1
|
Pesapane F, Rotili A, Scalco E, Pupo D, Carriero S, Corso F, De Marco P, Origgi D, Nicosia L, Ferrari F, Penco S, Pizzamiglio M, Rizzo G, Cassano E. Predictive value of tumoral and peritumoral radiomic features in neoadjuvant chemotherapy response for breast cancer: a retrospective study. LA RADIOLOGIA MEDICA 2025; 130:598-612. [PMID: 39992329 DOI: 10.1007/s11547-025-01969-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 02/05/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) improves surgical outcomes for breast cancer patients, with pathologic complete response (pCR) correlated with enhanced survival. The role of radiomics, particularly from peritumoral tissue, in predicting pCR remains under investigation. METHODS This retrospective study analyzed radiomic features from pretreatment dynamic contrast-enhanced breast MRI scans of 150 patients undergoing NACT. A proportional approach was used to define peritumoral zones, assessed both with a 10% and 30% extension, allowing more standardized assessments relative to the tumor size. Radiomic features were evaluated alongside clinical and biological data to predict pCR. The association of clinical/biological and radiomic features with pCR to NACT was evaluated using univariate and multivariate analysis, logistic regression, and a random forest model. A clinical/biological model, a radiomic model, and a combined clinical/biological and 4 radiomic models for predicting the response to NACT were constructed. Area under the curve (AUC) and 95% confidence intervals (CIs) were used to assess the performance of the models. RESULTS Ninety-five patients (average age 47 years) were finally included. HER2 + , basal-like molecular subtypes, and a high level of Ki67 (≥ 20%) were associated with a higher likelihood of pCR to NACT. The combined clinical-biological-radiomic model, especially with a 10% peritumoral extension, showed improved predictive accuracy (AUC 0.76, CI 0.65-0.85) compared to models using clinical-biological data alone (AUC 0.73, CI 0.63-0.83). CONCLUSIONS Integrating peritumoral radiomic features with clinical and biological data enhances the prediction of pCR to NACT, underscoring the potential of a multifaceted approach in treatment personalization.
Collapse
Affiliation(s)
- Filippo Pesapane
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | - Anna Rotili
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Elisa Scalco
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche (ITB-CNR), Segrate, MI, Italy
| | - Davide Pupo
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Serena Carriero
- Department of Radiology and Interventional Radiology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Corso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Paolo De Marco
- Medical Physics Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | - Luca Nicosia
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Ferrari
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Silvia Penco
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Pizzamiglio
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanna Rizzo
- Istituto di Sistemi e Tecnologie Industriali Intelligenti per il Manifatturiero Avanzato (STIIMA), CNR, Segrate, MI, Italy
| | - Enrico Cassano
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
2
|
Hachache R, Yahyaouy A, Riffi J, Tairi H, Abibou S, Adoui ME, Benjelloun M. Advancing personalized oncology: a systematic review on the integration of artificial intelligence in monitoring neoadjuvant treatment for breast cancer patients. BMC Cancer 2024; 24:1300. [PMID: 39434042 PMCID: PMC11495077 DOI: 10.1186/s12885-024-13049-0] [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: 06/21/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
PURPOSE Despite suffering from the same disease, each patient exhibits a distinct microbiological profile and variable reactivity to prescribed treatments. Most doctors typically use a standardized treatment approach for all patients suffering from a specific disease. Consequently, the challenge lies in the effectiveness of this standardized treatment and in adapting it to each individual patient. Personalized medicine is an emerging field in which doctors use diagnostic tests to identify the most effective medical treatments for each patient. Prognosis, disease monitoring, and treatment planning rely on manual, error-prone methods. Artificial intelligence (AI) uses predictive techniques capable of automating prognostic and monitoring processes, thus reducing the error rate associated with conventional methods. METHODS This paper conducts an analysis of current literature, encompassing the period from January 2015 to 2023, based on Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). RESULTS In assessing 25 pertinent studies concerning predicting neoadjuvant treatment (NAT) response in breast cancer (BC) patients, the studies explored various imaging modalities (Magnetic Resonance Imaging, Ultrasound, etc.), evaluating results based on accuracy, sensitivity, and area under the curve. Additionally, the technologies employed, such as machine learning (ML), deep learning (DL), statistics, and hybrid models, were scrutinized. The presentation of datasets used for predicting complete pathological response (PCR) was also considered. CONCLUSION This paper seeks to unveil crucial insights into the application of AI techniques in personalized oncology, particularly in the monitoring and prediction of responses to NAT for BC patients. Finally, the authors suggest avenues for future research into AI-based monitoring systems.
Collapse
Affiliation(s)
- Rachida Hachache
- Department of Computer Sciences, LISAC Laboratory, Sidi Mohammed Ben Abdellah University, Fez, Morocco.
| | - Ali Yahyaouy
- Department of Computer Sciences, LISAC Laboratory, Sidi Mohammed Ben Abdellah University, Fez, Morocco
- USPN, La Maison Des Sciences Numériques, Paris, France
| | - Jamal Riffi
- Department of Computer Sciences, LISAC Laboratory, Sidi Mohammed Ben Abdellah University, Fez, Morocco
| | - Hamid Tairi
- Department of Computer Sciences, LISAC Laboratory, Sidi Mohammed Ben Abdellah University, Fez, Morocco
| | - Soukayna Abibou
- Department of Computer Sciences, LISAC Laboratory, Sidi Mohammed Ben Abdellah University, Fez, Morocco
| | - Mohammed El Adoui
- Computer Science Unit, Faculty of Engineering, University of Mons, Place du Parc, 20, Mons, 7000, Belgium
| | - Mohammed Benjelloun
- Computer Science Unit, Faculty of Engineering, University of Mons, Place du Parc, 20, Mons, 7000, Belgium
| |
Collapse
|
3
|
Guo J, Chen B, Cao H, Dai Q, Qin L, Zhang J, Zhang Y, Zhang H, Sui Y, Chen T, Yang D, Gong X, Li D. Cross-modal deep learning model for predicting pathologic complete response to neoadjuvant chemotherapy in breast cancer. NPJ Precis Oncol 2024; 8:189. [PMID: 39237596 PMCID: PMC11377584 DOI: 10.1038/s41698-024-00678-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
Pathological complete response (pCR) serves as a critical measure of the success of neoadjuvant chemotherapy (NAC) in breast cancer, directly influencing subsequent therapeutic decisions. With the continuous advancement of artificial intelligence, methods for early and accurate prediction of pCR are being extensively explored. In this study, we propose a cross-modal multi-pathway automated prediction model that integrates temporal and spatial information. This model fuses digital pathology images from biopsy specimens and multi-temporal ultrasound (US) images to predict pCR status early in NAC. The model demonstrates exceptional predictive efficacy. Our findings lay the foundation for developing personalized treatment paradigms based on individual responses. This approach has the potential to become a critical auxiliary tool for the early prediction of NAC response in breast cancer patients.
Collapse
Affiliation(s)
- Jianming Guo
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Baihui Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Hongda Cao
- School of Computer, Beihang University, 100191, Beijing, China
| | - Quan Dai
- Medicine & Laboratory of Translational Research in Ultrasound Theranostics, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, 610041, Chengdu, China
- Department of Ultrasound, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, 610041, Chengdu, China
| | - Ling Qin
- Department of Pathology, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Jinfeng Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Youxue Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Huanyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Yuan Sui
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Tianyu Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Dongxu Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Xue Gong
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China
| | - Dalin Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150000, Harbin, China.
| |
Collapse
|
4
|
Mohamed RM, Panthi B, Adrada BE, Boge M, Candelaria RP, Chen H, Guirguis MS, Hunt KK, Huo L, Hwang KP, Korkut A, Litton JK, Moseley TW, Pashapoor S, Patel MM, Reed B, Scoggins ME, Son JB, Thompson A, Tripathy D, Valero V, Wei P, White J, Whitman GJ, Xu Z, Yang W, Yam C, Ma J, Rauch GM. Multiparametric MRI-based radiomic models for early prediction of response to neoadjuvant systemic therapy in triple-negative breast cancer. Sci Rep 2024; 14:16073. [PMID: 38992094 PMCID: PMC11239818 DOI: 10.1038/s41598-024-66220-9] [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: 07/26/2023] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is often treated with neoadjuvant systemic therapy (NAST). We investigated if radiomic models based on multiparametric Magnetic Resonance Imaging (MRI) obtained early during NAST predict pathologic complete response (pCR). We included 163 patients with stage I-III TNBC with multiparametric MRI at baseline and after 2 (C2) and 4 cycles of NAST. Seventy-eight patients (48%) had pCR, and 85 (52%) had non-pCR. Thirty-six multivariate models combining radiomic features from dynamic contrast-enhanced MRI and diffusion-weighted imaging had an area under the receiver operating characteristics curve (AUC) > 0.7. The top-performing model combined 35 radiomic features of relative difference between C2 and baseline; had an AUC = 0.905 in the training and AUC = 0.802 in the testing set. There was high inter-reader agreement and very similar AUC values of the pCR prediction models for the 2 readers. Our data supports multiparametric MRI-based radiomic models for early prediction of NAST response in TNBC.
Collapse
Affiliation(s)
- Rania M Mohamed
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bikash Panthi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Beatriz E Adrada
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
| | - Medine Boge
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
- Koc University Hospital, Istanbul, Turkey
| | - Rosalind P Candelaria
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
| | - Huiqin Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mary S Guirguis
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
| | - Kelly K Hunt
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken-Pin Hwang
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tanya W Moseley
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanaz Pashapoor
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
| | - Miral M Patel
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
| | - Brandy Reed
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marion E Scoggins
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
| | - Jong Bum Son
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gary J Whitman
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
| | - Zhan Xu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Yang
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA
| | - Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gaiane M Rauch
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, # 1473, Houston, TX, 77030, USA.
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
5
|
Li Z, Liu X, Gao Y, Lu X, Lei J. Ultrasound-based radiomics for early predicting response to neoadjuvant chemotherapy in patients with breast cancer: a systematic review with meta-analysis. LA RADIOLOGIA MEDICA 2024; 129:934-944. [PMID: 38630147 DOI: 10.1007/s11547-024-01783-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 01/10/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVE This study aims to evaluate the diagnostic accuracy of ultrasound imaging (US)-based radiomics for the early prediction of response to neoadjuvant chemotherapy (NAC) in breast cancer patients. METHODS We comprehensively searched PubMed, Cochrane Library, Embase, and Web of Science databases up to 1 January 2023 for eligible studies. We assessed the methodological quality of the enrolled studies with Radiomics Quality Score (RQS) and the Quality Assessment of Diagnostic Accuracy Studies-2 tools. We performed meta-analyses to summarize the diagnostic efficacy of US-based radiomics in response to NAC in breast cancer patients. RESULTS Eight studies proved eligible. Eligible studies exhibited an average RQS score of 12.88 (35.8% of the total score), with the RQS score ranging from 8 to 19. In the meta-analyses, the pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio were 0.87 (95% CI 0.81-0.92), 0.78 (95% CI 0.72-0.83), 4.02 (95% CI 3.18-5.08), 0.16 (95% CI 0.10-0.25), and 25.17 (95% CI 15.10-41.95), respectively. Results from subgroup analyses indicated that prospective studies apparently exhibited more optimal sensitivity than retrospective studies. Sensitivity analyses exhibited similar results to the primary analyses. CONCLUSION US-based radiomics may be a potentially crucial adjuvant method for evaluating the response of breast cancer to NAC. Due to limited data available and low quality of eligible studies, more multicenter prospective studies with rigorous methods are required to confirm our findings.
Collapse
Affiliation(s)
- Zhifan Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Xinran Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Ya Gao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Xingru Lu
- Department of Radiology, the First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Junqiang Lei
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China.
- Department of Radiology, the First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
6
|
Yu JB, DeStephano DM, Jeffers B, Horowitz DP, Soulos PR, Gross CP, Cheng SK. Updated Analysis of Comparative Toxicity of Proton and Photon Radiation for Prostate Cancer. J Clin Oncol 2024; 42:1943-1952. [PMID: 38507655 DOI: 10.1200/jco.23.01604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/30/2023] [Accepted: 01/17/2024] [Indexed: 03/22/2024] Open
Abstract
PURPOSE Previous comparative effectiveness studies have not demonstrated a benefit of proton beam therapy (PBT) compared with intensity-modulated radiation therapy (IMRT) for prostate cancer. An updated comparison of GI and genitourinary (GU) toxicity is needed. METHODS We investigated the SEER-Medicare linked database, identifying patients with localized prostate cancer diagnosed from 2010 to 2017. Procedure and diagnosis codes indicative of treatment-related toxicity were identified. As a sensitivity analysis, we also identified toxicity based only on procedure codes. Patients who underwent IMRT and PBT were matched 2:1 on the basis of clinical and sociodemographic characteristics. We then compared GI and GU toxicity at 6, 12, and 24 months after treatment. RESULTS The final sample included 772 PBT patients matched to 1,544 IMRT patients. The frequency of GI toxicity for IMRT versus PBT was 3.5% versus 2.5% at 6 months (P = .18), 9.5% versus 10.2% at 12 months (P = .18), and 20.5% versus 23.4% at 24 months (P = .11). The frequency of only procedure codes indicative of GI toxicity for IMRT versus PBT was too low to be reported and not significantly different. The frequency of GU toxicity for IMRT versus PBT was 6.8% versus 5.7% (P = .30), 14.3% versus 12.2% (P = .13), and 28.2% versus 25.8% (P = .21) at 6, 12, and 24 months, respectively. When looking only at procedure codes, the frequency of GU toxicity for IMRT was 1.0% at 6 months, whereas it was too infrequent to report for PBT (P = .64). GU toxicity for IMRT versus PBT was 3.3% versus 2.1% (P = .10), and 8.7% versus 6.7% (P = .10) at 12 and 24 months, respectively. CONCLUSION In this observational study, there were no statistically significant differences between PBT and IMRT in terms of GI or GU toxicity.
Collapse
Affiliation(s)
- James B Yu
- Smilow Cancer Center at St Francis Hospital, Hartford, CT
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center at Yale, New Haven, CT
| | - David M DeStephano
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Brian Jeffers
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - David P Horowitz
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Pamela R Soulos
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center at Yale, New Haven, CT
| | - Cary P Gross
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center at Yale, New Haven, CT
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Simon K Cheng
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| |
Collapse
|
7
|
Lo Gullo R, Marcus E, Huayanay J, Eskreis-Winkler S, Thakur S, Teuwen J, Pinker K. Artificial Intelligence-Enhanced Breast MRI: Applications in Breast Cancer Primary Treatment Response Assessment and Prediction. Invest Radiol 2024; 59:230-242. [PMID: 37493391 PMCID: PMC10818006 DOI: 10.1097/rli.0000000000001010] [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] [Indexed: 07/27/2023]
Abstract
ABSTRACT Primary systemic therapy (PST) is the treatment of choice in patients with locally advanced breast cancer and is nowadays also often used in patients with early-stage breast cancer. Although imaging remains pivotal to assess response to PST accurately, the use of imaging to predict response to PST has the potential to not only better prognostication but also allow the de-escalation or omission of potentially toxic treatment with undesirable adverse effects, the accelerated implementation of new targeted therapies, and the mitigation of surgical delays in selected patients. In response to the limited ability of radiologists to predict response to PST via qualitative, subjective assessments of tumors on magnetic resonance imaging (MRI), artificial intelligence-enhanced MRI with classical machine learning, and in more recent times, deep learning, have been used with promising results to predict response, both before the start of PST and in the early stages of treatment. This review provides an overview of the current applications of artificial intelligence to MRI in assessing and predicting response to PST, and discusses the challenges and limitations of their clinical implementation.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| | - Eric Marcus
- AI for Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jorge Huayanay
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
- Department of Radiology, National Institute of Neoplastic Diseases, Lima, Peru
| | - Sarah Eskreis-Winkler
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| | - Sunitha Thakur
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jonas Teuwen
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
- AI for Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| |
Collapse
|
8
|
Xu N, Guo X, Ouyang Z, Ran F, Li Q, Duan X, Zhu Y, Niu X, Liao C, Yang J. Multiparametric MRI-based radiomics combined with pathomics features for prediction of the efficacy of neoadjuvant chemotherapy in breast cancer. Heliyon 2024; 10:e24371. [PMID: 38298695 PMCID: PMC10827766 DOI: 10.1016/j.heliyon.2024.e24371] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 12/25/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
Purpose The aim of this study is to investigate a new method that combines radiological and pathological breast cancer information to predict discrepancies in pathological responses for individualized treatment planning. We used baseline multiparametric magnetic resonance imaging and hematoxylin and eosin-stained biopsy slides to extract quantitative feature information and predict the pathological response to neoadjuvant chemotherapy in breast cancer patients. Methods We retrospectively collected data from breast cancer patients who received neoadjuvant chemotherapy in our hospital from August 2016 to January 2018; multiparametric magnetic resonance imaging (contrast-enhanced T1-weighted imaging and diffusion-weighted imaging) and whole slide image of hematoxylin and eosin-stained biopsy sections were collected. Quantitative imaging features were extracted from the multiparametric magnetic resonance imaging and the whole slide image were used to construct a radiopathomics signature model powered by machine learning methods. Models based on multiparametric magnetic resonance imaging or whole slide image alone were also constructed for comparison and referred to as the radiomics signature and pathomics signature models, respectively. Four modeling methods were used to establish prediction models. Model performances were evaluated using receiver operating characteristic curve analysis and the area under the curve, accuracy, sensitivity, specificity, positive predictive value, and negative predictive value. Results The radiopathomics signature model had favourable performance for the prediction of pathological complete response in the training set (the best value: area under the curve 0.83, accuracy 0.84, and sensitivity 0.87), and in the test set (the best value: area under the curve 0.91, accuracy 0.90, and sensitivity 0.88). In the test set, the radiopathomics signature model also significantly outperformed the radiomics signature (the best value: area under the curve 0.83, accuracy 0.64, and sensitivity 0.62), pathomics signature (the best value: area under the curve 0.60, accuracy 0.74, and sensitivity 0.62) (p > 0.05). Decision curve analysis and calibration curves confirmed the excellent performance of these prediction models in discrimination, calibration, and clinical usefulness. Conclusions The results of this study suggest that radiopathomics, the combination of both radiological information regarding the whole tumor and pathological information at the cellular level, could potentially predict discrepancies in pathological response and provide evidence for rational treatment plans.
Collapse
Affiliation(s)
- Nan Xu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital/Center. No. 519 Kunzhou Road, Xishan District, Kunming 650118, Yunnan, PR China
| | - Xiaobin Guo
- Department of Radiology, Fuwai Central China Cardiovascular Hospital, Fuwai Road, Zhengzhou, Henan, 461464, PR China
| | - Zhiqiang Ouyang
- Department of Radiology, Kunming Yan’an Hospital (Yan’an Hospital Affiliated to Kunming Medical University), Kunming, PR China
| | - Fengming Ran
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital/Center. No. 519 Kunzhou Road, Xishan District, Kunming 650118, Yunnan, PR China
| | - Qinqing Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital/Center. No. 519 Kunzhou Road, Xishan District, Kunming 650118, Yunnan, PR China
| | - Xirui Duan
- Department of Radiology, Kunming Yan’an Hospital (Yan’an Hospital Affiliated to Kunming Medical University), Kunming, PR China
| | - Yu Zhu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital/Center. No. 519 Kunzhou Road, Xishan District, Kunming 650118, Yunnan, PR China
| | - Xiaofeng Niu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital/Center. No. 519 Kunzhou Road, Xishan District, Kunming 650118, Yunnan, PR China
| | - Chengde Liao
- Department of Radiology, Kunming Yan’an Hospital (Yan’an Hospital Affiliated to Kunming Medical University), Kunming, PR China
| | - Jun Yang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital/Center. No. 519 Kunzhou Road, Xishan District, Kunming 650118, Yunnan, PR China
| |
Collapse
|
9
|
Campana A, Gandomkar Z, Giannotti N, Reed W. The use of radiomics in magnetic resonance imaging for the pre-treatment characterisation of breast cancers: A scoping review. J Med Radiat Sci 2023; 70:462-478. [PMID: 37534540 PMCID: PMC10715343 DOI: 10.1002/jmrs.709] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023] Open
Abstract
Radiomics is an emerging field that aims to extract and analyse a comprehensive set of quantitative features from medical images. This scoping review is focused on MRI-based radiomic features for the molecular profiling of breast tumours and the implications of this work for predicting patient outcomes. A thorough systematic literature search and outcome extraction were performed to identify relevant studies published in MEDLINE/PubMed (National Centre for Biotechnology Information), EMBASE and Scopus from 2015 onwards. The following information was retrieved from each article: study purpose, study design, extracted radiomic features, machine learning technique(s), sample size/characteristics, statistical result(s) and implications on patient outcomes. Based on the study purpose, four key themes were identified in the included 63 studies: tumour subtype classification (n = 35), pathologically complete response (pCR) prediction (n = 15), lymph node metastasis (LNM) detection (n = 7) and recurrence rate prediction (n = 6). In all four themes, reported accuracies widely varied among the studies, for example, area under receiver characteristics curve (AUC) for detecting LNM ranged from 0.72 to 0.91 and the AUC for predicting pCR ranged from 0.71 to 0.99. In all four themes, combining radiomic features with clinical data improved the predictive models. Preliminary results of this study showed radiomics potential to characterise the whole tumour heterogeneity, with clear implications for individual-targeted treatment. However, radiomics is still in the pre-clinical phase, currently with an insufficient number of large multicentre studies and those existing studies are often limited by insufficient methodological transparency and standardised workflow. Consequently, the clinical translation of existing studies is currently limited.
Collapse
Affiliation(s)
- Annalise Campana
- Discipline of Medical Imaging Science, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Ziba Gandomkar
- Discipline of Medical Imaging Science, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Nicola Giannotti
- Discipline of Medical Imaging Science, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Warren Reed
- Discipline of Medical Imaging Science, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| |
Collapse
|
10
|
Elsayed B, Alksas A, Shehata M, Mahmoud A, Zaky M, Alghandour R, Abdelwahab K, Abdelkhalek M, Ghazal M, Contractor S, El-Din Moustafa H, El-Baz A. Exploring Neoadjuvant Chemotherapy, Predictive Models, Radiomic, and Pathological Markers in Breast Cancer: A Comprehensive Review. Cancers (Basel) 2023; 15:5288. [PMID: 37958461 PMCID: PMC10648987 DOI: 10.3390/cancers15215288] [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: 09/02/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer retains its position as the most prevalent form of malignancy among females on a global scale. The careful selection of appropriate treatment for each patient holds paramount importance in effectively managing breast cancer. Neoadjuvant chemotherapy (NACT) plays a pivotal role in the comprehensive treatment of this disease. Administering chemotherapy before surgery, NACT becomes a powerful tool in reducing tumor size, potentially enabling fewer invasive surgical procedures and even rendering initially inoperable tumors amenable to surgery. However, a significant challenge lies in the varying responses exhibited by different patients towards NACT. To address this challenge, researchers have focused on developing prediction models that can identify those who would benefit from NACT and those who would not. Such models have the potential to reduce treatment costs and contribute to a more efficient and accurate management of breast cancer. Therefore, this review has two objectives: first, to identify the most effective radiomic markers correlated with NACT response, and second, to explore whether integrating radiomic markers extracted from radiological images with pathological markers can enhance the predictive accuracy of NACT response. This review will delve into addressing these research questions and also shed light on the emerging research direction of leveraging artificial intelligence techniques for predicting NACT response, thereby shaping the future landscape of breast cancer treatment.
Collapse
Affiliation(s)
- Basma Elsayed
- Biomedical Engineering Program, Faculty of Engineering, Mansoura University, Mansoura 35516, Egypt;
| | - Ahmed Alksas
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA; (A.A.); (M.S.); (A.M.)
| | - Mohamed Shehata
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA; (A.A.); (M.S.); (A.M.)
| | - Ali Mahmoud
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA; (A.A.); (M.S.); (A.M.)
| | - Mona Zaky
- Diagnostic Radiology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Reham Alghandour
- Medical Oncology Department, Mansoura Oncology Center, Mansoura University, Mansoura 35516, Egypt;
| | - Khaled Abdelwahab
- Surgical Oncology Department, Mansoura Oncology Center, Mansoura University, Mansoura 35516, Egypt; (K.A.); (M.A.)
| | - Mohamed Abdelkhalek
- Surgical Oncology Department, Mansoura Oncology Center, Mansoura University, Mansoura 35516, Egypt; (K.A.); (M.A.)
| | - Mohammed Ghazal
- Electrical, Computer, and Biomedical Engineering Department, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates;
| | - Sohail Contractor
- Department of Radiology, University of Louisville, Louisville, KY 40202, USA;
| | | | - Ayman El-Baz
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA; (A.A.); (M.S.); (A.M.)
| |
Collapse
|
11
|
Huang JX, Shi J, Ding SS, Zhang HL, Wang XY, Lin SY, Xu YF, Wei MJ, Liu LZ, Pei XQ. Deep Learning Model Based on Dual-Modal Ultrasound and Molecular Data for Predicting Response to Neoadjuvant Chemotherapy in Breast Cancer. Acad Radiol 2023; 30 Suppl 2:S50-S61. [PMID: 37270368 DOI: 10.1016/j.acra.2023.03.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 06/05/2023]
Abstract
RATIONALE AND OBJECTIVES To carry out radiomics analysis/deep convolutional neural network (CNN) based on B-mode ultrasound (BUS) and shear wave elastography (SWE) to predict response to neoadjuvant chemotherapy (NAC) in breast cancer patients. MATERIALS AND METHODS In this prospective study, 255 breast cancer patients who received NAC between September 2016 and December 2021 were included. Radiomics models were designed using a support vector machine classifier based on US images obtained before treatment, including BUS and SWE. And CNN models also were developed using ResNet architecture. The final predictive model was developed by combining the dual-modal US and independently associated clinicopathologic characteristics. The predictive performances of the models were assessed with five-fold cross-validation. RESULTS Pretreatment SWE performed better than BUS in predicting the response to NAC for breast cancer for both the CNN and radiomics models (P < 0.001). The predictive results of the CNN models were significantly better than the radiomics models, with AUCs of 0.72 versus 0.69 for BUS and 0.80 versus 0.77 for SWE, respectively (P = 0.003). The CNN model based on the dual-modal US and molecular data exhibited outstanding performance in predicting NAC response, with an accuracy of 83.60% ± 2.63%, a sensitivity of 87.76% ± 6.44%, and a specificity of 77.45% ± 4.38%. CONCLUSION The pretreatment CNN model based on the dual-modal US and molecular data achieved excellent performance for predicting the response to chemotherapy in breast cancer. Therefore, this model has the potential to serve as a non-invasive objective biomarker to predict NAC response and aid clinicians with individual treatments.
Collapse
Affiliation(s)
- Jia-Xin Huang
- Department of Medical Ultrasound, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China (J.-X.H., X.-Y.W., Y.-F.X., M.-J.W., L.-Z.L., X.-Q.P.)
| | - Jun Shi
- School of Communication and Information Engineering, Shanghai University, Shanghai 200444, China (J.S., S.-S.D., H.-L.Z.)
| | - Sai-Sai Ding
- School of Communication and Information Engineering, Shanghai University, Shanghai 200444, China (J.S., S.-S.D., H.-L.Z.)
| | - Hui-Li Zhang
- School of Communication and Information Engineering, Shanghai University, Shanghai 200444, China (J.S., S.-S.D., H.-L.Z.)
| | - Xue-Yan Wang
- Department of Medical Ultrasound, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China (J.-X.H., X.-Y.W., Y.-F.X., M.-J.W., L.-Z.L., X.-Q.P.)
| | - Shi-Yang Lin
- Department of Medical Ultrasound, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China (S.-Y.L.)
| | - Yan-Fen Xu
- Department of Medical Ultrasound, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China (J.-X.H., X.-Y.W., Y.-F.X., M.-J.W., L.-Z.L., X.-Q.P.)
| | - Ming-Jie Wei
- Department of Medical Ultrasound, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China (J.-X.H., X.-Y.W., Y.-F.X., M.-J.W., L.-Z.L., X.-Q.P.)
| | - Long-Zhong Liu
- Department of Medical Ultrasound, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China (J.-X.H., X.-Y.W., Y.-F.X., M.-J.W., L.-Z.L., X.-Q.P.)
| | - Xiao-Qing Pei
- Department of Medical Ultrasound, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China (J.-X.H., X.-Y.W., Y.-F.X., M.-J.W., L.-Z.L., X.-Q.P.).
| |
Collapse
|
12
|
Chen Z, Huang M, Lyu J, Qi X, He F, Li X. Machine learning for predicting breast-conserving surgery candidates after neoadjuvant chemotherapy based on DCE-MRI. Front Oncol 2023; 13:1174843. [PMID: 37621690 PMCID: PMC10446166 DOI: 10.3389/fonc.2023.1174843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023] Open
Abstract
Purpose This study aimed to investigate a machine learning method for predicting breast-conserving surgery (BCS) candidates, from patients who received neoadjuvant chemotherapy (NAC) by using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) obtained before and after NAC. Materials and methods This retrospective study included 75 patients who underwent NAC and breast surgery. First, 3,390 features were comprehensively extracted from pre- and post-NAC DCE-MRIs. Then patients were then divided into two groups: type 1, patients with pathologic complete response (pCR) and single lesion shrinkage; type 2, major residual lesion with satellite foci, multifocal residual, stable disease (SD), and progressive disease (PD). The logistic regression (LR) was used to build prediction models to identify the two groups. Prediction performance was assessed using the area under the curve (AUC), accuracy, sensitivity, and specificity. Results Radiomics features were significantly related to breast cancer shrinkage after NAC. The combination model achieved an AUC of 0.82, and the pre-NAC model was 0.64, the post-NAC model was 0.70, and the pre-post-NAC model was 0.80. In the combination model, 15 features, including nine wavelet-based features, four Laplacian-of-Gauss (LoG) features, and two original features, were filtered. Among these selected were four features from pre-NAC DCE-MRI, six were from post-NAC DCE-MRI, and five were from pre-post-NAC features. Conclusion The model combined with pre- and post-NAC DCE-MRI can effectively predict candidates to undergo BCS and provide AI-based decision support for clinicians with ensured safety. High-order (LoG- and wavelet-based) features play an important role in our machine learning model. The features from pre-post-NAC DCE-MRI had better predictive performance.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiang Li
- Department of Radiology, the Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
13
|
Wang X, Hua H, Han J, Zhong X, Liu J, Chen J. Evaluation of Multiparametric MRI Radiomics-Based Nomogram in Prediction of Response to Neoadjuvant Chemotherapy in Breast Cancer: A Two-Center study. Clin Breast Cancer 2023:S1526-8209(23)00134-9. [PMID: 37321954 DOI: 10.1016/j.clbc.2023.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 06/17/2023]
Abstract
INTRODUCTION This study evaluated the performance of primary foci of breast cancer on multiparametric magnetic resonance imaging (MRI) contributing to establish and validate radiomics-based nomograms for predicting the different pathological outcome of breast cancer patients after neoadjuvant chemotherapy (NAC). MATERIALS AND METHODS Retrospectively collected 387 patients with locally advanced breast cancer, all treated with NAC and received breast dynamic contrast-enhanced MRI (DCE-MRI) before NAC. Radiomics signatures were extracted from region of interest (ROI) on multiparametric MRI to build rad score. Clinical-pathologic data and radiological features established the clinical model. The comprehensive model featured rad-score, predictive clinical-pathologic data and radiological features, which was ultimately displayed as a nomogram. Patients were grouped in 2 different ways in accordance with the Miller-Payne (MP) grading of surgical specimens. The first grouping method: 181 patients with pathological reaction grades Ⅳ∼Ⅴ were included in the significant remission group, while 206 patients with pathological reaction grades Ⅰ∼Ⅲ were included in the nonsignificant remission group. The second grouping method: 117 patients with pathological complete response (pCR) were assigned to the pCR group, and 270 patients who failed to meet pCR were assigned to in the non-pCR group. Two combined nomograms are created from 2 grouped data for predicting different pathological responses to NAC. The area under the curves (AUC) of the receiver operating characteristic curves (ROC) were used to evaluate the performance of each model. While decision curve analysis (DCA) and calibration curves were used for estimating the clinical application value of the nomogram. RESULTS Two combined nomograms embodying rad score and clinical-pathologic data outperformed, showing good calibrations for predicting response to NAC. The combined nomogram predicting pCR showed the best performance with the AUC values of 0.97, 0.90 and 0.86 in the training, testing, and external validation cohorts respectively. The AUC values of another combined nomogram predicting significant remission: 0.98, 0.88 0.80 in the training, testing and external validation cohorts. DCA showed the comprehensive model nomogram obtained the most clinical benefit. CONCLUSIONS The combined nomogram could preoperatively predict significant remission or even pCR to NAC in breast cancer based on multiparametric MRI and clinical-pathologic data.
Collapse
Affiliation(s)
- Xiaolin Wang
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Hua
- Department of Thyroid Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junqi Han
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xin Zhong
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Liu
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Chen
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
14
|
Bouhamama A, Leporq B, Faraz K, Foy JP, Boussageon M, Pérol M, Ortiz-Cuaran S, Ghiringhelli F, Saintigny P, Beuf O, Pilleul F. Radiomics combined with transcriptomics to predict response to immunotherapy from patients treated with PD-1/PD-L1 inhibitors for advanced NSCLC. FRONTIERS IN RADIOLOGY 2023; 3:1168448. [PMID: 37492391 PMCID: PMC10365090 DOI: 10.3389/fradi.2023.1168448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/31/2023] [Indexed: 07/27/2023]
Abstract
Introduction In this study, we aim to build radiomics and multiomics models based on transcriptomics and radiomics to predict the response from patients treated with the PD-L1 inhibitor. Materials and methods One hundred and ninety-five patients treated with PD-1/PD-L1 inhibitors were included. For all patients, 342 radiomic features were extracted from pretreatment computed tomography scans. The training set was built with 110 patients treated at the Léon Bérard Cancer Center. An independent validation cohort was built with the 85 patients treated in Dijon. The two sets were dichotomized into two classes, patients with disease control and those considered non-responders, in order to predict the disease control at 3 months. Various models were trained with different feature selection methods, and different classifiers were evaluated to build the models. In a second exploratory step, we used transcriptomics to enrich the database and develop a multiomic signature of response to immunotherapy in a 54-patient subgroup. Finally, we considered the HOT/COLD status. We first trained a radiomic model to predict the HOT/COLD status and then prototyped a hybrid model integrating radiomics and the HOT/COLD status to predict the response to immunotherapy. Results Radiomic signature for 3 months' progression-free survival (PFS) classification: The most predictive model had an area under the receiver operating characteristic curve (AUROC) of 0.94 on the training set and 0.65 on the external validation set. This model was obtained with the t-test selection method and with a support vector machine (SVM) classifier. Multiomic signature for PFS classification: The most predictive model had an AUROC of 0.95 on the training set and 0.99 on the validation set. Radiomic model to predict the HOT/COLD status: the most predictive model had an AUROC of 0.93 on the training set and 0.86 on the validation set. HOT/COLD radiomic hybrid model for PFS classification: the most predictive model had an AUROC of 0.93 on the training set and 0.90 on the validation set. Conclusion In conclusion, radiomics could be used to predict the response to immunotherapy in non-small-cell lung cancer patients. The use of transcriptomics or the HOT/COLD status, together with radiomics, may improve the working of the prediction models.
Collapse
Affiliation(s)
- Amine Bouhamama
- Department of Radiology, Centre Léon Bérard, Lyon, France
- Creatis, University Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Creatis, UMR 5220, U1206, Lyon, France
| | - Benjamin Leporq
- Creatis, University Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Creatis, UMR 5220, U1206, Lyon, France
| | - Khuram Faraz
- Creatis, University Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Creatis, UMR 5220, U1206, Lyon, France
| | - Jean-Philippe Foy
- Department of Oral and Maxillofacial Surgery, Sorbonne Université, Pitié-Salpêtrière Hospital, APHP, Paris, France
| | | | - Maurice Pérol
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Sandra Ortiz-Cuaran
- CRCL, University Lyon, Claude Bernard Lyon 1 University, Inserm 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | | | - Pierre Saintigny
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
- CRCL, University Lyon, Claude Bernard Lyon 1 University, Inserm 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Olivier Beuf
- Creatis, University Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Creatis, UMR 5220, U1206, Lyon, France
| | - Frank Pilleul
- Department of Radiology, Centre Léon Bérard, Lyon, France
- Creatis, University Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Creatis, UMR 5220, U1206, Lyon, France
| |
Collapse
|
15
|
Duan Y, Yang G, Miao W, Song B, Wang Y, Yan L, Wu F, Zhang R, Mao Y, Wang Z. Computed Tomography-Based Radiomics Analysis for Prediction of Response to Neoadjuvant Chemotherapy in Breast Cancer Patients. J Comput Assist Tomogr 2023; 47:199-204. [PMID: 36790871 DOI: 10.1097/rct.0000000000001426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
PURPOSE Previous studies have pointed out that magnetic resonance- and fluorodeoxyglucose positron emission tomography-based radiomics had a high predictive value for the response of the neoadjuvant chemotherapy (NAC) in breast cancer by respectively characterizing tumor heterogeneity of the relaxation time and the glucose metabolism. However, it is unclear whether computed tomography (CT)-based radiomics based on density heterogeneity can predict the response of NAC. This study aimed to develop and validate a CT-based radiomics nomogram to predict the response of NAC in breast cancer. METHODS A total of 162 breast cancer patients (110 in the training cohort and 52 in the validation cohort) who underwent CT scans before receiving NAC and had pathological response results were retrospectively enrolled. Grades 4 to 5 cases were classified as response to NAC. According to the Miller-Payne grading system, grades 1 to 3 cases were classified as nonresponse to NAC. Radiomics features were extracted, and the optimal radiomics features were obtained to construct a radiomics signature. Multivariate logistic regression was used to develop the clinical prediction model and the radiomics nomogram that incorporated clinical characteristics and radiomics score. We assessed the performance of different models, including calibration and clinical usefulness. RESULTS Eight optimal radiomics features were obtained. Human epidermal growth factor receptor 2 status and molecular subtype showed statistical differences between the response group and the nonresponse group. The radiomics nomogram had more favorable predictive efficacy than the clinical prediction model (areas under the curve, 0.82 vs 0.70 in the training cohort; 0.79 vs 0.71 in the validation cohort). The Delong test showed that there are statistical differences between the clinical prediction model and the radiomics nomogram ( z = 2.811, P = 0.005 in the training cohort). The decision curve analysis showed that the radiomics nomogram had higher overall net benefit than the clinical prediction model. CONCLUSION The radiomics nomogram based on CT radiomics signature and clinical characteristics has favorable predictive efficacy for the response of NAC in breast cancer.
Collapse
Affiliation(s)
- Yanli Duan
- From the Departments of Nuclear Medicine
| | | | | | - Bingxue Song
- Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong
| | | | - Lei Yan
- From the Departments of Nuclear Medicine
| | - Fengyu Wu
- From the Departments of Nuclear Medicine
| | - Ran Zhang
- Huiying Medical Technology Co, Ltd, Beijing
| | - Yan Mao
- Diagnosis and Treatment Centre of Breast Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | | |
Collapse
|
16
|
Pesapane F, De Marco P, Rapino A, Lombardo E, Nicosia L, Tantrige P, Rotili A, Bozzini AC, Penco S, Dominelli V, Trentin C, Ferrari F, Farina M, Meneghetti L, Latronico A, Abbate F, Origgi D, Carrafiello G, Cassano E. How Radiomics Can Improve Breast Cancer Diagnosis and Treatment. J Clin Med 2023; 12:jcm12041372. [PMID: 36835908 PMCID: PMC9963325 DOI: 10.3390/jcm12041372] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Recent technological advances in the field of artificial intelligence hold promise in addressing medical challenges in breast cancer care, such as early diagnosis, cancer subtype determination and molecular profiling, prediction of lymph node metastases, and prognostication of treatment response and probability of recurrence. Radiomics is a quantitative approach to medical imaging, which aims to enhance the existing data available to clinicians by means of advanced mathematical analysis using artificial intelligence. Various published studies from different fields in imaging have highlighted the potential of radiomics to enhance clinical decision making. In this review, we describe the evolution of AI in breast imaging and its frontiers, focusing on handcrafted and deep learning radiomics. We present a typical workflow of a radiomics analysis and a practical "how-to" guide. Finally, we summarize the methodology and implementation of radiomics in breast cancer, based on the most recent scientific literature to help researchers and clinicians gain fundamental knowledge of this emerging technology. Alongside this, we discuss the current limitations of radiomics and challenges of integration into clinical practice with conceptual consistency, data curation, technical reproducibility, adequate accuracy, and clinical translation. The incorporation of radiomics with clinical, histopathological, and genomic information will enable physicians to move forward to a higher level of personalized management of patients with breast cancer.
Collapse
Affiliation(s)
- Filippo Pesapane
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
- Correspondence: ; Tel.: +39-02-574891
| | - Paolo De Marco
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Anna Rapino
- Postgraduation School in Radiodiagnostics, University of Milan, 20122 Milan, Italy
| | - Eleonora Lombardo
- UOC of Diagnostic Imaging, Policlinico Tor Vergata University, 00133 Rome, Italy
| | - Luca Nicosia
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Priyan Tantrige
- Department of Radiology, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Anna Rotili
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Anna Carla Bozzini
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Silvia Penco
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Valeria Dominelli
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Chiara Trentin
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Federica Ferrari
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Mariagiorgia Farina
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenza Meneghetti
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Antuono Latronico
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Francesca Abbate
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Gianpaolo Carrafiello
- Department of Radiology, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Health Sciences, University of Milan, 20122 Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
17
|
Rossi EMC, Invento A, Pesapane F, Pagan E, Bagnardi V, Fusco N, Venetis K, Dominelli V, Trentin C, Cassano E, Gilardi L, Mazza M, Lazzeroni M, De Lorenzi F, Caldarella P, De Scalzi A, Girardi A, Sangalli C, Alberti L, Sacchini V, Galimberti V, Veronesi P. Diagnostic performance of image-guided vacuum-assisted breast biopsy after neoadjuvant therapy for breast cancer: prospective pilot study. Br J Surg 2023; 110:217-224. [PMID: 36477768 PMCID: PMC10364486 DOI: 10.1093/bjs/znac391] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/14/2022] [Accepted: 10/23/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Image-guided vacuum-assisted breast biopsy (VABB) of the tumour bed, performed after neoadjuvant therapy, is increasingly being used to assess residual cancer and to potentially identify to identify pathological complete response (pCR). In this study, the accuracy of preoperative VABB specimens was assessed and compared with surgical specimens in patients with triple-negative or human epidermal growth factor receptor 2 (HER2)-positive invasive ductal breast cancer after neoadjuvant therapy. As a secondary endpoint, the performance of contrast-enhanced MRI of the breast and PET-CT for response prediction was assessed. METHODS This single-institution prospective pilot study enrolled patients from April 2018 to April 2021 with a complete response on imaging (iCR) who subsequently underwent VABB before surgery. Those with a pCR at VABB were included in the primary analysis of the accuracy of VABB. The performance of imaging (MRI and PET-CT) was analysed for prediction of a pCR considering both patients with an iCR and those with residual disease at postneoadjuvant therapy imaging. RESULTS Twenty patients were included in the primary analysis. The median age was 44 (range 35-51) years. At surgery, 18 of 20 patients showed a complete response (accuracy 90 (95 per cent exact c.i. 68 to 99) per cent). Only two patients showed residual ductal intraepithelial neoplasia of grade 2 and 3 respectively. In the secondary analysis, accuracy was similar for MRI and PET-CT (77 versus 78 per cent; P = 0.76). CONCLUSION VABB in patients with an iCR might be a promising method to select patients for de-escalation of surgical treatment in triple-negative or HER2-positive breast cancer. The present results support such an approach and should inform the design of future trials on de-escalation of surgery.
Collapse
Affiliation(s)
| | - Alessandra Invento
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Filippo Pesapane
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Eleonora Pagan
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO European Institute of Oncology IRCSS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO European Institute of Oncology IRCSS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valeria Dominelli
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Trentin
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Laura Gilardi
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Manuelita Mazza
- Division of Medical Senology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Matteo Lazzeroni
- Division of Cancer Prevention and Genetics, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Francesca De Lorenzi
- Department of Plastic and Reconstructive Surgery, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Pietro Caldarella
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | | | - Antonia Girardi
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Claudia Sangalli
- Data Management, European Institute of Oncology IRCCS, Milan, Italy
| | - Luca Alberti
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Virgilio Sacchini
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Viviana Galimberti
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Paolo Veronesi
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
18
|
McAnena P, Moloney BM, Browne R, O’Halloran N, Walsh L, Walsh S, Sheppard D, Sweeney KJ, Kerin MJ, Lowery AJ. A radiomic model to classify response to neoadjuvant chemotherapy in breast cancer. BMC Med Imaging 2022; 22:225. [PMID: 36564734 PMCID: PMC9789647 DOI: 10.1186/s12880-022-00956-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Medical image analysis has evolved to facilitate the development of methods for high-throughput extraction of quantitative features that can potentially contribute to the diagnostic and treatment paradigm of cancer. There is a need for further improvement in the accuracy of predictive markers of response to neo-adjuvant chemotherapy (NAC). The aim of this study was to develop a radiomic classifier to enhance current approaches to predicting the response to NAC breast cancer. METHODS Data on patients treated for breast cancer with NAC prior to surgery who had a pre-NAC dynamic contrast enhanced breast MRI were included. Response to NAC was assessed using the Miller-Payne system on the excised tumor. Tumor segmentation was carried out manually under the supervision of a consultant breast radiologist. Features were selected using least absolute shrinkage selection operator regression. A support vector machine learning model was used to classify response to NAC. RESULTS 74 patients were included. Patients were classified as having a poor response to NAC (reduction in cellularity < 90%, n = 44) and an excellent response (> 90% reduction in cellularity, n = 30). 4 radiomics features (discretized kurtosis, NGDLM contrast, GLZLM_SZE and GLZLM_ZP) were identified as pertinent predictors of response to NAC. A SVM model using these features stratified patients into poor and excellent response groups producing an AUC of 0.75. Addition of estrogen receptor status improved the accuracy of the model with an AUC of 0.811. CONCLUSION This study identified a radiomic classifier incorporating 4 radiomics features to augment subtype based classification of response to NAC in breast cancer.
Collapse
Affiliation(s)
- Peter McAnena
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland
| | - Brian M. Moloney
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Robert Browne
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland
| | - Niamh O’Halloran
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Leon Walsh
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Sinead Walsh
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Declan Sheppard
- grid.412440.70000 0004 0617 9371Department of Radiology, University Hospital Galway, Galway, Ireland
| | - Karl J. Sweeney
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland
| | - Michael J. Kerin
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland ,grid.6142.10000 0004 0488 0789Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| | - Aoife J. Lowery
- grid.412440.70000 0004 0617 9371Department of Surgery, Clinical Sciences Institute, University Hospital Galway, Galway, Ireland ,grid.6142.10000 0004 0488 0789Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| |
Collapse
|
19
|
Jeon SH, Kim SW, Na K, Seo M, Sohn YM, Lim YJ. Radiomic models based on magnetic resonance imaging predict the spatial distribution of CD8 + tumor-infiltrating lymphocytes in breast cancer. Front Immunol 2022; 13:1080048. [PMID: 36601118 PMCID: PMC9806253 DOI: 10.3389/fimmu.2022.1080048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Infiltration of CD8+ T cells and their spatial contexture, represented by immunophenotype, predict the prognosis and therapeutic response in breast cancer. However, a non-surgical method using radiomics to evaluate breast cancer immunophenotype has not been explored. Here, we assessed the CD8+ T cell-based immunophenotype in patients with breast cancer undergoing upfront surgery (n = 182). We extracted radiomic features from the four phases of dynamic contrast-enhanced magnetic resonance imaging, and randomly divided the patients into training (n = 137) and validation (n = 45) cohorts. For predicting the immunophenotypes, radiomic models (RMs) that combined the four phases demonstrated superior performance to those derived from a single phase. For discriminating the inflamed tumor from the non-inflamed tumor, the feature-based combination model from the whole tumor (RM-wholeFC) showed high performance in both training (area under the receiver operating characteristic curve [AUC] = 0.973) and validation cohorts (AUC = 0.985). Similarly, the feature-based combination model from the peripheral tumor (RM-periFC) discriminated between immune-desert and excluded tumors with high performance in both training (AUC = 0.993) and validation cohorts (AUC = 0.984). Both RM-wholeFC and RM-periFC demonstrated good to excellent performance for every molecular subtype. Furthermore, in patients who underwent neoadjuvant chemotherapy (n = 64), pre-treatment images showed that tumors exhibiting complete response to neoadjuvant chemotherapy had significantly higher scores from RM-wholeFC and lower scores from RM-periFC. Our RMs predicted the immunophenotype of breast cancer based on the spatial distribution of CD8+ T cells with high accuracy. This approach can be used to stratify patients non-invasively based on the status of the tumor-immune microenvironment.
Collapse
Affiliation(s)
- Seung Hyuck Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - So-Woon Kim
- Department of Pathology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Kiyong Na
- Department of Pathology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Mirinae Seo
- Department of Radiology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Yu-Mee Sohn
- Department of Radiology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Yu Jin Lim
- Department of Radiation Oncology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea,*Correspondence: Yu Jin Lim,
| |
Collapse
|
20
|
O'Donnell J, Gasior S, Davey M, O'Malley E, Lowery A, McGarry J, O'Connell A, Kerin M, McCarthy P. The accuracy of breast MRI radiomic methodologies in predicting pathological complete response to neoadjuvant chemotherapy: A systematic review and network meta-analysis. Eur J Radiol 2022; 157:110561. [DOI: 10.1016/j.ejrad.2022.110561] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/13/2022] [Accepted: 10/11/2022] [Indexed: 11/03/2022]
|
21
|
Radiomic and Volumetric Measurements as Clinical Trial Endpoints—A Comprehensive Review. Cancers (Basel) 2022; 14:cancers14205076. [PMID: 36291865 PMCID: PMC9599928 DOI: 10.3390/cancers14205076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary The extraction of quantitative data from standard-of-care imaging modalities offers opportunities to improve the relevance and salience of imaging biomarkers used in drug development. This review aims to identify the challenges and opportunities for discovering new imaging-based biomarkers based on radiomic and volumetric assessment in the single-site solid tumor sites: breast cancer, rectal cancer, lung cancer and glioblastoma. Developing approaches to harmonize three essential areas: segmentation, validation and data sharing may expedite regulatory approval and adoption of novel cancer imaging biomarkers. Abstract Clinical trials for oncology drug development have long relied on surrogate outcome biomarkers that assess changes in tumor burden to accelerate drug registration (i.e., Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1) criteria). Drug-induced reduction in tumor size represents an imperfect surrogate marker for drug activity and yet a radiologically determined objective response rate is a widely used endpoint for Phase 2 trials. With the addition of therapies targeting complex biological systems such as immune system and DNA damage repair pathways, incorporation of integrative response and outcome biomarkers may add more predictive value. We performed a review of the relevant literature in four representative tumor types (breast cancer, rectal cancer, lung cancer and glioblastoma) to assess the preparedness of volumetric and radiomics metrics as clinical trial endpoints. We identified three key areas—segmentation, validation and data sharing strategies—where concerted efforts are required to enable progress of volumetric- and radiomics-based clinical trial endpoints for wider clinical implementation.
Collapse
|
22
|
Li C, Li W, Liu C, Zheng H, Cai J, Wang S. Artificial intelligence in multi-parametric magnetic resonance imaging: A review. Med Phys 2022; 49:e1024-e1054. [PMID: 35980348 DOI: 10.1002/mp.15936] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/06/2022] Open
Abstract
Multi-parametric magnetic resonance imaging (mpMRI) is an indispensable tool in the clinical workflow for the diagnosis and treatment planning of various diseases. Machine learning-based artificial intelligence (AI) methods, especially those adopting the deep learning technique, have been extensively employed to perform mpMRI image classification, segmentation, registration, detection, reconstruction, and super-resolution. The current availability of increasing computational power and fast-improving AI algorithms have empowered numerous computer-based systems for applying mpMRI to disease diagnosis, imaging-guided radiotherapy, patient risk and overall survival time prediction, and the development of advanced quantitative imaging technology for magnetic resonance fingerprinting. However, the wide application of these developed systems in the clinic is still limited by a number of factors, including robustness, reliability, and interpretability. This survey aims to provide an overview for new researchers in the field as well as radiologists with the hope that they can understand the general concepts, main application scenarios, and remaining challenges of AI in mpMRI. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Cheng Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wen Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Chenyang Liu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Shanshan Wang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,Peng Cheng Laboratory, Shenzhen, 518066, China.,Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| |
Collapse
|
23
|
Gu J, Jiang T. Ultrasound radiomics in personalized breast management: Current status and future prospects. Front Oncol 2022; 12:963612. [PMID: 36059645 PMCID: PMC9428828 DOI: 10.3389/fonc.2022.963612] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
Breast cancer is the most common cancer in women worldwide. Providing accurate and efficient diagnosis, risk stratification and timely adjustment of treatment strategies are essential steps in achieving precision medicine before, during and after treatment. Radiomics provides image information that cannot be recognized by the naked eye through deep mining of medical images. Several studies have shown that radiomics, as a second reader of medical images, can assist physicians not only in the detection and diagnosis of breast lesions but also in the assessment of risk stratification and prediction of treatment response. Recently, more and more studies have focused on the application of ultrasound radiomics in breast management. We summarized recent research advances in ultrasound radiomics for the diagnosis of benign and malignant breast lesions, prediction of molecular subtype, assessment of lymph node status, prediction of neoadjuvant chemotherapy response, and prediction of survival. In addition, we discuss the current challenges and future prospects of ultrasound radiomics.
Collapse
Affiliation(s)
- Jionghui Gu
- Department of Ultrasound, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
| | - Tian'an Jiang
- Department of Ultrasound, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- *Correspondence: Tian'an Jiang,
| |
Collapse
|
24
|
Pesapane F, Agazzi GM, Rotili A, Ferrari F, Cardillo A, Penco S, Dominelli V, D'Ecclesiis O, Vignati S, Raimondi S, Bozzini A, Pizzamiglio M, Petralia G, Nicosia L, Cassano E. Prediction of the Pathological Response to Neoadjuvant Chemotherapy in Breast Cancer Patients With MRI-Radiomics: A Systematic Review and Meta-analysis. Curr Probl Cancer 2022; 46:100883. [PMID: 35914383 DOI: 10.1016/j.currproblcancer.2022.100883] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/30/2022]
Abstract
We performed a systematic review and a meta-analysis of studies using MRI-radiomics for predicting the pathological complete response in breast cancer patients undergoing neoadjuvant therapy , and we evaluated their methodological quality using the radiomics-quality-score (RQS). Random effects meta-analysis was performed pooling area under the receiver operating characteristics curves. Publication-bias was assessed using the Egger's test and visually inspecting the funnel plot. Forty-three studies were included in the qualitative review and 34 in the meta-analysis. Summary area under the receiver operating characteristics curve was 0,78 (95%CI:0,74-0,81). Heterogeneity according to the I2 statistic was substantial (71%) and there was no evidence of publication bias (P-value = 0,2). The average RQS was 12,7 (range:-1-26), with an intra-class correlation coefficient of 0.93 (95%CI:0.61-0.97). Year of publication, field intensity and synthetic RQS score do not appear to be moderators of the effect (P-value = 0.36, P-value = 0.28 and P-value = 0.92, respectively). MRI-radiomics may predict response to neoadjuvant therapy in breast cancer patients but the heterogeneity of the current studies is still substantial.
Collapse
Affiliation(s)
- Filippo Pesapane
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | | | - Anna Rotili
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Ferrari
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Andrea Cardillo
- Radiology Department, Università degli studi di Torino, Turin, Italy
| | - Silvia Penco
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Valeria Dominelli
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Oriana D'Ecclesiis
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Silvano Vignati
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Raimondi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Anna Bozzini
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Pizzamiglio
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Petralia
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy; Division of Radiology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Luca Nicosia
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
25
|
Zhu X, Shen J, Zhang H, Wang X, Zhang H, Yu J, Zhang Q, Song D, Guo L, Zhang D, Zhu R, Wu J. A Novel Combined Nomogram Model for Predicting the Pathological Complete Response to Neoadjuvant Chemotherapy in Invasive Breast Carcinoma of No Specific Type: Real-World Study. Front Oncol 2022; 12:916526. [PMID: 35734603 PMCID: PMC9207207 DOI: 10.3389/fonc.2022.916526] [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: 04/09/2022] [Accepted: 05/02/2022] [Indexed: 12/03/2022] Open
Abstract
Objective To explore the value of a predictive model combining the multiparametric magnetic resonance imaging (mpMRI) radiomics score (RAD-score), clinicopathologic features, and morphologic features for the pathological complete response (pCR) to neoadjuvant chemotherapy (NAC) in invasive breast carcinoma of no specific type (IBC-NST). Methods We enrolled, retrospectively and consecutively, 206 women with IBC-NST who underwent surgery after NAC and obtained pathological results from August 2018 to October 2021. Four RAD-scores were constructed for predicting the pCR based on fat-suppression T2-weighted imaging (FS-T2WI), diffusion-weighted imaging (DWI), contrast-enhanced T1-weighted imaging (T1WI+C) and their combination, which was called mpMRI. The best RAD-score was combined with clinicopathologic and morphologic features to establish a nomogram model through binary logistic regression. The predictive performance of the nomogram was evaluated using the area under receiver operator characteristic (ROC) curve (AUC) and calibration curve. The clinical net benefit of the model was evaluated using decision curve analysis (DCA). Results The mpMRI RAD-score had the highest diagnostic performance, with AUC of 0.848 among the four RAD-scores. T stage, human epidermal growth factor receptor-2 (HER2) status, RAD-score, and roundness were independent factors for predicting the pCR (P < 0.05 for all). The combined nomogram model based on these factors achieved AUCs of 0.930 and 0.895 in the training cohort and validation cohort, respectively, higher than other models (P < 0.05 for all). The calibration curve showed that the predicted probabilities of the nomogram were in good agreement with the actual probabilities, and DCA indicated that it provided more net benefit than the treat-none or treat-all scheme by decision curve analysis in both training and validation datasets. Conclusion The combined nomogram model based on the mpMRI RAD-score combined with clinicopathologic and morphologic features may improve the predictive performance for the pCR of NAC in patients with IBC-NST.
Collapse
Affiliation(s)
- Xuelin Zhu
- Graduate School, Tianjin Medical University, Tianjin, China.,Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.,Department of Ultrasound, Qingzhou People's Hospital, Weifang, China
| | - Jing Shen
- Graduate School, Tianjin Medical University, Tianjin, China.,Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Huanlei Zhang
- Department of Radiology, Yidu Central Hospital of Weifang, Weifang, China
| | - Xiulin Wang
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China.,School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Huihui Zhang
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jing Yu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Qing Zhang
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Dongdong Song
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Liping Guo
- Department of Ultrasound, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Dianlong Zhang
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ruiping Zhu
- Department of Pathology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jianlin Wu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
26
|
Wu Z, Bian T, Dong C, Duan S, Fei H, Hao D, Xu W. Spinal MRI-Based Radiomics Analysis to Predict Treatment Response in Multiple Myeloma. J Comput Assist Tomogr 2022; 46:447-454. [PMID: 35405690 DOI: 10.1097/rct.0000000000001298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The aim of this study was to explore the clinical utility of spinal magnetic resonance imaging-based radiomics to predict treatment response (TR) in patients with multiple myeloma (MM). METHODS A total of 123 MM patients (85 in the training cohort and 38 in the test cohort) with complete response (CR) (n = 40) or non-CR (n = 83) were retrospectively enrolled in the study. Key feature selection and data dimension reduction were performed using the least absolute shrinkage and selection operator regression. A nomogram was built by combining radiomic signatures and independent clinical risk factors. The prediction performance of the nomogram was assessed using the area under the receiver operating characteristic curve (AUC), calibration curves, and decision curve analysis. Treatment response was assessed by determining the serum and urinary levels of M-proteins, serum-free light chain ratio, and the percentage of bone marrow plasma cells. RESULTS Thirteen features were selected to build a radiomic signature. The International Staging System (ISS) stage was selected as an independent clinical factor. The radiomic signature and nomogram showed better calibration and higher discriminatory capacity (AUC of 0.929 and 0.917 for the radiomics and nomogram in the training cohort, respectively, and 0.862 and 0.874 for the radiomics and nomogram in the test cohort, respectively) than the clinical model (AUC of 0.661 and 0.674 in the training and test cohort, respectively). Decision curve analysis confirmed the clinical utility of the radiomics model. CONCLUSIONS Nomograms incorporating a magnetic resonance imaging-based radiomic signature and ISS stage help predict the response to chemotherapy for MM and can be useful in clinical decision-making.
Collapse
Affiliation(s)
| | - Tiantian Bian
- Breast Disease Center, the Affiliated Hospital of Qingdao University, Qingdao, Shandong
| | | | | | - Hairong Fei
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | | | | |
Collapse
|
27
|
Luo Z, Li J, Liao Y, Liu R, Shen X, Chen W. Radiomics Analysis of Multiparametric MRI for Prediction of Synchronous Lung Metastases in Osteosarcoma. Front Oncol 2022; 12:802234. [PMID: 35273911 PMCID: PMC8901998 DOI: 10.3389/fonc.2022.802234] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/31/2022] [Indexed: 12/22/2022] Open
Abstract
Purpose To establish and verify a predictive model involving multiparameter MRI and clinical manifestations for predicting synchronous lung metastases (SLM) in osteosarcoma. Materials and Methods Seventy-eight consecutive patients with osteosarcoma (training dataset, n = 54; validation dataset, n = 24) were enrolled in our study. MRI features were extracted from the T1‐weighted image (T1WI), T2‐weighted image (T2WI), and contrast-enhanced T1-weighted image (CE-T1WI) of each patient. Least absolute shrinkage and selection operator (LASSO) regression and multifactor logistic regression were performed to select key features and build radiomics models in conjunction with logistic regression (LR) and support vector machine (SVM) classifiers. Eight individual models based on T1WI, T2WI, CE-T1WI, T1WI+T2WI, T1WI+CE-T1WI, T2WI+CE-T1WI, T1WI+T2WI+CE-T1WI, and clinical features, as well as two combined models, were built. The area under the receiver operating characteristic curve (AUC), sensitivity and specificity were employed to assess the different models. Results Tumor size was the most significant univariate clinical indicator (1). The AUC values of the LR predictive model based on T1WI, T2WI, CE-T1WI, T1WI+T2WI, T1WI+CE-T1WI, T2WI+CE-T1WI, and T1WI+T2WI+CE-T1WI were 0.686, 0.85, 0.87, 0.879, 0.736, 0.85, and 0.914, respectively (2). The AUC values of the SVM predictive model based on T1WI, T2WI, CE-T1WI, T1WI+T2WI, T1WI +CE-T1WI, T2WI +CE-T1WI, and T1WI+T2WI+CE-T1WI were 0.629, 0.829, 0.771, 0.879, 0.643, 0.829, and 0.929, respectively (3). The AUC values of the clinical, combined 1 (clinical and LR-radiomics) and combined 2 (clinical and SVM-radiomics) predictive models were 0.779, 0.957, and 0.943, respectively. Conclusion The combined model exhibited good performance in predicting osteosarcoma SLM and may be helpful in clinical decision-making.
Collapse
Affiliation(s)
- Zhendong Luo
- Department of Radiology, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Jing Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - YuTing Liao
- Department of Pharmaceuticals Diagnosis, GE Healthcare, Shanghai, China
| | - RengYi Liu
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinping Shen
- Department of Radiology, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Weiguo Chen
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Forouzannezhad P, Maes D, Hippe DS, Thammasorn P, Iranzad R, Han J, Duan C, Liu X, Wang S, Chaovalitwongse WA, Zeng J, Bowen SR. Multitask Learning Radiomics on Longitudinal Imaging to Predict Survival Outcomes following Risk-Adaptive Chemoradiation for Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:1228. [PMID: 35267535 PMCID: PMC8909466 DOI: 10.3390/cancers14051228] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/16/2022] Open
Abstract
Medical imaging provides quantitative and spatial information to evaluate treatment response in the management of patients with non-small cell lung cancer (NSCLC). High throughput extraction of radiomic features on these images can potentially phenotype tumors non-invasively and support risk stratification based on survival outcome prediction. The prognostic value of radiomics from different imaging modalities and time points prior to and during chemoradiation therapy of NSCLC, relative to conventional imaging biomarker or delta radiomics models, remains uncharacterized. We investigated the utility of multitask learning of multi-time point radiomic features, as opposed to single-task learning, for improving survival outcome prediction relative to conventional clinical imaging feature model benchmarks. Survival outcomes were prospectively collected for 45 patients with unresectable NSCLC enrolled on the FLARE-RT phase II trial of risk-adaptive chemoradiation and optional consolidation PD-L1 checkpoint blockade (NCT02773238). FDG-PET, CT, and perfusion SPECT imaging pretreatment and week 3 mid-treatment was performed and 110 IBSI-compliant pyradiomics shape-/intensity-/texture-based features from the metabolic tumor volume were extracted. Outcome modeling consisted of a fused Laplacian sparse group LASSO with component-wise gradient boosting survival regression in a multitask learning framework. Testing performance under stratified 10-fold cross-validation was evaluated for multitask learning radiomics of different imaging modalities and time points. Multitask learning models were benchmarked against conventional clinical imaging and delta radiomics models and evaluated with the concordance index (c-index) and index of prediction accuracy (IPA). FDG-PET radiomics had higher prognostic value for overall survival in test folds (c-index 0.71 [0.67, 0.75]) than CT radiomics (c-index 0.64 [0.60, 0.71]) or perfusion SPECT radiomics (c-index 0.60 [0.57, 0.63]). Multitask learning of pre-/mid-treatment FDG-PET radiomics (c-index 0.71 [0.67, 0.75]) outperformed benchmark clinical imaging (c-index 0.65 [0.59, 0.71]) and FDG-PET delta radiomics (c-index 0.52 [0.48, 0.58]) models. Similarly, the IPA for multitask learning FDG-PET radiomics (30%) was higher than clinical imaging (26%) and delta radiomics (15%) models. Radiomics models performed consistently under different voxel resampling conditions. Multitask learning radiomics for outcome modeling provides a clinical decision support platform that leverages longitudinal imaging information. This framework can reveal the relative importance of different imaging modalities and time points when designing risk-adaptive cancer treatment strategies.
Collapse
Affiliation(s)
- Parisa Forouzannezhad
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Dominic Maes
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
| | - Phawis Thammasorn
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Reza Iranzad
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Jie Han
- Department of Industrial, Manufacturing, and System Engineering, University of Texas, Arlington, TX 76019, USA; (J.H.); (S.W.)
| | - Chunyan Duan
- Department of Mechanical Engineering, Tongji University, Shanghai 200092, China;
| | - Xiao Liu
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Shouyi Wang
- Department of Industrial, Manufacturing, and System Engineering, University of Texas, Arlington, TX 76019, USA; (J.H.); (S.W.)
| | - W. Art Chaovalitwongse
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Jing Zeng
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Stephen R. Bowen
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
- Department of Radiology, School of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
29
|
Zhang X, Zhang Y, Zhang G, Qiu X, Tan W, Yin X, Liao L. Deep Learning With Radiomics for Disease Diagnosis and Treatment: Challenges and Potential. Front Oncol 2022; 12:773840. [PMID: 35251962 PMCID: PMC8891653 DOI: 10.3389/fonc.2022.773840] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
The high-throughput extraction of quantitative imaging features from medical images for the purpose of radiomic analysis, i.e., radiomics in a broad sense, is a rapidly developing and emerging research field that has been attracting increasing interest, particularly in multimodality and multi-omics studies. In this context, the quantitative analysis of multidimensional data plays an essential role in assessing the spatio-temporal characteristics of different tissues and organs and their microenvironment. Herein, recent developments in this method, including manually defined features, data acquisition and preprocessing, lesion segmentation, feature extraction, feature selection and dimension reduction, statistical analysis, and model construction, are reviewed. In addition, deep learning-based techniques for automatic segmentation and radiomic analysis are being analyzed to address limitations such as rigorous workflow, manual/semi-automatic lesion annotation, and inadequate feature criteria, and multicenter validation. Furthermore, a summary of the current state-of-the-art applications of this technology in disease diagnosis, treatment response, and prognosis prediction from the perspective of radiology images, multimodality images, histopathology images, and three-dimensional dose distribution data, particularly in oncology, is presented. The potential and value of radiomics in diagnostic and therapeutic strategies are also further analyzed, and for the first time, the advances and challenges associated with dosiomics in radiotherapy are summarized, highlighting the latest progress in radiomics. Finally, a robust framework for radiomic analysis is presented and challenges and recommendations for future development are discussed, including but not limited to the factors that affect model stability (medical big data and multitype data and expert knowledge in medical), limitations of data-driven processes (reproducibility and interpretability of studies, different treatment alternatives for various institutions, and prospective researches and clinical trials), and thoughts on future directions (the capability to achieve clinical applications and open platform for radiomics analysis).
Collapse
Affiliation(s)
- Xingping Zhang
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, China
| | - Yanchun Zhang
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, China
| | - Guijuan Zhang
- Department of Respiratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xingting Qiu
- Department of Radiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Wenjun Tan
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang, China
| | - Xiaoxia Yin
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
| | - Liefa Liao
- School of Information Engineering, Jiangxi University of Science and Technology, Ganzhou, China
| |
Collapse
|
30
|
Yang M, Liu H, Dai Q, Yao L, Zhang S, Wang Z, Li J, Duan Q. Treatment Response Prediction Using Ultrasound-Based Pre-, Post-Early, and Delta Radiomics in Neoadjuvant Chemotherapy in Breast Cancer. Front Oncol 2022; 12:748008. [PMID: 35198437 PMCID: PMC8859469 DOI: 10.3389/fonc.2022.748008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/10/2022] [Indexed: 12/21/2022] Open
Abstract
Objective To develop and validate a radiomics nomogram based on pre-treatment, early treatment ultrasound (US) radiomics features combined with clinical characteristics for early prediction of response to neoadjuvant chemotherapy (NAC) in breast cancer. Method A total of 217 patients with histological results of breast cancer receiving four to eight cycles of NAC before surgery from January 2018 to December 2020 were enrolled. Patients from the study population were randomly separated into a training set (n = 152) and a validation set (n = 65) at a ratio of 7:3. A total of 788 radiomics features were extracted from each region of interest in the US image at pre-treatment baseline (radiomic signature, RS1), early treatment (after completion of two cycles of NAC, RS2) and delta radiomics (calculated between the pre-treatment and post-treatment features, Delta RS). The Max-Relevance and Min-Redundancy (mRMR) and the least absolute shrinkage and selection operator (LASSO) regression were applied for feature selection. The predictive nomogram was built based on the radiomics signature combined with clinicopathological risk factors. Discrimination, calibration, and prediction performance were further evaluated in the validation set. Results Of the 217 breast masses, 127 (58.5%) were responsive to NAC and 90 (41.5%) were non-responsive. Following feature selection, nine features in RS1, 11 features in RS2, and eight features in Delta RS remained. With multivariate analysis, the RS1, RS2, Delta RS, and Ki-67 expression were independently associated with breast NAC response. However, the performance of the Delta RS (AUCDelta RS = 0.743) was not higher than RS1 (AUCRS1 = 0.722, PDelta vs RS1 = 0.086) and RS2 (AUCRS2 = 0.811, PDelta vs RS2 =0.173) with the Delong test. The nomogram incorporating RS1, RS2, and Ki-67 expression showed better predictive ability for NAC response with an area under the curve (AUC) of 0.866 in validation cohorts than either the single RS1 (AUC 0.725) or RS2 (AUC 0.793) or Ki-67 (AUC 0.643). Conclusion The nomogram incorporating pre-treatment and early-treatment US radiomics features and Ki-67 expression showed good performance in terms of NAC response in breast cancer, thereby providing valuable information for individual treatment and timely adjustment of chemotherapy regimens.
Collapse
Affiliation(s)
- Min Yang
- Department of Medical Imaging, the Affiliated Tumor Hospital of Guizhou Medical University, Guiyang, China
| | - Huan Liu
- Department of Advanced Application Team, GE Healthcare, Shanghai, China
| | - Qingli Dai
- Department of Medical Imaging, the Affiliated Tumor Hospital of Guizhou Medical University, Guiyang, China
| | - Ling Yao
- Department of Medical Imaging, the Affiliated Tumor Hospital of Guizhou Medical University, Guiyang, China
| | - Shun Zhang
- Department of Medical Imaging, the Affiliated Tumor Hospital of Guizhou Medical University, Guiyang, China
| | - Zhihong Wang
- Department of Breast Surgery, the Affiliated Tumor Hospital of Guizhou Medical University, Guiyang, China
| | - Jing Li
- Department of Medical Imaging, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qinghong Duan
- Department of Medical Imaging, the Affiliated Tumor Hospital of Guizhou Medical University, Guiyang, China
- *Correspondence: Qinghong Duan,
| |
Collapse
|
31
|
Wu L, Ye W, Liu Y, Chen D, Wang Y, Cui Y, Li Z, Li P, Li Z, Liu Z, Liu M, Liang C, Yang X, Xie Y, Wang Y. An integrated deep learning model for the prediction of pathological complete response to neoadjuvant chemotherapy with serial ultrasonography in breast cancer patients: a multicentre, retrospective study. Breast Cancer Res 2022; 24:81. [PMID: 36414984 PMCID: PMC9680135 DOI: 10.1186/s13058-022-01580-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/13/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The biological phenotype of tumours evolves during neoadjuvant chemotherapy (NAC). Accurate prediction of pathological complete response (pCR) to NAC in the early-stage or posttreatment can optimize treatment strategies or improve the breast-conserving rate. This study aimed to develop and validate an autosegmentation-based serial ultrasonography assessment system (SUAS) that incorporated serial ultrasonographic features throughout the NAC of breast cancer to predict pCR. METHODS A total of 801 patients with biopsy-proven breast cancer were retrospectively enrolled from three institutions and were split into a training cohort (242 patients), an internal validation cohort (197 patients), and two external test cohorts (212 and 150 patients). Three imaging signatures were constructed from the serial ultrasonographic features before (pretreatment signature), during the first-second cycle of (early-stage treatment signature), and after (posttreatment signature) NAC based on autosegmentation by U-net. The SUAS was constructed by subsequently integrating the pre, early-stage, and posttreatment signatures, and the incremental performance was analysed. RESULTS The SUAS yielded a favourable performance in predicting pCR, with areas under the receiver operating characteristic curve (AUCs) of 0.927 [95% confidence interval (CI) 0.891-0.963] and 0.914 (95% CI 0.853-0.976), compared with those of the clinicopathological prediction model [0.734 (95% CI 0.665-0.804) and 0.610 (95% CI 0.504-0.716)], and radiologist interpretation [0.632 (95% CI 0.570-0.693) and 0.724 (95% CI 0.644-0.804)] in the external test cohorts. Furthermore, similar results were also observed in the early-stage treatment of NAC [AUC 0.874 (0.793-0.955)-0.897 (0.851-0.943) in the external test cohorts]. CONCLUSIONS We demonstrate that autosegmentation-based SAUS integrating serial ultrasonographic features throughout NAC can predict pCR with favourable performance, which can facilitate individualized treatment strategies.
Collapse
Affiliation(s)
- Lei Wu
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China ,grid.413352.20000 0004 1760 3705Guangdong Cardiovascular Institute, 106 Zhongshan 2nd Road, Guangzhou, 510080 China
| | - Weitao Ye
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Yu Liu
- grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China ,grid.410643.4Department of Ultrasound, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China
| | - Dong Chen
- grid.452826.fDepartment of Medical Ultrasound, Yunnan Cancer Hospital, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 China
| | - Yuxiang Wang
- grid.263452.40000 0004 1798 4018Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013 China
| | - Yanfen Cui
- grid.263452.40000 0004 1798 4018Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013 China
| | - Zhenhui Li
- grid.452826.fDepartment of Radiology, Yunnan Cancer Hospital, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 China
| | - Pinxiong Li
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Zhen Li
- grid.452826.fDepartment of 3rd Breast Surgery, Yunnan Cancer Hospital, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 China
| | - Zaiyi Liu
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Min Liu
- grid.488530.20000 0004 1803 6191Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Changhong Liang
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Xiaotang Yang
- grid.263452.40000 0004 1798 4018Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013 China
| | - Yu Xie
- grid.452826.fDepartment of Radiology, Yunnan Cancer Hospital, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 China
| | - Ying Wang
- grid.470124.4Department of Medical Ultrasonics, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Guangzhou, 510120 China
| |
Collapse
|
32
|
Zheng RR, Cai MT, Lan L, Huang XW, Yang YJ, Powell M, Lin F. An MRI-based radiomics signature and clinical characteristics for survival prediction in early-stage cervical cancer. Br J Radiol 2022; 95:20210838. [PMID: 34797703 PMCID: PMC8722251 DOI: 10.1259/bjr.20210838] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES To investigate the prognostic role of magnetic resonance imaging (MRI)-based radiomics signature and clinical characteristics for overall survival (OS) and disease-free survival (DFS) in the early-stage cervical cancer. METHODS A total of 207 cervical cancer patients (training cohort: n = 144; validation cohort: n = 63) were enrolled. 792 radiomics features were extracted from T2W and diffusion-weighted imaging (DWI). 19 clinicopathological parameters were collected from the electronic medical record system. Least absolute shrinkage and selection operator (LASSO) regression analysis was used to select significant features to construct prognostic model for OS and DFS. Kaplan-Meier (KM) analysis and log-rank test were applied to identify the association between the radiomics score (Rad-score) and survival time. Nomogram discrimination and calibration were evaluated as well. Associations between radiomics features and clinical parameters were investigated by heatmaps. RESULTS A radiomics signature derived from joint T2W and DWI images showed better prognostic performance than that from either T2W or DWI image alone. Higher Rad-score was associated with worse OS (p < 0.05) and DFS (p < 0.05) in the training and validation set. The joint models outperformed both radiomics model and clinicopathological model alone for 3-year OS and DFS estimation. The calibration curves reached an agreement. Heatmap analysis demonstrated significant associations between radiomics features and clinical characteristics. CONCLUSIONS The MRI-based radiomics nomogram showed a good performance on survival prediction for the OS and DFS in the early-stage cervical cancer. The prediction of the prognostic models could be improved by combining with clinical characteristics, suggesting its potential for clinical application. ADVANCES IN KNOWLEDGE This is the first study to build the radiomics-derived models based on T2W and DWI images for the prediction of survival outcomes on the early-stage cervical cancer patients, and further construct a combined risk scoring system incorporating the clinical features.
Collapse
Affiliation(s)
- Ru-ru Zheng
- Department of Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Meng-ting Cai
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Li Lan
- Department of Ultrasound Imaging, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Xiao Wan Huang
- Department of Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Yun Jun Yang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Martin Powell
- Nottingham University Affiliated Hospital, Nottingham Treatment Centre, Nottingham, UK
| | - Feng Lin
- Department of Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| |
Collapse
|
33
|
Klimonda Z, Karwat P, Dobruch-Sobczak K, Piotrzkowska-Wróblewska H, Litniewski J. Assessment of breast cancer response to neoadjuvant chemotherapy based on ultrasound backscattering envelope statistics. Med Phys 2021; 49:1047-1054. [PMID: 34954844 DOI: 10.1002/mp.15428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Neoadjuvant chemotherapy (NAC) is used in breast cancer before tumor surgery to reduce the size of the tumor and the risk of spreading. Monitoring the effects of NAC is important because in a number of cases the response to therapy is poor and requires a change in treatment. A new method that uses quantitative ultrasound to assess tumor response to NAC has been presented. The aim was to detect NAC unresponsive tumors at an early stage of treatment. METHODS The method assumes that ultrasound scattering is different for responsive and non-responsive tumors. The assessment of the NAC effects was based on the differences between the histograms of the ultrasound echo amplitude recorded from the tumor after each NAC dose and from the tissue phantom, estimated using the Kolmogorov-Smirnov statistics (KSS) and the symmetrical Kullback-Leibler divergence (KLD). After therapy, tumors were resected and histopathologically evaluated. The percentage of residual malignant cells (RMC) was determined and was the basis for assessing the tumor response. The data set included ultrasound data obtained from 37 tumors. The performance of the methods was assessed by means of the area under the receiver operating characteristic curve (AUC). RESULTS For responding tumors a decrease in the mean KLD and KSS values was observed after subsequent doses of NAC. In non-responding tumors the KLD was higher and did not change in subsequent NAC courses. Classification based on the KSS or KLD parameters allowed to detect tumors not responding to NAC after the first dose of the drug, with AUC equal 0.83±0.06 and 0.84±0.07 respectively. After the third dose, the AUC increased to 0.90±0.05 and 0.91±0.04 respectively. CONCLUSIONS The results indicate the potential usefulness of the proposed parameters in assessing the effectiveness of the NAC and early detection of non-responding cases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ziemowit Klimonda
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, Warsaw, 02-106, Poland
| | - Piotr Karwat
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, Warsaw, 02-106, Poland
| | - Katarzyna Dobruch-Sobczak
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, Warsaw, 02-106, Poland.,Radiology Department II, Maria Skłodowska-Curie National Research Institute of Oncology, Wawelska 15B, Warsaw, 02-034, Poland
| | - Hanna Piotrzkowska-Wróblewska
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, Warsaw, 02-106, Poland
| | - Jerzy Litniewski
- Ultrasound Department, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, Warsaw, 02-106, Poland
| |
Collapse
|
34
|
Satake H, Ishigaki S, Ito R, Naganawa S. Radiomics in breast MRI: current progress toward clinical application in the era of artificial intelligence. Radiol Med 2021; 127:39-56. [PMID: 34704213 DOI: 10.1007/s11547-021-01423-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/14/2021] [Indexed: 12/11/2022]
Abstract
Breast magnetic resonance imaging (MRI) is the most sensitive imaging modality for breast cancer diagnosis and is widely used clinically. Dynamic contrast-enhanced MRI is the basis for breast MRI, but ultrafast images, T2-weighted images, and diffusion-weighted images are also taken to improve the characteristics of the lesion. Such multiparametric MRI with numerous morphological and functional data poses new challenges to radiologists, and thus, new tools for reliable, reproducible, and high-volume quantitative assessments are warranted. In this context, radiomics, which is an emerging field of research involving the conversion of digital medical images into mineable data for clinical decision-making and outcome prediction, has been gaining ground in oncology. Recent development in artificial intelligence has promoted radiomics studies in various fields including breast cancer treatment and numerous studies have been conducted. However, radiomics has shown a translational gap in clinical practice, and many issues remain to be solved. In this review, we will outline the steps of radiomics workflow and investigate clinical application of radiomics focusing on breast MRI based on published literature, as well as current discussion about limitations and challenges in radiomics.
Collapse
Affiliation(s)
- Hiroko Satake
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| | - Satoko Ishigaki
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Rintaro Ito
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
35
|
Deep learning radiomics of ultrasonography can predict response to neoadjuvant chemotherapy in breast cancer at an early stage of treatment: a prospective study. Eur Radiol 2021; 32:2099-2109. [PMID: 34654965 DOI: 10.1007/s00330-021-08293-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Breast cancer (BC) is the most common cancer in women worldwide, and neoadjuvant chemotherapy (NAC) is considered the standard of treatment for most patients with BC. However, response rates to NAC vary among patients, which leads to delays in appropriate treatment and affects the prognosis for patients who ineffectively respond to NAC. This study aimed to investigate the feasibility of deep learning radiomics (DLR) in the prediction of NAC response at an early stage. METHODS In total, 168 patients with clinicopathologically confirmed BC were enrolled in this prospective study, from March 2016 to December 2020. All patients completed NAC treatment and underwent ultrasonography (US) at three time points (before NAC, after the second course, and after the fourth course). We developed two DLR models, DLR-2 and DLR-4, for predicting responses after the second and fourth courses of NAC. Furthermore, a novel deep learning radiomics pipeline (DLRP) was proposed for stepwise prediction of response at different time points of NAC administration. RESULTS In the validation cohort, DLR-2 achieved an AUC of 0.812 (95% CI: 0.770-0.851) with an NPV of 83.3% (95% CI: 76.5-89.6). DLR-4 achieved an AUC of 0.937 (95% CI: 0.913-0.955) with a specificity of 90.5% (95% CI: 86.3-94.2). Moreover, 19 of 21 non-response patients were successfully identified by DLRP, suggesting that they could benefit from treatment strategy adjustment at an early stage of NAC. CONCLUSIONS The proposed DLRP strategy holds promise for effectively predicting NAC response at its early stage for BC patients. KEY POINTS • We proposed two novel deep learning radiomics (DLR) models to predict response to neoadjuvant chemotherapy (NAC) in breast cancer (BC) patients based on US images at different NAC time points. • Combining two DLR models, a deep learning radiomics pipeline (DLRP) was proposed for stepwise prediction of response to NAC. • The DLRP may provide BC patients and physicians with an effective and feasible tool to predict response to NAC at an early stage and to determine further personalized treatment options.
Collapse
|
36
|
Niu S, Wang X, Zhao N, Liu G, Kan Y, Dong Y, Cui EN, Luo Y, Yu T, Jiang X. Radiomic Evaluations of the Diagnostic Performance of DM, DBT, DCE MRI, DWI, and Their Combination for the Diagnosisof Breast Cancer. Front Oncol 2021; 11:725922. [PMID: 34568055 PMCID: PMC8461299 DOI: 10.3389/fonc.2021.725922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/23/2021] [Indexed: 12/29/2022] Open
Abstract
Objectives This study aims to evaluate digital mammography (DM), digital breast tomosynthesis (DBT), dynamic contrast-enhanced (DCE), and diffusion-weighted (DW) MRI, individually and combined, for the values in the diagnosis of breast cancer, and propose a visualized clinical-radiomics nomogram for potential clinical uses. Methods A total of 120 patients were enrolled between September 2017 and July 2018, all underwent preoperative DM, DBT, DCE, and DWI scans. Radiomics features were extracted and selected using the least absolute shrinkage and selection operator (LASSO) regression. A radiomics nomogram was constructed integrating the radiomics signature and important clinical predictors, and assessed with the receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA). Results The radiomics signature derived from DBT plus DM generated a lower area under the ROC curve (AUC) and sensitivity, but a higher specificity compared with that from DCE plus DWI. The nomogram integrating the combined radiomics signature, age, and menstruation status achieved the best diagnostic performance in the training (AUCs, nomogram vs. combined radiomics signature vs. clinical model, 0.975 vs. 0.964 vs. 0.782) and validation (AUCs, nomogram vs. combined radiomics signature vs. clinical model, 0.983 vs. 0.978 vs. 0.680) cohorts. DCA confirmed the potential clinical usefulness of the nomogram. Conclusions The DBT plus DM provided a lower AUC and sensitivity, but a higher specificity than DCE plus DWI for detecting breast cancer. The proposed clinical-radiomics nomogram has diagnostic advantages over each modality, and can be considered as an efficient tool for breast cancer screening.
Collapse
Affiliation(s)
- Shuxian Niu
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, China
| | - Xiaoyu Wang
- Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Nannan Zhao
- Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Guanyu Liu
- Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Yangyang Kan
- Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Yue Dong
- Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - E-Nuo Cui
- School of Computer Science and Engineering, Shenyang University, Shenyang, China
| | - Yahong Luo
- Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Tao Yu
- Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Xiran Jiang
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, China
| |
Collapse
|
37
|
Meyer‐Base A, Morra L, Tahmassebi A, Lobbes M, Meyer‐Base U, Pinker K. AI-Enhanced Diagnosis of Challenging Lesions in Breast MRI: A Methodology and Application Primer. J Magn Reson Imaging 2021; 54:686-702. [PMID: 32864782 PMCID: PMC8451829 DOI: 10.1002/jmri.27332] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/11/2022] Open
Abstract
Computer-aided diagnosis (CAD) systems have become an important tool in the assessment of breast tumors with magnetic resonance imaging (MRI). CAD systems can be used for the detection and diagnosis of breast tumors as a "second opinion" review complementing the radiologist's review. CAD systems have many common parts, such as image preprocessing, tumor feature extraction, and data classification that are mostly based on machine-learning (ML) techniques. In this review article, we describe applications of ML-based CAD systems in MRI covering the detection of diagnostically challenging lesions of the breast such as nonmass enhancing (NME) lesions, and furthermore discuss how multiparametric MRI and radiomics can be applied to the study of NME, including prediction of response to neoadjuvant chemotherapy (NAC). Since ML has been widely used in the medical imaging community, we provide an overview about the state-of-the-art and novel techniques applied as classifiers to CAD systems. The differences in the CAD systems in MRI of the breast for several standard and novel applications for NME are explained in detail to provide important examples, illustrating: 1) CAD for detection and diagnosis, 2) CAD in multiparametric imaging, 3) CAD in NAC, and 4) breast cancer radiomics. We aim to provide a comparison between these CAD applications and to illustrate a global view on intelligent CAD systems based on machine and deep learning in MRI of the breast. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Anke Meyer‐Base
- Department of Scientific ComputingFlorida State UniversityTallahasseeFloridaUSA
- Department of Radiology, Maastricht Medical CenterUniversity of MaastrichtMaastrichtNetherlands
| | - Lia Morra
- Department of Control and Computer EngineeringPolitecnico di TorinoTorinoItaly
| | | | - Marc Lobbes
- Department of Radiology, Maastricht Medical CenterUniversity of MaastrichtMaastrichtNetherlands
- GROW School for Oncology and Developmental BiologyMaastrichtNetherlands
- Zuyderland Medical Center, dep of Medical ImagingSittard‐GeleenNetherlands
| | - Uwe Meyer‐Base
- Department of Electrical and Computer EngineeringFlorida A&M University and Florida State UniversityTallahasseeFloridaUSA
| | - Katja Pinker
- Department of Radiology, Breast Imaging ServiceMemorial Sloan‐Kettering Cancer CenterNew YorkNew YorkUSA
- Department of Biomedical Imaging and Image‐Guided Therapy, Division of Molecular and Gender ImagingMedical University of ViennaViennaAustria
| |
Collapse
|
38
|
Radiomics of MRI for the Prediction of the Pathological Response to Neoadjuvant Chemotherapy in Breast Cancer Patients: A Single Referral Centre Analysis. Cancers (Basel) 2021; 13:cancers13174271. [PMID: 34503081 PMCID: PMC8428336 DOI: 10.3390/cancers13174271] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/19/2021] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Nowadays, the only widely recognized method for evaluating the efficacy of neoadjuvant chemotherapy is the assessment of the pathological response through surgery. However, delivering chemotherapy to not-responders could expose them to unnecessary drug toxicity with delayed access to other potentially effective therapies. Radiomics could be useful in the early detection of resistance to chemotherapy, which is crucial for switching treatment strategy. We determined whether tumor radiomic features extracted from a highly homogeneous database of breast MRI can improve the prediction of response to chemotherapy in patients with breast cancer, in addiction to biological characteristics, potentially avoiding unnecessary treatment. Abstract Objectives: We aimed to determine whether radiomic features extracted from a highly homogeneous database of breast MRI could non-invasively predict pathological complete responses (pCR) to neoadjuvant chemotherapy (NACT) in patients with breast cancer. Methods: One hundred patients with breast cancer receiving NACT in a single center (01/2017–06/2019) and undergoing breast MRI were retrospectively evaluated. For each patient, radiomic features were extracted within the biopsy-proven tumor on T1-weighted (T1-w) contrast-enhanced MRI performed before NACT. The pCR to NACT was determined based on the final surgical specimen. The association of clinical/biological and radiomic features with response to NACT was evaluated by univariate and multivariable analysis by using random forest and logistic regression. The performances of all models were assessed using the areas under the receiver operating characteristic curves (AUC) with 95% confidence intervals (CI). Results: Eighty-three patients (mean (SD) age, 47.26 (8.6) years) were included. Patients with HER2+, basal-like molecular subtypes and Ki67 ≥ 20% presented a pCR to NACT more frequently; the clinical/biological model’s AUC (95% CI) was 0.81 (0.71–0.90). Using 136 representative radiomics features selected through cluster analysis from the 1037 extracted features, a radiomic score was calculated to predict the response to NACT, with AUC (95% CI): 0.64 (0.51–0.75). After combining the clinical/biological and radiomics models, the AUC (95% CI) was 0.83 (0.73–0.92). Conclusions: MRI-based radiomic features slightly improved the pre-treatment prediction of pCR to NACT, in addiction to biological characteristics. If confirmed on larger cohorts, it could be helpful to identify such patients, to avoid unnecessary treatment.
Collapse
|
39
|
Recent Radiomics Advancements in Breast Cancer: Lessons and Pitfalls for the Next Future. ACTA ACUST UNITED AC 2021; 28:2351-2372. [PMID: 34202321 PMCID: PMC8293249 DOI: 10.3390/curroncol28040217] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
Radiomics is an emerging translational field of medicine based on the extraction of high-dimensional data from radiological images, with the purpose to reach reliable models to be applied into clinical practice for the purposes of diagnosis, prognosis and evaluation of disease response to treatment. We aim to provide the basic information on radiomics to radiologists and clinicians who are focused on breast cancer care, encouraging cooperation with scientists to mine data for a better application in clinical practice. We investigate the workflow and clinical application of radiomics in breast cancer care, as well as the outlook and challenges based on recent studies. Currently, radiomics has the potential ability to distinguish between benign and malignant breast lesions, to predict breast cancer’s molecular subtypes, the response to neoadjuvant chemotherapy and the lymph node metastases. Even though radiomics has been used in tumor diagnosis and prognosis, it is still in the research phase and some challenges need to be faced to obtain a clinical translation. In this review, we discuss the current limitations and promises of radiomics for improvement in further research.
Collapse
|
40
|
Granzier RWY, Ibrahim A, Primakov SP, Samiei S, van Nijnatten TJA, de Boer M, Heuts EM, Hulsmans FJ, Chatterjee A, Lambin P, Lobbes MBI, Woodruff HC, Smidt ML. MRI-Based Radiomics Analysis for the Pretreatment Prediction of Pathologic Complete Tumor Response to Neoadjuvant Systemic Therapy in Breast Cancer Patients: A Multicenter Study. Cancers (Basel) 2021; 13:cancers13102447. [PMID: 34070016 PMCID: PMC8157878 DOI: 10.3390/cancers13102447] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/23/2022] Open
Abstract
This retrospective study investigated the value of pretreatment contrast-enhanced Magnetic Resonance Imaging (MRI)-based radiomics for the prediction of pathologic complete tumor response to neoadjuvant systemic therapy in breast cancer patients. A total of 292 breast cancer patients, with 320 tumors, who were treated with neo-adjuvant systemic therapy and underwent a pretreatment MRI exam were enrolled. As the data were collected in two different hospitals with five different MRI scanners and varying acquisition protocols, three different strategies to split training and validation datasets were used. Radiomics, clinical, and combined models were developed using random forest classifiers in each strategy. The analysis of radiomics features had no added value in predicting pathologic complete tumor response to neoadjuvant systemic therapy in breast cancer patients compared with the clinical models, nor did the combined models perform significantly better than the clinical models. Further, the radiomics features selected for the models and their performance differed with and within the different strategies. Due to previous and current work, we tentatively attribute the lack of improvement in clinical models following the addition of radiomics to the effects of variations in acquisition and reconstruction parameters. The lack of reproducibility data (i.e., test-retest or similar) meant that this effect could not be analyzed. These results indicate the need for reproducibility studies to preselect reproducible features in order to properly assess the potential of radiomics.
Collapse
Affiliation(s)
- Renée W. Y. Granzier
- Department of Surgery, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (S.S.); (E.M.H.); (M.L.S.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- Correspondence: ; Tel.: +31-43-388-1575
| | - Abdalla Ibrahim
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands;
- The D-Lab, Department of Precision Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, University Hospital of Liège and GIGA CRC-In Vivo Imaging, University of Liège, 4000 Liege, Belgium
- Department of Nuclear Medicine and Comprehensive Diagnostic Center Aachen (CDCA), University Hospital RWTH Aachen University, 52074 Aachen, Germany
| | - Sergey P. Primakov
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- The D-Lab, Department of Precision Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Sanaz Samiei
- Department of Surgery, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (S.S.); (E.M.H.); (M.L.S.)
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands;
| | - Thiemo J. A. van Nijnatten
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands;
| | - Maaike de Boer
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- Department of Internal Medicine, Division of Medical Oncology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Esther M. Heuts
- Department of Surgery, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (S.S.); (E.M.H.); (M.L.S.)
| | - Frans-Jan Hulsmans
- Department of Medical Imaging, Zuyderland Medical Center, P.O. Box 5500, 6130 MB Sittard-Geleen, The Netherlands;
| | - Avishek Chatterjee
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- The D-Lab, Department of Precision Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Philippe Lambin
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands;
- The D-Lab, Department of Precision Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Marc B. I. Lobbes
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands;
- Department of Medical Imaging, Zuyderland Medical Center, P.O. Box 5500, 6130 MB Sittard-Geleen, The Netherlands;
| | - Henry C. Woodruff
- GROW-School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; (A.I.); (S.P.P.); (M.d.B.); (A.C.); (P.L.); (M.B.I.L.); (H.C.W.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands;
- The D-Lab, Department of Precision Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Marjolein L. Smidt
- Department of Surgery, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands; (S.S.); (E.M.H.); (M.L.S.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands;
| |
Collapse
|
41
|
Shang S, Sun J, Yue Z, Wang Y, Wang X, Luo Y, Zhao D, Yu T, Jiang X. Multi-parametric MRI based radiomics with tumor subregion partitioning for differentiating benign and malignant soft-tissue tumors. Biomed Signal Process Control 2021. [DOI: 10.1016/j.bspc.2021.102522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
42
|
Jiang M, Li CL, Luo XM, Chuan ZR, Lv WZ, Li X, Cui XW, Dietrich CF. Ultrasound-based deep learning radiomics in the assessment of pathological complete response to neoadjuvant chemotherapy in locally advanced breast cancer. Eur J Cancer 2021; 147:95-105. [PMID: 33639324 DOI: 10.1016/j.ejca.2021.01.028] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE The aim of the study was to develop and validate a deep learning radiomic nomogram (DLRN) for preoperatively assessing breast cancer pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) based on the pre- and post-treatment ultrasound. METHODS Patients with locally advanced breast cancer (LABC) proved by biopsy who proceeded to undergo preoperative NAC were enrolled from hospital #1 (training cohort, 356 cases) and hospital #2 (independent external validation cohort, 236 cases). Deep learning and handcrafted radiomic features reflecting the phenotypes of the pre-treatment (radiomic signature [RS] 1) and post-treatment tumour (RS2) were extracted. The minimum redundancy maximum relevance algorithm and the least absolute shrinkage and selection operator regression were used for feature selection and RS construction. A DLRN was then developed based on the RSs and independent clinicopathological risk factors. The performance of the model was assessed with regard to calibration, discrimination and clinical usefulness. RESULTS The DLRN predicted the pCR status with accuracy, yielded an area under the receiver operator characteristic curve of 0.94 (95% confidence interval, 0.91-0.97) in the validation cohort, with good calibration. The DLRN outperformed the clinical model and single RS within both cohorts (P < 0.05, as per the DeLong test) and performed better than two experts' prediction of pCR (both P < 0.01 for comparison of total accuracy). Besides, prediction within the hormone receptor-positive/human epidermal growth factor receptor 2 (HER2)-negative, HER2+ and triple-negative subgroups also achieved good discrimination performance, with an AUC of 0.90, 0.95 and 0.93, respectively, in the external validation cohort. Decision curve analysis confirmed that the model was clinically useful. CONCLUSION A deep learning-based radiomic nomogram had good predictive value for pCR in LABC, which could provide valuable information for individual treatment.
Collapse
Affiliation(s)
- Meng Jiang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Chang-Li Li
- Department of Geratology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, 11 Lingjiaohu Avenue, Wuhan, 430015, PR China
| | - Xiao-Mao Luo
- Department of Medical Ultrasound, Yunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, PR China
| | - Zhi-Rui Chuan
- Department of Medical Ultrasound, Yunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, PR China
| | - Wen-Zhi Lv
- Department of Artificial Intelligence, Julei Technology, Wuhan, 430030, PR China
| | - Xu Li
- School of Biomedical Engineering, South-Central University for Nationalities, 182 Minyuan Road, Wuhan, 430074, PR China
| | - Xin-Wu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Christoph F Dietrich
- Department of Internal Medicine, Hirslanden Clinic, Schänzlihalde 11, Bern, 3013, Switzerland
| |
Collapse
|
43
|
Aristei C, Perrucci E, Alì E, Marazzi F, Masiello V, Saldi S, Ingrosso G. Personalization in Modern Radiation Oncology: Methods, Results and Pitfalls. Personalized Interventions and Breast Cancer. Front Oncol 2021; 11:616042. [PMID: 33816246 PMCID: PMC8012886 DOI: 10.3389/fonc.2021.616042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/02/2021] [Indexed: 12/31/2022] Open
Abstract
Breast cancer, the most frequent malignancy in women worldwide, is a heterogeneous group of diseases, characterized by distinct molecular aberrations. In precision medicine, radiation oncology for breast cancer aims at tailoring treatment according to tumor biology and each patient’s clinical features and genetics. Although systemic therapies are personalized according to molecular sub-type [i.e. endocrine therapy for receptor-positive disease and anti-human epidermal growth factor receptor 2 (HER2) therapy for HER2-positive disease] and multi-gene assays, personalized radiation therapy has yet to be adopted in the clinical setting. Currently, attempts are being made to identify prognostic and/or predictive factors, biomarkers, signatures that could lead to personalized treatment in order to select appropriate patients who might, or might not, benefit from radiation therapy or whose radiation therapy might be escalated or de-escalated in dosages and volumes. This overview focuses on what has been achieved to date in personalized post-operative radiation therapy and individual patient radiosensitivity assessments by means of tumor sub-types and genetics.
Collapse
Affiliation(s)
- Cynthia Aristei
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| | | | - Emanuele Alì
- Radiation Oncology Section, University of Perugia, Perugia, Italy
| | - Fabio Marazzi
- Radiation Oncology Department, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Valeria Masiello
- Radiation Oncology Department, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Simonetta Saldi
- Radiation Oncology Section, Perugia General Hospital, Perugia, Italy
| | - Gianluca Ingrosso
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| |
Collapse
|
44
|
Wang Z, Lin F, Ma H, Shi Y, Dong J, Yang P, Zhang K, Guo N, Zhang R, Cui J, Duan S, Mao N, Xie H. Contrast-Enhanced Spectral Mammography-Based Radiomics Nomogram for the Prediction of Neoadjuvant Chemotherapy-Insensitive Breast Cancers. Front Oncol 2021; 11:605230. [PMID: 33692950 PMCID: PMC7937952 DOI: 10.3389/fonc.2021.605230] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose We developed and validated a contrast-enhanced spectral mammography (CESM)-based radiomics nomogram to predict neoadjuvant chemotherapy (NAC)-insensitive breast cancers prior to treatment. Methods We enrolled 117 patients with breast cancer who underwent CESM examination and NAC treatment from July 2017 to April 2019. The patients were grouped randomly into a training set (n = 97) and a validation set (n = 20) in a ratio of 8:2. 792 radiomics features were extracted from CESM images including low-energy and recombined images for each patient. Optimal radiomics features were selected by using analysis of variance (ANOVA) and least absolute shrinkage and selection operator (LASSO) regression with 10-fold cross-validation, to develop a radiomics score in the training set. A radiomics nomogram incorporating the radiomics score and independent clinical risk factors was then developed using multivariate logistic regression analysis. With regard to discrimination and clinical usefulness, radiomics nomogram was evaluated using the area under the receiver operator characteristic (ROC) curve (AUC) and decision curve analysis (DCA). Results The radiomics nomogram that incorporates 11 radiomics features and 3 independent clinical risk factors, including Ki-67 index, background parenchymal enhancement (BPE) and human epidermal growth factor receptor-2 (HER-2) status, showed an encouraging discrimination power with AUCs of 0.877 [95% confidence interval (CI) 0.816 to 0.924] and 0.81 (95% CI 0.575 to 0.948) in the training and validation sets, respectively. DCA revealed the increased clinical usefulness of this nomogram. Conclusion The proposed radiomics nomogram that integrates CESM-derived radiomics features and clinical parameters showed potential feasibility for predicting NAC-insensitive breast cancers.
Collapse
Affiliation(s)
- Zhongyi Wang
- School of Medical Imaging, Binzhou Medical University, Yantai, China.,Department of Radiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Fan Lin
- School of Medical Imaging, Binzhou Medical University, Yantai, China.,Department of Radiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Yinghong Shi
- Department of Radiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Jianjun Dong
- Department of Radiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Ping Yang
- Department of Pathology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Kun Zhang
- Department of Breast Surgery, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Na Guo
- Collaboration Department, Huiying Medical Technology Co., Ltd, Beijing, China
| | - Ran Zhang
- Collaboration Department, Huiying Medical Technology Co., Ltd, Beijing, China
| | - Jingjing Cui
- Collaboration Department, Huiying Medical Technology Co., Ltd, Beijing, China
| | - Shaofeng Duan
- Precision Health Institution, GE Healthcare, Shanghai, China
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Qingdao University, Yantai, China
| |
Collapse
|
45
|
Yan M, Wang W. A radiomics model of predicting tumor volume change of patients with stage III non-small cell lung cancer after radiotherapy. Sci Prog 2021; 104:36850421997295. [PMID: 33687294 PMCID: PMC10453712 DOI: 10.1177/0036850421997295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To predict the volume change of stage III NSCLC after radiotherapy with 60 Gy.This retrospective study included two independent cohorts, a train cohort of 192 patients, and a test cohort of 31 patients. We developed a radiomics model based on radiomics features and clinical variables. LIFEx package was used to extract radiomics texture features from CT images. The classification method was logistic regression analysis and feature selection was performed by correlation coefficients. Performance metrics of logistic regression include accuracy, precision, the receiver operating characteristic curves, and recall.The combination features of clinical variables and radiomics can predict the tumor volume change after radiotherapy with 88.7% accuracy (88.6% precision, 88.7% recall, and 88.7% ROC area).Radiomics features combined with medical knowledge have a great potential to predict accurately tumor volume change of stage III NSCLC after radiotherapy with 60 Gy.
Collapse
Affiliation(s)
- Mengmeng Yan
- Urban Vocational College of Sichuan, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Weidong Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Chengdu, China
- Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
46
|
Caballo M, Pangallo DR, Sanderink W, Hernandez AM, Lyu SH, Molinari F, Boone JM, Mann RM, Sechopoulos I. Multi-marker quantitative radiomics for mass characterization in dedicated breast CT imaging. Med Phys 2020; 48:313-328. [PMID: 33232521 PMCID: PMC7898616 DOI: 10.1002/mp.14610] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/07/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose To develop and evaluate the diagnostic performance of an algorithm for multi‐marker radiomic‐based classification of breast masses in dedicated breast computed tomography (bCT) images. Methods Over 1000 radiomic descriptors aimed at quantifying mass and border heterogeneity, morphology, and margin sharpness were developed and implemented. These included well‐established texture and shape feature descriptors, which were supplemented with additional approaches for contour irregularity quantification, spicule and lobe detection, characterization of degree of infiltration, and differences in peritumoral compartments. All descriptors were extracted from a training set of 202 bCT masses (133 benign and 69 malignant), and their individual diagnostic performance was investigated in terms of area under the receiver operating characteristics (ROC) curve (AUC) of single‐feature‐based linear discriminant analysis (LDA) classifiers. Subsequently, the most relevant descriptors were selected through a multiple‐step feature selection process (including stability analysis, statistical significance, evaluation of feature interaction, and dimensionality reduction), and used to develop a final LDA radiomic model for classification of benign and malignant masses, which was then tested on an independent test set of 82 cases (45 benign and 37 malignant). Results The majority of the individual radiomic descriptors showed, on the training set, an AUC value deriving from a linear decision boundary higher than 0.65, with the lower limit of the associated 95% confidence interval (C.I.) not overlapping with random chance (AUC = 0.5). The final LDA radiomic model resulted in a test set AUC of 0.90 (95% C.I. 0.80–0.96). Conclusions The proposed multi‐marker radiomic approach achieved high diagnostic accuracy in bCT mass classification, using a radiomic signature based on different feature types. While future studies with larger datasets are needed to further validate these results, quantitative radiomics applied to bCT shows potential to improve the breast cancer diagnosis pipeline.
Collapse
Affiliation(s)
- Marco Caballo
- Department of Medical Imaging, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Domenico R Pangallo
- Department of Medical Imaging, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands.,Biolab, Department of Electronics and Telecommunication, Politecnico di Torino, Torino, 10129, Italy
| | - Wendelien Sanderink
- Department of Medical Imaging, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Andrew M Hernandez
- Department of Radiology, University of California Davis, Sacramento, CA, 95817, USA
| | - Su Hyun Lyu
- Department of Biomedical Engineering, University of California Davis, Sacramento, CA, 95817, USA
| | - Filippo Molinari
- Biolab, Department of Electronics and Telecommunication, Politecnico di Torino, Torino, 10129, Italy
| | - John M Boone
- Department of Radiology, University of California Davis, Sacramento, CA, 95817, USA.,Department of Biomedical Engineering, University of California Davis, Sacramento, CA, 95817, USA
| | - Ritse M Mann
- Department of Medical Imaging, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Ioannis Sechopoulos
- Department of Medical Imaging, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands.,Dutch Expert Center for Screening (LRCB), PO Box 6873, Nijmegen, 6503 GJ, The Netherlands
| |
Collapse
|
47
|
Bian T, Wu Z, Lin Q, Wang H, Ge Y, Duan S, Fu G, Cui C, Su X. Radiomic signatures derived from multiparametric MRI for the pretreatment prediction of response to neoadjuvant chemotherapy in breast cancer. Br J Radiol 2020; 93:20200287. [PMID: 32822542 DOI: 10.1259/bjr.20200287] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objectives: To investigate the ability of radiomic signatures based on MRI to evaluate the response and efficiency of neoadjuvant chemotherapy (NAC) for treating breast cancers. Methods: 152 patients were included in this study at our institution between March 2017 and September 2019. All patients with breast cancer underwent a preoperative breast MRI and the Miller–Payne grading system was applied to evaluate response to NAC. Quantitative parameters were compared between patients with sensitive and insensitive responses to NAC and between those with pathological complete responses (pCR) and non-pCR. Four radiomic signatures were built based on T2W imaging, diffusion-weighted imaging, dynamic contrast-enhanced imaging and their combination, and radiomics scores (Rad-score) were calculated. The combination of the clinical factors and Rad-scores created a nomogram model. Multivariate logistic regression was performed to assess the association between MRI features and independent clinical risk factors. Results: 20 features and 18 features were selected to build the radiomic signature for evaluating sensitivity and the possibility of pCR, respectively. The combined radiomic signature and nomogram model showed a similar discrimination in the training (AUC 0.91, 0.92, 95% confidence interval [CI], 0.85–0.96, 0.86–0.98) and validation (AUC 0.93, 0.91, 95% CI, 0.86–1.00, 0.82–1.00) sets. The clinical factor model exhibited reduced performance (AUC 0.74, 0.64, 95% CI, 0.64–0.84, 0.46–0.82) in terms of NAC sensitivity and pCR. Conclusions: The combined radiomic signature and nomogram model exhibited potential predictive power for predicting effective NAC treatment which can aid in the prognosis and guidance of treatment regimens. Advances in knowledge: Identifying a means of assessing the efficacy of NAC before surgery can guide follow-up treatment and avoid chemotherapy-induced toxicity.
Collapse
Affiliation(s)
- Tiantian Bian
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Zengjie Wu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Qing Lin
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Haibo Wang
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yaqiong Ge
- GE Healthcare, Pudong, 210000, Shanghai, China
| | | | - Guangming Fu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Chunxiao Cui
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Xiaohui Su
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| |
Collapse
|
48
|
Zhuang X, Chen C, Liu Z, Zhang L, Zhou X, Cheng M, Ji F, Zhu T, Lei C, Zhang J, Jiang J, Tian J, Wang K. Multiparametric MRI-based radiomics analysis for the prediction of breast tumor regression patterns after neoadjuvant chemotherapy. Transl Oncol 2020; 13:100831. [PMID: 32759037 PMCID: PMC7399245 DOI: 10.1016/j.tranon.2020.100831] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Breast cancers show different regression patterns after neoadjuvant chemotherapy. Certain regression patterns are associated with more reliable margins in breast-conserving surgery. Our study aims to establish a nomogram based on radiomic features and clinicopathological factors to predict regression patterns in breast cancer patients. METHODS We retrospectively reviewed 144 breast cancer patients who received neoadjuvant chemotherapy and underwent definitive surgery in our center from January 2016 to December 2019. Tumor regression patterns were categorized as type 1 (concentric regression + pCR) and type 2 (multifocal residues + SD + PD) based on pathological results. We extracted 1158 multidimensional features from 2 sequences of MRI images. After feature selection, machine learning was applied to construct a radiomic signature. Clinical characteristics were selected by backward stepwise selection. The combined prediction model was built based on both the radiomic signature and clinical factors. The predictive performance of the combined prediction model was evaluated. RESULTS Two radiomic features were selected for constructing the radiomic signature. Combined with two significant clinical characteristics, the combined prediction model showed excellent prediction performance, with an area under the receiver operating characteristic curve of 0.902 (95% confidence interval 0.8343-0.9701) in the primary cohort and 0.826 (95% confidence interval 0.6774-0.9753) in the validation cohort. CONCLUSIONS Our study established a unique model combining a radiomic signature and clinicopathological factors to predict tumor regression patterns prior to the initiation of NAC. The early prediction of type 2 regression offers the opportunity to modify preoperative treatments or aids in determining surgical options.
Collapse
Key Words
- bcs, breast-conserving surgery
- er, estrogen receptor
- her-2, human epidermal growth factor receptor 2
- nac, neoadjuvant chemotherapy
- pc, primary cohort
- pcr, pathologic complete response
- pd, progressive disease
- pr, progesterone receptor
- sd, stable disease
- vc, validation cohort
Collapse
Affiliation(s)
- Xiaosheng Zhuang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Chi Chen
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhenyu Liu
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Science, Beijing 100080, China
| | - Liulu Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xuezhi Zhou
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Minyi Cheng
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Fei Ji
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Teng Zhu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Chuqian Lei
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Junsheng Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jingying Jiang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing 100191, China; Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing 100191, China.
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Science, Beijing 100080, China; Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing 100191, China; Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing 100191, China.
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| |
Collapse
|
49
|
Qiu X, Jiang Y, Zhao Q, Yan C, Huang M, Jiang T. Could Ultrasound-Based Radiomics Noninvasively Predict Axillary Lymph Node Metastasis in Breast Cancer? JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2020; 39:1897-1905. [PMID: 32329142 PMCID: PMC7540260 DOI: 10.1002/jum.15294] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES This work aimed to investigate whether quantitative radiomics imaging features extracted from ultrasound (US) can noninvasively predict breast cancer (BC) metastasis to axillary lymph nodes (ALNs). METHODS Presurgical B-mode US data of 196 patients with BC were retrospectively studied. The cases were divided into the training and validation cohorts (n = 141 versus 55). The elastic net regression technique was used for selecting features and building a signature in the training cohort. A linear combination of the selected features weighted by their respective coefficients produced a radiomics signature for each individual. A radiomics nomogram was established based on the radiomics signature and US-reported ALN status. In a receiver operating characteristic curve analysis, areas under the curves (AUCs) were determined for assessing the accuracy of the prediction model in predicting ALN metastasis in both cohorts. The clinical value was assessed by a decision curve analysis. RESULTS In all, 843 radiomics features per case were obtained from expert-delineated lesions on US imaging in this study. Through radiomics feature selection, 21 features were selected to constitute the radiomics signature for predicting ALN metastasis. Area under the curve values of 0.778 and 0.725 were obtained in the training and validation cohorts, respectively, indicating moderate predictive ability. The radiomics nomogram comprising the radiomics signature and US-reported ALN status showed the best performance for ALN detection in the training cohort (AUC, 0.816) but moderate performance in the validation cohort (AUC, 0.759). The decision curve showed that both the radiomics signature and nomogram displayed good clinical utility. CONCLUSIONS This pilot radiomics study provided a noninvasive method for predicting presurgical ALN metastasis status in BC.
Collapse
Affiliation(s)
- Xiaoying Qiu
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Yongluo Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Qiyu Zhao
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
- Hepatobiliary and Pancreatic SurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Chunhong Yan
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Min Huang
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Tian'an Jiang
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
- Hepatobiliary and Pancreatic SurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| |
Collapse
|
50
|
La Forgia D, Fanizzi A, Campobasso F, Bellotti R, Didonna V, Lorusso V, Moschetta M, Massafra R, Tamborra P, Tangaro S, Telegrafo M, Pastena MI, Zito A. Radiomic Analysis in Contrast-Enhanced Spectral Mammography for Predicting Breast Cancer Histological Outcome. Diagnostics (Basel) 2020; 10:E708. [PMID: 32957690 PMCID: PMC7555402 DOI: 10.3390/diagnostics10090708] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Contrast-Enhanced Spectral Mammography (CESM) is a recently introduced mammographic method with characteristics particularly suitable for breast cancer radiomic analysis. This work aims to evaluate radiomic features for predicting histological outcome and two cancer molecular subtypes, namely Human Epidermal growth factor Receptor 2 (HER2)-positive and triple-negative. From 52 patients, 68 lesions were identified and confirmed on histological examination. Radiomic analysis was performed on regions of interest (ROIs) selected from both low-energy (LE) and ReCombined (RC) CESM images. Fourteen statistical features were extracted from each ROI. Expression of estrogen receptor (ER) was significantly correlated with variation coefficient and variation range calculated on both LE and RC images; progesterone receptor (PR) with skewness index calculated on LE images; and Ki67 with variation coefficient, variation range, entropy and relative smoothness indices calculated on RC images. HER2 was significantly associated with relative smoothness calculated on LE images, and grading tumor with variation coefficient, entropy and relative smoothness calculated on RC images. Encouraging results for differentiation between ER+/ER-, PR+/PR-, HER2+/HER2-, Ki67+/Ki67-, High-Grade/Low-Grade and TN/NTN were obtained. Specifically, the highest performances were obtained for discriminating HER2+/HER2- (90.87%), ER+/ER- (83.79%) and Ki67+/Ki67- (84.80%). Our results suggest an interesting role for radiomics in CESM to predict histological outcomes and particular tumors' molecular subtype.
Collapse
Affiliation(s)
- Daniele La Forgia
- Struttura Semplice Dipartimentale di Radiologia Senologica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Annarita Fanizzi
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.F.); (V.D.); (P.T.)
| | - Francesco Campobasso
- Dipartimento di Economia e Finanza, Università degli Studi di Bari “Aldo Moro”, Largo Abbazia S. Scolastica, 70124 Bari, Italy;
| | - Roberto Bellotti
- Dipartimento Interateneo di Fisica, Università degli Studi di Bari “Aldo Moro”, Via Giovanni Amendola, 165/a, 70126 Bari, Italy;
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Via Giovanni Amendola, 165/a, 70126 Bari, Italy;
| | - Vittorio Didonna
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.F.); (V.D.); (P.T.)
| | - Vito Lorusso
- Unità Operativa Complessa di Oncologia Medica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Marco Moschetta
- Unità Operativa Semplice Dipartimentale Radiodiagnostica ad Indirizzo Senologico, Azienda Ospedaliero-Universitaria Consorziale Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy; (M.M.); (M.T.)
| | - Raffaella Massafra
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.F.); (V.D.); (P.T.)
| | - Pasquale Tamborra
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.F.); (V.D.); (P.T.)
| | - Sabina Tangaro
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Via Giovanni Amendola, 165/a, 70126 Bari, Italy;
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti, Università degli Studi di Bari Aldo Moro, 70121 Bari, Italy
| | - Michele Telegrafo
- Unità Operativa Semplice Dipartimentale Radiodiagnostica ad Indirizzo Senologico, Azienda Ospedaliero-Universitaria Consorziale Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy; (M.M.); (M.T.)
| | - Maria Irene Pastena
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (M.I.P.); (A.Z.)
| | - Alfredo Zito
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (M.I.P.); (A.Z.)
| |
Collapse
|