation (0.05-0.1% risk). Genetic and familial assessments further refine evaluations; for example, carriers of BRCA1/2 mutations face lifetime breast cancer risks exceeding 50-70%, justifying intensified screening like annual MRI from age 25, whereas average-risk individuals may experience harms outweighing benefits if screened prematurely. Emerging personalized approaches, including AI-enhanced models integrating imaging or multi-omics data, promise improved discrimination but require validation to avoid over-reliance on predictive tools with modest c-statistics (typically 0.6-0.8). Ultimately, assessments prioritize empirical trial data over population averages, recognizing that low-risk screening often yields minimal mortality benefits relative to cumulative harms, as evidenced by randomized trials showing overdiagnosis in up to one-third of detected breast cancers without corresponding life-years gained. Clinicians should document these discussions, incorporating patient values to mitigate biases in guideline interpretations that may underemphasize harms in favor of uptake.

Participation and Implementation

Global Attendance Patterns

Global cancer screening attendance varies widely by region, cancer type, and healthcare infrastructure, with participation rates typically exceeding 50% in organized programs in high-income countries (HICs) but falling below 10% in many low- and middle-income countries (LMICs). Data from the International Agency for Research on Cancer's (IARC) CanScreen5 repository, which compiles standardized metrics from over 100 countries for breast, cervical, and colorectal screening, reveal coverage ranging from under 2% in parts of sub-Saharan Africa and South Asia to over 80% in select European nations. These patterns reflect causal factors such as program organization (invitation-based vs. opportunistic), access to diagnostics, and public awareness, rather than inherent population differences. In HICs, breast cancer screening via mammography achieves high uptake where national programs exist; for instance, rates reached 74.4% in Finland and 67.2% in the Netherlands in recent Eurostat data for women aged 50-69. Cervical screening, often using Pap or HPV tests, shows similar trends, with OECD countries averaging 50-70% participation among eligible women. Colorectal screening, primarily through fecal immunochemical testing (FIT) or colonoscopy, lags slightly but exceeds 60% in leaders like Finland (79.4% in 2021). In contrast, LMICs report minimal coverage—e.g., 1.7% for breast in Bangladesh and 2.1% for cervical in Côte d'Ivoire—due to absent infrastructure and competing health priorities, as documented in CanScreen5 analyses.
Cancer TypeHigh Participation ExampleRateLow Participation ExampleRateSource
Breast, UK85.5%Bangladesh1.7%CanScreen5
Cervical~70%Côte d'Ivoire2.1%CanScreen5/OECD
Colorectal79.4%1.0%Global studies
Prostate and lung cancer screening exhibit even patchier global patterns, often opportunistic and guideline-restricted, with low overall uptake outside HICs; for example, PSA testing for prostate varies from routine in the U.S. (affecting ~50% of men over 50) to rare in LMICs. These disparities contribute to later-stage diagnoses in LMICs, where empirical evidence links higher attendance to reduced mortality in screened populations.

Barriers to Effective Screening

Barriers to effective cancer screening encompass patient-level factors, systemic limitations, and inherent test inaccuracies that reduce participation rates and overall utility. Low awareness and knowledge of screening benefits, coupled with emotional deterrents such as fear of results or perceived low personal risk, commonly impede uptake across multiple cancer types. In low- and middle-income countries (LMICs), economic constraints, geographic distance to facilities, and inadequate transportation exacerbate these issues, often resulting in late-stage diagnoses despite available programs. Socioeconomic disparities further compound non-participation, with lower education levels, rural residence, and lack of health insurance correlating to reduced adherence, particularly among ethnic minorities and unmarried individuals. Systemic healthcare challenges hinder screening efficacy, including insufficient monitoring systems for tracking participation, limited trained personnel, and insurance policies that fail to cover costs or provide timely reimbursements. In high-income settings like Germany, even free-of-charge programs face barriers such as opportunity costs and mistrust in public health initiatives, leading to attendance rates below 50% for certain screenings. For vulnerable populations, including refugees and those with intellectual disabilities, cultural stigma, language barriers, and discriminatory practices within healthcare systems further limit access. Test-related limitations undermine screening effectiveness by introducing harms that can deter future participation or cause net detriment. False-positive results trigger prolonged anxiety, depressive symptoms, and existential distress, persisting up to 14 years post-event in mammography screening. Overdiagnosis—detecting indolent cancers that would not progress to cause harm—leads to unnecessary biopsies, treatments, and associated physical, psychological, and financial burdens without improving outcomes. These issues are prevalent in breast, lung, and colorectal screening, where false positives and overdiagnosis rates can exceed 20-50% in some cohorts, eroding public trust and complicating risk-benefit assessments. Addressing these requires balancing detection gains against verifiable harms, as empirical data indicate that unmitigated overdiagnosis inflates perceived benefits while masking true screening value.

Controversies and Critiques

Net Benefit Debates

Debates over the net benefit of cancer screening center on weighing mortality reductions against harms such as overdiagnosis, overtreatment, false-positive results, and psychological distress. Overdiagnosis occurs when screening detects indolent cancers that would not have caused symptoms or death during a patient's lifetime, leading to unnecessary interventions like surgery or radiation that carry risks including morbidity and mortality from treatment itself. Systematic reviews have estimated overdiagnosis rates varying widely by cancer type and screening method; for instance, in breast cancer mammography screening, overdiagnosis may affect 1 in 5 to 1 in 3 detected cases, potentially inflating perceived benefits while imposing lifetime treatment burdens on healthy individuals. These harms can offset gains, as evidenced by modeling studies showing that even modest mortality reductions—such as the approximately 20% relative risk reduction in breast cancer death from screening—may not translate to overall survival gains if overdiagnosis leads to equivalent or greater life-years lost from overtreatment. In prostate cancer screening via PSA testing, long-term trials like the European Randomized Study of Screening for Prostate Cancer have demonstrated a 20% relative reduction in prostate cancer mortality after 16 years, but with substantial overdiagnosis rates exceeding 50% in some cohorts, prompting arguments that harms predominate in low-risk populations due to the slow progression of many detected tumors. Critics contend that contemporary protocols mitigate some overtreatment through active surveillance, yet empirical data from U.S. registries indicate persistent high rates of aggressive therapies, questioning the net positive impact beyond high-risk groups. For lung cancer low-dose CT screening in high-risk smokers, randomized trials report a 20% mortality reduction, but debates persist over false-positive rates (up to 25% per scan) necessitating invasive biopsies with complication risks, alongside incidental findings driving additional costs and anxiety without proportional survival benefits in broader populations. Systematic reviews highlight methodological inconsistencies fueling these debates, with some emphasizing absolute risk reductions (e.g., 1-2 breast cancer deaths averted per 1,000 screened women over a decade) while underreporting harms like radiation-induced cancers or quality-of-life decrements from false alarms. Organizations like the U.S. Preventive Services Task Force (USPSTF) have faced criticism for opaque balancing of benefits and harms, as in their biennial mammography recommendations for women aged 40-74, where evidence reviews acknowledge insufficient data on supplemental screening yet affirm net benefit amid unresolved overdiagnosis estimates. Proponents of screening argue that trial data from high-income settings understate benefits in diverse populations, while skeptics, drawing from first-principles analysis of lead-time and length biases, assert that inflated early-stage detections mask true efficacy, advocating personalized risk stratification over population-wide programs to maximize net gains. Recent updates, such as 2024 systematic reviews, reinforce that while screening reduces site-specific mortality, all-cause mortality improvements remain elusive, underscoring the need for refined eligibility criteria to avoid net harm in low-prevalence groups.

Policy and Ethical Concerns

Cancer screening policies often prioritize population-level mortality reductions, yet they raise concerns about individual harms from overdiagnosis and overtreatment, where indolent lesions are detected and treated unnecessarily, potentially affecting 20-50% of screen-detected prostate cancers and 19% of breast cancers. For instance, the European Randomized Study of Screening for Prostate Cancer (ERSPC) reported that while screening reduced prostate cancer mortality by about 20% over 13 years, it led to overdiagnosis in up to 50% of cases, resulting in treatments like radical prostatectomy that carry risks of incontinence and impotence without extending life. Policymakers face pressure to standardize recommendations, as seen in discrepancies between the U.S. Preventive Services Task Force (USPSTF), which advises against routine prostate-specific antigen (PSA) screening for most men due to harms outweighing benefits, and more aggressive endorsements from organizations like the American Cancer Society, highlighting how policy inertia can perpetuate interventions with marginal net gains. Ethical tensions center on non-maleficence and autonomy, as screening programs can inflict harms—such as anxiety from false positives (occurring in 10-20% of mammograms) and downstream procedures—without adequate disclosure, undermining informed consent. Critics argue that consent processes often emphasize benefits like early detection while downplaying overdiagnosis risks, which systematic reviews estimate at 6-67% across screening modalities, leading to overtreatment that violates the principle of "first, do no harm." In resource-limited settings, policies divert funds from proven treatments to screening infrastructure, exacerbating inequities; for example, low-income and racial minority populations in the U.S. exhibit 10-20% lower screening adherence for breast and colorectal cancers, compounded by access barriers rather than individual choice. Conflicts of interest further complicate guideline development, with studies indicating that panel members' financial ties to industry correlate with stronger screening endorsements, as observed in mammography recommendations where authors with industry funding were more likely to favor routine use despite evidence of limited mortality benefits. While some analyses find minimal direct influence on consensus statements, calls for stricter recusal policies persist, given that pharmaceutical and device manufacturers profit from diagnostic follow-ups and adjuvant therapies spurred by screening findings. Ethically, this underscores the need for transparency and independent oversight to ensure policies reflect causal evidence of net benefit rather than economic incentives, particularly when academic and media sources may underemphasize harms to align with public health optimism.

Future Directions

Multi-Cancer Detection Innovations

Multi-cancer early detection (MCED) tests represent a class of innovations designed to identify signals from multiple cancer types through a single non-invasive sample, typically blood, by analyzing circulating biomarkers such as cell-free DNA methylation patterns, protein markers, or multi-omics data. These tests aim to complement or expand beyond organ-specific screenings like mammography or colonoscopy, targeting cancers without routine screening options, which account for over 70% of cancer deaths. Unlike traditional methods, MCED approaches leverage machine learning algorithms to integrate disparate signals, potentially enabling earlier intervention, though large-scale randomized controlled trials demonstrating mortality reductions remain ongoing or pending. The GRAIL Galleri test, a laboratory-developed test (LDT) analyzing plasma cell-free DNA for methylation signatures across over 50 cancer types, has advanced through interventional studies. In the PATHFINDER 2 trial, reported on October 17, 2025, adding Galleri to standard U.S. Preventive Services Task Force-recommended screenings increased overall cancer detection more than seven-fold, with 73.7% episode sensitivity for 12 cancers responsible for two-thirds of U.S. cancer mortality; over half of detected cases were early-stage. The test identified a cancer signal in 0.93% of 6,587 asymptomatic participants aged 50+, confirming malignancy in 89% of signals via diagnostic workup. Despite these results, Galleri lacks FDA approval and operates under Clinical Laboratory Improvement Amendments (CLIA) regulation; GRAIL plans to submit for premarket approval using PATHFINDER 2 and NHS-Galleri data expected in 2026. Independent analyses note persistent challenges, including false-positive rates around 0.5-1% necessitating further biopsies, and the absence of direct evidence for improved survival outcomes. Other notable MCED innovations include Exact Sciences' Cancerguard, launched September 10, 2025, as an LDT detecting signals from cancers causing over 80% of annual U.S. deaths via blood-based multi-biomarker analysis. Guardant Health's Shield MCED test received FDA Breakthrough Device Designation on June 3, 2025, for its blood test targeting multiple cancers through cell-free DNA and other analytes, positioning it for expedited review pending clinical validation. Emerging AI-integrated tests, such as OncoSeek, validated in a multi-center study published October 8, 2025, combine seven protein biomarkers with machine learning for pan-cancer detection, achieving reported sensitivities exceeding 90% for certain stages in validation cohorts. Similarly, the Carcimun test demonstrated high specificity and sensitivity in a March 6, 2025, evaluation, minimizing false results across diverse cancers. As of October 2025, no MCED tests hold FDA approval for screening, limiting their use to high-risk or symptomatic contexts under CLIA oversight, with calls for Medicare coverage tied to proven net benefits. The global MCED market, valued at $1 billion in 2024, is projected to reach $4.3 billion by 2033, driven by technological refinements, yet experts emphasize the need for pragmatic trials to address overdiagnosis risks and cost-effectiveness, as early detection does not invariably translate to reduced mortality without causal reductions in late-stage incidence.

Technological Advances

Artificial intelligence (AI) has enhanced the precision of imaging-based cancer screening modalities, particularly in breast and lung cancer detection. In mammography, AI algorithms serve as independent second readers, improving the detection of clinically significant cancers while reducing false positives; a 2025 simulation study demonstrated that AI integration could increase screening efficiency by prioritizing high-risk cases for radiologist review. Similarly, the U.S. Food and Drug Administration (FDA) authorized an AI tool in 2025 capable of identifying subtle mammographic signs of breast cancer undetectable by human observers alone, potentially elevating early detection rates. For lung cancer, low-dose computed tomography (LDCT) screening benefits from AI models that analyze scan patterns to stratify risk, incorporating biomarkers like neutrophil-to-lymphocyte ratios for refined predictions. Liquid biopsy technologies, leveraging circulating tumor DNA (ctDNA) and other biomarkers in blood, represent a shift toward non-invasive, multi-analyte screening approaches. Advances in 2024-2025 have improved sensitivity for early-stage detection through methylation and fragmentation analysis of cell-free DNA, enabling real-time monitoring with reduced procedural risks compared to tissue biopsies. These methods detect genomic alterations associated with multiple cancers, with ongoing refinements addressing challenges like low ctDNA abundance in preclinical stages. Multi-cancer early detection (MCED) tests exemplify the convergence of liquid biopsy and AI, aiming to screen for over 50 cancer types from a single blood draw. The Galleri test, for instance, demonstrated in the 2025 PATHFINDER 2 study a seven-fold increase in cancer detection when added to standard USPSTF-recommended screenings for high-risk adults aged 50 and older, though it identifies signals rather than all cancers and requires confirmatory diagnostics. As of November 2024, no MCED tests have received FDA approval for population-wide screening, but initiatives like ARPA-H's POSEIDON program are developing at-home versions targeting stage I detection with high specificity. Biosensor integrations with AI further promise wearable devices for continuous biomarker tracking, though validation for screening efficacy remains pending large-scale trials.

References

  1. [1]
    Cancer Screening Overview - NCI - National Cancer Institute
    Oct 20, 2023 · Cancer screening is looking for cancer before a person has any symptoms. Screening tests can help find cancer at an early stage, before symptoms appear.
  2. [2]
    Cancer Screening Tests - CDC
    Jan 16, 2025 · CDC supports screening for breast, cervical, colorectal (colon), and lung cancers as recommended by the US Preventive Services Task Force (USPSTF).
  3. [3]
    Benefits and Harms of Breast Cancer Screening: A Systematic Review
    Oct 20, 2015 · This systematic review reports that among US women at average risk for breast cancer, mammographic screening is associated with reduction in ...
  4. [4]
    Estimated Lifetime Gained With Cancer Screening Tests
    The findings of this meta-analysis suggest that colorectal cancer screening with sigmoidoscopy may extend life by approximately 3 months.
  5. [5]
    Cervical Cancer Screening - NCI
    Feb 13, 2025 · The goal of screening for cervical cancer is to find precancerous cervical cell changes, when treatment can prevent cervical cancer from developing.
  6. [6]
    Recommendation: Breast Cancer: Screening - uspstf
    Apr 30, 2024 · The USPSTF recommends biennial screening mammography for women aged 40 to 74 years. B. Women 75 years or older, The USPSTF concludes that the ...Screening · Evidence Summary · Screening, 1996 · Screening, 2002
  7. [7]
    Cancer overdiagnosis: A challenge in the era of screening - PMC
    We provide here a qualitative review of cancer overdiagnosis and discuss specific examples due to extensive population-based screening.
  8. [8]
    Overdiagnosis of invasive breast cancer in population-based breast ...
    The risk of an overdiagnosed invasive breast cancer is < 1 in 1000 biennially screened women aged 50–69 with a 10-year follow-up time after screening stops.
  9. [9]
    Evidence categories in systematic assessment of cancer ...
    Cancer overdiagnosis occurs when a malignant tumour—that would never lead to any harm if left undetected—is found. This has been reported for several cancer ...
  10. [10]
    Cancer screening metrics: effective evaluation to balance benefits ...
    Apr 8, 2024 · Cancer screening metrics: effective evaluation to balance benefits and harms ... cancer screening: a systematic review and meta-analysis
  11. [11]
    Definition of screening - NCI Dictionary of Cancer Terms
    Checking for disease when there are no symptoms. Since screening may find diseases at an early stage, there may be a better chance of curing the disease.
  12. [12]
    Cancer Screening - StatPearls - NCBI Bookshelf - NIH
    Cancer screening is crucial in early detection and intervention for various types of cancer, including colon, lung, cervical, breast, and prostate cancer.Continuing Education Activity · Introduction · Function · Issues of Concern
  13. [13]
    Principles of Cancer Screening: Lessons from History and Study ...
    Cancer screening tests are more effective at identifying slower-growing lesions than rapidly advancing ones. Every tumor has its own window of time during which ...
  14. [14]
    Revisiting Wilson and Jungner in the genomic age - PubMed Central
    Box 1. Wilson and Jungner classic screening criteria. · 1. The condition sought should be an important health problem. · 2. There should be an accepted treatment ...
  15. [15]
    [PDF] PRINCIPLES AND PRACTICE OF SCREENING FOR DISEASE
    WILSON & JUNGNER. PRESENT SCREENING PRACTICE (INCLUDING MULTIPLE SCREENING) ... screening tests for all ages in pointing to unsuspected disease; Jungner ...<|separator|>
  16. [16]
    Principles of Cancer Screening - PMC - NIH
    A general principle of cancer screening is that the net benefit (benefits minus harms) of screening generally increases as the incidence rate of the cancer of ...
  17. [17]
    Cancer Screening Overview (PDQ®) - NCBI - NIH
    Oct 16, 2023 · This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about cancer screening.
  18. [18]
    Milestones in Cancer Research and Discovery - NCI
    Apr 18, 2025 · 1928: The Pap Smear​​ George Papanicolaou discovers that cervical cancer can be detected by examining cells from the vagina under a microscope. ...
  19. [19]
    The history of the Papanicolaou smear and the odyssey of ... - PubMed
    The Papanicolaou smear, a routine screening test for cancer of the uterine cervix, was reported in 1928, and its efficacy was proved by 1941.
  20. [20]
    The history of mammography - Siemens Healthineers MedMuseum
    Jul 1, 2022 · In 1913, the surgeon Albert Salomon started to examine diseases of the breast by using X-rays. As X-ray equipment was not designed for imaging breast tissue.
  21. [21]
    Mammography: a history of success and scientific enthusiasm - PMC
    In 1962, this author reports the first 53 cases of occult breast cancer detected at 2,000 mammograms. At that same time, John Martin and colleagues demonstrated ...
  22. [22]
    History of ACS Recommendations for the Early Detection of Cancer ...
    Nov 1, 2023 · The following tables give the history of cancer detection tests that have been recommended by the American Cancer Society for people who are at average risk ...
  23. [23]
    Noncancer-Related Mortality in Randomized Clinical Trials: A Meta ...
    Aug 25, 2025 · This meta-analysis assesses the association between cancer screening and noncancer-related mortality between screened and unscreened ...
  24. [24]
    What do Cochrane systematic reviews say about the clinical ...
    Meta-analyses on four RCTs showed that participants allocated to FOBT screening had a reduction in the risk of colorectal cancer mortality of 16%, compared with ...
  25. [25]
    Impact of low‐dose computed tomography (LDCT) screening on ...
    Aug 3, 2022 · The current evidence supports a reduction in lung cancer‐related mortality with the use of LDCT for lung cancer screening in high‐risk ...
  26. [26]
    Strong association between reduction of late-stage cancers and ...
    May 26, 2024 · Meta-regression of historical screening RCTs showed a strong linear association between reductions in late-stage cancer incidence and cancer mortality.
  27. [27]
    Review How effective are breast cancer screening programmes by ...
    Randomised controlled trials showed that breast cancer screening by mammography reduces breast cancer mortality in women over age 50 by 25–30%.
  28. [28]
    Meta-analysis of breast cancer mortality benefit and overdiagnosis ...
    Apr 28, 2016 · We present pooled estimates of the prevented fraction of mortality benefit and percentage risk of overdiagnosis for RCTs of screening ...
  29. [29]
    Systematic review and meta-analysis on the impact of lung cancer ...
    Seven RCTs were included in the meta-analysis which corresponds to 84,558 participants. A significant relative reduction of LC-specific mortality of 17% (risk ...<|separator|>
  30. [30]
    Impacts of prevention, screening, treatment on cancer deaths - NCI
    Dec 5, 2024 · Improvements in cancer prevention and screening have averted more deaths from five cancer types combined over the past 45 years than treatment advances.
  31. [31]
    Screening for prostate cancer: an updated Cochrane systematic ...
    Prostate cancer screening did not significantly decrease all-cause or prostate cancer-specific mortality in a combined meta-analysis of five RCTs.
  32. [32]
    Statistical issues in randomized trials of cancer screening - PMC
    Sep 19, 2002 · Lead-time bias is avoided by setting the time of randomization instead of the time of cancer detection as the zero time. Length-bias and ...
  33. [33]
    Methodologic Issues in the Evaluation of Early Detection Programs
    Overdiagnosis is an extreme example of length bias. Because screening is more likely than symptom recognition to yield lesions that might never become ...
  34. [34]
    Quantifying Overdiagnosis in Cancer Screening: A Systematic ...
    May 22, 2017 · This review aims to evaluate existing approaches to estimate the magnitude of overdiagnosis in cancer screening in order to gain insight into the strengths and ...
  35. [35]
    Quantification of overdiagnosis in randomised trials of cancer ...
    We found that trials were generally not designed to estimate overdiagnosis and many trials had high risk of biases that may draw the estimates of overdiagnosis ...Missing: challenges | Show results with:challenges
  36. [36]
    Self-Selection Bias in Randomized and Observational Studies ... - NIH
    May 1, 2025 · Observational studies aimed at evaluating the effectiveness of screening mammography are prone to self-selection due to differences in personal characteristics.
  37. [37]
    Self-Selection Bias in Randomized and Observational Studies on ...
    May 1, 2025 · Compared to women attending screening, the risk of breast cancer death was approximately two times greater in women not attending screening.
  38. [38]
    Correction for Self-Selection in Breast Cancer Screening ... - NIH
    Sep 15, 2023 · Studies looking at the reduction of breast cancer deaths from screenings that are not randomized may suffer bias because of “self selection”.
  39. [39]
    The effects of early treatment, lead time and length bias ... - PubMed
    Prognostic selection bias (a tendency of screen-detected cases to be relatively benign or relatively malignant) can be evaluated by comparing the case-mortality ...Missing: studies | Show results with:studies
  40. [40]
    Quantifying and monitoring overdiagnosis in cancer screening
    Jan 7, 2015 · It is possible to calculate an average lead time from randomized trials, though these estimates are biased because they include overdiagnosed ...
  41. [41]
    Correcting for Lead Time and Length Bias in Estimating the Effect of ...
    Aug 6, 2025 · The authors propose a simple correction for lead time, assuming an exponential distribution of the preclinical screen-detectable period.
  42. [42]
    Overdiagnosis is underestimated in screenings, a systematic review
    Apr 1, 2023 · For all trials and all types of cancer screening programs, estimates of overdiagnosis ranged from 6% to 67%. * For breast cancer screening ...
  43. [43]
    Study Estimates One in Seven U.S. Breast Cancers May Be Over ...
    Feb 28, 2022 · About one in seven breast cancers detected by mammography screening are over-diagnosed, according to a Duke Cancer Institute study.
  44. [44]
    Overdiagnosis: An Underrecognized Cause of Confusion and Harm ...
    However, overdiagnosis does not reduce disease-specific mortality because treating subjects with pseudodisease does not help those who have real disease.
  45. [45]
    Screening and Prostate-Cancer Mortality in a Randomized ...
    PSA-based screening reduced the rate of death from prostate cancer by 20% but was associated with a high risk of overdiagnosis.
  46. [46]
    Overdiagnosis of Prostate Cancer - PMC - NIH
    Using ERSPC data, Welch and Black estimated that 60% of such screen-detected cancers were overdiagnosed (6). Draisma et al. (24) assessed overdiagnosis in US ...
  47. [47]
    Lead Time and Overdiagnosis in Prostate-Specific Antigen Screening
    The original MISCAN model fitted to ERSPC Rotterdam data predicted a mean lead time of 7.9 years and an overdiagnosis estimate of 66%; in the model that was ...
  48. [48]
    Overdiagnosis in Colorectal Cancer Screening - Gastroenterology
    Aug 1, 2018 · Overtreatment of the cancer or precancerous condition itself, with all its short-term and long-term risks related to burden, side effects, and ...
  49. [49]
    An Estimate of Preventable Harms Associated With Screening ... - NIH
    The authors calculate that the annual incidence of serious harm from unnecessary colonoscopies ranges from 9,055 to 11,874. The annual incidence of bowel ...
  50. [50]
    Prostate-Specific Antigen–Based Screening for Prostate Cancer ...
    May 8, 2018 · PSA screening may reduce prostate cancer mortality risk but is associated with false-positive results, biopsy complications, and overdiagnosis.
  51. [51]
    Cancer Screening, Incidental Detection, and Overdiagnosis - PubMed
    Jan 4, 2024 · It is now possible to be diagnosed with a cancer not destined to cause symptoms or death-a phenomenon labeled as overdiagnosis.
  52. [52]
    Cumulative Incidence of False-Positive Results in Repeated ... - NIH
    The cumulative risk after 14 tests of undergoing an invasive diagnostic procedure prompted by a false-positive test is 28.5% (CI, 27.8%–29.3%) for men and 22.1% ...
  53. [53]
    False Positives in Cancer Screening: Key Facts
    May 14, 2025 · Mammograms: 10–12% per test; 50–60% cumulative risk over 10 years. PSA Tests: Up to 75% false positives. CT Lung Screens: 23–51% for initial ...
  54. [54]
    How do women experience a false-positive test result from breast ...
    Jul 23, 2019 · The review found that women who had received false-positive screening test results described feelings of fear and breast cancer-related worry, ...
  55. [55]
    Psychological effects of a suspicious prostate cancer screening test ...
    Psychological effects of a suspicious prostate cancer screening test ... Ten-year risk of false positive screening mammograms and clinical breast examinations.
  56. [56]
    Psychosocial consequences of false positives in the Danish Lung ...
    Results False positives experienced significantly more negative psychosocial consequences in seven outcomes at 1 week and in three outcomes at 1 month compared ...
  57. [57]
    Meta-analyses of the effect of false-positive mammograms ... - PubMed
    False positives were associated with only one of six generic outcomes (i.e. generalized anxiety), and this effect size was small. Conclusions: False-positive ...
  58. [58]
    Consequences of False-Positive Screening Mammograms | Radiology
    Apr 21, 2014 · Conclusions and Relevance False-positive mammograms were associated with increased short-term anxiety but not long-term anxiety, and there was ...
  59. [59]
    Long-term psychosocial consequences of false-positive screening ...
    Apr 26, 2023 · Our study suggests that a false-positive mammogram is associated with increased psychosocial consequences 12–14 years after the screening.
  60. [60]
    Reviewing the mental health impacts of cancer screening - LWW
    Sep 23, 2022 · The effects of false-positive cancer diagnosis results are mixed. Some studies have shown no effect of false positives on mental health.
  61. [61]
    Mammogram False Positives Affect Future Screening Behavior - NCI
    Oct 4, 2024 · False-positive screening test results can lead to fear, anxiety, and frustration with the health care system, noted Jennifer Croswell, M.D., ...
  62. [62]
    The Psychological Harms of Screening: the Evidence We Have ...
    Systematic reviews for the US Preventive Services Task Force have found less high-quality evidence on psychological than physical harms of screening.Study Selection · Figure 1 · Evidence By Design...
  63. [63]
    Cancer Screening Guidelines Lack Information on Harms - NCI
    Nov 23, 2022 · Physical harm: Screening tests can cause minor physical harm like bruising or discomfort, as well as serious physical harm like tearing the ...
  64. [64]
    Cancer Screening Overview (PDQ®) - PeaceHealth
    Oct 16, 2023 · The possibility of serious test-related complications, which may be immediate (e.g., perforation with colonoscopy) or delayed (e.g., potential ...
  65. [65]
    Radiation-Induced Breast Cancer Incidence and Mortality from ... - NIH
    Aug 16, 2016 · Radiation-induced breast cancer incidence and mortality from digital mammography screening are impacted by dose variability from screening and ...
  66. [66]
    Does Mammography Cause Cancer? - Princeton Radiology
    Jul 30, 2020 · However, with annual mammographic screenings, the risk of possibly causing 20-25 cancers per 100,000 women pales in comparison with the number ...
  67. [67]
    Understanding the risk of ionizing radiation in breast imaging
    The authors reported 125 cases of breast cancer attributed to radiation exposure, resulting in an additional 16 cancer-related deaths, relative to 968-breast ...
  68. [68]
    Navigating Updated Breast Imaging Guidelines: Balancing Early ...
    Sep 30, 2024 · A single screening mammogram poses the same risk as about 26 days of natural background radiation. This value is in comparison to a standard ...
  69. [69]
    Screening for Colorectal Cancer: Updated Evidence Report and ...
    May 18, 2021 · Serious harms of screening colonoscopy included perforations (3.1/10 000 procedures) and major bleeding (14.6/10 000 procedures). CT ...
  70. [70]
    Adverse events related to colonoscopy: Global trends and future ...
    Jan 14, 2019 · The rate of perforation in screening/surveillance colonoscopy was 0.010-0.067%. However, the rate of perforation in symptomatic/diagnostic ...
  71. [71]
    [PDF] Complications of colonoscopy - ASGE
    Colonic per- foration is a rare complication of endoscopic hemostasis. However, among patients undergoing treatment of angi- ectasia, particularly in the right ...
  72. [72]
    Outpatient Colonoscopy Complications in CDC's Colorectal Cancer ...
    Jul 24, 2025 · Three patients experienced bowel perforations (incidence of 0.89 per 1000 colonoscopies) that required surgery to repair the bowel. Two of the ...
  73. [73]
    Radiation Exposure to Low-Dose Computed Tomography for Lung ...
    Jan 24, 2023 · Analyzing the NLST data, an average total radiation dose is estimated at 8 mSv per participant, with a lifetime radiation-induced risk of cancer ...
  74. [74]
    Benefit‐to‐radiation‐risk of low‐dose computed tomography lung ...
    Nov 1, 2023 · Including only screening doses, NLST benefit-to-radiation-risk ratios are 12:1 for males, 19:1 for females, and 16:1 overall. Including both ...Abstract · INTRODUCTION · RESULTS · DISCUSSION
  75. [75]
    Exposure to low dose computed tomography for lung cancer ...
    Feb 8, 2017 · In the National Lung Screening Trial, the effective dose estimates were 1.6 mSv and 2.4 mSv for one low dose CT scan for men and women, ...
  76. [76]
    Pap smear - Mayo Clinic
    Jul 20, 2024 · Risks. A Pap smear is a safe way to screen for cervical cancer. Still, a Pap smear, also called a Pap test, isn't always accurate. It's ...
  77. [77]
    HARMS OF CERVICAL CANCER SCREENING IN THE UNITED ...
    We studied harms related to cervical cancer screening and management of screen-positive women in the United States (US) and the Netherlands.
  78. [78]
    Recommendation: Cervical Cancer: Screening - uspstf
    Aug 21, 2018 · The USPSTF recommends against screening for cervical cancer in women who have had a hysterectomy with removal of the cervix and do not have a ...Screening · Screening, 1996 · Draft Recommendation<|control11|><|separator|>
  79. [79]
    Effect of mammographic screening from age 40 years on breast ...
    A meta-analysis of all previous trials showed a 15% mortality reduction in invited women aged 40-49 years at study entry, but this finding could be due in part ...
  80. [80]
    Archived: Final Evidence Summary: Breast Cancer: Screening - uspstf
    Nov 15, 2009 · Our meta-analysis of mammography screening trials indicates breast cancer mortality benefit for all age groups from 39 to 69 years, with ...<|separator|>
  81. [81]
    Screening for Breast Cancer: Evidence Report and Systematic ...
    Apr 30, 2024 · Previous reviews of breast cancer screening effectiveness established the benefits and harms of mammography based primarily on large, long-term ...
  82. [82]
    Benefits and harms of mammography screening - PMC
    We aim at presenting the benefits and harms per 1,000 women invited to mammography screening who started screening at age 50 years and were screened every ...
  83. [83]
    Overdiagnosis of breast cancer in population screening - NIH
    Evidence from randomized trials and also from observational studies shows that mammography screening reduces the risk of BC death.
  84. [84]
    Estimating Breast Cancer Overdiagnosis After Screening ...
    Aug 8, 2023 · Continued breast cancer screening was associated with greater incidence of breast cancer, suggesting overdiagnosis may be common among older women.Missing: rates | Show results with:rates
  85. [85]
    Mammography in combination with breast ultrasonography versus ...
    Mar 31, 2023 · From a healthcare perspective, weighing benefits against harms is crucial when recommending screening tests. Whether the net effect of screening ...
  86. [86]
    Benefits and harms of breast cancer mammography screening for ...
    Feb 25, 2021 · Mammography screening is generally accepted in women aged 50-69, but the balance between benefits and harms remains controversial in other age groups.
  87. [87]
    Cervical Cancer Screening (PDQ®)–Health Professional Version
    Apr 18, 2025 · Magnitude of Effect: Regular Pap screening decreases cervix cancer incidence and mortality by at least 80%. Study Design: Population-based and ...Screening With the... · Screening With the Pap Test... · Screening With the Human...
  88. [88]
    Impact of cervical screening on cervical cancer mortality - NIH
    Sep 15, 2016 · It is estimated that in England screening currently prevents 70% (95% CI: 66–73%) of cervical cancer deaths (all ages).
  89. [89]
    Draft Recommendation: Cervical Cancer: Screening - uspstf
    Dec 10, 2024 · The USPSTF recommends screening for cervical cancer every 3 years with cervical cytology alone in women ages 21 to 29 years and then every 5 ...
  90. [90]
    Reducing overtreatment associated with overdiagnosis in cervical ...
    Aug 15, 2020 · A general concern exists that cervical cancer screening using human papillomavirus (HPV) testing may lead to considerable overtreatment.
  91. [91]
    The IARC Perspective on Cervical Cancer Screening
    Nov 10, 2021 · A single randomized, controlled trial conducted in India evaluated thebeneficial effects among women of nucleic acid testing for high-risk HPV ...
  92. [92]
    Effectiveness of Colorectal Cancer (CRC) Screening on All-Cause ...
    Mar 24, 2023 · Based on RCTs and model studies, biennial FIT/gFOBT, single and 5-yearly FS, and 10-yearly colonoscopy screening significantly reduces CRC-specific mortality.
  93. [93]
    Colonoscopy Screening vs Sigmoidoscopy Screening in Colorectal ...
    Feb 29, 2024 · In 4 large randomized clinical trials, sigmoidoscopy screening reduced CRC incidence and mortality predominantly in the distal colon and rectum.
  94. [94]
    Effect of Colonoscopy Screening on Risks of Colorectal Cancer and ...
    Oct 9, 2022 · In this randomized trial, the risk of colorectal cancer at 10 years was lower among participants who were invited to undergo screening colonoscopy.
  95. [95]
    Recommendation: Colorectal Cancer: Screening - uspstf
    May 18, 2021 · Serious harms from colonoscopy to follow-up positive screening results are estimated to be 17.5 serious bleeding events (95% CI, 7.6-27.5; 11 ...
  96. [96]
    Colorectal Cancer Guideline | How Often to Have Screening Tests
    Jan 29, 2024 · The American Cancer Society recommends that people at average risk of colorectal cancer start regular screening at age 45.
  97. [97]
    [PDF] ASGE Position on Blood-Based Colorectal Cancer Screening
    Aug 25, 2025 · ASGE has determined that blood-based tests are inferior to established screening options (i.e., colonoscopy and stool-based tests), especially ...
  98. [98]
    Risk of colon perforation during colonoscopy at Baylor University ...
    The risk of perforation at BUMC was 0.57 per 1000 procedures or 1 in 1750 colonoscopies. Continued efforts to make colonoscopy safer are needed.
  99. [99]
    Overdiagnosis in Colorectal Cancer Screening - Gastroenterology
    Aug 1, 2018 · Consequently, most precancerous polyps diagnosed during CRC screening interventions are removed with no clinical gain.
  100. [100]
    Prostate Cancer Screening - StatPearls - NCBI Bookshelf
    This continuing education activity focuses on prostate cancer screening and its crucial role in reducing morbidity and mortality from this common and ...
  101. [101]
    Recommendation: Prostate Cancer: Screening - uspstf
    May 8, 2018 · The American Urological Association recommends that men aged 55 to 69 years with a life expectancy of more than 10 to 15 years be informed of ...Screening, 2008 · Screening · Screening, 2002 · Screening, 1996
  102. [102]
    Extended mortality results for prostate cancer screening in the PLCO ...
    Dec 1, 2016 · In the current analysis, we update the mortality results of the PLCO trial with follow-up extended to up to 19 years from randomization.<|separator|>
  103. [103]
    Mortality reduction and cumulative excess incidence (CEI) in the ...
    Mar 9, 2024 · The full ERSPC study demonstrated a 31% mortality reduction with PSA screening after correcting for contamination and compliance16,17.
  104. [104]
    PSA Screening Slightly Reduces Prostate Cancer Death Risk at 15 ...
    Apr 10, 2024 · In the ERSPC trial, PSA screening every 2 to 4 years reduced prostate cancer mortality by 20% after 16 years. The PLCO trial found no prostate ...
  105. [105]
    Overdiagnosis and Overtreatment of Prostate Cancer - PMC
    Screening reduces spread and death from prostate cancer (PCa) but overdiagnoses some low-risk tumors that may not have caused harm.
  106. [106]
    Harm-to-Benefit of Three Decades of Prostate Cancer Screening in ...
    May 15, 2022 · For men of all races, we estimated 1.5 to 1.9 million (range between estimation approaches) overdiagnosed and 0.9 to 1.5 million overtreated ...
  107. [107]
    Complications Following Prostate Biopsy: Data from SEER-Medicare
    Jan 15, 2023 · The risk of hospitalization within 30 days of prostate biopsy is significantly higher than in a control population.
  108. [108]
    Early Detection of Prostate Cancer: AUA/SUO Guideline (2023)
    This guideline addresses early detection with an emphasis on PSA-based screening, considerations for initial and repeat biopsy, and biopsy technique.
  109. [109]
    American Cancer Society Recommendations for Prostate Cancer ...
    Nov 22, 2023 · Screening should be done yearly for men whose PSA level is 2.5 ng/mL or higher. Because prostate cancer often grows slowly, men without symptoms ...
  110. [110]
    Prostate cancer screening: Continued controversies and novel ... - NIH
    Aug 31, 2022 · However, screening via PSA has several drawbacks, including overdiagnosis and overtreatment of otherwise indolent cases, as well as the ...
  111. [111]
    Reduced Lung-Cancer Mortality with Low-Dose Computed ...
    Jun 29, 2011 · The rate of adherence to screening was more than 90%. The rate of positive screening tests was 24.2% with low-dose CT and 6.9% with radiography ...
  112. [112]
    Reduced Lung-Cancer Mortality with Volume CT Screening in a ...
    In the NELSON trial, an excess of 40 cases (344 vs. 304) was found among the male participants in the screening group 10 years after randomization (4.5 years ...
  113. [113]
    Lung Cancer Screening (PDQ®) - NCI
    Apr 17, 2025 · Lung cancer screening with low-dose spiral CT scans has been shown to decrease the risk of dying from lung cancer in heavy smokers.Evidence of Benefit Associated... · Evidence of No Benefit... · Overdiagnosis
  114. [114]
    Lung Cancer: Screening | United States Preventive Services Taskforce
    Mar 9, 2021 · Screen every year with low-dose CT. Stop screening once a person has not smoked for 15 years or has a health problem that limits life expectancy ...
  115. [115]
    Lung Cancer Screening Guidelines - American Cancer Society
    The American Cancer Society recommends yearly screening for lung cancer with a low-dose CT (LDCT) scan for people aged 50 to 80 years.
  116. [116]
    Lung Nodule Management in Low-Dose CT Screening for Lung ...
    Oct 22, 2024 · In the NELSON trial, participants with slow-growing nodules (VDT > 600 days) had a low 2-year lung cancer probability of 0.8%. Two-year lung ...Lung Nodule Management In... · Subsolid Lung Nodules · Ct-Based Nodule...
  117. [117]
    Overdiagnosis in Low-Dose Computed Tomography Screening for ...
    More than 18% of all lung cancers detected by LDCT in the NLST seem to be indolent, and overdiagnosis should be considered when describing the risks of LDCT ...
  118. [118]
    Lung Cancer Screening: Pros and Cons - AAFP
    Jun 15, 2019 · An estimated 18% to 67% of lung cancers detected with low-dose CT screening may be overdiagnosed, exposing many patients to unnecessary risk.Case Scenario · Clinical Commentary · Costs Of Lung Scans And...
  119. [119]
    Screening for Lung Cancer - CDC
    Oct 15, 2024 · During an LDCT scan, you lie on a table and an x-ray machine uses a low dose (amount) of radiation to make detailed images of your lungs. The ...
  120. [120]
    Evaluation of the Benefits and Harms of Lung Cancer Screening ...
    Mar 9, 2021 · Microsimulation modeling studies suggested that LDCT screening for lung cancer compared with no screening may increase lung cancer deaths averted and life- ...
  121. [121]
    Screening Low-Risk Individuals for Lung Cancer: The Need May Be ...
    With the mortality reduction recognized from annual low-radiation dose computed tomography by screening those at high risk, there has been consideration that ...
  122. [122]
    Recommendation: Ovarian Cancer: Screening - uspstf
    Feb 13, 2018 · The USPSTF found adequate evidence that screening for ovarian cancer can result in important harms, including many false-positive results.Missing: skin melanoma testicular oral liver esophageal gastric 2023-2025
  123. [123]
    Results - Screening for Ovarian Cancer - NCBI Bookshelf - NIH
    Four trials reported on the harms of ovarian cancer screening. False positive rates and surgical harms were highest for screening programs using transvaginal ...
  124. [124]
    Screening for Ovarian Cancer: Updated Evidence Report and ...
    Feb 13, 2018 · Screening trials have shown no effect on mortality and have documented harms; positive test results from screening asymptomatic women often ...
  125. [125]
    Recommendation: Pancreatic Cancer: Screening - uspstf
    Aug 6, 2019 · Screening Tests, The USPSTF does not recommend screening for pancreatic cancer in the general population using any method.Missing: ovarian skin melanoma testicular oral esophageal 2023-2025
  126. [126]
    Pancreas cancer screening in high-risk individuals
    Pancreas cancer screening in high-risk individuals should begin at age 50 years, or 10 years younger than the initial age of familial onset.
  127. [127]
    AGA Clinical Practice Update on Pancreas Cancer Screening in ...
    May 13, 2020 · Pancreas cancer screening in high-risk individuals should begin at age 50 years, or 10 years younger than the initial age of familial onset.
  128. [128]
    Skin Cancer: Screening | United States Preventive Services Taskforce
    Apr 18, 2023 · A visual skin examination is the most commonly proposed method for skin cancer screening and includes a survey of the body for skin lesions.Missing: ovarian testicular oral liver esophageal gastric 2023-2025
  129. [129]
    A and B Recommendations | United States Preventive ... - uspstf
    The USPSTF recommends that primary care clinicians assess women with a personal or family history of breast, ovarian, tubal, or peritoneal cancer or who have ...Breast Cancer: Screening · Colorectal Cancer: Screening · Lung Cancer: ScreeningMissing: pancreatic melanoma testicular liver esophageal gastric
  130. [130]
    Evaluation of Harms Reporting in United States Cancer Screening ...
    May 1, 2023 · Guidelines did not report all harms for any specific organ type nor for any category of harm across organ types. The most complete harms ...
  131. [131]
    Cancer Screening Guidelines | Detecting Cancer Early
    Nov 1, 2023 · Here are the American Cancer Society's recommendations to help guide you when you talk to your doctor about screening for certain cancers.
  132. [132]
  133. [133]
  134. [134]
    Detection, Prevention, and Risk Reduction - NCCN
    Explore NCCN Category 2 Guidelines for cancer detection, prevention, risk reduction, and screening strategies to improve patient outcomes.Breast Cancer Screening and ...Breast, Ovarian, PancreaticColorectal Cancer ScreeningGuidelinesBreast Cancer Risk Reduction
  135. [135]
    When is screening for cancer the right course of action?
    Feb 2, 2022 · Currently, WHO recommends screening programmes for only 3 types of cancer – breast, cervical and colorectal.
  136. [136]
    Congruence of cancer screening recommendations between the ...
    Mar 31, 2025 · The 2024 USPSTF guidelines recommend screening for women aged 40–74 every 2 years (grade B) with a conventional digital mammogram (DM) ± digital ...
  137. [137]
    Risk-Adjusted Cancer Screening and Prevention (RiskAP)
    Risk-adjusted screening thus comprises both individual risk assessment and early detection based on the outcome of that assessment. By exploiting all known ...
  138. [138]
    Implementation considerations for risk-tailored cancer screening in ...
    Risk-tailored screening has emerged as a promising approach to optimise the balance of benefits and harms of existing population cancer screening programs.
  139. [139]
    Breast Cancer Risk Assessment Tool: Online Calculator - NCI
    Nov 19, 2024 · The Breast Cancer Risk Assessment Tool (BCRAT) is an interactive calculator that estimates a woman's risk of developing breast cancer.Risk Calculator · About the Calculator · Health Professional Version
  140. [140]
    Lung cancer sceening using risk model-based strategies
    Feb 6, 2023 · Risk model-based lung cancer screening strategies, which select individuals based on personal risk, are more cost effective than current recommendations.<|separator|>
  141. [141]
    Identify Patients Likely to Benefit from Lung Cancer Screening | AAFP
    Jan 15, 2020 · Individualized risk assessment can be helpful because patients at higher risk of developing lung cancer are also more likely to benefit from ...
  142. [142]
    Shared Decision Making About Screening and Chemoprevention
    The USPSTF places a high value on informed and joint decisions about screening and chemoprevention; such decisions are essential for making recommendations to ...
  143. [143]
    The Effects of Shared Decision Making on Cancer Screening ... - NCBI
    The purpose of this review is to examine the effects of SDM interventions for cancer screening in adults on Decision Quality and Impact.
  144. [144]
    A Systematic Review: Shared Decision Making for Cancer Screening
    The review found moderate evidence that SDM interventions for prostate, breast, and colorectal cancer screening improve patient knowledge.
  145. [145]
    Benefit-Harm Analysis for Informed Decision Making on Participating ...
    The benefit-harm balance for a specific individual depends on CRC risk factors such as age, sex and lifestyle, the risk of potential harms of the screening ...
  146. [146]
    Cancer Genetics Risk Assessment and Counseling (PDQ®) - NCI
    Jan 3, 2025 · Cancer genetics risk assessment and genetic counseling includes family history, psychosocial assessments, and education on hereditary cancer ...
  147. [147]
    The Potential for Personalized Screening Using Image-based AI ...
    Aug 29, 2023 · Other AI-based models use imaging data to predict short-term risk by detecting early signs of breast cancer invisible to the human eye rather ...
  148. [148]
    A Clinical Risk Model for Personalized Screening and Prevention of ...
    Jun 19, 2023 · Conclusions: The lifestyle/familial-expanded AI risk model showed higher performance for both long-term and short-term risk assessment compared ...
  149. [149]
    A Simple Approach to Shared Decision Making in Cancer Screening
    Six steps to shared decision making · Invite the patient to participate. · Present the options. · Provide information on benefits and risks. · Assist patients in ...Introduction · What is shared decision... · Six steps to shared decision...
  150. [150]
    CanScreen5, a global repository for breast, cervical and colorectal ...
    Apr 27, 2023 · Reported screening coverage ranged from 1.7% (Bangladesh) to 85.5% (England, United Kingdom) for breast cancer, from 2.1% (Côte d'Ivoire) to ...
  151. [151]
    Cancer Screening in Five Continents (CanScreen5): a global data ...
    Apr 30, 2024 · Cancer Screening in Five Continents (CanScreen5): a global data repository for breast, cervical, and colorectal cancer screening programmes.
  152. [152]
    Cancer screening statistics - Statistics Explained - Eurostat
    The screening rate was highest in Finland at 74.4% (provisional), and was close to two thirds in the Netherlands (67.2%) and Sweden (64.7%). The screening rate ...Breast cancer screening · Cervical cancer screening · Colorectal cancer screening
  153. [153]
    Global colorectal cancer screening programs and coverage rate ...
    Jul 22, 2025 · The reported latest screening coverage rates varied greatly across countries, ranging from 1.0% (China, 2020) to 79.4% (Finland, 2021). The ...
  154. [154]
    Cancer screening: Health at a Glance 2023 - OECD
    In 2021, the highest rate was 79% in Sweden, followed by 75% in the Czech Republic, while the lowest rate was 3% in Costa Rica. Compared to breast and cervical ...
  155. [155]
    Cancer Screening in Low- and Middle-Income Countries
    Jun 6, 2024 · These rates are reported for 38 cancer sites in 20 world regions as defined by the United Nations and in 185 countries; Figure 1 depicts the ...
  156. [156]
    CanScreen5, a global repository for breast, cervical and colorectal ...
    Apr 27, 2023 · The CanScreen5 project is a global cancer screening data repository that aims to report the status and performance of breast, cervical and colorectal cancer ...
  157. [157]
    Facilitators and Barriers to Cancer Screening Participation ... - NIH
    Apr 3, 2025 · The commonly reported barriers to participation in cancer screening included poor knowledge, emotional factors, belief of low personal susceptibility, lack of ...
  158. [158]
    Barriers to Cervical Cancer Screening: A Systematic Review | Cureus
    Jul 28, 2024 · Common barriers identified across studies included lack of knowledge and awareness, economic constraints, access issues, cultural and religious ...
  159. [159]
    Barriers to Breast Cancer-Screening Adherence in Vulnerable ...
    Most of them present common barriers, such as race/ethnicity (47%), low socioeconomic (35.3%) and educational levels (29.4%), no family history of cancer and ...Barriers To Breast... · 3. Results · 4. Discussion
  160. [160]
    Barriers to cancer screening uptake and approaches to overcome ...
    Barriers to cancer screening included: being unmarried, experiencing higher deprivation, lower socioeconomic status and rural living conditions.
  161. [161]
    Assessment of barriers to cancer screening and interventions ...
    Apr 22, 2024 · The most prioritized barriers to increasing screening participation were not having an efficient information system for monitoring participation ...INTRODUCTION · METHODS · RESULTS · DISCUSSION
  162. [162]
    Facilitators and barriers of lung cancer screening participation
    Sep 1, 2025 · Challenges include health insurance policies that do not cover screening costs, delayed reimbursements, low payments, lack of coverage for ...
  163. [163]
    Barriers against utilization of population-based cancer screening ...
    Jul 14, 2025 · This study aimed to determine the attendance rates and barriers in accessing age- and gender-specific cancer screenings offered free-of-charge in Germany.
  164. [164]
    Barriers to cervical cancer screening among refugee women
    This review aimed to identify barriers hindering their access to cervical cancer screening. Five databases - Ovid MEDLINE, EMBASE, PsycINFO, CINAHL, and SCOPUS ...<|separator|>
  165. [165]
    Cervical screening participation and access facilitators and barriers ...
    This study systematically reviewed research on the rates of cervical screening participation among people with intellectual disability, and facilitators and ...
  166. [166]
    Overdiagnosis in lung cancer screening - Yankelevitz
    Thus, with the current definition an overdiagnosed cancer can only be diagnosed if the person does not die from the cancer, regardless of whether or not the ...
  167. [167]
    Addressing overdiagnosis and overtreatment in cancer - NIH
    These examples show the weaknesses of screening and how screening might lead to net harm while seeming to do good. An inflated perception of the benefit of ...Missing: debates | Show results with:debates
  168. [168]
    Overdiagnosis Due to Screening Mammography for Breast Cancer ...
    Mar 14, 2023 · So, overtreatment of overdiagnosed cases can increase mortality rates with no beneficial outcomes [15]. Overdiagnosed cases are subjected to ...
  169. [169]
    Determinants of benefits and harms of breast cancer screening - NIH
    Systematic reviews on mammography screening, mainly from high‐income countries, systematically disagree on the interpretation of the benefit‐to‐harm ratio.
  170. [170]
    Screening Low-Risk Individuals for Lung Cancer: The Need May Be ...
    Several observations reported from the NLST suggest that overdiagnosis may be a particular feature of LDCT-based lung cancer screening.
  171. [171]
    Conducting separate reviews of benefits and harms could improve ...
    Apr 15, 2023 · Systematic reviews may come to incorrect conclusions about harms when they exclude relevant study designs. For example, systematic reviews often ...
  172. [172]
    To Comply or Not to Comply With the US Preventive Services Task ...
    May 2, 2022 · The USPSTF has been subjected to outraged criticism (as has its Canadian precursor) for recommendations deemed unacceptable by the American ...
  173. [173]
    breaking the deadlock in the breast cancer screening debate - Nature
    Sep 13, 2017 · The overview of the Swedish trials of bi- or triennial screening showed a relative reduction in breast cancer mortality of 21%, with maximum ...
  174. [174]
    Overdiagnosis and breast cancer screening
    Aug 30, 2005 · The issues of current importance are not those referring to whether there is a net benefit, but how screening can be undertaken to optimise the ...Missing: debates | Show results with:debates
  175. [175]
    Screening for breast cancer: a systematic review update to inform ...
    Dec 19, 2024 · This systematic review update synthesized recent evidence on the benefits and harms of breast cancer screening in women aged ≥ 40 years<|separator|>
  176. [176]
    Crunching Numbers: What Cancer Screening Statistics Really Tell Us
    Jul 16, 2018 · For example, studies have estimated that 19% of screen-detected breast cancers and 20% to 50% of screen-detected prostate cancers are ...
  177. [177]
    Cancer overdiagnosis: a biological challenge and clinical dilemma
    Of those with a positive biopsy, an estimated 20–50% represent overdiagnosis (for example, 50.4% in the European Randomized Study of Screening for Prostate ...
  178. [178]
    Overdiagnosis and Overtreatment: Evaluation of What Physicians ...
    Oct 21, 2013 · The consequence of overdiagnosis is overtreatment—surgery, chemotherapy, or radiation—that provides the patient no benefits, but only adverse ...
  179. [179]
    Risks, Benefits, and Conundrums of Cancer Screening
    A patient's story represents an ethical dilemma of cancer screening: it can save lives but generates diagnostic morbidity and incurs costs.
  180. [180]
    Screening and Informed Consent | New England Journal of Medicine
    Feb 11, 1993 · Before they participate in a screening program, patients must give informed consent. To do so, they need to understand the risk of a false ...<|separator|>
  181. [181]
    Non-maleficence and the ethics of consent to cancer screening
    In this paper, I will explore whether an appeal to consent might justify overriding non-maleficence in cancer screening.Screening And... · Informed Consent · Patient Choice
  182. [182]
    Cancer Disparities and Health Equity: A Policy Statement From ... - NIH
    This updated statement affirms ASCO's commitment to moving beyond descriptions of differences in cancer outcomes toward achievement of cancer health equity.Missing: allocation | Show results with:allocation
  183. [183]
    Racial Disparities in Cancer Outcomes, Screening, and Treatment
    Feb 3, 2022 · This brief provides an overview of recent data on cancer incidence and mortality, risk factors, screening, treatment, and outcomes by race ...
  184. [184]
    Author's specialty and conflicts of interest contribute to ... - PubMed
    Objective: To examine the relationship between guideline panel members' conflicts of interest and guideline recommendations on screening mammography in ...
  185. [185]
    Financial Conflicts of Interest Among Authors of Clinical Practice ...
    Purpose: Financial conflicts of interest (FCOIs) may influence or undermine the credibility of clinical practice guidelines or society recommendations.<|control11|><|separator|>
  186. [186]
    Stronger Rules Needed for Oncology Financial Conflicts of Interest ...
    May 1, 2024 · Guideline panels, professional societies, and medical journals should set stronger rules to prevent bias stemming from financial conflicts of interest (COI) in ...
  187. [187]
    Mammography screening: A major issue in medicine - ScienceDirect
    Overdiagnosis is commonplace, representing 20% or more of all breast cancers among women invited to screening and 30–50% of screen-detected cancers.
  188. [188]
    Multicancer Early Detection Tests at a Crossroads
    Apr 22, 2025 · Multicancer early detection tests (MCEDs), sometimes referred to as liquid biopsies, are tests that can screen for multiple cancers by analyzing blood, urine, ...
  189. [189]
    Multi-cancer Detection Tests | MCD | GRAIL Galleri Test
    Oct 2, 2025 · The main goal of MCD tests is to find cancers earlier, ideally before symptoms appear. Cancer detected early is more likely to be treated ...Missing: innovations | Show results with:innovations
  190. [190]
    Multicancer Early Detection Tests — Keeping a High Bar for ...
    Jul 20, 2024 · The primary objective of the study is “to demonstrate a statistically significant reduction in the incidence rate of stage III and IV cancers ...<|separator|>
  191. [191]
    GRAIL PATHFINDER 2 Results Show Galleri® Multi-Cancer Early ...
    Oct 17, 2025 · Galleri is the only MCED test available that has been validated in an interventional trial in the screening population and could transform how ...Missing: innovations | Show results with:innovations
  192. [192]
  193. [193]
  194. [194]
    New Grail cancer test data show progress but raise questions | STAT
    Oct 17, 2025 · Grail says new data on multi-cancer screening test show improved performance. Findings strengthen biotech's push for FDA approval of Galleri ...
  195. [195]
    Exact Sciences Launches Cancerguard, First-of-Its-Kind Multi ...
    Exact Sciences Launches Cancerguard™, First-of-Its-Kind Multi-Cancer Early Detection Blood Test. By Exact Sciences / September 10, 2025. -Only MCED test ...
  196. [196]
    FDA Grants Breakthrough Device Designation to Guardant Health's ...
    Jun 3, 2025 · “This recognition by the FDA shows the promise of the Shield MCD test to detect multiple cancers at an early stage with just a single, routine ...
  197. [197]
    A large-scale, multi-centre validation study of an AI-empowered ...
    Oct 8, 2025 · In our previous study, we developed a blood-based test for multi-cancer early detection (MCED), named OncoSeek. By integrating a panel of seven ...
  198. [198]
    Evaluation of an innovative multi-cancer early detection test - NIH
    Mar 6, 2025 · The Carcimun test achieved high accuracy, sensitivity, and specificity, effectively identifying cancer patients while minimizing false positives and negatives.
  199. [199]
    Questions and Answers about Multi-Cancer Detection Tests
    However, no MCD tests have been authorized by the FDA. LDTs are regulated under the Clinical Laboratory Improvement Amendments of 1988 (CLIA). The federal ...About Cancer Screening · About Multi-Cancer Detection...
  200. [200]
    Multi Cancer Early Detection Market Forecast Report 2025-2033
    Aug 11, 2025 · The global multi cancer early detection market is set to grow from USD 1 billion in 2024 to USD 4.3 billion by 2033, driven by advancements ...
  201. [201]
    AI as an independent second reader in detection of clinically ...
    Aug 14, 2025 · Artificial intelligence (AI) offers potential to improve screening accuracy and efficiency. This study simulated different screening scenarios, ...
  202. [202]
  203. [203]
    Cancer screening: recent developments and future directions - Nature
    Mar 19, 2025 · An AI model that included age, neutrophil-to-lymphocyte ratio, and red blood cell count was developed, and a risk stratification model was ...
  204. [204]
    Liquid biopsy in cancer: current status, challenges and future ...
    Dec 2, 2024 · Compared with tissue biopsy, liquid biopsy has many advantages in the diagnosis and treatment of various types of cancer, including non-invasive ...
  205. [205]
    Recent progress in prompt molecular detection of liquid biopsy ...
    Dec 31, 2024 · In clinical oncology, liquid biopsy has become a noninvasive and early-detectable alternative to traditional biopsies over the last two decades.
  206. [206]
    Multicancer early detection testing: Guidance for primary care ...
    Apr 2, 2025 · As of November 2024, no MCED tests have yet been approved by the Food and Drug Administration (FDA). The FDA has granted a few MCED tests ...
  207. [207]
    ARPA-H launches program to develop at-home multi-cancer ...
    Aug 1, 2024 · POSEIDON intends to develop first-in-class, at-home, synthetic Multi-Cancer-Early Detection (MCED) tests for the most sensitive and specific stage I detection ...
  208. [208]
    First Decade of the National Cancer Institute's Affordable ... - NIH
    Jul 30, 2025 · Wearable and sensor-based technologies—Advancements in wearable health monitoring devices allow for continuous tracking of vital signs, early ...