Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Affiliation country
Publication year range
1.
Blood ; 142(9): 794-805, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37217183

ABSTRACT

Targeted therapies for cutaneous T-cell lymphoma (CTCL) are limited and curative approaches are lacking. Furthermore, relapses and drug induced side effects are major challenges in the therapeutic management of patients with CTCL, creating an urgent need for new and effective therapies. Pathologic constitutive NF-κB activity leads to apoptosis resistance in CTCL cells and, thus, represents a promising therapeutic target in CTCL. In a preclinical study we showed the potential of dimethyl fumarate (DMF) to block NF-κB and, specifically, kill CTCL cells. To translate these findings to applications in a clinical setting, we performed a multicentric phase 2 study evaluating oral DMF therapy in 25 patients with CTCL stages Ib to IV over 24 weeks (EudraCT number 2014-000924-11/NCT number NCT02546440). End points were safety and efficacy. We evaluated skin involvement (using a modified severity weighted assessment tool [mSWAT]), pruritus, quality of life, and blood involvement, if applicable, as well as translational data. Upon skin analysis, 7 of 23 (30.4%) patients showed a response with >50% reduction in the mSWAT score. Patients with high tumor burden in the skin and blood responded best to DMF therapy. Although not generally significant, DMF also improved pruritus in several patients. Response in the blood was mixed, but we confirmed the NF-κB-inhibiting mechanism of DMF in the blood. The overall tolerability of the DMF therapy was very favorable, with mostly mild side effects. In conclusion, our study presents DMF as an effective and excellently tolerable therapeutic option in CTCL to be further evaluated in a phase 3 study or real-life patient care as well as in combination therapies. This trial was registered at www.clinicaltrials.gov as #NCT02546440.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Skin Neoplasms , Humans , Dimethyl Fumarate/therapeutic use , NF-kappa B , Quality of Life , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Neoplasm Recurrence, Local/drug therapy , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/pathology , Pruritus/drug therapy
2.
World J Urol ; 41(8): 2233-2241, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37382622

ABSTRACT

PURPOSE: To develop and validate an interpretable deep learning model to predict overall and disease-specific survival (OS/DSS) in clear cell renal cell carcinoma (ccRCC). METHODS: Digitised haematoxylin and eosin-stained slides from The Cancer Genome Atlas were used as a training set for a vision transformer (ViT) to extract image features with a self-supervised model called DINO (self-distillation with no labels). Extracted features were used in Cox regression models to prognosticate OS and DSS. Kaplan-Meier for univariable evaluation and Cox regression analyses for multivariable evaluation of the DINO-ViT risk groups were performed for prediction of OS and DSS. For validation, a cohort from a tertiary care centre was used. RESULTS: A significant risk stratification was achieved in univariable analysis for OS and DSS in the training (n = 443, log rank test, p < 0.01) and validation set (n = 266, p < 0.01). In multivariable analysis, including age, metastatic status, tumour size and grading, the DINO-ViT risk stratification was a significant predictor for OS (hazard ratio [HR] 3.03; 95%-confidence interval [95%-CI] 2.11-4.35; p < 0.01) and DSS (HR 4.90; 95%-CI 2.78-8.64; p < 0.01) in the training set but only for DSS in the validation set (HR 2.31; 95%-CI 1.15-4.65; p = 0.02). DINO-ViT visualisation showed that features were mainly extracted from nuclei, cytoplasm, and peritumoural stroma, demonstrating good interpretability. CONCLUSION: The DINO-ViT can identify high-risk patients using histological images of ccRCC. This model might improve individual risk-adapted renal cancer therapy in the future.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Proportional Hazards Models , Risk Factors , Endoscopy , Prognosis
3.
BJU Int ; 128(3): 352-360, 2021 09.
Article in English | MEDLINE | ID: mdl-33706408

ABSTRACT

OBJECTIVE: To develop a new digital biomarker based on the analysis of primary tumour tissue by a convolutional neural network (CNN) to predict lymph node metastasis (LNM) in a cohort matched for already established risk factors. PATIENTS AND METHODS: Haematoxylin and eosin (H&E) stained primary tumour slides from 218 patients (102 N+; 116 N0), matched for Gleason score, tumour size, venous invasion, perineural invasion and age, who underwent radical prostatectomy were selected to train a CNN and evaluate its ability to predict LN status. RESULTS: With 10 models trained with the same data, a mean area under the receiver operating characteristic curve (AUROC) of 0.68 (95% confidence interval [CI] 0.678-0.682) and a mean balanced accuracy of 61.37% (95% CI 60.05-62.69%) was achieved. The mean sensitivity and specificity was 53.09% (95% CI 49.77-56.41%) and 69.65% (95% CI 68.21-71.1%), respectively. These results were confirmed via cross-validation. The probability score for LNM prediction was significantly higher on image sections from N+ samples (mean [SD] N+ probability score 0.58 [0.17] vs 0.47 [0.15] N0 probability score, P = 0.002). In multivariable analysis, the probability score of the CNN (odds ratio [OR] 1.04 per percentage probability, 95% CI 1.02-1.08; P = 0.04) and lymphovascular invasion (OR 11.73, 95% CI 3.96-35.7; P < 0.001) proved to be independent predictors for LNM. CONCLUSION: In our present study, CNN-based image analyses showed promising results as a potential novel low-cost method to extract relevant prognostic information directly from H&E histology to predict the LN status of patients with prostate cancer. Our ubiquitously available technique might contribute to an improved LN status prediction.


Subject(s)
Deep Learning , Lymphatic Metastasis , Neural Networks, Computer , Prostatic Neoplasms/pathology , Aged , Humans , Male , Middle Aged , Neoplasm Grading , Prognosis , Retrospective Studies
4.
J Med Internet Res ; 23(3): e21695, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33764307

ABSTRACT

BACKGROUND: Studies have shown that artificial intelligence achieves similar or better performance than dermatologists in specific dermoscopic image classification tasks. However, artificial intelligence is susceptible to the influence of confounding factors within images (eg, skin markings), which can lead to false diagnoses of cancerous skin lesions. Image segmentation can remove lesion-adjacent confounding factors but greatly change the image representation. OBJECTIVE: The aim of this study was to compare the performance of 2 image classification workflows where images were either segmented or left unprocessed before the subsequent training and evaluation of a binary skin lesion classifier. METHODS: Separate binary skin lesion classifiers (nevus vs melanoma) were trained and evaluated on segmented and unsegmented dermoscopic images. For a more informative result, separate classifiers were trained on 2 distinct training data sets (human against machine [HAM] and International Skin Imaging Collaboration [ISIC]). Each training run was repeated 5 times. The mean performance of the 5 runs was evaluated on a multi-source test set (n=688) consisting of a holdout and an external component. RESULTS: Our findings showed that when trained on HAM, the segmented classifiers showed a higher overall balanced accuracy (75.6% [SD 1.1%]) than the unsegmented classifiers (66.7% [SD 3.2%]), which was significant in 4 out of 5 runs (P<.001). The overall balanced accuracy was numerically higher for the unsegmented ISIC classifiers (78.3% [SD 1.8%]) than for the segmented ISIC classifiers (77.4% [SD 1.5%]), which was significantly different in 1 out of 5 runs (P=.004). CONCLUSIONS: Image segmentation does not result in overall performance decrease but it causes the beneficial removal of lesion-adjacent confounding factors. Thus, it is a viable option to address the negative impact that confounding factors have on deep learning models in dermatology. However, the segmentation step might introduce new pitfalls, which require further investigations.


Subject(s)
Melanoma , Skin Neoplasms , Algorithms , Artificial Intelligence , Dermoscopy , Humans , Neural Networks, Computer , Skin Neoplasms/diagnostic imaging
5.
J Med Internet Res ; 22(9): e18091, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32915161

ABSTRACT

BACKGROUND: Early detection of melanoma can be lifesaving but this remains a challenge. Recent diagnostic studies have revealed the superiority of artificial intelligence (AI) in classifying dermoscopic images of melanoma and nevi, concluding that these algorithms should assist a dermatologist's diagnoses. OBJECTIVE: The aim of this study was to investigate whether AI support improves the accuracy and overall diagnostic performance of dermatologists in the dichotomous image-based discrimination between melanoma and nevus. METHODS: Twelve board-certified dermatologists were presented disjoint sets of 100 unique dermoscopic images of melanomas and nevi (total of 1200 unique images), and they had to classify the images based on personal experience alone (part I) and with the support of a trained convolutional neural network (CNN, part II). Additionally, dermatologists were asked to rate their confidence in their final decision for each image. RESULTS: While the mean specificity of the dermatologists based on personal experience alone remained almost unchanged (70.6% vs 72.4%; P=.54) with AI support, the mean sensitivity and mean accuracy increased significantly (59.4% vs 74.6%; P=.003 and 65.0% vs 73.6%; P=.002, respectively) with AI support. Out of the 10% (10/94; 95% CI 8.4%-11.8%) of cases where dermatologists were correct and AI was incorrect, dermatologists on average changed to the incorrect answer for 39% (4/10; 95% CI 23.2%-55.6%) of cases. When dermatologists were incorrect and AI was correct (25/94, 27%; 95% CI 24.0%-30.1%), dermatologists changed their answers to the correct answer for 46% (11/25; 95% CI 33.1%-58.4%) of cases. Additionally, the dermatologists' average confidence in their decisions increased when the CNN confirmed their decision and decreased when the CNN disagreed, even when the dermatologists were correct. Reported values are based on the mean of all participants. Whenever absolute values are shown, the denominator and numerator are approximations as every dermatologist ended up rating a varying number of images due to a quality control step. CONCLUSIONS: The findings of our study show that AI support can improve the overall accuracy of the dermatologists in the dichotomous image-based discrimination between melanoma and nevus. This supports the argument for AI-based tools to aid clinicians in skin lesion classification and provides a rationale for studies of such classifiers in real-life settings, wherein clinicians can integrate additional information such as patient age and medical history into their decisions.


Subject(s)
Artificial Intelligence/standards , Dermatologists/standards , Dermoscopy/methods , Diagnostic Imaging/classification , Melanoma/diagnostic imaging , Skin Neoplasms/diagnostic imaging , Humans , Internet , Melanoma/diagnosis , Skin Neoplasms/diagnosis , Surveys and Questionnaires
6.
Hautarzt ; 71(11): 887-897, 2020 Nov.
Article in German | MEDLINE | ID: mdl-32728813

ABSTRACT

BACKGROUND: Teledermatology addresses the problems associated with the lack of specialists and the often long waiting time for an appointment with a dermatologist. The research project Online Dermatologist-AppDoc enables a fast anonymous expert opinion and was approved on 22 October 2018 by the Landesärztekammer Baden-Württemberg for 2 years as a model project. OBJECTIVES: The aim of the present work is the presentation of the first real healthcare data for German teledematology within the framework of the external quality assurance of the model project Online Dermatologist-AppDoc. MATERIALS AND METHODS: Anonymous data records submitted to Online Dermatologist-AppDoc between 21 November 2018 and 1 August 2019 were analyzed qualitatively and quantitatively at the Department of Dermatology of the University Hospital Essen. In addition to the evaluation of the data records submitted so far, 100 cases submitted underwent a second assessment by a board-certified dermatologist to assess concordance. RESULTS: A total of 1364 cases (60.4% men, 39.6% women) were included in the current first external scientific evaluation. In 90.3% of the cases, remote diagnosis was possible. The two most frequent diagnoses were different forms of eczema (n = 270) and nevi (n = 163). Almost two thirds of the patients (64.3%) could be treated teledermatologically only. The random second examination of 100 cases resulted in an agreement of the diagnosis including the differential diagnosis/diagnoses in 97% of the cases. CONCLUSIONS: The first external scientific evaluation of the teledermatological model project Online Dermatologist-AppDoc indicates that the reduction of spatial and temporal barriers of a dermatological examination as well as the teledermatological triage have been so far successful.


Subject(s)
Dermatology , Skin Diseases , Skin Neoplasms , Telemedicine , Dermatologists , Female , Germany , Humans , Male , Skin Diseases/diagnosis , Skin Diseases/therapy
11.
Nat Commun ; 15(1): 524, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38225244

ABSTRACT

Artificial intelligence (AI) systems have been shown to help dermatologists diagnose melanoma more accurately, however they lack transparency, hindering user acceptance. Explainable AI (XAI) methods can help to increase transparency, yet often lack precise, domain-specific explanations. Moreover, the impact of XAI methods on dermatologists' decisions has not yet been evaluated. Building upon previous research, we introduce an XAI system that provides precise and domain-specific explanations alongside its differential diagnoses of melanomas and nevi. Through a three-phase study, we assess its impact on dermatologists' diagnostic accuracy, diagnostic confidence, and trust in the XAI-support. Our results show strong alignment between XAI and dermatologist explanations. We also show that dermatologists' confidence in their diagnoses, and their trust in the support system significantly increase with XAI compared to conventional AI. This study highlights dermatologists' willingness to adopt such XAI systems, promoting future use in the clinic.


Subject(s)
Melanoma , Trust , Humans , Artificial Intelligence , Dermatologists , Melanoma/diagnosis , Diagnosis, Differential
12.
JAMA Dermatol ; 160(3): 303-311, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38324293

ABSTRACT

Importance: The development of artificial intelligence (AI)-based melanoma classifiers typically calls for large, centralized datasets, requiring hospitals to give away their patient data, which raises serious privacy concerns. To address this concern, decentralized federated learning has been proposed, where classifier development is distributed across hospitals. Objective: To investigate whether a more privacy-preserving federated learning approach can achieve comparable diagnostic performance to a classical centralized (ie, single-model) and ensemble learning approach for AI-based melanoma diagnostics. Design, Setting, and Participants: This multicentric, single-arm diagnostic study developed a federated model for melanoma-nevus classification using histopathological whole-slide images prospectively acquired at 6 German university hospitals between April 2021 and February 2023 and benchmarked it using both a holdout and an external test dataset. Data analysis was performed from February to April 2023. Exposures: All whole-slide images were retrospectively analyzed by an AI-based classifier without influencing routine clinical care. Main Outcomes and Measures: The area under the receiver operating characteristic curve (AUROC) served as the primary end point for evaluating the diagnostic performance. Secondary end points included balanced accuracy, sensitivity, and specificity. Results: The study included 1025 whole-slide images of clinically melanoma-suspicious skin lesions from 923 patients, consisting of 388 histopathologically confirmed invasive melanomas and 637 nevi. The median (range) age at diagnosis was 58 (18-95) years for the training set, 57 (18-93) years for the holdout test dataset, and 61 (18-95) years for the external test dataset; the median (range) Breslow thickness was 0.70 (0.10-34.00) mm, 0.70 (0.20-14.40) mm, and 0.80 (0.30-20.00) mm, respectively. The federated approach (0.8579; 95% CI, 0.7693-0.9299) performed significantly worse than the classical centralized approach (0.9024; 95% CI, 0.8379-0.9565) in terms of AUROC on a holdout test dataset (pairwise Wilcoxon signed-rank, P < .001) but performed significantly better (0.9126; 95% CI, 0.8810-0.9412) than the classical centralized approach (0.9045; 95% CI, 0.8701-0.9331) on an external test dataset (pairwise Wilcoxon signed-rank, P < .001). Notably, the federated approach performed significantly worse than the ensemble approach on both the holdout (0.8867; 95% CI, 0.8103-0.9481) and external test dataset (0.9227; 95% CI, 0.8941-0.9479). Conclusions and Relevance: The findings of this diagnostic study suggest that federated learning is a viable approach for the binary classification of invasive melanomas and nevi on a clinically representative distributed dataset. Federated learning can improve privacy protection in AI-based melanoma diagnostics while simultaneously promoting collaboration across institutions and countries. Moreover, it may have the potential to be extended to other image classification tasks in digital cancer histopathology and beyond.


Subject(s)
Dermatology , Melanoma , Nevus , Skin Neoplasms , Humans , Melanoma/diagnosis , Artificial Intelligence , Retrospective Studies , Skin Neoplasms/diagnosis , Nevus/diagnosis
13.
Front Oncol ; 13: 1111119, 2023.
Article in English | MEDLINE | ID: mdl-36959781

ABSTRACT

Mutations in the NRAS gene are common alterations in malignant melanoma. However, there are no specific treatment options approved for NRAS-mutated melanoma patients besides immune checkpoint inhibition. Since preclinical data suggests a synergistic effect of a MEK inhibitor (MEKi) and the oncolytic virus talimogene laherparepvec (T-VEC), we have treated three melanoma patients with this combination. All of the three patients had been suffering from recurring cutaneous and subcutaneous in-transit metastases. Upon treatment one patient (case 1) presented full regression of locoregional metastases and remained progression-free until date, for almost three years. The second patient (case 2) showed a partial regression of painful gluteal satellite metastases but died from brain metastases. The third patient (case 3) showed a durable response of locoregional metastases for seven months. The combination treatment was well tolerated with common adverse events known for each single agent. This report is the first case series presenting a clinical benefit of the combined T-VEC and MEKi treatment. We suggest the combination of T-VEC and MEKi as an off-label treatment option for patients with NRAS mutations, especially with recurrent in-transit or satellite metastases.

14.
Eur J Cancer ; 183: 131-138, 2023 04.
Article in English | MEDLINE | ID: mdl-36854237

ABSTRACT

BACKGROUND: In machine learning, multimodal classifiers can provide more generalised performance than unimodal classifiers. In clinical practice, physicians usually also rely on a range of information from different examinations for diagnosis. In this study, we used BRAF mutation status prediction in melanoma as a model system to analyse the contribution of different data types in a combined classifier because BRAF status can be determined accurately by sequencing as the current gold standard, thus nearly eliminating label noise. METHODS: We trained a deep learning-based classifier by combining individually trained random forests of image, clinical and methylation data to predict BRAF-V600 mutation status in primary and metastatic melanomas of The Cancer Genome Atlas cohort. RESULTS: With our multimodal approach, we achieved an area under the receiver operating characteristic curve of 0.80, whereas the individual classifiers yielded areas under the receiver operating characteristic curve of 0.63 (histopathologic image data), 0.66 (clinical data) and 0.66 (methylation data) on an independent data set. CONCLUSIONS: Our combined approach can predict BRAF status to some extent by identifying BRAF-V600 specific patterns at the histologic, clinical and epigenetic levels. The multimodal classifiers have improved generalisability in predicting BRAF mutation status.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Proto-Oncogene Proteins B-raf/genetics , Melanoma/pathology , Skin Neoplasms/pathology , Mutation , Epigenesis, Genetic
15.
Clin Imaging ; 83: 72-76, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34990983

ABSTRACT

PURPOSE: Pulmonary embolism (PE) occurs frequently in patients with malignant melanoma (MM). The aim of this study is to determine the incidence of PE in patients with MM and to assess the clinical characteristics and mortality of MM patients with PE. MATERIAL AND METHODS: Medical records from 381 MM patients who underwent contrast-enhanced computed tomography were evaluated. Imaging parameters including location of PE and measurements of right heart dysfunction and clinical parameters including D-Dimer levels, local and distant tumor stage and time of death were analyzed. RESULTS: PE was found in 23/381 (6%) MM patients, whereby 17/23 (74%) were detected incidentally and only 6/23 (26%) were symptomatic. The presence of PE significantly correlated with elevated D-Dimers (p < 0.001), right ventricular dysfunction (p = 0.04), higher local tumor stage (≥T3) (p = 0.05), presence of visceral (p = 0.02) or cerebral metastases (p = 0.03) and increased mortality (p = 0.05). Further, patients with central PE showed an increased mortality compared to peripheral PE (p = 0.03), but no correlation was found between the localization of PE and the occurrence of clinical symptoms (p = 0.36). CONCLUSION: PE in patients with MM often occurs without clinical symptoms and is indicative for advanced disease and a poorer prognosis.


Subject(s)
Melanoma , Pulmonary Embolism , Ventricular Dysfunction, Right , Humans , Incidence , Melanoma/diagnostic imaging , Pulmonary Embolism/diagnostic imaging , Pulmonary Embolism/epidemiology , Retrospective Studies , Ventricular Dysfunction, Right/etiology
16.
PLoS One ; 17(8): e0272656, 2022.
Article in English | MEDLINE | ID: mdl-35976907

ABSTRACT

For clear cell renal cell carcinoma (ccRCC) risk-dependent diagnostic and therapeutic algorithms are routinely implemented in clinical practice. Artificial intelligence-based image analysis has the potential to improve outcome prediction and thereby risk stratification. Thus, we investigated whether a convolutional neural network (CNN) can extract relevant image features from a representative hematoxylin and eosin-stained slide to predict 5-year overall survival (5y-OS) in ccRCC. The CNN was trained to predict 5y-OS in a binary manner using slides from TCGA and validated using an independent in-house cohort. Multivariable logistic regression was used to combine of the CNNs prediction and clinicopathological parameters. A mean balanced accuracy of 72.0% (standard deviation [SD] = 7.9%), sensitivity of 72.4% (SD = 10.6%), specificity of 71.7% (SD = 11.9%) and area under receiver operating characteristics curve (AUROC) of 0.75 (SD = 0.07) was achieved on the TCGA training set (n = 254 patients / WSIs) using 10-fold cross-validation. On the external validation cohort (n = 99 patients / WSIs), mean accuracy, sensitivity, specificity and AUROC were 65.5% (95%-confidence interval [CI]: 62.9-68.1%), 86.2% (95%-CI: 81.8-90.5%), 44.9% (95%-CI: 40.2-49.6%), and 0.70 (95%-CI: 0.69-0.71). A multivariable model including age, tumor stage and metastasis yielded an AUROC of 0.75 on the TCGA cohort. The inclusion of the CNN-based classification (Odds ratio = 4.86, 95%-CI: 2.70-8.75, p < 0.01) raised the AUROC to 0.81. On the validation cohort, both models showed an AUROC of 0.88. In univariable Cox regression, the CNN showed a hazard ratio of 3.69 (95%-CI: 2.60-5.23, p < 0.01) on TCGA and 2.13 (95%-CI: 0.92-4.94, p = 0.08) on external validation. The results demonstrate that the CNN's image-based prediction of survival is promising and thus this widely applicable technique should be further investigated with the aim of improving existing risk stratification in ccRCC.


Subject(s)
Carcinoma, Renal Cell , Deep Learning , Kidney Neoplasms , Artificial Intelligence , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Neural Networks, Computer , Retrospective Studies
17.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35074902

ABSTRACT

BACKGROUND: Based on its viral-associated or UV-associated carcinogenesis, Merkel cell carcinoma (MCC) is a highly immunogenic skin cancer. Thus, clinically evident MCC occurs either in immuno-compromised patients or based on tumor-intrinsic immune escape mechanisms. This notion may explain that although advanced MCC can be effectively restrained by treatment with PD-1/PD-L1 immune checkpoint inhibitors (ICIs), a considerable percentage of patients does not benefit from ICI therapy. Biomarkers predicting ICI treatment response are currently not available. METHODS: The present multicenter retrospective study investigated clinical and molecular characteristics in 114 patients with unresectable MCC at baseline before treatment with ICI for their association with therapy response (best overall response, BOR). In a subset of 21 patients, pretreatment tumor tissue was analyzed for activation, differentiation and spatial distribution of tumor infiltrating lymphocytes (TIL). RESULTS: Of the 114 patients, n=74 (65%) achieved disease control (BOR=complete response/partial response/stable disease) on ICI. A Bayesian cumulative ordinal regression model revealed absence of immunosuppression and a limited number of tumor-involved organ systems was highly associated with a favorable therapy response. Unimpaired overall performance status, high age, normal serum lactate dehydrogenase and normal serum C reactive protein were moderately associated with disease control. While neither tumor Merkel cell polyomavirus nor tumor PD-L1 status showed a correlation with therapy response, treatment with anti-PD-1 antibodies was associated with a higher probability of disease control than treatment with anti-PD-L1 antibodies. Multiplexed immunohistochemistry demonstrated the predominance of CD8+ effector and central memory T cells (TCM) in close proximity to tumor cells in patients with a favorable therapy response. CONCLUSIONS: Our findings indicate the absence of immunosuppression, a limited number of tumor-affected organs, and a predominance of CD8+ TCM among TIL, as baseline parameters associated with a favorable response to PD-1/PD-L1 ICI therapy of advanced MCC. These factors should be considered when making treatment decisions in MCC patients.


Subject(s)
Carcinoma, Merkel Cell/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Merkel Cell/immunology , Carcinoma, Merkel Cell/mortality , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Memory T Cells/immunology , Middle Aged , Retrospective Studies , Skin Neoplasms/immunology , Skin Neoplasms/mortality
18.
Eur J Cancer ; 167: 54-69, 2022 05.
Article in English | MEDLINE | ID: mdl-35390650

ABSTRACT

BACKGROUND: Due to their ability to solve complex problems, deep neural networks (DNNs) are becoming increasingly popular in medical applications. However, decision-making by such algorithms is essentially a black-box process that renders it difficult for physicians to judge whether the decisions are reliable. The use of explainable artificial intelligence (XAI) is often suggested as a solution to this problem. We investigate how XAI is used for skin cancer detection: how is it used during the development of new DNNs? What kinds of visualisations are commonly used? Are there systematic evaluations of XAI with dermatologists or dermatopathologists? METHODS: Google Scholar, PubMed, IEEE Explore, Science Direct and Scopus were searched for peer-reviewed studies published between January 2017 and October 2021 applying XAI to dermatological images: the search terms histopathological image, whole-slide image, clinical image, dermoscopic image, skin, dermatology, explainable, interpretable and XAI were used in various combinations. Only studies concerned with skin cancer were included. RESULTS: 37 publications fulfilled our inclusion criteria. Most studies (19/37) simply applied existing XAI methods to their classifier to interpret its decision-making. Some studies (4/37) proposed new XAI methods or improved upon existing techniques. 14/37 studies addressed specific questions such as bias detection and impact of XAI on man-machine-interactions. However, only three of them evaluated the performance and confidence of humans using CAD systems with XAI. CONCLUSION: XAI is commonly applied during the development of DNNs for skin cancer detection. However, a systematic and rigorous evaluation of its usefulness in this scenario is lacking.


Subject(s)
Artificial Intelligence , Skin Neoplasms , Algorithms , Humans , Neural Networks, Computer , Skin Neoplasms/diagnosis
19.
Eur J Cancer ; 173: 307-316, 2022 09.
Article in English | MEDLINE | ID: mdl-35973360

ABSTRACT

BACKGROUND: Image-based cancer classifiers suffer from a variety of problems which negatively affect their performance. For example, variation in image brightness or different cameras can already suffice to diminish performance. Ensemble solutions, where multiple model predictions are combined into one, can improve these problems. However, ensembles are computationally intensive and less transparent to practitioners than single model solutions. Constructing model soups, by averaging the weights of multiple models into a single model, could circumvent these limitations while still improving performance. OBJECTIVE: To investigate the performance of model soups for a dermoscopic melanoma-nevus skin cancer classification task with respect to (1) generalisation to images from other clinics, (2) robustness against small image changes and (3) calibration such that the confidences correspond closely to the actual predictive uncertainties. METHODS: We construct model soups by fine-tuning pre-trained models on seven different image resolutions and subsequently averaging their weights. Performance is evaluated on a multi-source dataset including holdout and external components. RESULTS: We find that model soups improve generalisation and calibration on the external component while maintaining performance on the holdout component. For robustness, we observe performance improvements for pertubated test images, while the performance on corrupted test images remains on par. CONCLUSIONS: Overall, souping for skin cancer classifiers has a positive effect on generalisation, robustness and calibration. It is easy for practitioners to implement and by combining multiple models into a single model, complexity is reduced. This could be an important factor in achieving clinical applicability, as less complexity generally means more transparency.


Subject(s)
Melanoma , Skin Neoplasms , Dermoscopy/methods , Humans , Melanoma/diagnostic imaging , Sensitivity and Specificity , Skin Neoplasms/diagnostic imaging , Melanoma, Cutaneous Malignant
20.
Eur J Cancer ; 155: 200-215, 2021 09.
Article in English | MEDLINE | ID: mdl-34391053

ABSTRACT

BACKGROUND: Gastrointestinal cancers account for approximately 20% of all cancer diagnoses and are responsible for 22.5% of cancer deaths worldwide. Artificial intelligence-based diagnostic support systems, in particular convolutional neural network (CNN)-based image analysis tools, have shown great potential in medical computer vision. In this systematic review, we summarise recent studies reporting CNN-based approaches for digital biomarkers for characterization and prognostication of gastrointestinal cancer pathology. METHODS: Pubmed and Medline were screened for peer-reviewed papers dealing with CNN-based gastrointestinal cancer analyses from histological slides, published between 2015 and 2020.Seven hundred and ninety titles and abstracts were screened, and 58 full-text articles were assessed for eligibility. RESULTS: Sixteen publications fulfilled our inclusion criteria dealing with tumor or precursor lesion characterization or prognostic and predictive biomarkers: 14 studies on colorectal or rectal cancer, three studies on gastric cancer and none on esophageal cancer. These studies were categorised according to their end-points: polyp characterization, tumor characterization and patient outcome. Regarding the translation into clinical practice, we identified several studies demonstrating generalization of the classifier with external tests and comparisons with pathologists, but none presenting clinical implementation. CONCLUSIONS: Results of recent studies on CNN-based image analysis in gastrointestinal cancer pathology are promising, but studies were conducted in observational and retrospective settings. Large-scale trials are needed to assess performance and predict clinical usefulness. Furthermore, large-scale trials are required for approval of CNN-based prediction models as medical devices.


Subject(s)
Deep Learning/standards , Gastrointestinal Neoplasms/classification , Gastrointestinal Neoplasms/pathology , Humans , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL