Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Radiology ; 311(1): e232535, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38591971

ABSTRACT

Background Mammographic density measurements are used to identify patients who should undergo supplemental imaging for breast cancer detection, but artificial intelligence (AI) image analysis may be more effective. Purpose To assess whether AISmartDensity-an AI-based score integrating cancer signs, masking, and risk-surpasses measurements of mammographic density in identifying patients for supplemental breast imaging after a negative screening mammogram. Materials and Methods This retrospective study included randomly selected individuals who underwent screening mammography at Karolinska University Hospital between January 2008 and December 2015. The models in AISmartDensity were trained and validated using nonoverlapping data. The ability of AISmartDensity to identify future cancer in patients with a negative screening mammogram was evaluated and compared with that of mammographic density models. Sensitivity and positive predictive value (PPV) were calculated for the top 8% of scores, mimicking the proportion of patients in the Breast Imaging Reporting and Data System "extremely dense" category. Model performance was evaluated using area under the receiver operating characteristic curve (AUC) and was compared using the DeLong test. Results The study population included 65 325 examinations (median patient age, 53 years [IQR, 47-62 years])-64 870 examinations in healthy patients and 455 examinations in patients with breast cancer diagnosed within 3 years of a negative screening mammogram. The AUC for detecting subsequent cancers was 0.72 and 0.61 (P < .001) for AISmartDensity and the best-performing density model (age-adjusted dense area), respectively. For examinations with scores in the top 8%, AISmartDensity identified 152 of 455 (33%) future cancers with a PPV of 2.91%, whereas the best-performing density model (age-adjusted dense area) identified 57 of 455 (13%) future cancers with a PPV of 1.09% (P < .001). AISmartDensity identified 32% (41 of 130) and 34% (111 of 325) of interval and next-round screen-detected cancers, whereas the best-performing density model (dense area) identified 16% (21 of 130) and 9% (30 of 325), respectively. Conclusion AISmartDensity, integrating cancer signs, masking, and risk, outperformed traditional density models in identifying patients for supplemental imaging after a negative screening mammogram. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Kim and Chang in this issue.


Subject(s)
Breast Neoplasms , Early Detection of Cancer , Humans , Middle Aged , Female , Breast Neoplasms/diagnostic imaging , Artificial Intelligence , Retrospective Studies , Mammography
2.
BMJ Open ; 14(2): e084014, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38355190

ABSTRACT

BACKGROUND: Understanding women's perspectives can help to create an effective and acceptable artificial intelligence (AI) implementation for triaging mammograms, ensuring a high proportion of screening-detected cancer. This study aimed to explore Swedish women's perceptions and attitudes towards the use of AI in mammography. METHOD: Semistructured interviews were conducted with 16 women recruited in the spring of 2023 at Capio S:t Görans Hospital, Sweden, during an ongoing clinical trial of AI in screening (ScreenTrustCAD, NCT04778670) with Philips equipment. The interview transcripts were analysed using inductive thematic content analysis. RESULTS: In general, women viewed AI as an excellent complementary tool to help radiologists in their decision-making, rather than a complete replacement of their expertise. To trust the AI, the women requested a thorough evaluation, transparency about AI usage in healthcare, and the involvement of a radiologist in the assessment. They would rather be more worried because of being called in more often for scans than risk having overlooked a sign of cancer. They expressed substantial trust in the healthcare system if the implementation of AI was to become a standard practice. CONCLUSION: The findings suggest that the interviewed women, in general, hold a positive attitude towards the implementation of AI in mammography; nonetheless, they expect and demand more from an AI than a radiologist. Effective communication regarding the role and limitations of AI is crucial to ensure that patients understand the purpose and potential outcomes of AI-assisted healthcare.


Subject(s)
Breast Neoplasms , Neoplasms , Female , Humans , Artificial Intelligence , Sweden , Qualitative Research , Mammography , Breast Neoplasms/diagnostic imaging
3.
Eur Radiol ; 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38165430

ABSTRACT

OBJECTIVES: The aim of our study was to examine how breast radiologists would be affected by high cancer prevalence and the use of artificial intelligence (AI) for decision support. MATERIALS AND METHOD: This reader study was based on selection of screening mammograms, including the original radiologist assessment, acquired in 2010 to 2013 at the Karolinska University Hospital, with a ratio of 1:1 cancer versus healthy based on a 2-year follow-up. A commercial AI system generated an exam-level positive or negative read, and image markers. Double-reading and consensus discussions were first performed without AI and later with AI, with a 6-week wash-out period in between. The chi-squared test was used to test for differences in contingency tables. RESULTS: Mammograms of 758 women were included, half with cancer and half healthy. 52% were 40-55 years; 48% were 56-75 years. In the original non-enriched screening setting, the sensitivity was 61% (232/379) at specificity 98% (323/379). In the reader study, the sensitivity without and with AI was 81% (307/379) and 75% (284/379) respectively (p < 0.001). The specificity without and with AI was 67% (255/379) and 86% (326/379) respectively (p < 0.001). The tendency to change assessment from positive to negative based on erroneous AI information differed between readers and was affected by type and number of image signs of malignancy. CONCLUSION: Breast radiologists reading a list with high cancer prevalence performed at considerably higher sensitivity and lower specificity than the original screen-readers. Adding AI information, calibrated to a screening setting, decreased sensitivity and increased specificity. CLINICAL RELEVANCE STATEMENT: Radiologist screening mammography assessments will be biased towards higher sensitivity and lower specificity by high-risk triaging and nudged towards the sensitivity and specificity setting of AI reads. After AI implementation in clinical practice, there is reason to carefully follow screening metrics to ensure the impact is desired. KEY POINTS: • Breast radiologists' sensitivity and specificity will be affected by changes brought by artificial intelligence. • Reading in a high cancer prevalence setting markedly increased sensitivity and decreased specificity. • Reviewing the binary reads by AI, negative or positive, biased screening radiologists towards the sensitivity and specificity of the AI system.

4.
Radiology ; 309(1): e222691, 2023 10.
Article in English | MEDLINE | ID: mdl-37874241

ABSTRACT

Background Despite variation in performance characteristics among radiologists, the pairing of radiologists for the double reading of screening mammograms is performed randomly. It is unknown how to optimize pairing to improve screening performance. Purpose To investigate whether radiologist performance characteristics can be used to determine the optimal set of pairs of radiologists to double read screening mammograms for improved accuracy. Materials and Methods This retrospective study was performed with reading outcomes from breast cancer screening programs in Sweden (2008-2015), England (2012-2014), and Norway (2004-2018). Cancer detection rates (CDRs) and abnormal interpretation rates (AIRs) were calculated, with AIR defined as either reader flagging an examination as abnormal. Individual readers were divided into performance categories based on their high and low CDR and AIR. The performance of individuals determined the classification of pairs. Random pair performance, for which any type of pair was equally represented, was compared with the performance of specific pairing strategies, which consisted of pairs of readers who were either opposite or similar in AIR and/or CDR. Results Based on a minimum number of examinations per reader and per pair, the final study sample consisted of 3 592 414 examinations (Sweden, n = 965 263; England, n = 837 048; Norway, n = 1 790 103). The overall AIRs and CDRs for all specific pairing strategies (Sweden AIR range, 45.5-56.9 per 1000 examinations and CDR range, 3.1-3.6 per 1000; England AIR range, 68.2-70.5 per 1000 and CDR range, 8.9-9.4 per 1000; Norway AIR range, 81.6-88.1 per 1000 and CDR range, 6.1-6.8 per 1000) were not significantly different from the random pairing strategy (Sweden AIR, 54.1 per 1000 examinations and CDR, 3.3 per 1000; England AIR, 69.3 per 1000 and CDR, 9.1 per 1000; Norway AIR, 84.1 per 1000 and CDR, 6.3 per 1000). Conclusion Pairing a set of readers based on different pairing strategies did not show a significant difference in screening performance when compared with random pairing. © RSNA, 2023.


Subject(s)
Mammography , Physical Examination , Humans , Retrospective Studies , England , Radiologists
5.
Br J Radiol ; 96(1151): 20230210, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37660400

ABSTRACT

OBJECTIVE: We aimed to evaluate the false interpretations between artificial intelligence (AI) and radiologists in screening mammography to get a better understanding of how the distribution of diagnostic mistakes might change when moving from entirely radiologist-driven to AI-integrated breast cancer screening. METHODS AND MATERIALS: This retrospective case-control study was based on a mammography screening cohort from 2008 to 2015. The final study population included screening examinations for 714 women diagnosed with breast cancer and 8029 randomly selected healthy controls. Oversampling of controls was applied to attain a similar cancer proportion as in the source screening cohort. We examined how false-positive (FP) and false-negative (FN) assessments by AI, the first reader (RAD 1) and the second reader (RAD 2), were associated with age, density, tumor histology and cancer invasiveness in a single- and double-reader setting. RESULTS: For each reader, the FN assessments were distributed between low- and high-density females with 53 (42%) and 72 (58%) for AI; 59 (36%) and 104 (64%) for RAD 1 and 47 (36%) and 84 (64%) for RAD 2. The corresponding numbers for FP assessments were 1820 (47%) and 2016 (53%) for AI; 1568 (46%) and 1834 (54%) for RAD 1 and 1190 (43%) and 1610 (58%) for RAD 2. For ductal cancer, the FN assessments were 79 (77%) for AI CAD; with 120 (83%) for RAD 1 and with 96 (16%) for RAD 2. For the double-reading simulation, the FP assessments were distributed between younger and older females with 2828 (2.5%) and 1554 (1.4%) for RAD 1 + RAD 2; 3850 (3.4%) and 2940 (2.6%) for AI+RAD 1 and 3430 (3%) and 2772 (2.5%) for AI+RAD 2. CONCLUSION: The most pronounced decrease in FN assessments was noted for females over the age of 55 and for high density-women. In conclusion, AI could have an important complementary role when combined with radiologists to increase sensitivity for high-density and older females. ADVANCES IN KNOWLEDGE: Our results highlight the potential impact of integrating AI in breast cancer screening, particularly to improve interpretation accuracy. The use of AI could enhance screening outcomes for high-density and older females.


Subject(s)
Breast Neoplasms , Mammography , Female , Humans , Mammography/methods , Breast Neoplasms/diagnostic imaging , Artificial Intelligence , Retrospective Studies , Case-Control Studies , Sensitivity and Specificity , Early Detection of Cancer/methods , Radiologists , Mass Screening/methods
6.
Lancet Digit Health ; 5(10): e703-e711, 2023 10.
Article in English | MEDLINE | ID: mdl-37690911

ABSTRACT

BACKGROUND: Artificial intelligence (AI) as an independent reader of screening mammograms has shown promise, but there are few prospective studies. Our aim was to conduct a prospective clinical trial to examine how AI affects cancer detection and false positive findings in a real-world setting. METHODS: ScreenTrustCAD was a prospective, population-based, paired-reader, non-inferiority study done at the Capio Sankt Göran Hospital in Stockholm, Sweden. Consecutive women without breast implants aged 40-74 years participating in population-based screening in the geographical uptake area of the study hospital were included. The primary outcome was screen-detected breast cancer within 3 months of mammography, and the primary analysis was to assess non-inferiority (non-inferiority margin of 0·15 relative reduction in breast cancer diagnoses) of double reading by one radiologist plus AI compared with standard-of-care double reading by two radiologists. We also assessed single reading by AI alone and triple reading by two radiologists plus AI compared with standard-of-care double reading by two radiologists. This study is registered with ClinicalTrials.gov, NCT04778670. FINDINGS: From April 1, 2021, to June 9, 2022, 58 344 women aged 40-74 years underwent regular mammography screening, of whom 55 581 were included in the study. 269 (0·5%) women were diagnosed with screen-detected breast cancer based on an initial positive read: double reading by one radiologist plus AI was non-inferior for cancer detection compared with double reading by two radiologists (261 [0·5%] vs 250 [0·4%] detected cases; relative proportion 1·04 [95% CI 1·00-1·09]). Single reading by AI (246 [0·4%] vs 250 [0·4%] detected cases; relative proportion 0·98 [0·93-1·04]) and triple reading by two radiologists plus AI (269 [0·5%] vs 250 [0·4%] detected cases; relative proportion 1·08 [1·04-1·11]) were also non-inferior to double reading by two radiologists. INTERPRETATION: Replacing one radiologist with AI for independent reading of screening mammograms resulted in a 4% higher non-inferior cancer detection rate compared with radiologist double reading. Our study suggests that AI in the study setting has potential for controlled implementation, which would include risk management and real-world follow-up of performance. FUNDING: Swedish Research Council, Swedish Cancer Society, Region Stockholm, and Lunit.


Subject(s)
Breast Neoplasms , Mammography , Female , Humans , Male , Artificial Intelligence , Breast Neoplasms/diagnostic imaging , Early Detection of Cancer/methods , Mammography/methods , Prospective Studies , Sweden
8.
Radiology ; 307(5): e222639, 2023 06.
Article in English | MEDLINE | ID: mdl-37219445

ABSTRACT

Background There is considerable interest in the potential use of artificial intelligence (AI) systems in mammographic screening. However, it is essential to critically evaluate the performance of AI before it can become a modality used for independent mammographic interpretation. Purpose To evaluate the reported standalone performances of AI for interpretation of digital mammography and digital breast tomosynthesis (DBT). Materials and Methods A systematic search was conducted in PubMed, Google Scholar, Embase (Ovid), and Web of Science databases for studies published from January 2017 to June 2022. Sensitivity, specificity, and area under the receiver operating characteristic curve (AUC) values were reviewed. Study quality was assessed using the Quality Assessment of Diagnostic Accuracy Studies 2 and Comparative (QUADAS-2 and QUADAS-C, respectively). A random effects meta-analysis and meta-regression analysis were performed for overall studies and for different study types (reader studies vs historic cohort studies) and imaging techniques (digital mammography vs DBT). Results In total, 16 studies that include 1 108 328 examinations in 497 091 women were analyzed (six reader studies, seven historic cohort studies on digital mammography, and four studies on DBT). Pooled AUCs were significantly higher for standalone AI than radiologists in the six reader studies on digital mammography (0.87 vs 0.81, P = .002), but not for historic cohort studies (0.89 vs 0.96, P = .152). Four studies on DBT showed significantly higher AUCs in AI compared with radiologists (0.90 vs 0.79, P < .001). Higher sensitivity and lower specificity were seen for standalone AI compared with radiologists. Conclusion Standalone AI for screening digital mammography performed as well as or better than radiologists. Compared with digital mammography, there is an insufficient number of studies to assess the performance of AI systems in the interpretation of DBT screening examinations. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Scaranelo in this issue.


Subject(s)
Artificial Intelligence , Breast Neoplasms , Female , Humans , Breast Neoplasms/diagnostic imaging , Early Detection of Cancer/methods , Mammography/methods , Breast/diagnostic imaging , Retrospective Studies
9.
J Med Imaging (Bellingham) ; 10(Suppl 2): S22405, 2023 Feb.
Article in English | MEDLINE | ID: mdl-37035276

ABSTRACT

Purpose: In double-reading of screening mammograms, artificial intelligence (AI) algorithms hold promise as a potential replacement for one of the two readers. The choice of operating point, or abnormality threshold, for the AI algorithm will affect cancer detection and workload. In our retrospective study, the baseline approach was based on matching stand-alone reader sensitivity, while the alternative approach was based on matching the combined-reader sensitivity of two humans and of AI plus human. Approach: Full-field digital screening mammograms within the Stockholm County area between February 1, 2012, and December 30, 2015, acquired on Philips equipment, were collected. All exams of women with breast cancer within 23 months of screening and a random selection of healthy controls were included. An exam-level continuous AI abnormality score was generated (Insight MMG from Lunit Inc). Sensitivity and abnormal interpretation rates were estimated for operating points defined by the standalone-reader approach and the combined-reader approach. Results: The study population included 1684 exams of women with breast cancer and 5024 exams of healthy women. Observations of healthy women were up-sampled to attain a realistic proportion of cancer. The observed sensitivity for reader 1, 2 and 1+2 was 69.7%, 75.6%, and 78.6%, respectively, at an abnormal interpretation rate of 4.4%, 4.6%, and 6.1%, respectively. For the combination of reader 1 + AI we estimated a sensitivity of 82.4% for standalone-reader matching and 78.6% for combined-reader matching, at an abnormal interpretation rate of 12.6% and 7.0%, respectively. Conclusions: Setting the operating point by matching stand-alone AI stand-alone with a radiologist will nearly double the downstream workload compared to a modest increase of 15% for the alternative method of matching sensitivity between the combination of AI and a radiologist with two radiologists.

10.
J Med Imaging (Bellingham) ; 10(6): 061404, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36949901

ABSTRACT

Purpose: Multiple vendors are currently offering artificial intelligence (AI) computer-aided systems for triage detection, diagnosis, and risk prediction of breast cancer based on screening mammography. There is an imminent need to establish validation platforms that enable fair and transparent testing of these systems against external data. Approach: We developed validation of artificial intelligence for breast imaging (VAI-B), a platform for independent validation of AI algorithms in breast imaging. The platform is a hybrid solution, with one part implemented in the cloud and another in an on-premises environment at Karolinska Institute. Cloud services provide the flexibility of scaling the computing power during inference time, while secure on-premises clinical data storage preserves their privacy. A MongoDB database and a python package were developed to store and manage the data on-premises. VAI-B requires four data components: radiological images, AI inferences, radiologist assessments, and cancer outcomes. Results: To pilot test VAI-B, we defined a case-control population based on 8080 patients diagnosed with breast cancer and 36,339 healthy women based on the Swedish national quality registry for breast cancer. Images and radiological assessments from more than 100,000 mammography examinations were extracted from hospitals in three regions of Sweden. The images were processed by AI systems from three vendors in a virtual private cloud to produce abnormality scores related to signs of cancer in the images. A total of 105,706 examinations have been processed and stored in the database. Conclusions: We have created a platform that will allow downstream evaluation of AI systems for breast cancer detection, which enables faster development cycles for participating vendors and safer AI adoption for participating hospitals. The platform was designed to be scalable and ready to be expanded should a new vendor want to evaluate their system or should a new hospital wish to obtain an evaluation of different AI systems on their images.

11.
J Natl Compr Canc Netw ; 21(2): 143-152.e4, 2023 02.
Article in English | MEDLINE | ID: mdl-36791753

ABSTRACT

BACKGROUND: We aimed to identify factors associated with false-positive recalls in mammography screening compared with women who were not recalled and those who received true-positive recalls. METHODS: We included 29,129 women, aged 40 to 74 years, who participated in the Karolinska Mammography Project for Risk Prediction of Breast Cancer (KARMA) between 2011 and 2013 with follow-up until the end of 2017. Nonmammographic factors were collected from questionnaires, mammographic factors were generated from mammograms, and genotypes were determined using the OncoArray or an Illumina custom array. By the use of conditional and regular logistic regression models, we investigated the association between breast cancer risk factors and risk models and false-positive recalls. RESULTS: Women with a history of benign breast disease, high breast density, masses, microcalcifications, high Tyrer-Cuzick 10-year risk scores, KARMA 2-year risk scores, and polygenic risk scores were more likely to have mammography recalls, including both false-positive and true-positive recalls. Further analyses restricted to women who were recalled found that women with a history of benign breast disease and dense breasts had a similar risk of having false-positive and true-positive recalls, whereas women with masses, microcalcifications, high Tyrer-Cuzick 10-year risk scores, KARMA 2-year risk scores, and polygenic risk scores were more likely to have true-positive recalls than false-positive recalls. CONCLUSIONS: We found that risk factors associated with false-positive recalls were also likely, or even more likely, to be associated with true-positive recalls in mammography screening.


Subject(s)
Breast Neoplasms , Calcinosis , Female , Humans , Mammography , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Breast Density , Risk Factors , Early Detection of Cancer , Mass Screening , False Positive Reactions
12.
JAMA Netw Open ; 5(12): e2244212, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36454573

ABSTRACT

Importance: A discrepancy on current guidelines and clinical practice exists regarding routine imaging surveillance after mastectomy, mainly regarding the lack of adequate evidence for imaging in this setting. Objective: To investigate the usefulness of imaging surveillance in terms of cancer detection and interval cancer rates after mastectomy with or without reconstruction for patients with prior breast cancer. Data Sources: A comprehensive literature search was conducted in 3 electronic databases-PubMed, ISI Web of Science, and Scopus-without year restriction. References from relevant reviews and eligible studies were also manually searched. Study Selection: Eligible studies were defined as those conducting surveillance imaging (mammography, ultrasonography, or magnetic resonance imaging [MRI]) of patients with prior breast cancer after mastectomy with or without reconstruction that presented adequate data to calculate cancer detection rates for each surveillance method. Data Extraction and Synthesis: Independent data extraction by 2 investigators with consensus on discrepant results was performed. A quality assessment of studies was performed using the QUADAS-2 (Quality Assessment of Diagnostic Accuracy Studies-2) template. The generalized linear mixed model framework with both fixed-effects and random-effects models was used to meta-analyze the proportion of cases across studies including 3 variables: surveillance method, reconstruction after mastectomy, and surveillance measure. Main Outcomes and Measures: Three outcome measures were calculated for each eligible study and each surveillance imaging method within studies: overall cancer detection (defined as ipsilateral cancer, both palpable and nonpalpable) rate per 1000 examinations, clinically occult (nonpalpable) cancer detection rate per 1000 examinations, and interval cancer rate per 1000 examinations. Results: In total, 16 studies were eligible for the meta-analysis. The pooled overall cancer detection rates per 1000 examinations were 1.86 (95% CI, 1.05-3.30) for mammography, 2.66 (95% CI, 1.48-4.76) for ultrasonography, and 5.17 (95% CI, 1.49-17.75) for MRI. For mastectomy without reconstruction, the rate of clinically occult (nonpalpable) cancer per 1000 examinations (2.96; 95% CI, 1.38-6.32) and the interval cancer rate per 1000 examinations (3.73; 95% CI, 0.84-3.98) were lower than the overall cancer detection rate (including both palpable and nonpalpable lesions) per 1000 examinations (6.41; 95% CI, 3.09-13.25) across all imaging modalities. The interval cancer rate per 1000 examinations for mastectomy with reconstruction (3.73; 95% CI, 0.41-2.73) was comparable to the pooled cancer detection rate per 1000 examinations (4.73; 95% CI, 2.32-9.63) across all imaging modalities. In all clinical scenarios and imaging modalities, lower rates of clinically occult cancer compared with cancer detection rates were observed. Conclusions and Relevance: Lower detection rates of clinically occult-compared with overall-cancer across all 3 imaging modalities challenge the use of imaging surveillance after mastectomy, with or without reconstruction. Findings suggest that imaging surveillance in this context is unnecessary in clinical practice, at least until further studies demonstrate otherwise. Future studies should consider using the clinically occult cancer detection rate as a more clinically relevant measure in this setting.


Subject(s)
Breast Neoplasms , Mastectomy , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/surgery , Mammography , Physical Examination , Consensus
13.
Nat Med ; 28(1): 136-143, 2022 01.
Article in English | MEDLINE | ID: mdl-35027757

ABSTRACT

Screening programs must balance the benefit of early detection with the cost of overscreening. Here, we introduce a novel reinforcement learning-based framework for personalized screening, Tempo, and demonstrate its efficacy in the context of breast cancer. We trained our risk-based screening policies on a large screening mammography dataset from Massachusetts General Hospital (MGH; USA) and validated this dataset in held-out patients from MGH and external datasets from Emory University (Emory; USA), Karolinska Institute (Karolinska; Sweden) and Chang Gung Memorial Hospital (CGMH; Taiwan). Across all test sets, we find that the Tempo policy combined with an image-based artificial intelligence (AI) risk model is significantly more efficient than current regimens used in clinical practice in terms of simulated early detection per screen frequency. Moreover, we show that the same Tempo policy can be easily adapted to a wide range of possible screening preferences, allowing clinicians to select their desired trade-off between early detection and screening costs without training new policies. Finally, we demonstrate that Tempo policies based on AI-based risk models outperform Tempo policies based on less accurate clinical risk models. Altogether, our results show that pairing AI-based risk models with agile AI-designed screening policies has the potential to improve screening programs by advancing early detection while reducing overscreening.


Subject(s)
Artificial Intelligence , Breast Neoplasms/diagnosis , Mammography/methods , Early Detection of Cancer/methods , Female , Humans
14.
J Clin Oncol ; 40(16): 1732-1740, 2022 06 01.
Article in English | MEDLINE | ID: mdl-34767469

ABSTRACT

PURPOSE: Accurate risk assessment is essential for the success of population screening programs in breast cancer. Models with high sensitivity and specificity would enable programs to target more elaborate screening efforts to high-risk populations, while minimizing overtreatment for the rest. Artificial intelligence (AI)-based risk models have demonstrated a significant advance over risk models used today in clinical practice. However, the responsible deployment of novel AI requires careful validation across diverse populations. To this end, we validate our AI-based model, Mirai, across globally diverse screening populations. METHODS: We collected screening mammograms and pathology-confirmed breast cancer outcomes from Massachusetts General Hospital, USA; Novant, USA; Emory, USA; Maccabi-Assuta, Israel; Karolinska, Sweden; Chang Gung Memorial Hospital, Taiwan; and Barretos, Brazil. We evaluated Uno's concordance index for Mirai in predicting risk of breast cancer at one to five years from the mammogram. RESULTS: A total of 128,793 mammograms from 62,185 patients were collected across the seven sites, of which 3,815 were followed by a cancer diagnosis within 5 years. Mirai obtained concordance indices of 0.75 (95% CI, 0.72 to 0.78), 0.75 (95% CI, 0.70 to 0.80), 0.77 (95% CI, 0.75 to 0.79), 0.77 (95% CI, 0.73 to 0.81), 0.81 (95% CI, 0.79 to 0.82), 0.79 (95% CI, 0.76 to 0.83), and 0.84 (95% CI, 0.81 to 0.88) at Massachusetts General Hospital, Novant, Emory, Maccabi-Assuta, Karolinska, Chang Gung Memorial Hospital, and Barretos, respectively. CONCLUSION: Mirai, a mammography-based risk model, maintained its accuracy across globally diverse test sets from seven hospitals across five countries. This is the broadest validation to date of an AI-based breast cancer model and suggests that the technology can offer broad and equitable improvements in care.


Subject(s)
Breast Neoplasms , Artificial Intelligence , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/epidemiology , Early Detection of Cancer , Female , Humans , Mammography , Mass Screening
15.
Front Oncol ; 12: 1044496, 2022.
Article in English | MEDLINE | ID: mdl-36755853

ABSTRACT

Computer-aided detection systems based on deep learning have shown good performance in breast cancer detection. However, high-density breasts show poorer detection performance since dense tissues can mask or even simulate masses. Therefore, the sensitivity of mammography for breast cancer detection can be reduced by more than 20% in dense breasts. Additionally, extremely dense cases reported an increased risk of cancer compared to low-density breasts. This study aims to improve the mass detection performance in high-density breasts using synthetic high-density full-field digital mammograms (FFDM) as data augmentation during breast mass detection model training. To this end, a total of five cycle-consistent GAN (CycleGAN) models using three FFDM datasets were trained for low-to-high-density image translation in high-resolution mammograms. The training images were split by breast density BI-RADS categories, being BI-RADS A almost entirely fatty and BI-RADS D extremely dense breasts. Our results showed that the proposed data augmentation technique improved the sensitivity and precision of mass detection in models trained with small datasets and improved the domain generalization of the models trained with large databases. In addition, the clinical realism of the synthetic images was evaluated in a reader study involving two expert radiologists and one surgical oncologist.

17.
BMC Nephrol ; 22(1): 297, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34465289

ABSTRACT

BACKGROUND: Kidney disease and renal failure are associated with hospital deaths in patients with COVID - 19. We aimed to test if contrast enhancement affects short-term renal function in hospitalized COVID - 19 patients. METHODS: Plasma creatinine (P-creatinine) was measured on the day of computed tomography (CT) and 24 h, 48 h, and 4-10 days after CT. Contrast-enhanced (n = 142) and unenhanced (n = 24) groups were subdivided, based on estimated glomerular filtration rates (eGFR), > 60 and ≤ 60 ml/min/1.73 m2. Contrast-induced acute renal failure (CI-AKI) was defined as ≥27 µmol/L increase or a > 50% rise in P-creatinine from CT or initiation of renal replacement therapy during follow-up. Patients with renal replacement therapy were studied separately. We evaluated factors associated with a > 50% rise in P-creatinine at 48 h and at 4-10 days after contrast-enhanced CT. RESULTS: Median P-creatinine at 24-48 h and days 4-10 post-CT in patients with eGFR> 60 and eGFR≥30-60 in contrast-enhanced and unenhanced groups did not differ from basal values. CI-AKI was observed at 48 h and at 4-10 days post contrast administration in 24 and 36% (n = 5/14) of patients with eGFR≥30-60. Corresponding figures in the eGFR> 60 contrast-enhanced CT group were 5 and 5% respectively, (p < 0.037 and p < 0.001, Pearson χ2 test). In the former group, four of the five patients died within 30 days. Odds ratio analysis showed that an eGFR≥30-60 and 30-day mortality were associated with CK-AKI both at 48 h and 4-10 days after contrast-enhanced CT. CONCLUSION: Patients with COVID - 19 and eGFR≥30-60 had a high frequency of CK-AKI at 48 h and at 4-10 days after contrast administration, which was associated with increased 30-day mortality. For patients with eGFR≥30-60, we recommend strict indications are practiced for contrast-enhanced CT. Contrast-enhanced CT had a modest effect in patients with eGFR> 60.


Subject(s)
Acute Kidney Injury/chemically induced , COVID-19/complications , Contrast Media/adverse effects , Creatinine/blood , Iodine/adverse effects , Kidney/drug effects , Acute Kidney Injury/blood , Acute Kidney Injury/mortality , Acute Kidney Injury/therapy , Aged , COVID-19/blood , COVID-19/mortality , COVID-19/physiopathology , Female , Glomerular Filtration Rate , Humans , Kidney/diagnostic imaging , Kidney/physiopathology , Male , Middle Aged , Odds Ratio , Regression Analysis , Renal Replacement Therapy , Retrospective Studies , Time Factors , Tomography, X-Ray Computed
18.
Radiology ; 301(2): 295-308, 2021 11.
Article in English | MEDLINE | ID: mdl-34427465

ABSTRACT

Background Suppression of background parenchymal enhancement (BPE) is commonly observed after neoadjuvant chemotherapy (NAC) at contrast-enhanced breast MRI. It was hypothesized that nonsuppressed BPE may be associated with inferior response to NAC. Purpose To investigate the relationship between lack of BPE suppression and pathologic response. Materials and Methods A retrospective review was performed for women with menopausal status data who were treated for breast cancer by one of 10 drug arms (standard NAC with or without experimental agents) between May 2010 and November 2016 in the Investigation of Serial Studies to Predict Your Therapeutic Response with Imaging and Molecular Analysis 2, or I-SPY 2 TRIAL (NCT01042379). Patients underwent MRI at four points: before treatment (T0), early treatment (T1), interregimen (T2), and before surgery (T3). BPE was quantitatively measured by using automated fibroglandular tissue segmentation. To test the hypothesis effectively, a subset of examinations with BPE with high-quality segmentation was selected. BPE change from T0 was defined as suppressed or nonsuppressed for each point. The Fisher exact test and the Z tests of proportions with Yates continuity correction were used to examine the relationship between BPE suppression and pathologic complete response (pCR) in hormone receptor (HR)-positive and HR-negative cohorts. Results A total of 3528 MRI scans from 882 patients (mean age, 48 years ± 10 [standard deviation]) were reviewed and the subset of patients with high-quality BPE segmentation was determined (T1, 433 patients; T2, 396 patients; T3, 380 patients). In the HR-positive cohort, an association between lack of BPE suppression and lower pCR rate was detected at T2 (nonsuppressed vs suppressed, 11.8% [six of 51] vs 28.9% [50 of 173]; difference, 17.1% [95% CI: 4.7, 29.5]; P = .02) and T3 (nonsuppressed vs suppressed, 5.3% [two of 38] vs 27.4% [48 of 175]; difference, 22.2% [95% CI: 10.9, 33.5]; P = .003). In the HR-negative cohort, patients with nonsuppressed BPE had lower estimated pCR rate at all points, but the P values for the association were all greater than .05. Conclusions In hormone receptor-positive breast cancer, lack of background parenchymal enhancement suppression may indicate inferior treatment response. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Philpotts in this issue.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Chemotherapy, Adjuvant/methods , Contrast Media , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Neoadjuvant Therapy/methods , Adult , Aged , Breast/diagnostic imaging , Cohort Studies , Female , Humans , Middle Aged , Retrospective Studies , Treatment Outcome , Young Adult
19.
Front Med (Lausanne) ; 8: 666723, 2021.
Article in English | MEDLINE | ID: mdl-34268322

ABSTRACT

Purpose: Severe COVID-19 is associated with inflammation, thromboembolic disease, and high mortality. We studied factors associated with fatal outcomes in consecutive COVID-19 patients examined by computed tomography pulmonary angiogram (CTPA). Methods: This retrospective, single-center cohort analysis included 130 PCR-positive patients hospitalized for COVID-19 [35 women and 95 men, median age 57 years (interquartile range 51-64)] with suspected pulmonary embolism based on clinical suspicion. The presence and extent of embolism and parenchymal abnormalities on CTPA were recorded. The severity of pulmonary parenchymal involvement was stratified by two experienced radiologists into two groups: lesions affecting ≤50% or >50% of the parenchyma. Patient characteristics, radiological aspects, laboratory parameters, and 60-day mortality data were collected. Results: Pulmonary embolism was present in 26% of the patients. Most emboli were small and peripheral. Patients with widespread parenchymal abnormalities, with or without pulmonary embolism, had increased main pulmonary artery diameter (p < 0.05) and higher C-reactive protein (p < 0.01), D-dimer (p < 0.01), and troponin T (p < 0.001) and lower hemoglobin (p < 0.001). A wider main pulmonary artery diameter correlated positively with C-reactive protein (r = 0.28, p = 0.001, and n = 130) and procalcitonin. In a multivariant analysis, D-dimer >7.2 mg/L [odds ratio (±95% confidence interval) 4.1 (1.4-12.0)] and ICU stay were significantly associated with embolism (p < 0.001). The highest 60-day mortality was found in patients with widespread parenchymal abnormalities combined with pulmonary embolism (36%), followed by patients with widespread parenchymal abnormalities without pulmonary embolism (26%). In multivariate analysis, high troponin T, D-dimer, and plasma creatinine and widespread parenchymal abnormalities on CT were associated with 60-day mortality. Conclusions: Pulmonary embolism combined with widespread parenchymal abnormalities contributed to mortality risk in COVID-19. Elevated C-reactive protein, D-dimer, troponin-T, P-creatinine, and enlarged pulmonary artery were associated with a worse outcome and may mirror a more severe systemic disease. A liberal approach to radiological investigation should be recommended at clinical deterioration, when the situation allows it. Computed tomography imaging, even without intravenous contrast to assess the severity of pulmonary infiltrates, are of value to predict outcome in COVID-19. Better radiological techniques with higher resolution could potentially improve the detection of microthromboses. This could influence anticoagulant treatment strategies, preventing clinical detoriation.

SELECTION OF CITATIONS
SEARCH DETAIL
...