1
|
Li Y, Tao X, Ye Y, Tang Y, Xu Z, Tian Y, Liu Z, Zhao J. Prognostic nomograms for young breast cancer: A retrospective study based on the SEER and METABRIC databases. CANCER INNOVATION 2024; 3:e152. [PMID: 39464427 PMCID: PMC11503687 DOI: 10.1002/cai2.152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 10/29/2024]
Abstract
Background Young breast cancer (YBC) is a subset of breast cancer that is often more aggressive, but less is known about its prognosis. In this study, we aimed to generate nomograms to predict the overall survival (OS) and breast cancer-specific survival (BCSS) of YBC patients. Methods Data of women diagnosed with YBC between 2010 and 2020 were obtained from the Surveillance, Epidemiology, and End Results (SEER) database. The patients were randomly allocated into a training cohort (n = 15,227) and internal validation cohort (n = 6,526) at a 7:3 ratio. With the Cox regression models, significant prognostic factors were identified and used to construct 3-, 5-, and 10-year nomograms of OS and BCSS. Data from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) database were used as an external validation cohort (n = 90). Results We constructed nomograms incorporating 10 prognostic factors for OS and BCSS. These nomograms demonstrated strong predictive accuracy for OS and BCSS in the training cohort, with C-indexes of 0.806 and 0.813, respectively. The calibration curves verified that the nomograms have good prediction accuracy. Decision curve analysis demonstrated their practical clinical value for predicting YBC patient survival rates. Additionally, we provided dynamic nomograms to improve the operability of the results. The risk stratification ability assessment also showed that the OS and BCSS rates of the low-risk group were significantly better than those of the high-risk group. Conclusions Here, we generated and validated more comprehensive and accurate OS and BCSS nomograms than models previously developed for YBC. These nomograms can help clinicians evaluate patient prognosis and make clinical decisions.
Collapse
Affiliation(s)
- Yongxin Li
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai UniversityXiningQinghaiChina
| | - Xinlong Tao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai UniversityXiningQinghaiChina
| | - Yinyin Ye
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai UniversityXiningQinghaiChina
| | - Yuyao Tang
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai UniversityXiningQinghaiChina
| | | | - Yaming Tian
- Department of ImagingAffiliated Hospital of Qinghai UniversityXiningQinghaiChina
| | - Zhen Liu
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai UniversityXiningQinghaiChina
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai UniversityXiningQinghaiChina
| |
Collapse
|
2
|
Herrmann K, Gafita A, de Bono JS, Sartor O, Chi KN, Krause BJ, Rahbar K, Tagawa ST, Czernin J, El-Haddad G, Wong CC, Zhang Z, Wilke C, Mirante O, Morris MJ, Fizazi K. Multivariable models of outcomes with [ 177Lu]Lu-PSMA-617: analysis of the phase 3 VISION trial. EClinicalMedicine 2024; 77:102862. [PMID: 39430616 PMCID: PMC11490806 DOI: 10.1016/j.eclinm.2024.102862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 10/22/2024] Open
Abstract
Background [177Lu]Lu-PSMA-617 (177Lu-PSMA-617) prolonged life in patients with metastatic castration-resistant prostate cancer (mCRPC) in VISION (NCT03511664). However, distinguishing between patients likely and unlikely to respond remains a clinical challenge. We present the first multivariable models of outcomes with 177Lu-PSMA-617 built using data from VISION, a large prospective phase 3 clinical trial powered for overall survival. Methods Adults with progressive post androgen receptor pathway inhibitor and taxane prostate-specific membrane antigen (PSMA)-positive mCRPC received 177Lu-PSMA-617 plus protocol-permitted standard of care (SoC) or SoC alone. In this post hoc analysis, multivariable Cox proportional hazards models of overall survival (OS) and radiographic progression-free survival (rPFS), and a logistic regression model of prostate-specific antigen response (≥50% decline; PSA50) were constructed and evaluated using C-index or receiver operating characteristic (ROC) analyses with bootstrapping validation. Nomograms were constructed for visualisation. Findings Patients were randomised between June 2018 and October 2019. Data from all 551 patients in the 177Lu-PSMA-617 arm were analysed in multivariable modelling. The OS nomogram (C-index, 0.73; 95% confidence interval [CI], 0.70-0.76) included whole-body maximum standardised uptake value (SUVmax), time since diagnosis, opioid analgesic use, aspartate aminotransferase, haemoglobin, lymphocyte count, presence of PSMA-positive lesions in lymph nodes, lactate dehydrogenase (LDH), alkaline phosphatase (ALP), and neutrophil count. The rPFS nomogram (C-index, 0.68; 0.65-0.72) included SUVmax, time since diagnosis, opioid analgesic use, lymphocyte count, presence of liver metastases by computed tomography, LDH, and ALP. The PSA50 nomogram (area under ROC curve, 0.72; 95% CI, 0.68-0.77) included SUVmax, lymphocyte count and ALP. Performances of the OS and rPFS models were maintained when they were reconstructed excluding SUVmax. Interpretation These models of outcomes with 177Lu-PSMA-617 are the first built using prospective phase 3 data. They show that a combination of pretreatment laboratory, clinical, and imaging parameters, reflecting both patient and tumour status, influences outcomes. These models are important for aiding treatment selection, patient management, and clinical trial design. Funding Novartis.
Collapse
Affiliation(s)
- Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK), Essen University Hospital, Essen, Germany
| | - Andrei Gafita
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Johann S. de Bono
- Division of Clinical Studies, The Institute of Cancer Research and the Royal Marsden Hospital, London, UK
| | - Oliver Sartor
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Kim N. Chi
- Division of Medical Oncology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Bernd J. Krause
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Kambiz Rahbar
- Department of Nuclear Medicine, Münster University Hospital, Münster, Germany
| | - Scott T. Tagawa
- Department of Medicine, Division of Hematology and Medical Oncology and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology and Institute of Urologic Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ghassan El-Haddad
- Department of Diagnostic Imaging and Interventional Radiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Connie C. Wong
- Novartis Pharmaceuticals Corporation, Cambridge, MA, USA
| | - Zhaojie Zhang
- Novartis Pharmaceuticals Corporation, Cambridge, MA, USA
| | | | - Osvaldo Mirante
- Advanced Accelerator Applications, A Novartis Company, Geneva, Switzerland
| | - Michael J. Morris
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karim Fizazi
- Medical Oncology Department, Institut Gustave Roussy, University of Paris-Saclay, Villejuif, France
| |
Collapse
|
3
|
van den Ende T, Kuijper SC, Widaatalla Y, Noortman WA, van Velden FHP, Woodruff HC, van der Pol Y, Moldovan N, Pegtel DM, Derks S, Bijlsma MF, Mouliere F, de Geus-Oei PLF, Lambin PP, van Laarhoven PHWM. Integrating Clinical Variables, Radiomics, and Tumor-derived Cell-free DNA for Enhanced Prediction of Resectable Esophageal Adenocarcinoma Outcomes. Int J Radiat Oncol Biol Phys 2024:S0360-3016(24)03468-0. [PMID: 39424077 DOI: 10.1016/j.ijrobp.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/13/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The value of integrating clinical variables, radiomics, and tumor-derived cell-free DNA (cfDNA) for the prediction of survival and response to chemoradiation of resectable esophageal adenocarcinoma (rEAC) patients is not yet known. Our aim was to investigate if radiomics and cfDNA metrics combined with clinical variables can improve personalized predictions. METHODS A cohort of 111 rEAC patients from two centers treated with neoadjuvant chemoradiotherapy was used for exploratory retrospective analyses. Models combining the clinical variables of the SOURCE survival model with radiomic features and cfDNA, were built using elastic net regression and internally validated using 5-fold cross validation. Model performance for overall survival (OS) and time to progression (TTP) were evaluated with the C-index and the area under the curve (AUC) for pathological complete response (pCR) RESULTS: The best performing baseline models for OS and TTP were based on the combination of SOURCE-cfDNA which reached a C-index of 0.55 and 0.59 compared to 0.44-0.45 with SOURCE alone. The addition of re-staging PET radiomics to SOURCE was the most promising addition for predicting OS (C-index: 0.65) and TTP (C-index: 0.60). Baseline risk-stratification was achieved for OS and TTP by combining SOURCE with radiomics or cfDNA, log-rank p<0.01. The best performing combination model for the prediction of pCR reached an AUC of 0.61 compared to 0.47 with SOURCE variables alone. CONCLUSIONS The addition of radiomics and cfDNA can improve the performance of an established survival model. External validity needs to be further assessed in future studies together with the optimization of radiomic pipelines.
Collapse
Affiliation(s)
- Tom van den Ende
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Steven C Kuijper
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| | - Yousif Widaatalla
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Wyanne A Noortman
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands; TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Floris H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6200 Maastricht, The Netherlands
| | - Ymke van der Pol
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Norbert Moldovan
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - D Michiel Pegtel
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Sarah Derks
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Oncology, De Boelelaan 1117, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Maarten F Bijlsma
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Oncode Institute, Utrecht, the Netherlands; Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - Florent Mouliere
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Prof Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands; TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands; Department of Radiation Science & Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Prof Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6200 Maastricht, The Netherlands
| | - Prof Hanneke W M van Laarhoven
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Avendano D, Marino MA, Bosques-Palomo BA, Dávila-Zablah Y, Zapata P, Avalos-Montes PJ, Armengol-García C, Sofia C, Garza-Montemayor M, Pinker K, Cardona-Huerta S, Tamez-Peña J. Validation of the Mirai model for predicting breast cancer risk in Mexican women. Insights Imaging 2024; 15:244. [PMID: 39387984 PMCID: PMC11466924 DOI: 10.1186/s13244-024-01808-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/01/2024] [Indexed: 10/12/2024] Open
Abstract
OBJECTIVES To validate the performance of Mirai, a mammography-based deep learning model, in predicting breast cancer risk over a 1-5-year period in Mexican women. METHODS This retrospective single-center study included mammograms in Mexican women who underwent screening mammography between January 2014 and December 2016. For women with consecutive mammograms during the study period, only the initial mammogram was included. Pathology and imaging follow-up served as the reference standard. Model performance in the entire dataset was evaluated, including the concordance index (C-Index) and area under the receiver operating characteristic curve (AUC). Mirai's performance in terms of AUC was also evaluated between mammography systems (Hologic versus IMS). Clinical utility was evaluated by determining a cutoff point for Mirai's continuous risk index based on identifying the top 10% of patients in the high-risk category. RESULTS Of 3110 patients (median age 52.6 years ± 8.9), throughout the 5-year follow-up period, 3034 patients remained cancer-free, while 76 patients developed breast cancer. Mirai achieved a C-index of 0.63 (95% CI: 0.6-0.7) for the entire dataset. Mirai achieved a higher mean C-index in the Hologic subgroup (0.63 [95% CI: 0.5-0.7]) versus the IMS subgroup (0.55 [95% CI: 0.4-0.7]). With a Mirai index score > 0.029 (10% threshold) to identify high-risk individuals, the study revealed that individuals in the high-risk group had nearly three times the risk of developing breast cancer compared to those in the low-risk group. CONCLUSIONS Mirai has a moderate performance in predicting future breast cancer among Mexican women. CRITICAL RELEVANCE STATEMENT Prospective efforts should refine and apply the Mirai model, especially to minority populations and women aged between 30 and 40 years who are currently not targeted for routine screening. KEY POINTS The applicability of AI models to non-White, minority populations remains understudied. The Mirai model is linked to future cancer events in Mexican women. Further research is needed to enhance model performance and establish usage guidelines.
Collapse
Affiliation(s)
- Daly Avendano
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| | - Maria Adele Marino
- Department of Biomedical Sciences and Morphologic and Functional Imaging, Policlinico Universitario "G. Martino," University of Messina, Messina, Italy
| | | | | | - Pedro Zapata
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| | - Pablo J Avalos-Montes
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| | - Cecilio Armengol-García
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| | - Carmelo Sofia
- Department of Biomedical Sciences and Morphologic and Functional Imaging, Policlinico Universitario "G. Martino," University of Messina, Messina, Italy
| | | | - Katja Pinker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Servando Cardona-Huerta
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Nuevo León, México.
| | - José Tamez-Peña
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| |
Collapse
|
5
|
Wang J, Jiang X, Ning J. Evaluating dynamic and predictive discrimination for recurrent event models: use of a time-dependent C-index. Biostatistics 2024; 25:1140-1155. [PMID: 37952117 PMCID: PMC11471962 DOI: 10.1093/biostatistics/kxad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023] Open
Abstract
Interest in analyzing recurrent event data has increased over the past few decades. One essential aspect of a risk prediction model for recurrent event data is to accurately distinguish individuals with different risks of developing a recurrent event. Although the concordance index (C-index) effectively evaluates the overall discriminative ability of a regression model for recurrent event data, a local measure is also desirable to capture dynamic performance of the regression model over time. Therefore, in this study, we propose a time-dependent C-index measure for inferring the model's discriminative ability locally. We formulated the C-index as a function of time using a flexible parametric model and constructed a concordance-based likelihood for estimation and inference. We adapted a perturbation-resampling procedure for variance estimation. Extensive simulations were conducted to investigate the proposed time-dependent C-index's finite-sample performance and estimation procedure. We applied the time-dependent C-index to three regression models of a study of re-hospitalization in patients with colorectal cancer to evaluate the models' discriminative capability.
Collapse
Affiliation(s)
- Jian Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 7007 Bertner Ave, 1MC12.3557, Houston, TX 77030, United States
| | - Xinyang Jiang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 7007 Bertner Ave, 1MC12.3557, Houston, TX 77030, United States
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 7007 Bertner Ave, 1MC12.3557, Houston, TX 77030, United States
| |
Collapse
|
6
|
da Fonsecaa LG, de Melob MAZ, da Silveirac THM, Yamamotod VJ, Hashizumee PHS, Sabbagaf J. Prognostic role of albumin-bilirubin (ALBI) score and Child-Pugh classification in patients with advanced hepatocellular carcinoma under systemic treatment. Ecancermedicalscience 2024; 18:1748. [PMID: 39421189 PMCID: PMC11484683 DOI: 10.3332/ecancer.2024.1748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Indexed: 10/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a lethal malignancy associated with cirrhosis and liver dysfunction. The aim of this study is to characterize a cohort of patients with advanced HCC according to liver function-related variables and evaluate the prognostic significance of Child-Pugh (CP) and albumin-bilirubin (ALBI) scores. A database of 406 HCC patients treated between 2009 and 2023 was retrospectively evaluated. Clinical and laboratory parameters were collected to classify patients into ALBI and CP scores. Survival was estimated using the Kaplan-Meier method and multivariate models were used to evaluate prognosis prediction. In this cohort, 337 (83%) patients were classified as CP-A, while 69 (17%) as CP-B. Additionally, according to ALBI score, 159 (39.2%) individuals were categorised as ALBI-1, 233 (57.4%) as ALBI-2 and 14 (3.4%) as ALBI-3. A statistically significant association between both classifications was observed (p < 0.001). CP and ALBI scores were independently associated with prognosis (Hazard ratio = 2.93 and 1.66, respectively), with better survival for patients with CP-A (versus B) and ALBI-1 (versus -2 and -3). ALBI score showed better predictive performance versus CP (c Harrell´s C index = 0.65 versus 0.62; p = 0.008) and ALBI evolution during the first month of treatment was associated with overall survival. Additionally, ALBI score was able to define distinct prognostic subgroups within CP-A patients. In conclusion, liver function scores, such as ALBI and CP, have a clinically relevant prognostic role in patients with advanced HCC under systemic treatment. ALBI score is a more granular scoring scale than CP, and enables a more precise evaluation of patients with CP-A.
Collapse
Affiliation(s)
- Leonardo G da Fonsecaa
- Department of Oncology, ICESP - Instituto do Cancer do Estado de Sao Paulo, University of Sao Paulo School of Medicine, Sao Paulo, SP 01246-000, Brazil
- https://orcid.org/0000-0002-0216-3618
| | - Marina Acevedo Zarzar de Melob
- Department of Oncology, ICESP - Instituto do Cancer do Estado de Sao Paulo, University of Sao Paulo School of Medicine, Sao Paulo, SP 01246-000, Brazil
- https://orcid.org/0000-0002-3031-7928
| | - Thamires Haick Martins da Silveirac
- Department of Oncology, ICESP - Instituto do Cancer do Estado de Sao Paulo, University of Sao Paulo School of Medicine, Sao Paulo, SP 01246-000, Brazil
- https://orcid.org/0009-0000-8427-8592
| | - Victor Junji Yamamotod
- Department of Oncology, ICESP - Instituto do Cancer do Estado de Sao Paulo, University of Sao Paulo School of Medicine, Sao Paulo, SP 01246-000, Brazil
- https://orcid.org/0000-0002-1422-0042
| | - Pedro Henrique Shimiti Hashizumee
- Department of Oncology, ICESP - Instituto do Cancer do Estado de Sao Paulo, University of Sao Paulo School of Medicine, Sao Paulo, SP 01246-000, Brazil
- https://orcid.org/0000-0002-9159-6756
| | - Jorge Sabbagaf
- Department of Oncology, ICESP - Instituto do Cancer do Estado de Sao Paulo, University of Sao Paulo School of Medicine, Sao Paulo, SP 01246-000, Brazil
- https://orcid.org/0000-0003-0715-4670
| |
Collapse
|
7
|
Escribano-Serrat S, Rodríguez-Lobato LG, Suárez-Lledó M, Pedraza A, Charry P, Cid J, Lozano M, Esteve J, Rosiñol L, Fernández-Avilés F, Carreras E, Díaz-Ricart M, Martínez C, Rovira M, Salas MQ. Improving the EASIX' predictive power for NRM in adults undergoing allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 2024; 59:1022-1024. [PMID: 38521886 DOI: 10.1038/s41409-024-02267-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/25/2024]
Affiliation(s)
- Silvia Escribano-Serrat
- Hematopathology, Pathology Department, CDB, IDIBAPS, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Luis Gerardo Rodríguez-Lobato
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - María Suárez-Lledó
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Alexandra Pedraza
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Paola Charry
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Joan Cid
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Miquel Lozano
- Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, ICMHO, IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Jordi Esteve
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Laura Rosiñol
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Francesc Fernández-Avilés
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Enric Carreras
- Fundació i Institut de Recerca Josep Carreras contra la leucèmia (Campus Clínic), Barcelona, Spain
| | - Maribel Díaz-Ricart
- Hematopathology, Pathology Department, CDB, IDIBAPS, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Carmen Martínez
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - Montserrat Rovira
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain
| | - María Queralt Salas
- Hematopoietic Transplantation Unit and Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), IDIBAPS, Hospital Clínic, Barcelona, Spain.
| |
Collapse
|
8
|
Amorim AM, Piochi LF, Gaspar AT, Preto A, Rosário-Ferreira N, Moreira IS. Advancing Drug Safety in Drug Development: Bridging Computational Predictions for Enhanced Toxicity Prediction. Chem Res Toxicol 2024; 37:827-849. [PMID: 38758610 PMCID: PMC11187637 DOI: 10.1021/acs.chemrestox.3c00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
The attrition rate of drugs in clinical trials is generally quite high, with estimates suggesting that approximately 90% of drugs fail to make it through the process. The identification of unexpected toxicity issues during preclinical stages is a significant factor contributing to this high rate of failure. These issues can have a major impact on the success of a drug and must be carefully considered throughout the development process. These late-stage rejections or withdrawals of drug candidates significantly increase the costs associated with drug development, particularly when toxicity is detected during clinical trials or after market release. Understanding drug-biological target interactions is essential for evaluating compound toxicity and safety, as well as predicting therapeutic effects and potential off-target effects that could lead to toxicity. This will enable scientists to predict and assess the safety profiles of drug candidates more accurately. Evaluation of toxicity and safety is a critical aspect of drug development, and biomolecules, particularly proteins, play vital roles in complex biological networks and often serve as targets for various chemicals. Therefore, a better understanding of these interactions is crucial for the advancement of drug development. The development of computational methods for evaluating protein-ligand interactions and predicting toxicity is emerging as a promising approach that adheres to the 3Rs principles (replace, reduce, and refine) and has garnered significant attention in recent years. In this review, we present a thorough examination of the latest breakthroughs in drug toxicity prediction, highlighting the significance of drug-target binding affinity in anticipating and mitigating possible adverse effects. In doing so, we aim to contribute to the development of more effective and secure drugs.
Collapse
Affiliation(s)
- Ana M.
B. Amorim
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PhD
Programme in Biosciences, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PURR.AI,
Rua Pedro Nunes, IPN Incubadora, Ed C, 3030-199 Coimbra, Portugal
| | - Luiz F. Piochi
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Ana T. Gaspar
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - António
J. Preto
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- PhD Programme
in Experimental Biology and Biomedicine, Institute for Interdisciplinary
Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Nícia Rosário-Ferreira
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Irina S. Moreira
- Department
of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-UC—Center
for Neuroscience and Cell Biology, University
of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CIBB—Centre
for Innovative Biomedicine and Biotechnology, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| |
Collapse
|
9
|
Kim C, Kwon JM, Lee J, Jo H, Gwon D, Jang JH, Sung MK, Park SW, Kim C, Oh MY. Deep learning model integrating radiologic and clinical data to predict mortality after ischemic stroke. Heliyon 2024; 10:e31000. [PMID: 38826743 PMCID: PMC11141274 DOI: 10.1016/j.heliyon.2024.e31000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
Objective Most prognostic indexes for ischemic stroke mortality lack radiologic information. We aimed to create and validate a deep learning-based mortality prediction model using brain diffusion weighted imaging (DWI), apparent diffusion coefficient (ADC), and clinical factors. Methods Data from patients with ischemic stroke who admitted to tertiary hospital during acute periods from 2013 to 2019 were collected and split into training (n = 1109), validation (n = 437), and internal test (n = 654). Data from patients from secondary cardiovascular center was used for external test set (n = 507). The algorithm for predicting mortality, based on DWI and ADC (DLP_DWI), was initially trained. Subsequently, important clinical factors were integrated into this model to create the integrated model (DLP_INTG). The performance of DLP_DWI and DLP_INTG was evaluated by using time-dependent area under the receiver operating characteristic curves (TD AUCs) and Harrell concordance index (C-index) at one-year mortality. Results The TD AUC of DLP_DWI was 0.643 in internal test set, and 0.785 in the external dataset. DLP_INTG had a higher performance at predicting one-year mortality than premise score in internal dataset (TD- AUC: 0.859 vs. 0.746; p = 0.046), and in external dataset (TD- AUC: 0.876 vs. 0.808; p = 0.007). DLP_DWI and DLP_INTG exhibited strong discrimination for the high-risk group for one-year mortality. Interpretation A deep learning model using brain DWI, ADC and the clinical factors was capable of predicting mortality in patients with ischemic stroke.
Collapse
Affiliation(s)
- Changi Kim
- Department of Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Joon-myoung Kwon
- Medical Research Team, Medical AI Inc, DC, USA
- Department of Critical Care Emergency Medicine, Incheon Sejong Hospital, Incheon, Republic of Korea
- Artificial Intelligence and Big Data Research Center, Sejong Medical Research Institute, Bucheon, Republic of Korea
| | - Jiyeong Lee
- Department of Neurology, Bucheon Sejong Hospital, Bucheon, Republic of Korea
| | | | - Dowan Gwon
- Department of Digital&Biohealth, Group of AI/DX Business, KT, Seoul, Republic of Korea
| | - Jae Hoon Jang
- Department of Family Medicine, College of Medicine, KyungHee University, Seoul, Republic of Korea
| | - Min Kyu Sung
- Department of Family Medicine, College of Medicine, KyungHee University, Seoul, Republic of Korea
| | - Sang Won Park
- Department of Medical Informatics, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
- Institute of Medical Science, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Chulho Kim
- Department of Neurology, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Mi-Young Oh
- Department of Neurology, Bucheon Sejong Hospital, Bucheon, Republic of Korea
| |
Collapse
|
10
|
Mandla R, Schroeder P, Porneala B, Florez JC, Meigs JB, Mercader JM, Leong A. Polygenic scores for longitudinal prediction of incident type 2 diabetes in an ancestrally and medically diverse primary care physician network: a patient cohort study. Genome Med 2024; 16:63. [PMID: 38671457 PMCID: PMC11046943 DOI: 10.1186/s13073-024-01337-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The clinical utility of genetic information for type 2 diabetes (T2D) prediction with polygenic scores (PGS) in ancestrally diverse, real-world US healthcare systems is unclear, especially for those at low clinical phenotypic risk for T2D. METHODS We tested the association of PGS with T2D incidence in patients followed within a primary care practice network over 16 years in four hypothetical scenarios that varied by clinical data availability (N = 14,712): (1) age and sex; (2) age, sex, body mass index (BMI), systolic blood pressure, and family history of T2D; (3) all variables in (2) and random glucose; and (4) all variables in (3), HDL, total cholesterol, and triglycerides, combined in a clinical risk score (CRS). To determine whether genetic effects differed by baseline clinical risk, we tested for interaction with the CRS. RESULTS PGS was associated with incident T2D in all models. Adjusting for age and sex only, the Hazard Ratio (HR) per PGS standard deviation (SD) was 1.76 (95% CI 1.68, 1.84) and the HR of top 5% of PGS vs interquartile range (IQR) was 2.80 (2.39, 3.28). Adjusting for the CRS, the HR per SD was 1.48 (1.40, 1.57) and HR of the top 5% of PGS vs IQR was 2.09 (1.72, 2.55). Genetic effects differed by baseline clinical risk ((PGS-CRS interaction p = 0.05; CRS below the median: HR 1.60 (1.43, 1.79); CRS above the median: HR 1.45 (1.35, 1.55)). CONCLUSIONS Genetic information can help identify high-risk patients even among those perceived to be low risk in a clinical evaluation.
Collapse
Affiliation(s)
- Ravi Mandla
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Philip Schroeder
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Bianca Porneala
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital, 100 Cambridge St. Fl. 16, Boston, MA, 02114, USA
| | - Jose C Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - James B Meigs
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital, 100 Cambridge St. Fl. 16, Boston, MA, 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Josep M Mercader
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Aaron Leong
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Division of General Internal Medicine, Department of Medicine, Massachusetts General Hospital, 100 Cambridge St. Fl. 16, Boston, MA, 02114, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Sun X, Nong M, Meng F, Sun X, Jiang L, Li Z, Zhang P. Architecting the metabolic reprogramming survival risk framework in LUAD through single-cell landscape analysis: three-stage ensemble learning with genetic algorithm optimization. J Transl Med 2024; 22:353. [PMID: 38622716 PMCID: PMC11017668 DOI: 10.1186/s12967-024-05138-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/27/2024] [Indexed: 04/17/2024] Open
Abstract
Recent studies have increasingly revealed the connection between metabolic reprogramming and tumor progression. However, the specific impact of metabolic reprogramming on inter-patient heterogeneity and prognosis in lung adenocarcinoma (LUAD) still requires further exploration. Here, we introduced a cellular hierarchy framework according to a malignant and metabolic gene set, named malignant & metabolism reprogramming (MMR), to reanalyze 178,739 single-cell reference profiles. Furthermore, we proposed a three-stage ensemble learning pipeline, aided by genetic algorithm (GA), for survival prediction across 9 LUAD cohorts (n = 2066). Throughout the pipeline of developing the three stage-MMR (3 S-MMR) score, double training sets were implemented to avoid over-fitting; the gene-pairing method was utilized to remove batch effect; GA was harnessed to pinpoint the optimal basic learner combination. The novel 3 S-MMR score reflects various aspects of LUAD biology, provides new insights into precision medicine for patients, and may serve as a generalizable predictor of prognosis and immunotherapy response. To facilitate the clinical adoption of the 3 S-MMR score, we developed an easy-to-use web tool for risk scoring as well as therapy stratification in LUAD patients. In summary, we have proposed and validated an ensemble learning model pipeline within the framework of metabolic reprogramming, offering potential insights for LUAD treatment and an effective approach for developing prognostic models for other diseases.
Collapse
Affiliation(s)
- Xinti Sun
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Minyu Nong
- School of Clinical Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Fei Meng
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaojuan Sun
- Department of Oncology, Qingdao University Affiliated Hospital, Qingdao, Shandong, China
| | - Lihe Jiang
- School of Clinical Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Zihao Li
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhang
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
12
|
Zeng J, Song D, Li K, Cao F, Zheng Y. Deep learning model for predicting postoperative survival of patients with gastric cancer. Front Oncol 2024; 14:1329983. [PMID: 38628668 PMCID: PMC11018873 DOI: 10.3389/fonc.2024.1329983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Background Prognostic prediction for surgical treatment of gastric cancer remains valuable in clinical practice. This study aimed to develop survival models for postoperative gastric cancer patients. Methods Eleven thousand seventy-five patients from the Surveillance, Epidemiology, and End Results (SEER) database were included, and 122 patients from the Chinese database were used for external validation. The training cohort was created to create three separate models, including Cox regression, RSF, and DeepSurv, using data from the SEER database split into training and test cohorts with a 7:3 ratio. Test cohort was used to evaluate model performance using c-index, Brier scores, calibration, and the area under the curve (AUC). The new risk stratification based on the best model will be compared with the AJCC stage on the test and Chinese cohorts using decision curve analysis (DCA), the net reclassification index (NRI), and integrated discrimination improvement (IDI). Results It was discovered that the DeepSurv model predicted postoperative gastric cancer patients' overall survival (OS) with a c-index of 0.787; the area under the curve reached 0.781, 0.798, 0.868 at 1-, 3- and 5- years, respectively; the Brier score was below 0.25 at different time points; showing an advantage over the Cox and RSF models. The results are also validated in the China cohort. The calibration plots demonstrated good agreement between the DeepSurv model's forecast and actual results. The NRI values (test cohort: 0.399, 0.288, 0.267 for 1-, 3- and 5-year OS prediction; China cohort:0.399, 0.288 for 1- and 3-year OS prediction) and IDI (test cohort: 0.188, 0.169, 0.157 for 1-, 3- and 5-year OS prediction; China cohort: 0.189, 0.169 for 1- and 3-year OS prediction) indicated that the risk score stratification performed significantly better than the AJCC staging alone (P < 0.05). DCA showed that the risk score stratification was clinically useful and had better discriminative ability than the AJCC staging. Finally, an interactive native web-based prediction tool was constructed for the survival prediction of patients with postoperative gastric cancer. Conclusion In this study, a high-performance prediction model for the postoperative prognosis of gastric cancer was developed using DeepSurv, which offers essential benefits for risk stratification and prognosis prediction for each patient.
Collapse
Affiliation(s)
| | | | | | | | - Yongbin Zheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
13
|
Hartrampf PE, Hüttmann T, Seitz AK, Kübler H, Serfling SE, Higuchi T, Schlötelburg W, Michalski K, Gafita A, Rowe SP, Pomper MG, Buck AK, Werner RA. Prognostic Performance of RECIP 1.0 Based on [ 18F]PSMA-1007 PET in Prostate Cancer Patients Treated with [ 177Lu]Lu-PSMA I&T. J Nucl Med 2024; 65:560-565. [PMID: 38453363 DOI: 10.2967/jnumed.123.266702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/05/2024] [Indexed: 03/09/2024] Open
Abstract
In metastatic castration-resistant prostate cancer (mCRPC) patients treated with prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT), the recently proposed criteria for evaluating response to PSMA PET (RECIP 1.0) based on 68Ga- and 18F-labeled PET agents provided prognostic information in addition to changes in prostate-specific antigen (PSA) levels. Our aim was to evaluate the prognostic performance of this framework for overall survival (OS) in patients undergoing RLT and imaged with [18F]PSMA-1007 PET/CT and compare the prognostic performance with the PSA-based response assessment. Methods: In total, 73 patients with mCRPC who were scanned with [18F]PSMA-1007 PET/CT before and after 2 cycles of RLT were retrospectively analyzed. We calculated the changes in serum PSA levels (ΔPSA) and quantitative PET parameters for the whole-body tumor burden (SUVmean, SUVmax, PSMA tumor volume, and total lesion PSMA). Men were also classified following the Prostate Cancer Working Group 3 (PCWG3) criteria for ΔPSA and RECIP 1.0 for PET imaging response. We performed univariable Cox regression analysis, followed by multivariable and Kaplan-Meier analyses. Results: Median OS was 15 mo with a median follow-up time of 14 mo. Univariable Cox regression analysis provided significant associations with OS for ΔPSA (per percentage, hazard ratio [HR], 1.004; 95% CI, 1.002-1.007; P < 0.001) and PSMA tumor volume (per unit, HR, 1.003; 95% CI, 1.000-1.005; P = 0.03). Multivariable Cox regression analysis confirmed ΔPSA (per percentage, HR, 1.004; 95% CI, 1.001-1.006; P = 0.006) as an independent prognosticator for OS. Kaplan-Meier analyses provided significant segregation between individuals with versus those without any PSA response (19 mo vs. 14 mo; HR, 2.00; 95% CI, 0.95-4.18; P = 0.04). Differentiation between patients with or without progressive disease (PD) was also feasible when applying PSA-based PCWG3 (19 mo vs. 9 mo for non-PD and PD, respectively; HR, 2.29; 95% CI, 1.03-5.09; P = 0.01) but slightly failed when applying RECIP 1.0 (P = 0.08). A combination of both response systems (PCWG3 and RECIP 1.0), however, yielded the best discrimination between individuals without versus those with PD (19 mo vs. 8 mo; HR, 2.78; 95% CI, 1.32-5.86; P = 0.002). Conclusion: In patients with mCRPC treated with RLT and imaged with [18F]PSMA-1007, frameworks integrating both the biochemical (PCWG3) and PET-based response (RECIP 1.0) may best assist in identifying subjects prone to disease progression.
Collapse
Affiliation(s)
- Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany;
| | - Thomas Hüttmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Anna Katharina Seitz
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Würzburg, Germany
| | - Hubert Kübler
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Würzburg, Germany
| | | | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Wiebke Schlötelburg
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Kerstin Michalski
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andrei Gafita
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Steven P Rowe
- Division of Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, North Carolina; and
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, Maryland
- Division of Nuclear Medicine, Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
14
|
Zeremski V, Adolph L, Beer S, Berisha M, Jacobs B, Kahl C, Koenecke C, Kropf S, Panse J, Petersen J, Schmidt-Hieber M, Schneider J, Vucinic V, Walter J, Weigert O, Witte HM, Mougiakakos D. Relevance of different prognostic scores in primary CNS lymphoma in the era of intensified treatment regimens: A retrospective, multicenter analysis of 174 patients. Eur J Haematol 2024; 112:641-649. [PMID: 38164819 DOI: 10.1111/ejh.14159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVES Treatment intensification (including consolidative high-dose chemotherapy with autologous stem cell transplantation [HDT-ASCT]) significantly improved outcome in primary central nervous system lymphoma (PCNSL) patients. METHODS We conducted a multicenter, retrospective analysis of newly diagnosed PCNSL patients, treated with intensified treatment regimens. The following scores were evaluated in terms of overall survival (OS) and progression-free survival (PFS): Memorial Sloan-Kettering Cancer Center (MSKCC), International Extranodal Lymphoma Study Group (IELSG), and three-factor (3F) prognostic score. Further, all scores were comparatively investigated for model quality and concordance. RESULTS Altogether, 174 PCNSL patients were included. One hundred and five patients (60.3%) underwent HDT-ASCT. Two-year OS and 2-year PFS for the entire population were 73.3% and 48.5%, respectively. The MSKCC (p = .003) and 3F score (p < .001), but not the IELSG score (p = .06), had the discriminatory power to identify different risk groups for OS. In regard to concordance, the 3F score (C-index [0.71]) outperformed both the MSKCC (C-index [0.64]) and IELSG (C-index [0.53]) score. Moreover, the superiority of the 3F score was shown for PFS, successfully stratifying patients in three risk groups, which also resulted in the highest C-index (0.66). CONCLUSION The comparative analysis of established PCNSL risk scores affirm the clinical utility of the 3F score stratifying the widest prognostic spectrum among PCNSL patients treated with intensified treatment approaches.
Collapse
Affiliation(s)
- Vanja Zeremski
- Department of Hematology and Oncology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Louisa Adolph
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Sina Beer
- Department of Hematology and Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Mirjeta Berisha
- Department of Hematology and Oncology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Department of Internal Medicine 5, Hematology and Clinical Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Benedikt Jacobs
- Department of Internal Medicine 5, Hematology and Clinical Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Christoph Kahl
- Department of Hematology, Oncology and Palliative Care, Klinikum Magdeburg, Magdeburg, Germany
- Department of Hematology, Oncology, and Palliative Care, University Medical Center, University of Rostock, Rostock, Germany
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Siegfried Kropf
- Department of Biometry and Medical Informatics, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Jens Panse
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Aachen, Germany
| | - Judith Petersen
- Department of Hematology, Cell Therapy, Hemostaseology and Infectious Diseases, Leipzig University Medical Center, Leipzig, Germany
| | - Martin Schmidt-Hieber
- Clinic of Hematology, Oncology, Pneumology and Nephrology, Carl-Thiem-Hospital Cottbus, Cottbus, Germany
| | - Jessica Schneider
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Vladan Vucinic
- Department of Hematology, Cell Therapy, Hemostaseology and Infectious Diseases, Leipzig University Medical Center, Leipzig, Germany
| | - Jeanette Walter
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Aachen, Germany
| | - Oliver Weigert
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Hanno M Witte
- Department of Hematology and Oncology, Federal Armed Hospital Ulm, Ulm, Germany
| | - Dimitrios Mougiakakos
- Department of Hematology and Oncology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Department of Internal Medicine 5, Hematology and Clinical Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
15
|
Carrillo-Perez F, Pizurica M, Zheng Y, Nandi TN, Madduri R, Shen J, Gevaert O. Generation of synthetic whole-slide image tiles of tumours from RNA-sequencing data via cascaded diffusion models. Nat Biomed Eng 2024:10.1038/s41551-024-01193-8. [PMID: 38514775 DOI: 10.1038/s41551-024-01193-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/29/2024] [Indexed: 03/23/2024]
Abstract
Training machine-learning models with synthetically generated data can alleviate the problem of data scarcity when acquiring diverse and sufficiently large datasets is costly and challenging. Here we show that cascaded diffusion models can be used to synthesize realistic whole-slide image tiles from latent representations of RNA-sequencing data from human tumours. Alterations in gene expression affected the composition of cell types in the generated synthetic image tiles, which accurately preserved the distribution of cell types and maintained the cell fraction observed in bulk RNA-sequencing data, as we show for lung adenocarcinoma, kidney renal papillary cell carcinoma, cervical squamous cell carcinoma, colon adenocarcinoma and glioblastoma. Machine-learning models pretrained with the generated synthetic data performed better than models trained from scratch. Synthetic data may accelerate the development of machine-learning models in scarce-data settings and allow for the imputation of missing data modalities.
Collapse
Affiliation(s)
- Francisco Carrillo-Perez
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
- Internet technology and Data science Lab (IDLab), Ghent University, Ghent, Belgium
| | - Yuanning Zheng
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
| | - Tarak Nath Nandi
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - Ravi Madduri
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - Jeanne Shen
- Department of Pathology, Stanford University, School of Medicine, Palo Alto, CA, USA
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA.
- Department of Biomedical Data Science, Stanford University, School of Medicine, Stanford, CA, USA.
| |
Collapse
|
16
|
Cerono G, Melaiu O, Chicco D. Clinical Feature Ranking Based on Ensemble Machine Learning Reveals Top Survival Factors for Glioblastoma Multiforme. JOURNAL OF HEALTHCARE INFORMATICS RESEARCH 2024; 8:1-18. [PMID: 38273986 PMCID: PMC10805687 DOI: 10.1007/s41666-023-00138-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 01/27/2024]
Abstract
Glioblastoma multiforme (GM) is a malignant tumor of the central nervous system considered to be highly aggressive and often carrying a terrible survival prognosis. An accurate prognosis is therefore pivotal for deciding a good treatment plan for patients. In this context, computational intelligence applied to data of electronic health records (EHRs) of patients diagnosed with this disease can be useful to predict the patients' survival time. In this study, we evaluated different machine learning models to predict survival time in patients suffering from glioblastoma and further investigated which features were the most predictive for survival time. We applied our computational methods to three different independent open datasets of EHRs of patients with glioblastoma: the Shieh dataset of 84 patients, the Berendsen dataset of 647 patients, and the Lammer dataset of 60 patients. Our survival time prediction techniques obtained concordance index (C-index) = 0.583 in the Shieh dataset, C-index = 0.776 in the Berendsen dataset, and C-index = 0.64 in the Lammer dataset, as best results in each dataset. Since the original studies regarding the three datasets analyzed here did not provide insights about the most predictive clinical features for survival time, we investigated the feature importance among these datasets. To this end, we then utilized Random Survival Forests, which is a decision tree-based algorithm able to model non-linear interaction between different features and might be able to better capture the highly complex clinical and genetic status of these patients. Our discoveries can impact clinical practice, aiding clinicians and patients alike to decide which therapy plan is best suited for their unique clinical status.
Collapse
Affiliation(s)
- Gabriel Cerono
- Department of Neurology, University of California San Francisco, San Francisco, CA USA
| | | | - Davide Chicco
- Dipartimento di Informatica Sistemistica e Comunicazione, Università di Milano-Bicocca, Milan, Italy
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario Canada
| |
Collapse
|
17
|
Liu M, Lu J, Yu C, Zhao J, Wang L, Hu Y, Chen L, Han R, Liu Y, Sun M, Wei G, Wu S. Differentiation Potential of Hypodifferentiated Subsets of Nephrogenic Rests and Its Relationship to Prognosis in Wilms Tumor. Fetal Pediatr Pathol 2024; 43:123-139. [PMID: 38217324 DOI: 10.1080/15513815.2024.2303081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024]
Abstract
Background Wilms tumor (WT) is highly curable, although anaplastic histology or relapse imparts a worse prognosis. Nephrogenic rests (NR) associated with a high risk of developing WT are abnormally retained embryonic kidney precursor cells. Methods After pseudo-time analysis using single-cell RNA sequencing (scRNA-seq) data, we generated and validated a WT differentiation-related gene (WTDRG) signature to predict overall survival (OS) in children with a poor OS. Results A differentiation trajectory from NR to WT was identified and showed that hypodifferentiated subsets of NR could differentiate into WT. Classification of WT children with anaplastic histology or relapse based on the expression patterns of WTDRGs suggested that patients with relatively high levels of hypodifferentiated NR presented a poorer prognosis. A WTDRG-based risk model and a clinically applicable nomogram was developed. Conclusions These findings may inform oncogenesis of WT and interventions directed toward poor prognosis in WT children of anaplastic histology or relapse.
Collapse
Affiliation(s)
- Maolin Liu
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jiandong Lu
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chengjun Yu
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Zhao
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Hu
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Long Chen
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Rong Han
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Liu
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Miao Sun
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Guanghui Wei
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shengde Wu
- Department of Urology, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Swolinsky JS, Hinz RM, Markus CE, Singer E, Bachmann F, Halleck F, Kron S, Naik MG, Schmidt D, Obermeier M, Gebert P, Rauch G, Kropf S, Haase M, Budde K, Eckardt KU, Westhoff TH, Schmidt-Ott KM. Plasma NGAL levels in stable kidney transplant recipients and the risk of allograft loss. Nephrol Dial Transplant 2024; 39:483-495. [PMID: 37858309 PMCID: PMC11024820 DOI: 10.1093/ndt/gfad226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND The objective of this study was to investigate the utility of neutrophil gelatinase-associated lipocalin (NGAL) and calprotectin (CPT) to predict long-term graft survival in stable kidney transplant recipients (KTR). METHODS A total of 709 stable outpatient KTR were enrolled >2 months post-transplant. The utility of plasma and urinary NGAL (pNGAL, uNGAL) and plasma and urinary CPT at enrollment to predict death-censored graft loss was evaluated during a 58-month follow-up. RESULTS Among biomarkers, pNGAL showed the best predictive ability for graft loss and was the only biomarker with an area under the curve (AUC) > 0.7 for graft loss within 5 years. Patients with graft loss within 5 years (n = 49) had a median pNGAL of 304 [interquartile range (IQR) 235-358] versus 182 (IQR 128-246) ng/mL with surviving grafts (P < .001). Time-dependent receiver operating characteristic analyses at 58 months indicated an AUC for pNGAL of 0.795, serum creatinine-based Chronic Kidney Disease Epidemiology Collaboration estimated glomerular filtration rate (eGFR) had an AUC of 0.866. pNGAL added to a model based on conventional risk factors for graft loss with death as competing risk (age, transplant age, presence of donor-specific antibodies, presence of proteinuria, history of delayed graft function) had a strong independent association with graft loss {subdistribution hazard ratio (sHR) for binary log-transformed pNGAL [log2(pNGAL)] 3.4, 95% confidence interval (CI) 2.24-5.15, P < .0001}. This association was substantially attenuated when eGFR was added to the model [sHR for log2(pNGAL) 1.63, 95% CI 0.92-2.88, P = .095]. Category-free net reclassification improvement of a risk model including log2(pNGAL) in addition to conventional risk factors and eGFR was 54.3% (95% CI 9.2%-99.3%) but C-statistic did not improve significantly. CONCLUSIONS pNGAL was an independent predictor of renal allograft loss in stable KTR from one transplant center but did not show consistent added value when compared with baseline predictors including the conventional marker eGFR. Future studies in larger cohorts are warranted.
Collapse
Affiliation(s)
- Jutta S Swolinsky
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ricarda M Hinz
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Carolin E Markus
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Eugenia Singer
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Susanne Kron
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Marcel G Naik
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin
| | - Danilo Schmidt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | | | - Pimrapat Gebert
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology
| | - Geraldine Rauch
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology
| | - Siegfried Kropf
- Institute of Biometry and Medical Informatics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Michael Haase
- Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
- Diaverum Renal Services, MVZ Potsdam, Potsdam, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Timm H Westhoff
- Medical Department I, Marien Hospital Herne, Universitätsklinikum der Ruhr-Universität Bochum, Bochum, Germany
| | - Kai M Schmidt-Ott
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
19
|
Mendiburu‐Eliçabe M, García‐Sancha N, Corchado‐Cobos R, Martínez‐López A, Chang H, Hua Mao J, Blanco‐Gómez A, García‐Casas A, Castellanos‐Martín A, Salvador N, Jiménez‐Navas A, Pérez‐Baena MJ, Sánchez‐Martín MA, Abad‐Hernández MDM, Carmen SD, Claros‐Ampuero J, Cruz‐Hernández JJ, Rodríguez‐Sánchez CA, García‐Cenador MB, García‐Criado FJ, Vicente RS, Castillo‐Lluva S, Pérez‐Losada J. NCAPH drives breast cancer progression and identifies a gene signature that predicts luminal a tumour recurrence. Clin Transl Med 2024; 14:e1554. [PMID: 38344872 PMCID: PMC10859882 DOI: 10.1002/ctm2.1554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/01/2024] [Accepted: 01/09/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Luminal A tumours generally have a favourable prognosis but possess the highest 10-year recurrence risk among breast cancers. Additionally, a quarter of the recurrence cases occur within 5 years post-diagnosis. Identifying such patients is crucial as long-term relapsers could benefit from extended hormone therapy, while early relapsers might require more aggressive treatment. METHODS We conducted a study to explore non-structural chromosome maintenance condensin I complex subunit H's (NCAPH) role in luminal A breast cancer pathogenesis, both in vitro and in vivo, aiming to identify an intratumoural gene expression signature, with a focus on elevated NCAPH levels, as a potential marker for unfavourable progression. Our analysis included transgenic mouse models overexpressing NCAPH and a genetically diverse mouse cohort generated by backcrossing. A least absolute shrinkage and selection operator (LASSO) multivariate regression analysis was performed on transcripts associated with elevated intratumoural NCAPH levels. RESULTS We found that NCAPH contributes to adverse luminal A breast cancer progression. The intratumoural gene expression signature associated with elevated NCAPH levels emerged as a potential risk identifier. Transgenic mice overexpressing NCAPH developed breast tumours with extended latency, and in Mouse Mammary Tumor Virus (MMTV)-NCAPHErbB2 double-transgenic mice, luminal tumours showed increased aggressiveness. High intratumoural Ncaph levels correlated with worse breast cancer outcome and subpar chemotherapy response. A 10-gene risk score, termed Gene Signature for Luminal A 10 (GSLA10), was derived from the LASSO analysis, correlating with adverse luminal A breast cancer progression. CONCLUSIONS The GSLA10 signature outperformed the Oncotype DX signature in discerning tumours with unfavourable outcomes, previously categorised as luminal A by Prediction Analysis of Microarray 50 (PAM50) across three independent human cohorts. This new signature holds promise for identifying luminal A tumour patients with adverse prognosis, aiding in the development of personalised treatment strategies to significantly improve patient outcomes.
Collapse
|
20
|
Eidex Z, Ding Y, Wang J, Abouei E, Qiu RLJ, Liu T, Wang T, Yang X. Deep learning in MRI-guided radiation therapy: A systematic review. J Appl Clin Med Phys 2024; 25:e14155. [PMID: 37712893 PMCID: PMC10860468 DOI: 10.1002/acm2.14155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023] Open
Abstract
Recent advances in MRI-guided radiation therapy (MRgRT) and deep learning techniques encourage fully adaptive radiation therapy (ART), real-time MRI monitoring, and the MRI-only treatment planning workflow. Given the rapid growth and emergence of new state-of-the-art methods in these fields, we systematically review 197 studies written on or before December 31, 2022, and categorize the studies into the areas of image segmentation, image synthesis, radiomics, and real time MRI. Building from the underlying deep learning methods, we discuss their clinical importance and current challenges in facilitating small tumor segmentation, accurate x-ray attenuation information from MRI, tumor characterization and prognosis, and tumor motion tracking. In particular, we highlight the recent trends in deep learning such as the emergence of multi-modal, visual transformer, and diffusion models.
Collapse
Affiliation(s)
- Zach Eidex
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
- School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Yifu Ding
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Jing Wang
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Elham Abouei
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Richard L. J. Qiu
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
| | - Tian Liu
- Department of Radiation OncologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Tonghe Wang
- Department of Medical PhysicsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Xiaofeng Yang
- Department of Radiation Oncology and Winship Cancer InstituteEmory UniversityAtlantaGeorgiaUSA
- School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| |
Collapse
|
21
|
Mahootiha M, Qadir HA, Aghayan D, Fretland ÅA, von Gohren Edwin B, Balasingham I. Deep learning-assisted survival prognosis in renal cancer: A CT scan-based personalized approach. Heliyon 2024; 10:e24374. [PMID: 38298725 PMCID: PMC10828686 DOI: 10.1016/j.heliyon.2024.e24374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/19/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
This paper presents a deep learning (DL) approach for predicting survival probabilities of renal cancer patients based solely on preoperative CT imaging. The proposed approach consists of two networks: a classifier- and a survival- network. The classifier attempts to extract features from 3D CT scans to predict the ISUP grade of Renal cell carcinoma (RCC) tumors, as defined by the International Society of Urological Pathology (ISUP). Our classifier is a 3D convolutional neural network to avoid losing crucial information on the interconnection of slides in 3D images. We employ multiple procedures, including image augmentation, preprocessing, and concatenation, to improve the performance of the classifier. Given the strong correlation between ISUP grading and renal cancer prognosis in the clinical context, we use the ISUP grading features extracted by the classifier as the input to the survival network. By leveraging this clinical association and the classifier network, we are able to model our survival analysis using a simple DL-based network. We adopt a discrete LogisticHazard-based loss to extract intrinsic survival characteristics of RCC tumors from CT images. This allows us to build a completely parametric survival model that varies with patients' tumor characteristics and predicts non-proportional survival probability curves for different patients. Our results demonstrated that the proposed method could predict the future course of renal cancer with reasonable accuracy from the CT scans. The proposed method obtained an average concordance index of 0.72, an integrated Brier score of 0.15, and an area under the curve value of 0.71 on the test cohorts.
Collapse
Affiliation(s)
- Maryamalsadat Mahootiha
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
- Faculty of Medicine, University of Oslo, Oslo, 0372, Norway
| | - Hemin Ali Qadir
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
| | - Davit Aghayan
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
| | | | - Bjørn von Gohren Edwin
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
- Faculty of Medicine, University of Oslo, Oslo, 0372, Norway
| | - Ilangko Balasingham
- The Intervention Centre, Oslo University Hospital, Oslo, 0372, Norway
- Department of Electronic Systems, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
22
|
Babaei Rikan S, Sorayaie Azar A, Naemi A, Bagherzadeh Mohasefi J, Pirnejad H, Wiil UK. Survival prediction of glioblastoma patients using modern deep learning and machine learning techniques. Sci Rep 2024; 14:2371. [PMID: 38287149 PMCID: PMC10824760 DOI: 10.1038/s41598-024-53006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 01/25/2024] [Indexed: 01/31/2024] Open
Abstract
In this study, we utilized data from the Surveillance, Epidemiology, and End Results (SEER) database to predict the glioblastoma patients' survival outcomes. To assess dataset skewness and detect feature importance, we applied Pearson's second coefficient test of skewness and the Ordinary Least Squares method, respectively. Using two sampling strategies, holdout and five-fold cross-validation, we developed five machine learning (ML) models alongside a feed-forward deep neural network (DNN) for the multiclass classification and regression prediction of glioblastoma patient survival. After balancing the classification and regression datasets, we obtained 46,340 and 28,573 samples, respectively. Shapley additive explanations (SHAP) were then used to explain the decision-making process of the best model. In both classification and regression tasks, as well as across holdout and cross-validation sampling strategies, the DNN consistently outperformed the ML models. Notably, the accuracy were 90.25% and 90.22% for holdout and five-fold cross-validation, respectively, while the corresponding R2 values were 0.6565 and 0.6622. SHAP analysis revealed the importance of age at diagnosis as the most influential feature in the DNN's survival predictions. These findings suggest that the DNN holds promise as a practical auxiliary tool for clinicians, aiding them in optimal decision-making concerning the treatment and care trajectories for glioblastoma patients.
Collapse
Affiliation(s)
| | | | - Amin Naemi
- SDU Health Informatics and Technology, The Maersk Mc-Kinney Moller Institute, University of Southern Denmark, Odense, Denmark
| | | | - Habibollah Pirnejad
- Erasmus School of Health Policy and Management (ESHPM), Erasmus University Rotterdam, Rotterdam, The Netherlands.
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Uffe Kock Wiil
- SDU Health Informatics and Technology, The Maersk Mc-Kinney Moller Institute, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
23
|
Zhao K, Liu L, Zhou X, Wang G, Zhang J, Gao X, Yang L, Rao K, Guo C, Zhang Y, Huang C, Liu H, Li S, Chen Y. Re-exploration of prognosis in type B thymomas: establishment of a predictive nomogram model. World J Surg Oncol 2024; 22:26. [PMID: 38263144 PMCID: PMC10804589 DOI: 10.1186/s12957-023-03293-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
OBJECTIVE To explore the risk factors for disease progression after initial treatment of type B thymomas using a predictive nomogram model. METHODS A single-center retrospective study of patients with type B thymoma was performed. The Cox proportional hazard model was used for univariate and multivariate analyses. Variables with statistical and clinical significance in the multivariate Cox regression were integrated into a nomogram to establish a predictive model for disease progression. RESULTS A total of 353 cases with type B thymoma were retrieved between January 2012 and December 2021. The median follow-up was 58 months (range: 1-128 months). The 10-year progression-free survival (PFS) was 91.8%. The final nomogram model included R0 resection status and Masaoka stage, with a concordance index of 0.880. Non-R0 resection and advanced Masaoka stage were negative prognostic factors for disease progression (p < 0.001). No benefits of postoperative radiotherapy (PORT) were observed in patients with advanced stage and non-R0 resection (p = 0.114 and 0.284, respectively). CONCLUSION The best treatment strategy for type B thymoma is the detection and achievement of R0 resection as early as possible. Long-term follow-up is necessary, especially for patients with advanced Masaoka stage and who have not achieved R0 resection. No prognostic benefits were observed for PORT.
Collapse
Affiliation(s)
- Ke Zhao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Lei Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Xiaoyun Zhou
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Guige Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Jiaqi Zhang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Xuehan Gao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Libing Yang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Ke Rao
- Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Ye Zhang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Cheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Hongsheng Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Yeye Chen
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
24
|
Zhong L, Yang F, Sun S, Wang L, Yu H, Nie X, Liu A, Xu N, Zhang L, Zhang M, Qi Y, Ji H, Liu G, Zhao H, Jiang Y, Li J, Song C, Yu X, Yang L, Yu J, Feng H, Guo X, Yang F, Xue F. Predicting lung cancer survival prognosis based on the conditional survival bayesian network. BMC Med Res Methodol 2024; 24:16. [PMID: 38254038 PMCID: PMC10801949 DOI: 10.1186/s12874-023-02043-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/25/2023] [Indexed: 01/24/2024] Open
Abstract
Lung cancer is a leading cause of cancer deaths and imposes an enormous economic burden on patients. It is important to develop an accurate risk assessment model to determine the appropriate treatment for patients after an initial lung cancer diagnosis. The Cox proportional hazards model is mainly employed in survival analysis. However, real-world medical data are usually incomplete, posing a great challenge to the application of this model. Commonly used imputation methods cannot achieve sufficient accuracy when data are missing, so we investigated novel methods for the development of clinical prediction models. In this article, we present a novel model for survival prediction in missing scenarios. We collected data from 5,240 patients diagnosed with lung cancer at the Weihai Municipal Hospital, China. Then, we applied a joint model that combined a BN and a Cox model to predict mortality risk in individual patients with lung cancer. The established prognostic model achieved good predictive performance in discrimination and calibration. We showed that combining the BN with the Cox proportional hazards model is highly beneficial and provides a more efficient tool for risk prediction.
Collapse
Affiliation(s)
- Lu Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Hainan Center for Disease Control and Prevention, Institute for Prevention and Control of Tropical Diseases and Chronic Noninfectious Diseases, Haikou, Hainan, China.
| | - Fan Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Institute for Medical Dataology, Shandong University, Jinan, China.
| | - Shanshan Sun
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Lijie Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Shandong University, Jinan, China
| | - Hong Yu
- Chongqing Key Laboratory of Computational Intelligence, Chongqing University of Posts and Telecommunications, Chongqing, China
| | - Xiushan Nie
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, China
| | - Ailing Liu
- Department of Pulmonary and Critical Care Medicine, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Ning Xu
- Department of Pulmonary and Critical Care Medicine, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Lanfang Zhang
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Mingjuan Zhang
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Yue Qi
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Huaijun Ji
- Department of Thoracic Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Guiyuan Liu
- Department of Radiology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Huan Zhao
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, China
| | - Yinan Jiang
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Jingyi Li
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Chengcun Song
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Xin Yu
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Liu Yang
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Jinchao Yu
- Department of Radiology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Hu Feng
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Xiaolei Guo
- The Department for Chronic and Non-communicable Disease Control and Prevention, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Fujun Yang
- Department of Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China.
| | - Fuzhong Xue
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Institute for Medical Dataology, Shandong University, Jinan, China.
| |
Collapse
|
25
|
Moore JL, Santaolalla A, Van Hemelrijck M, North B, Davies AR. Reply to R. Sun et al. J Clin Oncol 2024; 42:367-368. [PMID: 37988643 DOI: 10.1200/jco.23.02131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 11/23/2023] Open
Affiliation(s)
- Jonathan L Moore
- Jonathan L. Moore, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; Aida Santaolalla, PhD, Mieke Van Hemelrijck, PhD, and Bernard North, PhD, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; and Andrew R. Davies, MD, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Aida Santaolalla
- Jonathan L. Moore, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; Aida Santaolalla, PhD, Mieke Van Hemelrijck, PhD, and Bernard North, PhD, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; and Andrew R. Davies, MD, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Mieke Van Hemelrijck
- Jonathan L. Moore, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; Aida Santaolalla, PhD, Mieke Van Hemelrijck, PhD, and Bernard North, PhD, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; and Andrew R. Davies, MD, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Bernard North
- Jonathan L. Moore, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; Aida Santaolalla, PhD, Mieke Van Hemelrijck, PhD, and Bernard North, PhD, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; and Andrew R. Davies, MD, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Andrew R Davies
- Jonathan L. Moore, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; Aida Santaolalla, PhD, Mieke Van Hemelrijck, PhD, and Bernard North, PhD, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; and Andrew R. Davies, MD, FRCS, Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
26
|
Escribano-Serrat S, Rodríguez-Lobato LG, Charry P, Martínez-Cibrian N, Suárez-Lledó M, Rivero A, Moreno-Castaño AB, Solano MT, Arcarons J, Nomdedeu M, Cid J, Lozano M, Pedraza A, Rosiñol L, Esteve J, Urbano-Ispizua Á, Palomo M, Fernández-Avilés F, Martínez C, Díaz-Ricart M, Carreras E, Rovira M, Salas MQ. Endothelial Activation and Stress Index in adults undergoing allogeneic hematopoietic cell transplantation with post-transplant cyclophosphamide-based prophylaxis. Cytotherapy 2024; 26:73-80. [PMID: 37952139 DOI: 10.1016/j.jcyt.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/09/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND AIMS Post-transplant cyclophosphamide (PTCY)-based prophylaxis is becoming widespread for allogeneic hematopoietic cell transplantation (allo-HCT) performed independently of the selected donor source. In parallel, use of the Endothelial Activation and Stress Index (EASIX)-considered a surrogate parameter of endothelial activation-for predicting patient outcomes and clinical complications is gaining popularity in the allo-HCT setting. METHODS We first investigated whether the dynamics of EASIX after allo-HCT differ between patients receiving PTCY and patients receiving other prophylaxis. We then investigated whether the predictive capacity of EASIX persists in PTCY-based allo-HCT. A total of 328 patients transplanted between 2014 and 2020 were included, and 201 (61.2%) received PTCY. RESULTS EASIX trends differed significantly between the groups. Compared with patients receiving other prophylaxis, patients receiving PTCY had lower EASIX on day 0 and higher values between day 7 and day 100. In patients receiving PTCY, higher EASIX correlated significantly with higher non-relapse mortality (NRM) and lower overall survival (OS) when measured before and during the first 180 days after allo-HCT. In addition, higher EASIX scores measured at specific time points were predictors of veno-occlusive disease (VOD), transplant-associated thrombotic microangiopathy (TA-TMA) and grade 2-4 acute graft-versus-host disease (aGVHD) risk. CONCLUSIONS This study demonstrates how EASIX trends vary during the first 180 days after allo-HCT in patients receiving PTCY and those not receiving PTCY and validates the utility of this index for predicting NRM, OS and risk of VOD, TA-TMA and grade 2-4 aGVHD in patients receiving PTCY.
Collapse
Affiliation(s)
- Silvia Escribano-Serrat
- Department of Hematology and Hemotherapy, Hospital Clínico San Carlos, IdiSSC, Madrid, Spain; Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Luis Gerardo Rodríguez-Lobato
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Paola Charry
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Nuria Martínez-Cibrian
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - María Suárez-Lledó
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Andrea Rivero
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Ana Belén Moreno-Castaño
- Hematopathology, Pathology Department, CDB, Hospital Clínic Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - María Teresa Solano
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Jordi Arcarons
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Meritxell Nomdedeu
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Joan Cid
- Apheresis and Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Miquel Lozano
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Alexandra Pedraza
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Laura Rosiñol
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Jordi Esteve
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Álvaro Urbano-Ispizua
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Marta Palomo
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Francesc Fernández-Avilés
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Carmen Martínez
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Maribel Díaz-Ricart
- Hematopathology, Pathology Department, CDB, Hospital Clínic Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Enric Carreras
- Fundació i Institut de Recerca Josep Carreras Contra la Leucèmia (Campus Clínic), Barcelona, Spain
| | - Montserrat Rovira
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - María Queralt Salas
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain.
| |
Collapse
|
27
|
Burnazovic E, Yee A, Levy J, Gore G, Abbasgholizadeh Rahimi S. Application of Artificial intelligence in COVID-19-related geriatric care: A scoping review. Arch Gerontol Geriatr 2024; 116:105129. [PMID: 37542917 DOI: 10.1016/j.archger.2023.105129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND Older adults have been disproportionately affected by the COVID-19 pandemic. This scoping review aimed to summarize the current evidence of artificial intelligence (AI) use in the screening/monitoring, diagnosis, and/or treatment of COVID-19 among older adults. METHOD The review followed the Joanna Briggs Institute and Arksey and O'Malley frameworks. An information specialist performed a comprehensive search from the date of inception until May 2021, in six bibliographic databases. The selected studies considered all populations, and all AI interventions that had been used in COVID-19-related geriatric care. We focused on patient, healthcare provider, and healthcare system-related outcomes. The studies were restricted to peer-reviewed English publications. Two authors independently screened the titles and abstracts of the identified records, read the selected full texts, and extracted data from the included studies using a validated data extraction form. Disagreements were resolved by consensus, and if this was not possible, the opinion of a third reviewer was sought. RESULTS Six databases were searched , yielding 3,228 articles, of which 10 were included. The majority of articles used a single AI model to assess the association between patients' comorbidities and COVID-19 outcomes. Articles were mainly conducted in high-income countries, with limited representation of females in study participants, and insufficient reporting of participants' race and ethnicity. DISCUSSION This review highlighted how the COVID-19 pandemic has accelerated the application of AI to protect older populations, with most interventions in the pilot testing stage. Further work is required to measure effectiveness of these technologies in a larger scale, use more representative datasets for training of AI models, and expand AI applications to low-income countries.
Collapse
Affiliation(s)
- Emina Burnazovic
- Integrated Biomedical Engineering and Health Sciences, Department of Computing and Software, Faculty of Engineering, McMaster University, Hamilton, ON, Canada
| | - Amanda Yee
- Department of Family Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Joshua Levy
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Genevieve Gore
- Schulich Library of Physical Sciences, Life Sciences and Engineering, McGill University, Montreal, QC, Canada
| | - Samira Abbasgholizadeh Rahimi
- Department of Family Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Mila-Quebec Artificial Intelligence Institute, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
28
|
Bayraktar E, Graf T, Ayuk FA, Beutel G, Penack O, Luft T, Brueder N, Castellani G, Reinhardt HC, Kröger N, Beelen DW, Turki AT. Data-driven grading of acute graft-versus-host disease. Nat Commun 2023; 14:7799. [PMID: 38017035 PMCID: PMC10684603 DOI: 10.1038/s41467-023-43372-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 11/08/2023] [Indexed: 11/30/2023] Open
Abstract
Despite advances in allogeneic hematopoietic cell transplantation, acute graft-versus-host disease (aGVHD) remains its leading complication, yet with heterogeneous outcomes. Here, we analyzed aGVHD phenotypes and clinical classifications in depth in large, multicenter cohorts involving 3019 patients and addressed prevailing gaps by developing data-driven models. We compared, tested and verified these along with all conventional classifications in independent cohorts and found that data-driven grading outperformed conventional grading in Akaike information criterion and concordance index metrics. Data-driven classifications refined aGVHD assessment with up to 12 severity grades, which were associated with distinct nonrelapse mortality (NRM) and confirmed the key role of intestinal aGVHD. We developed an online calculator for physicians to implement principal component-derived grading (PC1). These results provide substantial insight into the evaluation of aGVHD phenotypes and multiorgan involvement, which relegates the exclusive reporting of overall aGVHD severity grades in transplant registries and clinical trials. Data-driven aGVHD grading provides an expandable platform to refine classification and transplant risk assessment.
Collapse
Affiliation(s)
- Evren Bayraktar
- Computational Hematology Lab, West-German Cancer Center, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
- Chair III of Applied Mathematics, TU Dortmund University of Applied Sciences, Vogelpothsweg 87, 44227, Dortmund, Germany
| | - Theresa Graf
- Computational Hematology Lab, West-German Cancer Center, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Francis A Ayuk
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20251, Hamburg, Germany
| | - Gernot Beutel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Olaf Penack
- Department of Hematology, Oncology and Tumorimmunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Nicole Brueder
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Gastone Castellani
- Department of Medical and Surgical Sciences- DIMEC, Applied Physics and Biophysics group, University of Bologna, Via Zamboni 33, 40126, Bologna, Italy
| | - H Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
- German Cancer Consortium (DKTK), Partner sites Essen/Düsseldorf, Hufelandstr. 55, 45122, Essen, Germany
- Cancer Research Center Cologne Essen (CCCE), Partner site Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Nicolaus Kröger
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße 52, 20251, Hamburg, Germany
| | - Dietrich W Beelen
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany
- German Cancer Consortium (DKTK), Partner sites Essen/Düsseldorf, Hufelandstr. 55, 45122, Essen, Germany
| | - Amin T Turki
- Computational Hematology Lab, West-German Cancer Center, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany.
- Department of Hematology and Stem Cell Transplantation, West-German Cancer Center, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany.
- German Cancer Consortium (DKTK), Partner sites Essen/Düsseldorf, Hufelandstr. 55, 45122, Essen, Germany.
- Cancer Research Center Cologne Essen (CCCE), Partner site Essen, Hufelandstr. 55, 45122, Essen, Germany.
- Department of Hematology and Oncology, Marienhospital University Hospital, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
- Institute for Experimental Cellular Therapy, University Hospital Essen, Hufelandstr. 55, 45122, Essen, Germany.
| |
Collapse
|
29
|
Hua Y, Cai D, Shirley CA, Mo S, Chen R, Gao F, Chen F. A prognostic model for ovarian neoplasms established by an integrated analysis of 1580 transcriptomic profiles. Sci Rep 2023; 13:19429. [PMID: 37940688 PMCID: PMC10632395 DOI: 10.1038/s41598-023-45410-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023] Open
Abstract
Even after debulking surgery combined with chemotherapy or new adjuvant chemotherapy paired with internal surgery, the average year of disease free survival in advanced ovarian cancer was approximately 1.7 years1. The development of a molecular predictor of early recurrence would allow for the identification of ovarian cancer (OC) patients with high risk of relapse. The Ovarian Cancer Disease Free Survival Predictor (ODFSP), a predictive model constructed from a special set of 1580 OC tumors in which gene expression was assessed using both microarray and sequencing platforms, was created by our team. To construct gene expression barcodes that were resistant to biases caused by disparate profiling platforms and batch effects, we employed a meta-analysis methodology that was based on the binary gene pair technique. We demonstrate that ODFSP is a reliable single-sample predictor of early recurrence (1 year or less) using the largest pool of OC transcriptome data sets available to date. The ODFSP model showed significantly high prognostic value for binary recurrence prediction unaffected by clinicopathologic factors, with a meta-estimate of the area under the receiver operating curve of 0.64 (P = 4.6E-05) and a D-index (robust hazard ratio) of 1.67 (P = 9.2E-06), respectively. GO analysis of ODFSP's 2040 gene pairs (collapsed to 886 distinct genes) revealed the involvement in small molecular catabolic process, sulfur compound metabolic process, organic acid catabolic process, sulfur compound biosynthetic process, glycosaminoglycan metabolic process and aminometabolic process. Kyoto encyclopedia of genes and genomes pathway analysis of ODFSP's signature genes identified prominent pathways that included cAMP signaling pathway and FoxO signaling pathway. By identifying individuals who might benefit from a more aggressive treatment plan or enrolment in a clinical trial but who will not benefit from standard surgery or chemotherapy, ODFSP could help with treatment decisions.
Collapse
Affiliation(s)
- Yanjiao Hua
- The Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Du Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong Province, China
| | - Cole Andrea Shirley
- Sun Yat-Sen University, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Sien Mo
- The Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Ruyun Chen
- Sun Yat-Sen University, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Feng Gao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong Province, China
| | - Fangying Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| |
Collapse
|
30
|
Li W, Huang Q, Peng Y, Pan S, Hu M, Wang P, He Y. A deep learning approach based on multi-omics data integration to construct a risk stratification prediction model for skin cutaneous melanoma. J Cancer Res Clin Oncol 2023; 149:15923-15938. [PMID: 37673824 DOI: 10.1007/s00432-023-05358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/26/2023] [Indexed: 09/08/2023]
Abstract
PURPOSE Skin cutaneous melanoma (SKCM) is a highly aggressive melanocytic carcinoma whose high heterogeneity and complex etiology make its prognosis difficult to predict. This study aimed to construct a risk subtype typing model for SKCM. METHODS The study proposes a deep learning framework combining early fusion feature autoencoder (AE) and late fusion feature AE for risk subtype prediction of SKCM. The deep learning framework integrates mRNA, miRNA, and DNA methylation data of SKCM patients from The Cancer Genome Atlas (TCGA), and clusters the screened multi-omics features associated with survival prognosis to identify risk subtypes. Differential expression analysis and functional enrichment analysis were performed between risk subtypes, while SVM classifiers were constructed between differentially expressed genes (DEGs) obtained by Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression screening and risk subtype labels inferred from multi-omics data, and the predictive robustness of risk subtypes inferred from the risk subtype classification prediction model was validated using two independent datasets. RESULTS The deep learning framework that combined early fusion feature AE with late fusion feature AE distinguished the two best risk subtypes compared to the multi-omics integration approach with single strategy AE or PCA. A promising C-index (C-index = 0.748) and a significant difference in survival (log-rank P value = 4.61 × 10-9) were found between the identified risk subtypes. The DEGs with the top significance values together with differentially expressed miRNAs provided the biological interpretation of risk subtypes on SKCM. Finally, the framework was applied to predict risk subtypes in two independent test datasets of SKCM patients, all of which showed good predictive power (C-index > 0.680) and significant survival differences (log-rank P value < 0.01). CONCLUSION The SKCM risk subtypes identified by integrating multi-omics data based on deep learning can not only improve the understanding of the molecular mechanisms of SKCM, but also provide clinicians with assistance in treatment decisions.
Collapse
Affiliation(s)
- Weijia Li
- Department of Epidemiology and Medical Statistics, Institute of Medical Systems Biology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Qiao Huang
- Department of Epidemiology and Medical Statistics, Institute of Medical Systems Biology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yi Peng
- Department of Epidemiology and Medical Statistics, Institute of Medical Systems Biology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Suyue Pan
- Department of Epidemiology and Medical Statistics, Institute of Medical Systems Biology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Min Hu
- Department of Epidemiology and Medical Statistics, Institute of Medical Systems Biology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Pu Wang
- Department of Epidemiology and Medical Statistics, Institute of Medical Systems Biology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yuqing He
- Department of Epidemiology and Medical Statistics, Institute of Medical Systems Biology, Guangdong Medical University, Dongguan, Guangdong, China.
- Dongguan Liaobu Hospital, Dongguan, Guangdong, China.
| |
Collapse
|
31
|
Schmid M, Friede T, Klein N, Weinhold L. Accounting for time dependency in meta-analyses of concordance probability estimates. Res Synth Methods 2023; 14:807-823. [PMID: 37429580 DOI: 10.1002/jrsm.1655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/21/2023] [Accepted: 06/19/2023] [Indexed: 07/12/2023]
Abstract
Recent years have seen the development of many novel scoring tools for disease prognosis and prediction. To become accepted for use in clinical applications, these tools have to be validated on external data. In practice, validation is often hampered by logistical issues, resulting in multiple small-sized validation studies. It is therefore necessary to synthesize the results of these studies using techniques for meta-analysis. Here we consider strategies for meta analyzing the concordance probability for time-to-event data ("C-index"), which has become a popular tool to evaluate the discriminatory power of prediction models with a right-censored outcome. We show that standard meta-analysis of the C-index may lead to biased results, as the magnitude of the concordance probability depends on the length of the time interval used for evaluation (defined e.g., by the follow-up time, which might differ considerably between studies). To address this issue, we propose a set of methods for random-effects meta-regression that incorporate time directly as covariate in the model equation. In addition to analyzing nonlinear time trends via fractional polynomial, spline, and exponential decay models, we provide recommendations on suitable transformations of the C-index before meta-regression. Our results suggest that the C-index is best meta-analyzed using fractional polynomial meta-regression with logit-transformed C-index values. Classical random-effects meta-analysis (not considering time as covariate) is demonstrated to be a suitable alternative when follow-up times are small. Our findings have implications for the reporting of C-index values in future studies, which should include information on the length of the time interval underlying the calculations.
Collapse
Affiliation(s)
- Matthias Schmid
- Department of Medical Biometry, Informatics, and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Nadja Klein
- Research Center for Trustworthy Data Science and Security, UA Ruhr/Department of Statistics, Technische Universität Dortmund, Dortmund, Germany
| | - Leonie Weinhold
- Department of Medical Biometry, Informatics, and Epidemiology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
32
|
Bose G, Healy BC, Saxena S, Saleh F, Glanz BI, Bakshi R, Weiner HL, Chitnis T. Increasing Neurofilament and Glial Fibrillary Acidic Protein After Treatment Discontinuation Predicts Multiple Sclerosis Disease Activity. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200167. [PMID: 37813595 PMCID: PMC10574823 DOI: 10.1212/nxi.0000000000200167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/17/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND AND OBJECTIVES Stable patients with multiple sclerosis (MS) may discontinue treatment, but the risk of disease activity is unknown. Serum neurofilament light chain (sNfL) and serum glial fibrillary acidic protein (sGFAP) are biomarkers of subclinical disease activity and may help risk stratification. In this study, sNfL and sGFAP levels in stable patients were evaluated before and after treatment discontinuation to determine association with disease activity. METHODS This observational study included patients enrolled in the Comprehensive Longitudinal Investigation in MS at the Brigham and Women's Hospital who discontinued treatment after >2 years disease activity-free. Two serum samples within 2 years, before and after treatment stop, were sent for sNfL and sGFAP measurements by single-molecule array. Biannual neurologic examinations and yearly MRI scans determined disease activity by 3 time-to-event outcomes: 6-month confirmed disability worsening (CDW), clinical attacks, and MRI activity (new T2 or contrast-enhancing lesions). Associations between each outcome and log-transformed sNfL and sGFAP levels pretreatment stop and posttreatment stop and the percent change were estimated using multivariable Cox regression analysis adjusting for age, disability, disease duration, and duration from attack before treatment stop. RESULTS Seventy-eight patients (92% female) discontinued treatment at a median (interquartile range) age of 48.5 years (39.0-55.7) and disease duration of 12.3 years (7.5-18.8) and were followed up for 6.3 years (4.2-8.5). CDW occurred in 27 patients (35%), new attacks in 19 (24%), and new MRI activity in 26 (33%). Higher posttreatment stop sNfL level was associated with CDW (adjusted hazard ratio (aHR) 2.80, 95% CI 1.36-5.76, p = 0.005) and new MRI activity (aHR 3.09, 95% CI 1.42-6.70, p = 0.004). Patients who had >100% increase in sNfL level from pretreatment stop to posttreatment stop had greater risk of CDW (HR 3.87, 95% CI 1.4-10.7, p = 0.009) and developing new MRI activity (HR 4.02, 95% CI 1.51-10.7, p = 0.005). Patients who had >50% increase in sGFAP level also had greater risk of CDW (HR 5.34, 95% CI 1.4-19.9, p = 0.012) and developing new MRI activity (HR 5.16, 95% CI 1.71-15.6, p = 0.004). DISCUSSION Stable patients who discontinue treatment may be risk stratified by sNfL and sGFAP levels measured before and after discontinuing treatment. Further studies are needed to validate findings and determine whether resuming treatment in patients with increasing biomarker levels reduces risk of subsequent disease activity.
Collapse
Affiliation(s)
- Gauruv Bose
- From the Department of Neurology (G.B., B.C.H., S.S., F.S., B.I.G., R.B., H.L.W., T.C.), Brigham and Women's Hospital, Boston, MA; Harvard Medical School (G.B., B.C.H., B.I.G., R.B., H.L.W., T.C.), Boston, MA; The University of Ottawa and Ottawa Hospital Research Institute (G.B.), Ottawa, Canada
| | - Brian C Healy
- From the Department of Neurology (G.B., B.C.H., S.S., F.S., B.I.G., R.B., H.L.W., T.C.), Brigham and Women's Hospital, Boston, MA; Harvard Medical School (G.B., B.C.H., B.I.G., R.B., H.L.W., T.C.), Boston, MA; The University of Ottawa and Ottawa Hospital Research Institute (G.B.), Ottawa, Canada
| | - Shrishti Saxena
- From the Department of Neurology (G.B., B.C.H., S.S., F.S., B.I.G., R.B., H.L.W., T.C.), Brigham and Women's Hospital, Boston, MA; Harvard Medical School (G.B., B.C.H., B.I.G., R.B., H.L.W., T.C.), Boston, MA; The University of Ottawa and Ottawa Hospital Research Institute (G.B.), Ottawa, Canada
| | - Fermisk Saleh
- From the Department of Neurology (G.B., B.C.H., S.S., F.S., B.I.G., R.B., H.L.W., T.C.), Brigham and Women's Hospital, Boston, MA; Harvard Medical School (G.B., B.C.H., B.I.G., R.B., H.L.W., T.C.), Boston, MA; The University of Ottawa and Ottawa Hospital Research Institute (G.B.), Ottawa, Canada
| | - Bonnie I Glanz
- From the Department of Neurology (G.B., B.C.H., S.S., F.S., B.I.G., R.B., H.L.W., T.C.), Brigham and Women's Hospital, Boston, MA; Harvard Medical School (G.B., B.C.H., B.I.G., R.B., H.L.W., T.C.), Boston, MA; The University of Ottawa and Ottawa Hospital Research Institute (G.B.), Ottawa, Canada
| | - Rohit Bakshi
- From the Department of Neurology (G.B., B.C.H., S.S., F.S., B.I.G., R.B., H.L.W., T.C.), Brigham and Women's Hospital, Boston, MA; Harvard Medical School (G.B., B.C.H., B.I.G., R.B., H.L.W., T.C.), Boston, MA; The University of Ottawa and Ottawa Hospital Research Institute (G.B.), Ottawa, Canada
| | - Howard L Weiner
- From the Department of Neurology (G.B., B.C.H., S.S., F.S., B.I.G., R.B., H.L.W., T.C.), Brigham and Women's Hospital, Boston, MA; Harvard Medical School (G.B., B.C.H., B.I.G., R.B., H.L.W., T.C.), Boston, MA; The University of Ottawa and Ottawa Hospital Research Institute (G.B.), Ottawa, Canada
| | - Tanuja Chitnis
- From the Department of Neurology (G.B., B.C.H., S.S., F.S., B.I.G., R.B., H.L.W., T.C.), Brigham and Women's Hospital, Boston, MA; Harvard Medical School (G.B., B.C.H., B.I.G., R.B., H.L.W., T.C.), Boston, MA; The University of Ottawa and Ottawa Hospital Research Institute (G.B.), Ottawa, Canada.
| |
Collapse
|
33
|
Li YJ, Chou HH, Lin PC, Shen MR, Hsieh SY. A novel deep learning-based algorithm combining histopathological features with tissue areas to predict colorectal cancer survival from whole-slide images. J Transl Med 2023; 21:731. [PMID: 37848862 PMCID: PMC10580604 DOI: 10.1186/s12967-023-04530-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 09/15/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Many methodologies for selecting histopathological images, such as sample image patches or segment histology from regions of interest (ROIs) or whole-slide images (WSIs), have been utilized to develop survival models. With gigapixel WSIs exhibiting diverse histological appearances, obtaining clinically prognostic and explainable features remains challenging. Therefore, we propose a novel deep learning-based algorithm combining tissue areas with histopathological features to predict cancer survival. METHODS The Cancer Genome Atlas Colon Adenocarcinoma (TCGA-COAD) dataset was used in this investigation. A deep convolutional survival model (DeepConvSurv) extracted histopathological information from the image patches of nine different tissue types, including tumors, lymphocytes, stroma, and mucus. The tissue map of the WSIs was segmented using image processing techniques that involved localizing and quantifying the tissue region. Six survival models with the concordance index (C-index) were used as the evaluation metrics. RESULTS We extracted 128 histopathological features from four histological types and five tissue area features from WSIs to predict colorectal cancer survival. Our method performed better in six distinct survival models than the Whole Slide Histopathological Images Survival Analysis framework (WSISA), which adaptively sampled patches using K-means from WSIs. The best performance using histopathological features was 0.679 using LASSO-Cox. Compared to histopathological features alone, tissue area features increased the C-index by 2.5%. Based on histopathological features and tissue area features, our approach achieved performance of 0.704 with RIDGE-Cox. CONCLUSIONS A deep learning-based algorithm combining histopathological features with tissue area proved clinically relevant and effective for predicting cancer survival.
Collapse
Affiliation(s)
- Yan-Jun Li
- Institute of Medical Informatics, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Hsin-Hung Chou
- Department of Computer Science and Information Engineering, National Chi Nan University, Nantou, 545301, Taiwan
| | - Peng-Chan Lin
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| | - Meng-Ru Shen
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Sun-Yuan Hsieh
- Institute of Medical Informatics, National Cheng Kung University, Tainan, 70101, Taiwan
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| |
Collapse
|
34
|
Salas MQ, Rodríguez-Lobato LG, Charry P, Suárez-Lledó M, Pedraza A, Solano MT, Arcarons J, Cid J, Lozano M, Rosiñol L, Esteve J, Carreras E, Fernández-Avilés F, Martínez C, Rovira M. Applicability and validation of different prognostic scores in allogeneic hematopoietic cell transplant (HCT) in the post-transplant cyclophosphamide era. Hematol Transfus Cell Ther 2023:S2531-1379(23)00162-1. [PMID: 37891074 DOI: 10.1016/j.htct.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/21/2023] [Indexed: 10/29/2023] Open
Abstract
We investigated the predictive capacity of six prognostic indices [Karnofsky Performance Status (KPS), Hematopoietic Cell Transplant-Specific Comorbidity Index (HCT-CI), Disease Risk Index (DRI), European Bone Marrow Transplantation (EBMT) and Revised Pre-Transplantation Assessment of Mortality (rPAM) Scores and Endothelial Activation and Stress Index (EASIX)] in 205 adults undergoing post-transplant cyclophosphamide (PTCy)-based allo-HCT. KPS, HCT-CI, DRI and EASIX grouped patients into higher and lower risk strata. KPS and EASIX maintained appropriate discrimination for OS prediction across the first 2 years after allo-HCT [receiver operating characteristic curve (area under the curve (AUC) > 55 %)]. The discriminative capacity of DRI and HCT-CI increased during the post-transplant period, with a peak of prediction at 2 years (AUC of 61.1 % and 61.8 %). The maximum rPAM discriminative capacity was at 1 year (1-year AUC of 58.2 %). The predictive capacity of the EBMT score was not demonstrated. This study validates the discrimination capacity for OS prediction of KPS, HCT-CI, DRI and EASIX in PTCy-based allo-HCT.
Collapse
Affiliation(s)
- María Queralt Salas
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain.
| | - Luis Gerardo Rodríguez-Lobato
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Paola Charry
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Maria Suárez-Lledó
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Alexandra Pedraza
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain
| | - María Teresa Solano
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Jordi Arcarons
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Joan Cid
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Josep Carreras Leukemia Research Institute (Clinic Campus), Barcela, Spain
| | - Miquel Lozano
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Josep Carreras Leukemia Research Institute (Clinic Campus), Barcela, Spain; University of Barcelona, Barcelona, Spain
| | - Laura Rosiñol
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Jordi Esteve
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Enric Carreras
- Josep Carreras Leukemia Research Institute (Clinic Campus), Barcela, Spain
| | - Francesc Fernández-Avilés
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Carmen Martínez
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Josep Carreras Leukemia Research Institute (Clinic Campus), Barcela, Spain; University of Barcelona, Barcelona, Spain
| | - Montserrat Rovira
- Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Josep Carreras Leukemia Research Institute (Clinic Campus), Barcela, Spain; University of Barcelona, Barcelona, Spain
| |
Collapse
|
35
|
Tschodu D, Lippoldt J, Gottheil P, Wegscheider AS, Käs JA, Niendorf A. Re-evaluation of publicly available gene-expression databases using machine-learning yields a maximum prognostic power in breast cancer. Sci Rep 2023; 13:16402. [PMID: 37798300 PMCID: PMC10556090 DOI: 10.1038/s41598-023-41090-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/22/2023] [Indexed: 10/07/2023] Open
Abstract
Gene expression signatures refer to patterns of gene activities and are used to classify different types of cancer, determine prognosis, and guide treatment decisions. Advancements in high-throughput technology and machine learning have led to improvements to predict a patient's prognosis for different cancer phenotypes. However, computational methods for analyzing signatures have not been used to evaluate their prognostic power. Contention remains on the utility of gene expression signatures for prognosis. The prevalent approaches include random signatures, expert knowledge, and machine learning to construct an improved signature. We unify these approaches to evaluate their prognostic power. Re-evaluation of publicly available gene-expression data from 8 databases with 9 machine-learning models revealed previously unreported results. Gene-expression signatures are confirmed to be useful in predicting a patient's prognosis. Convergent evidence from [Formula: see text] 10,000 signatures implicates a maximum prognostic power. By calculating the concordance index, which measures how well patients with different prognoses can be discriminated, we show that a signature can correctly discriminate patients' prognoses no more than 80% of the time. Additionally, we show that more than 50% of the potentially available information is still missing at this value. We surmise that an accurate prognosis must incorporate molecular, clinical, histological, and other complementary factors.
Collapse
Affiliation(s)
- Dimitrij Tschodu
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103, Leipzig, Germany.
| | - Jürgen Lippoldt
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103, Leipzig, Germany
| | - Pablo Gottheil
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103, Leipzig, Germany
| | - Anne-Sophie Wegscheider
- Institute for Histology, Cytology and Molecular Diagnostics, MVZ Prof. Dr. med. A. Niendorf Pathologie Hamburg-West GmbH, 22767, Hamburg, Germany
| | - Josef A Käs
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103, Leipzig, Germany.
| | - Axel Niendorf
- Institute for Histology, Cytology and Molecular Diagnostics, MVZ Prof. Dr. med. A. Niendorf Pathologie Hamburg-West GmbH, 22767, Hamburg, Germany.
| |
Collapse
|
36
|
Yukhnenko D, Blackwood N, Lichtenstein P, Fazel S. Association of substance use and other psychiatric disorders with all-cause and external-cause mortality in individuals given community sentences in Sweden: a national cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2023; 33:100703. [PMID: 37954004 PMCID: PMC10636268 DOI: 10.1016/j.lanepe.2023.100703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 11/14/2023]
Abstract
Background Consistently high rates of premature mortality have been reported in individuals who receive community sentences. However, few studies have explored potential modifiable risk factors for these rates, particularly mental health. We examined the association of substance use and other psychiatric disorders with all-cause and external-cause mortality in individuals convicted of a criminal offence and given a community sentence. Methods We did a longitudinal cohort study of 109,751 individuals given community sentences in Sweden using population-based registers. We calculated mortality rates for all-cause and external-cause mortality, hazard ratios for the association between psychiatric disorders and mortality, and population attributable fractions to quantify the contribution of psychiatric disorders to mortality risk. Findings During the follow-up, 5749 (5.2%) individuals died, including 2709 (2.5%) from external causes. Individuals with pre-existing substance use and other psychiatric disorders had an increased mortality risk from any cause (aHR = 2.28 [95% CI 2.15-2.42]) and from external causes (3.11 [2.85-3.40]) compared to individuals without known psychiatric or substance use disorders. Suicide was the most common cause of death in younger persons. Interpretation In individuals given community sentences, substance use and other psychiatric disorders were associated with an increased risk of premature death with suicide being the leading cause of death. Community supervision represents an opportunity to provide sentenced individuals with access to evidence-based treatment targeting substance misuse and psychiatric disorders to prevent potentially preventable deaths. Funding Wellcome Trust.
Collapse
Affiliation(s)
| | - Nigel Blackwood
- Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, UK
| | - Paul Lichtenstein
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| | - Seena Fazel
- Department of Psychiatry, University of Oxford, Oxford, UK
| |
Collapse
|
37
|
Pentenero M, Castagnola P, Castillo FV, Isaevska E, Sutera S, Gandolfo S. Predictors of malignant transformation in oral leukoplakia and proliferative verrucous leukoplakia: An observational prospective study including the DNA ploidy status. Head Neck 2023; 45:2589-2604. [PMID: 37563936 DOI: 10.1002/hed.27483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/23/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND This prospective observational study investigated the determinants of malignant transformation (MT) in localized oral leukoplakia (OL) and proliferative verrucous leukoplakia (PVL). METHODS Demographic, clinical, histological, and DNA ploidy status data were collected at enrolment. Survival analysis was performed (MT being the event of interest). RESULTS One-hundred and thirty-three patients with OL and 20 patients with PVL entered the study over 6 years (mean follow-up 7.8 years). The presence of OED, DNA ploidy, clinical presentation, and lesion site were associated with MT in patients with OL in a univariate analysis. In a multivariate model, OED was the strongest predictor of MT in patients with OL. Adding DNA ploidy increased the model's predictive power. None of the assessed predictors was associated with MT in patients with PVL. CONCLUSIONS DNA ploidy might identify a subset OL with low risk or minimal risk of MT, but it does not seem to be a reliable predictor in patients with PVL.
Collapse
Affiliation(s)
- Monica Pentenero
- Oral Medicine and Oral Oncology Unit, Department of Oncology, University of Turin, Turin, Italy
| | | | | | - Elena Isaevska
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Samuele Sutera
- Oral Medicine and Oral Oncology Unit, Department of Oncology, University of Turin, Turin, Italy
| | - Sergio Gandolfo
- Oral Medicine and Oral Oncology Unit, Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
38
|
Stabellini N, Cullen J, Moore JX, Dent S, Sutton AL, Shanahan J, Montero AJ, Guha A. Social Determinants of Health Data Improve the Prediction of Cardiac Outcomes in Females with Breast Cancer. Cancers (Basel) 2023; 15:4630. [PMID: 37760599 PMCID: PMC10526347 DOI: 10.3390/cancers15184630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality among breast cancer (BC) patients aged 50 and above. Machine Learning (ML) models are increasingly utilized as prediction tools, and recent evidence suggests that incorporating social determinants of health (SDOH) data can enhance its performance. This study included females ≥ 18 years diagnosed with BC at any stage. The outcomes were the diagnosis and time-to-event of major adverse cardiovascular events (MACEs) within two years following a cancer diagnosis. Covariates encompassed demographics, risk factors, individual and neighborhood-level SDOH, tumor characteristics, and BC treatment. Race-specific and race-agnostic Extreme Gradient Boosting ML models with and without SDOH data were developed and compared based on their C-index. Among 4309 patients, 11.4% experienced a 2-year MACE. The race-agnostic models exhibited a C-index of 0.78 (95% CI 0.76-0.79) and 0.81 (95% CI 0.80-0.82) without and with SDOH data, respectively. In non-Hispanic Black women (NHB; n = 765), models without and with SDOH data achieved a C-index of 0.74 (95% CI 0.72-0.76) and 0.75 (95% CI 0.73-0.78), respectively. Among non-Hispanic White women (n = 3321), models without and with SDOH data yielded a C-index of 0.79 (95% CI 0.77-0.80) and 0.79 (95% CI 0.77-0.80), respectively. In summary, including SDOH data improves the predictive performance of ML models in forecasting 2-year MACE among BC females, particularly within NHB.
Collapse
Affiliation(s)
- Nickolas Stabellini
- Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Hematology-Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo 05652-900, SP, Brazil
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Jennifer Cullen
- Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Justin X. Moore
- Center for Health Equity Transformation, Department of Behavioral Science, Department of Internal Medicine, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, NC 27708, USA
| | - Arnethea L. Sutton
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - John Shanahan
- Cancer Informatics, Seidman Cancer Center, University Hospitals of Cleveland, Cleveland, OH 44106, USA
| | - Alberto J. Montero
- Department of Hematology-Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Avirup Guha
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Cardio-Oncology Program, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
39
|
Mandla R, Schroeder P, Porneala B, Florez JC, Meigs JB, Mercader JM, Leong A. Polygenic Scores for Longitudinal Prediction of Incident Type 2 Diabetes in an Ancestrally and Medically Diverse Primary Care Network. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.08.23295276. [PMID: 37732255 PMCID: PMC10508788 DOI: 10.1101/2023.09.08.23295276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
OBJECTIVE The clinical utility of genetic information for type 2 diabetes (T2D) prediction with polygenic score (PGS) in ancestrally diverse, real-world US healthcare systems is unclear, especially for those at low clinical phenotypic risk for T2D. RESEARCH DESIGN AND METHODS We tested the association of PGS with T2D incidence in patients followed within a primary care practice network over 16 years in four hypothetical scenarios that varied by clinical data availability (N = 14,712): 1) age and sex, 2) age, sex, BMI, systolic blood pressure, and family history of diabetes; 3) all variables in (2) and random glucose; 4) all variables in (3), HDL, total cholesterol, and triglycerides, combined in a clinical risk score (CRS). To determine whether genetic effects differed by baseline clinical risk, we tested for interaction with the CRS. RESULTS PGS was associated with incident diabetes in all models. Adjusting for age and sex only, the Hazard Ratio (HR) per PGS standard deviation (SD) was 1.76 (95% CI 1.68, 1.84) and the HR of top 5% of PGS vs interquartile range (IQR) was 2.80 (2.39, 3.28). Adjusting for the CRS, the HR per SD was 1.48 (1.40, 1.57) and HR of top 5% of PGS vs IQR was 2.09 (1.72, 2.55). Genetic effects differed by baseline clinical risk [(PGS-CRS interaction p =0.05; CRS below the median: HR 1.60 (1.43, 1.79); CRS above the median: HR 1.45 (1.35, 1.55)]. CONCLUSIONS Genetic information can help identify high-risk patients even among those perceived to be low risk in a clinical evaluation.
Collapse
|
40
|
Choby G, Geltzeiler M, Almeida JP, Champagne PO, Chan E, Ciporen J, Chaskes MB, Fernandez-Miranda J, Gardner P, Hwang P, Ji KSY, Kalyvas A, Kong KA, McMillan R, Nayak J, O’Byrne J, Patel C, Patel Z, Peris Celda M, Pinheiro-Neto C, Sanusi O, Snyderman C, Thorp BD, Van Gompel JJ, Young SC, Zenonos G, Zwagerman NT, Wang EW. Multicenter Survival Analysis and Application of an Olfactory Neuroblastoma Staging Modification Incorporating Hyams Grade. JAMA Otolaryngol Head Neck Surg 2023; 149:837-844. [PMID: 37535372 PMCID: PMC10401389 DOI: 10.1001/jamaoto.2023.1939] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/08/2023] [Indexed: 08/04/2023]
Abstract
Importance Current olfactory neuroblastoma (ONB) staging systems inadequately delineate locally advanced tumors, do not incorporate tumor grade, and poorly estimate survival and recurrence. Objective The primary aims of this study were to (1) examine the clinical covariates associated with survival and recurrence of ONB in a modern-era multicenter cohort and (2) incorporate Hyams tumor grade into existing staging systems to assess its ability to estimate survival and recurrence. Design, Setting, and Participants This retrospective, multicenter, case-control study included patients with ONB who underwent treatment between January 1, 2005, and December 31, 2021, at 9 North American academic medical centers. Intervention Standard-of-care ONB treatment. Main Outcome and Measures The main outcomes were overall survival (OS), disease-free survival (DFS), and disease-specific survival (DSS) as C statistics for model prediction. Results A total of 256 patients with ONB (mean [SD] age, 52.0 [15.6] years; 115 female [44.9%]; 141 male [55.1%]) were included. The 5-year rate for OS was 83.5% (95% CI, 78.3%-89.1%); for DFS, 70.8% (95% CI, 64.3%-78.0%); and for DSS, 94.1% (95% CI, 90.5%-97.8%). On multivariable analysis, age, American Joint Committee on Cancer (AJCC) stage, involvement of bilateral maxillary sinuses, and positive margins were associated with OS. Only AJCC stage was associated with DFS. Only N stage was associated with DSS. When assessing the ability of staging systems to estimate OS, the best-performing model was the novel modification of the Dulguerov system (C statistic, 0.66; 95% CI, 0.59-0.76), and the Kadish system performed most poorly (C statistic, 0.57; 95% CI, 0.50-0.63). Regarding estimation of DFS, the modified Kadish system performed most poorly (C statistic, 0.55; 95% CI, 0.51-0.66), while the novel modification of the AJCC system performed the best (C statistic, 0.70; 95% CI, 0.66-0.80). Regarding estimation of DSS, the modified Kadish system was the best-performing model (C statistic, 0.79; 95% CI, 0.70-0.94), and the unmodified Kadish performed the worst (C statistic, 0.56; 95% CI, 0.51-0.68). The ability for novel ONB staging systems to estimate disease progression across stages was also assessed. In the novel Kadish staging system, patients with stage VI disease were approximately 7 times as likely to experience disease progression as patients with stage I disease (hazard ratio [HR], 6.84; 95% CI, 1.60-29.20). Results were similar for the novel modified Kadish system (HR, 8.99; 95% CI, 1.62-49.85) and the novel Dulguerov system (HR, 6.86; 95% CI, 2.74-17.18). Conclusions and Relevance The study findings indicate that 5-year OS for ONB is favorable and that incorporation of Hyams grade into traditional ONB staging systems is associated with improved estimation of disease progression.
Collapse
Affiliation(s)
- Garret Choby
- Department of Otolaryngology–Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Mathew Geltzeiler
- Department of Otolaryngology–Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
| | | | | | - Erik Chan
- Department of Otolaryngology–Head and Neck Surgery, Stanford University, Palo Alto, California
| | - Jeremy Ciporen
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| | - Mark B. Chaskes
- Department of Otolaryngology–Head and Neck Surgery, University of North Carolina at Chapel Hill
| | | | - Paul Gardner
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Peter Hwang
- Department of Otolaryngology–Head and Neck Surgery, Stanford University, Palo Alto, California
| | - Keven Seung Yong Ji
- Department of Otolaryngology–Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
| | | | - Keonho A. Kong
- Department of Otolaryngology–Head and Neck Surgery, University of North Carolina at Chapel Hill
| | - Ryan McMillan
- Department of Otolaryngology–Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Jayakar Nayak
- Department of Otolaryngology–Head and Neck Surgery, Stanford University, Palo Alto, California
| | - Jamie O’Byrne
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Chirag Patel
- Department of Otolaryngology–Head and Neck Surgery, Loyola University, Maywood, Illinois
| | - Zara Patel
- Department of Otolaryngology–Head and Neck Surgery, Stanford University, Palo Alto, California
| | - Maria Peris Celda
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota
| | - Carlos Pinheiro-Neto
- Department of Otolaryngology–Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Olabisi Sanusi
- Department of Otolaryngology–Head and Neck Surgery, University of North Carolina at Chapel Hill
| | - Carl Snyderman
- Department of Otolaryngology–Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Brian D. Thorp
- Department of Otolaryngology–Head and Neck Surgery, University of North Carolina at Chapel Hill
| | | | - Sarah C. Young
- Department of Neurological Surgery, University of Wisconsin, Milwaukee, Wisconsin
| | - Georgios Zenonos
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Nathan T. Zwagerman
- Department of Neurological Surgery, University of Wisconsin, Milwaukee, Wisconsin
| | - Eric W. Wang
- Department of Otolaryngology–Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
41
|
Deng L, Jiang H. Decreased Expression of GLYATL1 Predicts Poor Prognosis in Patients with Clear Cell Renal Cell Carcinoma. Int J Gen Med 2023; 16:3757-3768. [PMID: 37649851 PMCID: PMC10464902 DOI: 10.2147/ijgm.s419301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023] Open
Abstract
Background GLYATL1 is a member of the glycine-N-acyltransferase family, which catalyses acyl group transfer. The role of GLYATL1 in cancer is largely unknown; therefore, the potential value of GLYATL1 in clear cell renal cell carcinoma (ccRCC) was explored. Methods The ccRCC gene expression profiles and clinical data were obtained from the University of California Santa Cruz Xena platform. Differential expression and survival analysis were performed using R software. Samples from the TIMER public database and real-world were used for validation. The potential molecular mechanism of GLYATL1 in ccRCC was explored using gene set enrichment analysis (GSEA). Results GLYATL1 was downregulated, indicating a poor prognosis in ccRCC. Low expression of GLYATL1 was significantly associated with advanced stage and higher histological grade ccRCC. The differential expression of GLYATL1 was validated at the protein level using clinical samples and tissue microarray. The results of GSEA showed that multiple crucial signalling pathways including fatty acid metabolism, adipogenesis, oxidative phosphorylation and epithelial-mesenchymal transition were enriched. Conclusion This study demonstrated that GLYATL1 downregulation has an unfavourable impact on the survival of patients with ccRCC. The resulting data indicated that GLYATL1 could be a potential new target for ccRCC therapy, which may be helpful for the personalized treatment of such individuals.
Collapse
Affiliation(s)
- Limin Deng
- Department of Urology, Meizhou Academy of Medical Sciences, Meizhou People’s Hospital, Guangdong Medical University, Meizhou, Guangdong Province, People’s Republic of China
| | - Huiming Jiang
- Department of Urology, Meizhou Academy of Medical Sciences, Meizhou People’s Hospital, Meizhou, Guangdong Province, People's Republic of China
| |
Collapse
|
42
|
Wang X, Zhao M, Zhang C, Chen H, Liu X, An Y, Zhang L, Guo X. Establishment and Clinical Application of the Nomogram Related to Risk or Prognosis of Hepatocellular Carcinoma: A Review. J Hepatocell Carcinoma 2023; 10:1389-1398. [PMID: 37637500 PMCID: PMC10460189 DOI: 10.2147/jhc.s417123] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent primary liver malignancy, accounting for approximately 90% of all primary liver cancers, with high mortality and a poor prognosis. A large number of predictive models have been applied that integrate multiple clinical factors and biomarkers to predict the prognosis of HCC. Nomograms, as easy-to-use prognostic predictive models, are widely used to predict the probability of clinical outcomes. We searched PubMed with the keywords "hepatocellular carcinoma" and "nomogram", and 974 relative literatures were retrieved. According to the construction methodology and the real validity of the nomograms, in this study, 97 nomograms for HCC were selected in 77 publications. These 97 nomograms were established based on more than 100,000 patients, covering seven main prognostic outcomes. The research data of 56 articles are from hospital-based HCC patients, and 13 articles provided external validation results of the nomogram. In addition to AFP, tumor size, tumor number, stage, vascular invasion, age, and other common prognostic risk factors are included in the HCC-related nomogram, more and more biomarkers, including gene mRNA expression, gene polymorphisms, and gene signature, etc. were also included in the nomograms. The establishment, assessment and validation of these nomograms are also discussed in depth. This study would help clinicians construct and select appropriate nomograms to guide precise judgment and appropriate treatments.
Collapse
Affiliation(s)
- Xiangze Wang
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Minghui Zhao
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Chensheng Zhang
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Haobo Chen
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Xingyu Liu
- School of Computer and Information Engineering, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Yang An
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Lu Zhang
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Xiangqian Guo
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 475004, People’s Republic of China
| |
Collapse
|
43
|
Moreno-Sánchez PA. Improvement of a prediction model for heart failure survival through explainable artificial intelligence. Front Cardiovasc Med 2023; 10:1219586. [PMID: 37600061 PMCID: PMC10434534 DOI: 10.3389/fcvm.2023.1219586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Cardiovascular diseases and their associated disorder of heart failure (HF) are major causes of death globally, making it a priority for doctors to detect and predict their onset and medical consequences. Artificial Intelligence (AI) allows doctors to discover clinical indicators and enhance their diagnoses and treatments. Specifically, "eXplainable AI" (XAI) offers tools to improve the clinical prediction models that experience poor interpretability of their results. This work presents an explainability analysis and evaluation of two HF survival prediction models using a dataset that includes 299 patients who have experienced HF. The first model utilizes survival analysis, considering death events and time as target features, while the second model approaches the problem as a classification task to predict death. The model employs an optimization data workflow pipeline capable of selecting the best machine learning algorithm as well as the optimal collection of features. Moreover, different post hoc techniques have been used for the explainability analysis of the model. The main contribution of this paper is an explainability-driven approach to select the best HF survival prediction model balancing prediction performance and explainability. Therefore, the most balanced explainable prediction models are Survival Gradient Boosting model for the survival analysis and Random Forest for the classification approach with a c-index of 0.714 and balanced accuracy of 0.74 (std 0.03) respectively. The selection of features by the SCI-XAI in the two models is similar where "serum_creatinine", "ejection_fraction", and "sex" are selected in both approaches, with the addition of "diabetes" for the survival analysis model. Moreover, the application of post hoc XAI techniques also confirm common findings from both approaches by placing the "serum_creatinine" as the most relevant feature for the predicted outcome, followed by "ejection_fraction". The explainable prediction models for HF survival presented in this paper would improve the further adoption of clinical prediction models by providing doctors with insights to better understand the reasoning behind usually "black-box" AI clinical solutions and make more reasonable and data-driven decisions.
Collapse
|
44
|
Chen K, Zheng T, Chen C, Liu L, Guo Z, Peng Y, Zhang X, Yang Z. Pregnancy Zone Protein Serves as a Prognostic Marker and Favors Immune Infiltration in Lung Adenocarcinoma. Biomedicines 2023; 11:1978. [PMID: 37509617 PMCID: PMC10377424 DOI: 10.3390/biomedicines11071978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is a public enemy with a very high incidence and mortality rate, for which there is no specific detectable biomarker. Pregnancy zone protein (PZP) is an immune-related protein; however, the functions of PZP in LUAD are unclear. In this study, a series of bioinformatics methods, combined with immunohistochemistry (IHC), four-color multiplex fluorescence immunohistochemistry (mIHC), quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA), were utilized to explore the prognostic value and potential role of PZP in LUAD. Our data revealed that PZP expression was markedly reduced in LUAD tissues, tightly correlated with clinical stage and could be an independent unfavorable prognostic factor. In addition, pathway analysis revealed that high expression of PZP in LUAD was mainly involved in immune-related molecules. Tumor immune infiltration analysis by CIBERSORT showed a significant correlation between PZP expression and several immune cell infiltrations, and IHC further confirmed a positive correlation with CD4+ T-cell infiltration and a negative correlation with CD68+ M0 macrophage infiltration. Furthermore, mIHC demonstrated that PZP expression gave rise to an increase in CD86+ M1 macrophages and a decrease in CD206+ M2 macrophages. Therefore, PZP can be used as a new biomarker for the prediction of prognosis and may be a promising immune-related molecular target for LUAD.
Collapse
Affiliation(s)
- Kehong Chen
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Taihao Zheng
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Cai Chen
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Liangzhong Liu
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhengjun Guo
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yuan Peng
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaoyue Zhang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhenzhou Yang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
45
|
Zheng Y, Carrillo-Perez F, Pizurica M, Heiland DH, Gevaert O. Spatial cellular architecture predicts prognosis in glioblastoma. Nat Commun 2023; 14:4122. [PMID: 37433817 DOI: 10.1038/s41467-023-39933-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023] Open
Abstract
Intra-tumoral heterogeneity and cell-state plasticity are key drivers for the therapeutic resistance of glioblastoma. Here, we investigate the association between spatial cellular organization and glioblastoma prognosis. Leveraging single-cell RNA-seq and spatial transcriptomics data, we develop a deep learning model to predict transcriptional subtypes of glioblastoma cells from histology images. Employing this model, we phenotypically analyze 40 million tissue spots from 410 patients and identify consistent associations between tumor architecture and prognosis across two independent cohorts. Patients with poor prognosis exhibit higher proportions of tumor cells expressing a hypoxia-induced transcriptional program. Furthermore, a clustering pattern of astrocyte-like tumor cells is associated with worse prognosis, while dispersion and connection of the astrocytes with other transcriptional subtypes correlate with decreased risk. To validate these results, we develop a separate deep learning model that utilizes histology images to predict prognosis. Applying this model to spatial transcriptomics data reveal survival-associated regional gene expression programs. Overall, our study presents a scalable approach to unravel the transcriptional heterogeneity of glioblastoma and establishes a critical connection between spatial cellular architecture and clinical outcomes.
Collapse
Affiliation(s)
- Yuanning Zheng
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, 94305, USA
| | - Francisco Carrillo-Perez
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, 94305, USA
- Department of Architecture and Computer Technology (ATC), University of Granada, Granada, 18014, Spain
| | - Marija Pizurica
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, 94305, USA
- Internet technology and Data science Lab (IDLab), Ghent University, Technologiepark-Zwijnaarde 126, Ghent, 9052, Gent, Belgium
| | - Dieter Henrik Heiland
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, 79106, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, 79106, Germany
| | - Olivier Gevaert
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, 94305, USA.
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
46
|
Pu CC, Yin L, Yan JM. Risk factors and survival prediction of young breast cancer patients with liver metastases: a population-based study. Front Endocrinol (Lausanne) 2023; 14:1158759. [PMID: 37424855 PMCID: PMC10328090 DOI: 10.3389/fendo.2023.1158759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Background The risk and prognosis of young breast cancer (YBC) with liver metastases (YBCLM) remain unclear. Thus, this study aimed to determine the risk and prognostic factors in these patients and construct predictive nomogram models. Methods This population-based retrospective study was conducted using data of YBCLM patients from the Surveillance, Epidemiology, and End Results database between 2010 and 2019. Multivariate logistic and Cox regression analyses were used to identify independent risk and prognostic factors, which were used to construct the diagnostic and prognostic nomograms. The concordance index (C-index), calibration plot, receiver operating characteristic (ROC) curve, and decision curve analysis (DCA) were used to assess the performances of the established nomogram models. Propensity score matching (PSM) analysis was used to balance the baseline characteristics between the YBCLM patients and non-young patients with BCLM when comparing overall survival (OS) and cancer-specific survival (CSS). Results A total of 18,275 YBC were identified, of whom 400 had LM. T stage, N stage, molecular subtypes, and bone, lung, and brain metastases were independent risk factors for LM developing in YBC. The established diagnostic nomogram showed that bone metastases contributed the most risk of LM developing, with a C-index of 0.895 (95% confidence interval 0.877-0.913) for this nomogram model. YBCLM had better survival than non-young patients with BCLM in unmatched and matched cohorts after propensity score matching analysis. The multivariate Cox analysis demonstrated that molecular subtypes, surgery and bone, lung, and brain metastases were independently associated with OS and CSS, chemotherapy was an independent prognostic factor for OS, and marital status and T stage were independent prognostic factors for CSS. The C-indices for the OS- and CSS-specific nomograms were 0.728 (0.69-0.766) and 0.74 (0.696-0.778), respectively. The ROC analysis indicated that these models had excellent discriminatory power. The calibration curve also showed that the observed results were consistent with the predicted results. DCA showed that the developed nomogram models would be effective in clinical practice. Conclusion The present study determined the risk and prognostic factors of YBCLM and further developed nomograms that can be used to effectively identify high-risk patients and predict survival outcomes.
Collapse
Affiliation(s)
- Chen-Chen Pu
- Department of Breast and Thyroid Surgery, The First People’s Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, Jiangsu, China
| | - Lei Yin
- Department of Breast and Thyroid Surgery, Wuzhong People’s Hospital of Suzhou City, Suzhou, Jiangsu, China
| | - Jian-Ming Yan
- Department of Breast and Thyroid Surgery, The First People’s Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Taicang, Jiangsu, China
| |
Collapse
|
47
|
Hartrampf PE, Mihatsch PW, Seitz AK, Solnes LB, Rowe SP, Pomper MG, Kübler H, Bley TA, Buck AK, Werner RA. Elevated Body Mass Index Is Associated with Improved Overall Survival in Castration-Resistant Prostate Cancer Patients Undergoing Prostate-Specific Membrane Antigen-Directed Radioligand Therapy. J Nucl Med 2023:jnumed.122.265379. [PMID: 37290794 DOI: 10.2967/jnumed.122.265379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/21/2023] [Indexed: 06/10/2023] Open
Abstract
In patients with prostate cancer scheduled for systemic treatment, being overweight is linked to prolonged overall survival (OS), whereas sarcopenia is associated with shorter OS. We investigated fat-related and body composition parameters in patients undergoing prostate-specific membrane antigen (PSMA)-directed radioligand therapy (RLT) to assess their predictive value for OS. Methods: Body mass index (BMI, in kg/m2) and CT-derived body composition parameters (total, subcutaneous, visceral fat area, and psoas muscle area at the L3-L4 level) were determined for 171 patients scheduled for PSMA-directed RLT. After normalization for stature, the psoas muscle index was used to define sarcopenia. Outcome analysis was performed using Kaplan-Meier curves and Cox regression including fat-related and other clinical parameters (Gleason score, C-reactive protein [CRP], lactate dehydrogenase [LDH], hemoglobin, and prostate-specific antigen levels). The Harrell C-index was used for goodness-of-fit analysis. Results: Sixty-five patients (38%) had sarcopenia, and 98 patients (57.3%) had increased BMI. Relative to the 8-mo OS in normal-weight men (BMI < 25), overweight men (25 ≥ BMI > 30) and obese men (BMI ≥ 30) achieved a longer OS of 14 mo (hazard ratio [HR], 0.63; 95% CI, 0.40-0.99; P = 0.03) and 13 mo (HR, 0.47; 95% CI, 0.29-0.77; P = 0.004), respectively. Sarcopenia showed no impact on OS (11 vs. 12 mo; HR, 1.4; 95% CI, 0.91-2.1; P = 0.09). Most of the body composition parameters were tightly linked to OS on univariable analyses, with the highest C-index for BMI. In multivariable analysis, a higher BMI (HR, 0.91; 95% CI, 0.86-0.97; P = 0.006), lower CRP (HR, 1.09; 95% CI, 1.03-1.14; P < 0.001), lower LDH (HR, 1.08; 95% CI, 1.03-1.14; P < 0.001), and longer interval between initial diagnosis and RLT (HR, 0.95; 95% CI, 0.91-0.99; P = 0.02) were significant predictors of OS. Conclusion: Increased fat reserves assessed by BMI, CRP, LDH, and interval between initial diagnosis and RLT, but not CT-derived body composition parameters, were relevant predictors for OS. As BMI can be altered, future research should investigate whether a high-calorie diet before or during PSMA RLT may improve OS.
Collapse
Affiliation(s)
- Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany;
| | - Patrick W Mihatsch
- Department of Diagnostic and Interventional Radiology, University Hospital of Würzburg, Würzburg, Germany
| | - Anna Katharina Seitz
- Department of Urology and Paediatric Urology, University Hospital of Würzburg, Würzburg, Germany; and
| | - Lilja B Solnes
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hubert Kübler
- Department of Urology and Paediatric Urology, University Hospital of Würzburg, Würzburg, Germany; and
| | - Thorsten A Bley
- Department of Diagnostic and Interventional Radiology, University Hospital of Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
48
|
Deleuze C, Dickinson L, Orczyk C. Re: Thomas Bommelaere, Arnauld Villers, Philippe Puech, et al. Risk Estimation of Metastatic Recurrence After Prostatectomy: A Model Using Preoperative Magnetic Resonance Imaging and Targeted Biopsy. Eur Urol Open Sci 2022;41:24-34. EUR UROL SUPPL 2023; 52:135-136. [PMID: 37213239 PMCID: PMC10196329 DOI: 10.1016/j.euros.2023.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2023] [Indexed: 05/23/2023] Open
Affiliation(s)
- Claire Deleuze
- Division of Surgery and Interventional Sciences, University College London, London, UK
| | - Louise Dickinson
- Department of Radiology, University College London Hospitals, London, UK
| | - Clement Orczyk
- Division of Surgery and Interventional Sciences, University College London, London, UK
- Department of Urology, University College London Hospitals, London, UK
- Corresponding author. Division of Surgery and Interventional Sciences, University College London, Charles Bell House, 43–45 Foley Street, London W1W 7TS, UK. Tel. +44 7 958550727; Fax: +44 207 6799511.
| |
Collapse
|
49
|
Han CH, Park M, Kim H, Roh YY, Kim SY, Kim JD, Kim MJ, Lee YJ, Kim KW, Kim YH. Radiologic Assessment of Lung Edema Score as a Predictor of Clinical Outcome in Children with Acute Respiratory Distress Syndrome. Yonsei Med J 2023; 64:384-394. [PMID: 37226565 DOI: 10.3349/ymj.2022.0653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 05/26/2023] Open
Abstract
PURPOSE The radiographic assessment of lung edema (RALE) score enables objective quantification of lung edema and is a valuable prognostic marker of adult acute respiratory distress syndrome (ARDS). We aimed to evaluate the validity of RALE score in children with ARDS. MATERIALS AND METHODS The RALE score was measured for its reliability and correlation to other ARDS severity indices. ARDS-specific mortality was defined as death from severe pulmonary dysfunction or the need for extracorporeal membrane oxygenation therapy. The C-index of the RALE score and other ARDS severity indices were compared via survival analyses. RESULTS Among 296 children with ARDS, 88 did not survive, and there were 70 ARDS-specific non-survivors. The RALE score showed good reliability with an intraclass correlation coefficient of 0.809 [95% confidence interval (CI), 0.760-0.848]. In univariable analysis, the RALE score had a hazard ratio (HR) of 1.19 (95% CI, 1.18-3.11), and the significance was maintained in multivariable analysis adjusting with age, ARDS etiology, and comorbidity, with an HR of 1.77 (95% CI, 1.05-2.91). The RALE score was a good predictor of ARDS-specific mortality, with a C-index of 0.607 (95% CI, 0.519-0.695). CONCLUSION The RALE score is a reliable measure for ARDS severity and a useful prognostic marker of mortality in children, especially for ARDS-specific mortality. This score provides information that clinicians can use to decide the proper time of aggressive therapy targeting severe lung injury and to appropriately manage the fluid balance of children with ARDS.
Collapse
Affiliation(s)
- Chang Hoon Han
- Department of Pediatrics, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Mireu Park
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea
| | - Hamin Kim
- Department of Pediatrics, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea
| | - Yun Young Roh
- Department of Pediatrics, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea
| | - Soo Yeon Kim
- Department of Pediatrics, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea
| | - Jong Deok Kim
- Department of Pediatrics, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea
| | - Min Jung Kim
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea
- Department of Pediatrics, Yongin Severance Hospital, Yongin, Korea
| | - Yong Ju Lee
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea
- Department of Pediatrics, Yongin Severance Hospital, Yongin, Korea
| | - Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea
| | - Yoon Hee Kim
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Seoul, Korea.
| |
Collapse
|
50
|
Saumarez R, Silberbauer J, Scannell J, Pytkowski M, Behr ER, Betts T, Della Bella P, Peters NS. Should lethal arrhythmias in hypertrophic cardiomyopathy be predicted using non-electrophysiological methods? Europace 2023; 25:euad045. [PMID: 36942430 PMCID: PMC10227650 DOI: 10.1093/europace/euad045] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/20/2023] [Indexed: 03/23/2023] Open
Abstract
While sudden cardiac death (SCD) in hypertrophic cardiomyopathy (HCM) is due to arrhythmias, the guidelines for prediction of SCD are based solely on non-electrophysiological methods. This study aims to stimulate thinking about whether the interests of patients with HCM are better served by using current, 'risk factor', methods of prediction or by further development of electrophysiological methods to determine arrhythmic risk. Five published predictive studies of SCD in HCM, which contain sufficient data to permit analysis, were analysed to compute receiver operating characteristics together with their confidence bounds to compare their formal prediction either by bootstrapping or Monte Carlo analysis. Four are based on clinical risk factors, one with additional MRI analysis, and were regarded as exemplars of the risk factor approach. The other used an electrophysiological method and directly compared this method to risk factors in the same patients. Prediction methods that use conventional clinical risk factors and MRI have low predictive capacities that will only detect 50-60% of patients at risk with a 15-30% false positive rate [area under the curve (AUC) = ∼0.7], while the electrophysiological method detects 90% of events with a 20% false positive rate (AUC = ∼0.89). Given improved understanding of complex arrhythmogenesis, arrhythmic SCD is likely to be more accurately predictable using electrophysiologically based approaches as opposed to current guidelines and should drive further development of electrophysiologically based methods.
Collapse
Affiliation(s)
| | - John Silberbauer
- Department Cardiology, Royal Sussex Hospital, Eastern Road, Brighton BN2 5BE, UK
| | - Jack Scannell
- The Bayes Centre, University of Edinburgh, Edinburgh EH8 9BT, UK
| | - Mariusz Pytkowski
- Department of Cardiology, Narodowy Instytut Kardiologii, ul Alpejska 42, 04-628 Warsaw, Poland
| | | | - Timothy Betts
- Department of Cardiology, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Paulo Della Bella
- Department of Cardiology, San Raffaele Hospital, IT 20133, Milan, Italy
| | - Nicholas S Peters
- Department of Cardiology, Hammersmith Hospital, Imperial College, London W12 0HS, UK
| |
Collapse
|