1
|
Pimentel JM, Zhou JY, Wu GS. Autophagy and cancer therapy. Cancer Lett 2024; 605:217285. [PMID: 39395780 DOI: 10.1016/j.canlet.2024.217285] [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: 08/02/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Autophagy is an intracellular degradation process that sequesters cytoplasmic components in double-membrane vesicles known as autophagosomes, which are degraded upon fusion with lysosomes. This pathway maintains the integrity of proteins and organelles while providing energy and nutrients to cells, particularly under nutrient deprivation. Deregulation of autophagy can cause genomic instability, low protein quality, and DNA damage, all of which can contribute to cancer. Autophagy can also be overactivated in cancer cells to aid in cancer cell survival and drug resistance. Emerging evidence indicates that autophagy has functions beyond cargo degradation, including roles in tumor immunity and cancer stem cell survival. Additionally, autophagy can also influence the tumor microenvironment. This feature warrants further investigation of the role of autophagy in cancer, in which autophagy manipulation can improve cancer therapies, including cancer immunotherapy. This review discusses recent findings on the regulation of autophagy and its role in cancer therapy and drug resistance.
Collapse
Affiliation(s)
- Julio M Pimentel
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA; Institutional Research Academic Career Development Award Program, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jun Ying Zhou
- Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, 48201, USA; Department of Oncology, Wayne State University, Detroit, MI, 48201, USA
| | - Gen Sheng Wu
- Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, 48201, USA; Department of Oncology, Wayne State University, Detroit, MI, 48201, USA; Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
2
|
Wang Y, Zhao H, Fu P, Tian L, Su Y, Lyu Z, Gu W, Wang Y, Liu S, Wang X, Zheng H, Du J, Zhang R. Preoperative prediction of lymph node metastasis in colorectal cancer using 18F-FDG PET/CT peritumoral radiomics analysis. Med Phys 2024; 51:5214-5225. [PMID: 38801340 DOI: 10.1002/mp.17193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Radiomics has been used in the diagnosis of tumor lymph node metastasis (LNM). However, to date, most studies have been based on intratumoral radiomics. Few studies have focused on the use of 18F-fluorodeoxyglucose positron emission computed tomography (18F-FDG PET/CT) peritumoral radiomics for the diagnosis of LNM in colorectal cancer (CRC). PURPOSE Determining the value of radiomics features extracted from 18F-FDG PET/CT images of the peritumoral region in predicting LNM in patients with CRC. METHODS The clinical data and preoperative 18F-FDG PET/CT images of 244 CRC patients were retrospectively analyzed. Intratumoral and peritumoral radiomics features were screened using the mutual information method, and least absolute shrinkage and selection operator regression. Based on the selected radiomics features, a radiomics score (Rad-score) was calculated, and independent risk factors obtained from univariate and multivariate logistic regression analyses were used to construct clinical and combined (Radiomics + Clinical) models. The performance of these models was evaluated using the DeLong test, while their clinical utility was assessed by decision curve analysis. Finally, a nomogram was constructed to visualize the predictive model. RESULTS The most optimal set of features retained by the feature filtering process were all peritumoral radiomic features. Carcinoembryonic antigen levels, PET/CT-reported lymph node status and Rad-score were found to be independent risk factors for LNM. All three LNM risk assessment models exhibited good predictive performance, with the combined model showing the best classification results, with areas under the curve of 0.85 and 0.76 in the training and validation groups, respectively. The DeLong test revealed that the performance of the combined model was superior to that of the clinical and radiomics models in both the training and validation groups, although this difference was only statistically significant in the training group. DCA indicated that the combined model displayed better clinical utility. CONCLUSIONS 18F-FDG PET/CT peritumoral radiomics is uniquely suited to predict the presence of LNM in patients with CRC. In particular, the predictive efficacy of LNM for precision therapy and individualized patient management can be improved by using a combination of clinical risk factors.
Collapse
Affiliation(s)
- Yan Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Fu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Tian
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yexin Su
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhehao Lyu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenchao Gu
- Department of Diagnostic and Interventional Radiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xi Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Han Zheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jingjing Du
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Rui Zhang
- Department of Magnetic Resonance, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, China
| |
Collapse
|
3
|
Zhang B, Zhou Q, Xue C, Ke X, Zhang P, Han T, Deng L, Jing M, Zhou J. Nomogram of magnetic resonance imaging (MRI) histogram analysis to predict telomerase reverse transcriptase promoter mutation status in glioblastoma. Quant Imaging Med Surg 2024; 14:4840-4854. [PMID: 39022283 PMCID: PMC11250314 DOI: 10.21037/qims-24-71] [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: 01/13/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024]
Abstract
Background Telomerase reverse transcriptase promoter (pTERT) status is a strong biomarker to diagnose and predict the prognosis of glioblastoma (GBM). In this study, we explored the predictive value of preoperative magnetic resonance imaging (MRI) histogram analysis in the form of nomogram for evaluating pTERT mutation status in GBM. Methods The clinical and imaging data of 181 patients with GBM at our hospital between November 2018 and April 2023 were retrospectively assessed. We used the molecular sequencing results to classify the datasets into pTERT mutations (C228T and C250T) and pTERT-wildtype groups. FireVoxel software was used to extract preoperative T1-weighted contrast-enhanced (T1C) histogram parameters of GBM patients. The T1C histogram parameters were compared between groups. Univariate and multivariate logistic regression analyses were used to construct the nomogram, and the predictive efficacy of model was evaluated using calibration and decision curves. Receiver operating characteristic curve was used to assess model performance. Results Patient age and percentage of unenhanced tumor area showed statistically significant differences between the pTERT mutation and pTERT-wildtype groups (P<0.001). Among the T1C histogram features, the maximum, standard deviation (SD), variance, coefficient of variation (CV), skewness, 5th, 10th, 25th, 95th and 99th percentiles were statistically significantly different between groups (P=0.000-0.040). Multivariate logistic regression analysis showed that age, percentage of unenhanced tumor area, SD and CV were independent risk factors for predicting pTERT mutation status in GBM patients. The logistic regression model based on these four features showed a better sample predictive performance, and the area under the curve (AUC) [95% confidence interval (CI)], accuracy, sensitivity, specificity were 0.842 (0.767-0.917), 0.796, 0.820, and 0.729, respectively. There were no significant differences in the T1C histogram parameters between the C228T and C250T groups (P=0.055-0.854). Conclusions T1C histogram parameters can be used to evaluate pTERT mutations status in GBM. A nomogram based on conventional MRI features and T1C histogram parameters is a reliable tool for the pTERT mutation status, allowing for non-invasive radiological prediction before surgery.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Peng Zhang
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, China
| | - Tao Han
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Liangna Deng
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Mengyuan Jing
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| |
Collapse
|
4
|
Skirzynska A, Xue C, Shoichet MS. Engineering Biomaterials to Model Immune-Tumor Interactions In Vitro. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310637. [PMID: 38349174 DOI: 10.1002/adma.202310637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Engineered biomaterial scaffolds are becoming more prominent in research laboratories to study drug efficacy for oncological applications in vitro, but do they have a place in pharmaceutical drug screening pipelines? The low efficacy of cancer drugs in phase II/III clinical trials suggests that there are critical mechanisms not properly accounted for in the pre-clinical evaluation of drug candidates. Immune cells associated with the tumor may account for some of these failures given recent successes with cancer immunotherapies; however, there are few representative platforms to study immune cells in the context of cancer as traditional 2D culture is typically monocultures and humanized animal models have a weakened immune composition. Biomaterials that replicate tumor microenvironmental cues may provide a more relevant model with greater in vitro complexity. In this review, the authors explore the pertinent microenvironmental cues that drive tumor progression in the context of the immune system, discuss how these cues can be incorporated into hydrogel design to culture immune cells, and describe progress toward precision oncological drug screening with engineered tissues.
Collapse
Affiliation(s)
- Arianna Skirzynska
- Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
| | - Chang Xue
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
- Institute for Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
| | - Molly S Shoichet
- Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
- Institute for Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Department of Chemistry, University of Toronto, 80 College Street, Toronto, ON, M5S 3H4, Canada
| |
Collapse
|
5
|
Hirschel J, Barcos-Munoz F, Chalard F, Chiodini F, Epiney M, Fluss J, Rougemont AL. Perinatal arterial ischemic stroke: how informative is the placenta? Virchows Arch 2024; 484:815-825. [PMID: 38502326 PMCID: PMC11106178 DOI: 10.1007/s00428-024-03780-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/31/2024] [Accepted: 03/05/2024] [Indexed: 03/21/2024]
Abstract
Neuroplacentology is an expanding field of interest that addresses the placental influence on fetal and neonatal brain lesions and on further neurodevelopment. The objective of this study was to clarify the link between placental pathology and perinatal arterial ischemic stroke (PAIS). Prior publications have reported different types of perinatal stroke with diverse methodologies precluding firm conclusions. We report here the histological placental findings in a series of 16 neonates with radiologically confirmed PAIS. Findings were grouped into 3 categories of lesions: (1) inflammation, (2) placental and fetal hypoxic lesions, and (3) placentas with a high birthweight/placenta weight ratio. Matched control placentas were compared to the pathological placentas when feasible. The eight term singleton placentas were compared to a series of 20 placentas from a highly controlled amniotic membrane donation program; in three twin pregnancies, the placental portions from the affected twin and unaffected co-twin were compared. Slightly more than half (9/16, 56%) had histopathological features belonging to more than one category, a feature shared by the singleton control placentas (13/20, 65%). More severe and extensive lesions were however observed in the pathological placentas. One case occurring in the context of SARS-CoV-2 placentitis further expands the spectrum of COVID-related perinatal disease. Our study supports the assumption that PAIS can result from various combinations and interplay of maternal and fetal factors and confirms the value of placenta examination. Yet, placental findings must be interpreted with caution given their prevalence in well-designed controls.
Collapse
Affiliation(s)
- Jessica Hirschel
- Division of Neonatal and Intensive Care, Department of Pediatrics, Gynecology and Obstetrics, University Hospitals of Geneva, Geneva, Switzerland
| | - Francisca Barcos-Munoz
- Division of Neonatal and Intensive Care, Department of Pediatrics, Gynecology and Obstetrics, University Hospitals of Geneva, Geneva, Switzerland
| | - François Chalard
- Unit of Pediatric Radiology, Department of Radiology, University Hospitals of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Florence Chiodini
- Therapeutic Tissue Biobank, University Hospitals of Geneva, Geneva, Switzerland
| | - Manuella Epiney
- Obstetrics Unit Department of Obstetrics and Gynecology, University Hospitals of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Joel Fluss
- Pediatric Neurology Unit, University Hospitals of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anne-Laure Rougemont
- Division of Clinical Pathology, Diagnostic Department, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
6
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
7
|
Li H, Chai L, Pu H, Yin LL, Li M, Zhang X, Liu YS, Pang MH, Lu T. T2WI-based MRI radiomics for the prediction of preoperative extranodal extension and prognosis in resectable rectal cancer. Insights Imaging 2024; 15:57. [PMID: 38411722 PMCID: PMC10899552 DOI: 10.1186/s13244-024-01625-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 01/18/2024] [Indexed: 02/28/2024] Open
Abstract
OBJECTIVE To investigate whether T2-weighted imaging (T2WI)-based intratumoral and peritumoral radiomics can predict extranodal extension (ENE) and prognosis in patients with resectable rectal cancer. METHODS One hundred sixty-seven patients with resectable rectal cancer including T3T4N + cases were prospectively included. Radiomics features were extracted from intratumoral, peritumoral 3 mm, and peritumoral-mesorectal fat on T2WI images. Least absolute shrinkage and selection operator regression were used for feature selection. A radiomics signature score (Radscore) was built with logistic regression analysis. The area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of each Radscore. A clinical-radiomics nomogram was constructed by the most predictive radiomics signature and clinical risk factors. A prognostic model was constructed by Cox regression analysis to identify 3-year recurrence-free survival (RFS). RESULTS Age, cT stage, and lymph node-irregular border and/or adjacent fat invasion were identified as independent clinical risk factors to construct a clinical model. The nomogram incorporating intratumoral and peritumoral 3 mm Radscore and independent clinical risk factors achieved a better AUC than the clinical model in the training (0.799 vs. 0.736) and validation cohorts (0.723 vs. 0.667). Nomogram-based ENE (hazard ratio [HR] = 2.625, 95% CI = 1.233-5.586, p = 0.012) and extramural vascular invasion (EMVI) (HR = 2.523, 95% CI = 1.247-5.106, p = 0.010) were independent risk factors for predicting 3-year RFS. The prognostic model constructed by these two indicators showed good performance for predicting 3-year RFS in the training (AUC = 0.761) and validation cohorts (AUC = 0.710). CONCLUSION The nomogram incorporating intratumoral and peritumoral 3 mm Radscore and clinical risk factors could predict preoperative ENE. Combining nomogram-based ENE and MRI-reported EMVI may be useful in predicting 3-year RFS. CRITICAL RELEVANCE STATEMENT A clinical-radiomics nomogram could help preoperative predict ENE, and a prognostic model constructed by the nomogram-based ENE and MRI-reported EMVI could predict 3-year RFS in patients with resectable rectal cancer. KEY POINTS • Intratumoral and peritumoral 3 mm Radscore showed the most capability for predicting ENE. • Clinical-radiomics nomogram achieved the best predictive performance for predicting ENE. • Combining clinical-radiomics based-ENE and EMVI showed good performance for 3-year RFS.
Collapse
Affiliation(s)
- Hang Li
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Li Chai
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong Pu
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Long-Lin Yin
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
- Institute of Radiation Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mou Li
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Xin Zhang
- Pharmaceutical Diagnostic Team, GE Healthcare, Beijing, 100176, China
| | - Yi-Sha Liu
- Department of Pathology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Ming-Hui Pang
- Department of Geriatric Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China
| | - Tao Lu
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610070, China.
| |
Collapse
|
8
|
Li H, Chen XL, Liu H, Liu YS, Li ZL, Pang MH, Pu H. MRI-based multiregional radiomics for preoperative prediction of tumor deposit and prognosis in resectable rectal cancer: a bicenter study. Eur Radiol 2023; 33:7561-7572. [PMID: 37160427 DOI: 10.1007/s00330-023-09723-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE To build T2WI-based multiregional radiomics for predicting tumor deposit (TD) and prognosis in patients with resectable rectal cancer. MATERIALS AND METHODS A total of 208 patients with pathologically confirmed rectal cancer from two hospitals were prospectively enrolled. Intra- and peritumoral features were extracted separately from T2WI images and the least absolute shrinkage and selection operator was used to screen the most valuable radiomics features. Clinical-radiomics nomogram was developed by radiomics signatures and the most predictive clinical parameters. Prognostic model for 3-year recurrence-free survival (RFS) was constructed using univariate and multivariate Cox analysis. RESULTS For TD, the area under the receiver operating characteristic curve (AUC) for intratumoral radiomics model was 0.956, 0.823, and 0.860 in the training cohort, test cohort, and external validation cohort, respectively. AUC for the peritumoral radiomics model was 0.929, 0.906, and 0.773 in the training cohort, test cohort, and external validation cohort, respectively. The AUC for combined intra- and peritumoral radiomics model was 0.976, 0.918, and 0.874 in the training cohort, test cohort, and external validation cohort, respectively. The AUC for clinical-radiomics nomogram was 0.989, 0.777, and 0.870 in the training cohort, test cohort, and external validation cohort, respectively. The prognostic model constructed by combining intra- and peritumoral radiomics signature score (radscore)-based TD and MRI-reported lymph nodes metastasis (LNM) indicated good performance for predicting 3-year RFS, with AUC of 0.824, 0.865, and 0.738 in the training cohort, test cohort and external validation cohort, respectively. CONCLUSION Combined intra- and peritumoral radiomics model showed good performance for predicting TD. Combining intra- and peritumoral radscore-based TD and MRI-reported LNM indicated the recurrence risk. CLINICAL RELEVANCE STATEMENT Combined intra- and peritumoral radiomics model could help accurately predict tumor deposits. Combining this predictive model-based tumor deposits with MRI-reported lymph node metastasis was associated with relapse risk of rectal cancer after surgery. KEY POINTS • Combined intra- and peritumoral radiomics model provided better diagnostic performance than that of intratumoral and peritumoral radiomics model alone for predicting TD in rectal cancer. • The predictive performance of the clinical-radiomics nomogram was not improved compared with the combined intra- and peritumoral radiomics model for predicting TD. • The prognostic model constructed by combining intra- and peritumoral radscore-based TD and MRI-reported LNM showed good performance for assessing 3-year RFS.
Collapse
Affiliation(s)
- Hang Li
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China
| | - Xiao-Li Chen
- Department of Radiology, Affiliated Cancer Hospital of Medical School, University of Electronic Science and Technology of China, Sichuan Cancer Hospital, Chengdu, 610000, China
| | | | - Yi-Sha Liu
- Department of Pathology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China
| | - Zhen-Lin Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ming-Hui Pang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China
| | - Hong Pu
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China.
| |
Collapse
|
9
|
Qin S, Lu S, Liu K, Zhou Y, Wang Q, Chen Y, Zhang E, Wang H, Lang N. Radiomics from Mesorectal Blood Vessels and Lymph Nodes: A Novel Prognostic Predictor for Rectal Cancer with Neoadjuvant Therapy. Diagnostics (Basel) 2023; 13:1987. [PMID: 37370882 DOI: 10.3390/diagnostics13121987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/24/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
The objective of our study is to investigate the predictive value of various combinations of radiomic features from intratumoral and different peritumoral regions of interest (ROIs) for achieving a good pathological response (pGR) following neoadjuvant chemoradiotherapy (nCRT) in patients with locally advanced rectal cancer (LARC). This retrospective study was conducted using data from LARC patients who underwent nCRT between 2013 and 2021. Patients were divided into training and validation cohorts at a ratio of 4:1. Intratumoral ROIs (ROIITU) were segmented on T2-weighted imaging, while peritumoral ROIs were segmented using two methods: ROIPTU_2mm, ROIPTU_4mm, and ROIPTU_6mm, obtained by dilating the boundary of ROIITU by 2 mm, 4 mm, and 6 mm, respectively; and ROIMR_F and ROIMR_BVLN, obtained by separating the fat and blood vessels + lymph nodes in the mesorectum. After feature extraction and selection, 12 logistic regression models were established using radiomics features derived from different ROIs or ROI combinations, and five-fold cross-validation was performed. The average area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of the models. The study included 209 patients, consisting of 118 pGR and 91 non-pGR patients. The model that integrated ROIITU and ROIMR_BVLN features demonstrated the highest predictive ability, with an AUC (95% confidence interval) of 0.936 (0.904-0.972) in the training cohort and 0.859 (0.745-0.974) in the validation cohort. This model outperformed models that utilized ROIITU alone (AUC = 0.779), ROIMR_BVLN alone (AUC = 0.758), and other models. The radscore derived from the optimal model can predict the treatment response and prognosis after nCRT. Our findings validated that the integration of intratumoral and peritumoral radiomic features, especially those associated with mesorectal blood vessels and lymph nodes, serves as a potent predictor of pGR to nCRT in patients with LARC. Pending further corroboration in future research, these insights could provide novel imaging markers for refining therapeutic strategies.
Collapse
Affiliation(s)
- Siyuan Qin
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Siyi Lu
- Department of General Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Ke Liu
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Yan Zhou
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Qizheng Wang
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Yongye Chen
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Enlong Zhang
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
- Department of Radiology, Peking University International Hospital, Life Park Road No. 1 Life Science Park of Zhong Guancun, Chang Ping District, Beijing 102206, China
| | - Hao Wang
- Department of Radiation Oncology, Cancer Center, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Ning Lang
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| |
Collapse
|
10
|
Hong Y, Xia Z, Sun Y, Lan Y, Di T, Yang J, Sun J, Qiu M, Luo Q, Yang D. A Comprehensive Pan-Cancer Analysis of the Regulation and Prognostic Effect of Coat Complex Subunit Zeta 1. Genes (Basel) 2023; 14:genes14040889. [PMID: 37107648 PMCID: PMC10137353 DOI: 10.3390/genes14040889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/26/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
The Coatomer protein complex Zeta 1 (COPZ1) has been reported to play an essential role in maintaining the survival of some types of tumors. In this study, we sought to explore the molecular characteristics of COPZ1 and its clinical prognostic value through a pan-cancers bioinformatic analysis. We found that COPZ1 was extremely prevalent in a variety of cancer types, and high expression of COPZ1 was linked to poor overall survival in many cancers, while low expression in LAML and PADC was correlated with tumorigenesis. Besides, the CRISPR Achilles' knockout analysis revealed that COPZ1 was vital for many tumor cells' survival. We further demonstrated that the high expression level of COPZ1 in tumors was regulated in multi-aspects, including abnormal CNV, DNA-methylation, transcription factor and microRNAs. As for the functional exploration of COPZ1, we found a positive relationship between COPZ1's expression and stemness and hypoxia signature, especially the contribution of COPZ1 on EMT ability in SARC. GSEA analysis revealed that COPZ1 was associated with many immune response pathways. Further investigation demonstrated that COPZ expression was negatively correlated with immune score and stromal score, and low expression of COPZ1 has been associated to more antitumor immune cell infiltration and pro-inflammatory cytokines. The further analysis of COPZ1 expression and anti-inflammatory M2 cells showed a consistent result. Finally, we verified the expression of COPZ1 in HCC cells, and proved its ability of sustaining tumor growth and invasion with biological experiments. Our study provides a multi-dimensional pan-cancer analysis of COPZ and demonstrates that COPZ1 can serve as both a prospective target for the treatment of cancer and a prognostic marker for a variety of cancer types.
Collapse
Affiliation(s)
- Ye Hong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Zengfei Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yuting Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yingxia Lan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Tian Di
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Jing Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Jian Sun
- Department of Clinical Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510060, China
| | - Miaozhen Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Qiuyun Luo
- Department of Cancer Research, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China
| | - Dajun Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
11
|
Li H, Chen XL, Liu H, Lu T, Li ZL. MRI-based multiregional radiomics for predicting lymph nodes status and prognosis in patients with resectable rectal cancer. Front Oncol 2023; 12:1087882. [PMID: 36686763 PMCID: PMC9846353 DOI: 10.3389/fonc.2022.1087882] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
Purpose To establish and evaluate multiregional T2-weighted imaging (T2WI)-based clinical-radiomics model for predicting lymph node metastasis (LNM) and prognosis in patients with resectable rectal cancer. Methods A total of 346 patients with pathologically confirmed rectal cancer from two hospitals between January 2019 and December 2021 were prospectively enrolled. Intra- and peritumoral features were extracted separately, and least absolute shrinkage and selection operator regression was applied for feature selection. Radiomics signatures were built using the selected features from different regions. The clinical-radiomic nomogram was developed by combining the intratumoral and peritumoral radiomics signatures score (radscore) and the most predictive clinical parameters. The diagnostic performances of the nomogram and clinical model were evaluated using the area under the receiver operating characteristic curve (AUC). The prognostic model for 3-year recurrence-free survival (RFS) was constructed using univariate and multivariate Cox analysis. Results The intratumoral radscore (radscore 1) included four features, the peritumoral radscore (radscore 2) included five features, and the combined intratumoral and peritumoural radscore (radscore 3) included ten features. The AUCs for radscore 3 were higher than that of radscore 1 in training cohort (0.77 vs. 0.71, P=0.182) and internal validation cohort (0.76 vs. 0.64, P=0.041). The AUCs for radscore 3 were higher than that of radscore 2 in training cohort (0.77 vs. 0.74, P=0.215) and internal validation cohort (0.76 vs. 0.68, P=0.083). A clinical-radiomic nomogram showed a higher AUC compared with the clinical model in training cohort (0.84 vs. 0.67, P<0.001) and internal validation cohort (0.78 vs. 0.64, P=0.038) but not in external validation (0.72 vs. 0.76, P=0.164). Multivariate Cox analysis showed MRI-reported extramural vascular invasion (EMVI) (HR=1.099, 95%CI: 0.462-2.616; P=0.031) and clinical-radiomic nomogram-based LNM (HR=2.232, 95%CI:1.238-7.439; P=0.017) were independent risk factors for assessing 3-year RFS. Combined clinical-radiomic nomogram based LNM and MRI-reported EMVI showed good performance in training cohort (AUC=0.748), internal validation cohort (AUC=0.706) and external validation (AUC=0.688) for predicting 3-year RFS. Conclusion A clinical-radiomics nomogram exhibits good performance for predicting preoperative LNM. Combined clinical-radiomic nomogram based LNM and MRI-reported EMVI showed clinical potential for assessing 3-year RFS.
Collapse
Affiliation(s)
- Hang Li
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Xiao-li Chen
- Department of Radiology, Affiliated Cancer Hospital of Medical School, University of Electronic Science and Technology of China, Sichuan Cancer Hospital, Chengdu, China
| | | | - Tao Lu
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China,*Correspondence: Tao Lu, ; Zhen-lin Li,
| | - Zhen-lin Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Tao Lu, ; Zhen-lin Li,
| |
Collapse
|
12
|
Xue C, Zhou Q, Zhang P, Zhang B, Sun Q, Li S, Deng J, Liu X, Zhou J. MRI histogram analysis of tumor-infiltrating CD8+ T cell levels in patients with glioblastoma. Neuroimage Clin 2023; 37:103353. [PMID: 36812768 PMCID: PMC9958466 DOI: 10.1016/j.nicl.2023.103353] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/05/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
OBJECTIVE To investigate the utility of preoperative magnetic resonance imaging histogram analysis for evaluating tumor-infiltrating CD8+ T cells in patients with glioblastoma (GBM). METHODS We retrospectively analyzed the pathological and imaging data of 61 patients with GBM confirmed by surgery and pathology. Moreover, the levels of tumor-infiltrating CD8+ T cells in tumor tissue samples obtained from the patients were quantified through immunohistochemical staining and evaluated with respect to overall survival. The patients were divided into the high and low CD8 expression groups. Preoperative T1-weighted contrast-enhanced (T1C) histogram parameters of patients with GBM were extracted using Firevoxel software. We investigated the correlation between the histogram feature parameters and CD8+ T cells. We performed statistical analyses of the T1C histogram parameters in both groups and identified characteristic parameters with significant between-group differences. Additionally, we performed a receiver operating characteristic curve (ROC) analysis to determine the predictive utility of these parameters. RESULTS The levels of tumor-infiltrating CD8+ T cells were positively associated with overall survival in patients with GBM (P = 0.0156). Among the T1C histogram features, the mean, 5th, 10th, 25th, and 50th percentiles were negatively correlated with the levels of CD8+ T cells. Moreover, the coefficient of variation (CV) was positively correlated with the levels of CD8+ T cells (all P < 0.05). There was a significant between-group difference in the CV, 1st, 5th, 10th, 25th, and 50th percentiles (all p < 0.05). The ROC curve analysis revealed that the CV had the highest AUC value (0.783; 95% confidence interval: 0.658-0.878), with sensitivity and specificity values of 0.784 and 0.750, respectively, for distinguishing between the groups. CONCLUSIONS The preoperative T1C histogram have additional value for the levels of tumor-infiltrating CD8+ T cells in patients with GBM.
Collapse
Affiliation(s)
- Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Peng Zhang
- Department of Pathology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Qiu Sun
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Shenglin Li
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Juan Deng
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Xianwang Liu
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China.
| |
Collapse
|
13
|
Andersen MB, Harders SW, Thygesen J, Ganeshan B, Torp Madsen HH, Rasmussen F. Potential impact of texture analysis in contrast enhanced CT in non-small cell lung cancer as a marker of survival: A retrospective feasibility study. Medicine (Baltimore) 2022; 101:e31855. [PMID: 36482650 PMCID: PMC9726401 DOI: 10.1097/md.0000000000031855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The objective of this feasibility study was to assess computed tomography (CT) texture analysis (CTTA) of pulmonary lesions as a predictor of overall survival in patients with suspected lung cancer on contrast-enhanced computed tomography (CECT). In a retrospective pilot study, 94 patients (52 men and 42 women; mean age, 67.2 ± 10.8 yrs) from 1 center with non-small cell lung cancer (NSCLC) underwent CTTA on the primary lesion by 2 individual readers. Both simple and multivariate Cox regression analyses correlating textural parameters with overall survival were performed. Statistically significant parameters were selected, and optimal cutoff values were determined. Kaplan-Meier plots based on these results were produced. Simple Cox regression analysis showed that normalized uniformity had a hazard ratio (HR) of 16.059 (3.861-66.788, P < .001), and skewness had an HR of 1.914 (1.330-2.754, P < .001). The optimal cutoff values for both parameters were 0.8602 and 0.1554, respectively. Normalized uniformity, clinical stage, and skewness were found to be prognostic factors for overall survival in multivariate analysis. Tumor heterogeneity, assessed by normalized uniformity and skewness on CECT may be a prognostic factor for overall survival.
Collapse
Affiliation(s)
- Michael Brun Andersen
- Department of Radiology, Aarhus University Hospital, Skejby, Denmark
- Department of Radiology, Copenhagen University Hospital, Gentofte, Denmark
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
- * Correspondence: Michael Brun Andersen, Department of Radiology, Copenhagen University Hospital, Gentofte Hospitalsvej 1, Hellerup 2900, Denmark (e-mail: )
| | | | - Jesper Thygesen
- Department of Clinical Engineering c/o Aarhus University Hospital, Central Denmark Region, Skejby, Denmark
| | - Balaji Ganeshan
- Institute of Nuclear Medicine, University College London, London, UK
| | | | - Finn Rasmussen
- Department of Radiology, Aarhus University Hospital, Skejby, Denmark
| |
Collapse
|
14
|
Chen R, Fu Y, Yi X, Pei Q, Zai H, Chen BT. Application of Radiomics in Predicting Treatment Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer: Strategies and Challenges. JOURNAL OF ONCOLOGY 2022; 2022:1590620. [PMID: 36471884 PMCID: PMC9719428 DOI: 10.1155/2022/1590620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 08/01/2023]
Abstract
Neoadjuvant chemoradiotherapy (nCRT) followed by total mesorectal excision is the standard treatment for locally advanced rectal cancer (LARC). A noninvasive preoperative prediction method should greatly assist in the evaluation of response to nCRT and for the development of a personalized strategy for patients with LARC. Assessment of nCRT relies on imaging and radiomics can extract valuable quantitative data from medical images. In this review, we examined the status of radiomic application for assessing response to nCRT in patients with LARC and indicated a potential direction for future research.
Collapse
Affiliation(s)
- Rui Chen
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yan Fu
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiaoping Yi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Qian Pei
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Hongyan Zai
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Bihong T. Chen
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
15
|
Guo F, Li Q, Gao F, Huang C, Zhang F, Xu J, Xu Y, Li Y, Sun J, Jiang L. Evaluation of the peritumoral features using radiomics and deep learning technology in non-spiculated and noncalcified masses of the breast on mammography. Front Oncol 2022; 12:1026552. [PMID: 36479079 PMCID: PMC9721450 DOI: 10.3389/fonc.2022.1026552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/18/2022] [Indexed: 09/05/2023] Open
Abstract
OBJECTIVE To assess the significance of peritumoral features based on deep learning in classifying non-spiculated and noncalcified masses (NSNCM) on mammography. METHODS We retrospectively screened the digital mammography data of 2254 patients who underwent surgery for breast lesions in Harbin Medical University Cancer Hospital from January to December 2018. Deep learning and radiomics models were constructed. The classification efficacy in ROI and patient levels of AUC, accuracy, sensitivity, and specificity were compared. Stratified analysis was conducted to analyze the influence of primary factors on the AUC of the deep learning model. The image filter and CAM were used to visualize the radiomics and depth features. RESULTS For 1298 included patients, 771 (59.4%) were benign, and 527 (40.6%) were malignant. The best model was the deep learning combined model (2 mm), in which the AUC was 0.884 (P < 0.05); especially the AUC of breast composition B reached 0.941. All the deep learning models were superior to the radiomics models (P < 0.05), and the class activation map (CAM) showed a high expression of signals around the tumor of the deep learning model. The deep learning model achieved higher AUC for large size, age >60 years, and breast composition type B (P < 0.05). CONCLUSION Combining the tumoral and peritumoral features resulted in better identification of malignant NSNCM on mammography, and the performance of the deep learning model exceeded the radiomics model. Age, tumor size, and the breast composition type are essential for diagnosis.
Collapse
Affiliation(s)
- Fei Guo
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Qiyang Li
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Fei Gao
- Deepwise Artificial Intelligence Lab, Beijing Deepwise and League of PHD Technology Co., Ltd, Beijing, China
| | - Chencui Huang
- Deepwise Artificial Intelligence Lab, Beijing Deepwise and League of PHD Technology Co., Ltd, Beijing, China
| | - Fandong Zhang
- Deepwise Artificial Intelligence Lab, Beijing Deepwise and League of PHD Technology Co., Ltd, Beijing, China
| | - Jingxu Xu
- Deepwise Artificial Intelligence Lab, Beijing Deepwise and League of PHD Technology Co., Ltd, Beijing, China
| | - Ye Xu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yuanzhou Li
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jianghong Sun
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Li Jiang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
16
|
Zhao FH, Fan HJ, Shan KF, Zhou L, Pang ZZ, Fu CL, Yang ZB, Wu MK, Sun JH, Yang XM, Huang ZH. Predictive Efficacy of a Radiomics Random Forest Model for Identifying Pathological Subtypes of Lung Adenocarcinoma Presenting as Ground-Glass Nodules. Front Oncol 2022; 12:872503. [PMID: 35646675 PMCID: PMC9133455 DOI: 10.3389/fonc.2022.872503] [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: 02/09/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose To establish and verify the ability of a radiomics prediction model to distinguish invasive adenocarcinoma (IAC) and minimal invasive adenocarcinoma (MIA) presenting as ground-glass nodules (GGNs). Methods We retrospectively analyzed 118 lung GGN images and clinical data from 106 patients in our hospital from March 2016 to April 2019. All pathological classifications of lung GGN were confirmed as IAC or MIA by two pathologists. R language software (version 3.5.1) was used for the statistical analysis of the general clinical data. ITK-SNAP (version 3.6) and A.K. software (Analysis Kit, American GE Company) were used to manually outline the regions of interest of lung GGNs and collect three-dimensional radiomics features. Patients were randomly divided into training and verification groups (ratio, 7:3). Random forest combined with hyperparameter tuning was used for feature selection and prediction modeling. The receiver operating characteristic curve and the area under the curve (AUC) were used to evaluate model prediction efficacy. The calibration curve was used to evaluate the calibration effect. Results There was no significant difference between IAC and MIA in terms of age, gender, smoking history, tumor history, and lung GGN location in both the training and verification groups (P>0.05). For each lung GGN, the collected data included 396 three-dimensional radiomics features in six categories. Based on the training cohort, nine optimal radiomics features in three categories were finally screened out, and a prediction model was established. We found that the training group had a high diagnostic efficacy [accuracy, sensitivity, specificity, and AUC of the training group were 0.89 (95%CI, 0.73 - 0.99), 0.98 (95%CI, 0.78 - 1.00), 0.81 (95%CI, 0.59 - 1.00), and 0.97 (95%CI, 0.92-1.00), respectively; those of the validation group were 0.80 (95%CI, 0.58 - 0.93), 0.82 (95%CI, 0.55 - 1.00), 0.78 (95%CI, 0.57 - 1.00), and 0.92 (95%CI, 0.83 - 1.00), respectively]. The model calibration curve showed good consistency between the predicted and actual probabilities. Conclusions The radiomics prediction model established by combining random forest with hyperparameter tuning effectively distinguished IAC from MIA presenting as GGNs and represents a noninvasive, low-cost, rapid, and reproducible preoperative prediction method for clinical application.
Collapse
Affiliation(s)
- Fen-hua Zhao
- Department of Radiology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Hong-jie Fan
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kang-fei Shan
- Department of Radiology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Long Zhou
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhen-zhu Pang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chun-long Fu
- Department of Radiology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Ze-bin Yang
- Department of Radiology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Mei-kang Wu
- Department of Radiology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Ji-hong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-ming Yang
- Image-Guided Bio-Molecular Intervention Research, Department of Radiology, University of Washington School of Medicine, Seattle, WA, United States
| | - Zhao-hui Huang
- Department of Radiology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| |
Collapse
|
17
|
Yildirim M, Baykara M. Differentiation of progressive disease from pseudoprogression using MRI histogram analysis in patients with treated glioblastoma. Acta Neurol Belg 2022; 122:363-368. [PMID: 33555560 DOI: 10.1007/s13760-021-01607-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/18/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE Conventional magnetic resonance imaging (MRI) technics are insufficient in the differentiation of tumor progression from post-treatment changes in patients with treated glioblastoma. Previous studies have suggested that histogram analysis is a useful tool in the assessment of treatment response in different cancer types. The aim of the study was to to evaluate the effectiveness of MRI histogram analysis in the differentiation of tumor progression from pseudoprogression in patients with treated glioblastoma. METHODS Forty-six patients with glioblastoma who newly developed enhancing lesions following chemoradiation treatment were included in this retrospective study. Histogram analysis was performed from new enhancing lesions on T1-weighted contrast-enhanced MRI. Histogram analysis findings of patients with progression (23) and pseudoprogression (23) were compared. RESULTS Mean, minimum, median, maximum, standard deviation, variance, entropy, skewness, uniformity values were found to be significantly higher in progressive disease (p < 0.05). A receiver-operating characteristic (ROC) curve analysis was performed for mean value, and area under the curve (AUC) was found as 0.975. When the threshold value was selected as 528.86, two groups could be differentiated with 95.7% sensitivity and 87.0% specificity. CONCLUSION MRI histogram analysis can be used for the differentiation of progressive disease from pseudoprogression.
Collapse
|
18
|
The application of apparent diffusion coefficients derived from intratumoral and peritumoral zones for assessing pathologic prognostic factors in rectal cancer. Eur Radiol 2022; 32:5106-5118. [PMID: 35320412 DOI: 10.1007/s00330-022-08717-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 02/12/2022] [Accepted: 03/05/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To investigate the diagnostic performance of the apparent diffusion coefficient (ADC) derived from intratumoral and peritumoral zones for assessing pathologic prognostic factors in rectal cancer. MATERIALS AND METHODS One hundred forty-six patients with rectal cancer who underwent preoperative MRI were prospectively enrolled. Two radiologists independently placed free-hand regions of interest (ROIs) in the largest tumor cross section and three small ROIs on the peritumoral zone adjacent to the tumor contour. Maximum values of tumor ADC (ADCtmax), minimum values of tumor ADC (ADCtmin), mean values of tumor ADC (ADCtmean), mean values of peritumor ADC (ADCpmean), and ADCpmean/ADCtmean (ADC ratio) were obtained on ADC maps and correlated with prognostic factors using uni- and multivariate logistic regression, and receiver operating characteristic curve (ROC) analysis. RESULTS Interobserver agreement was excellent for ADCtmax and ADCtmean (intraclass correlation coefficient [ICC], 0.915-0.958), and were good for ADCtmin, ADCpmean, and ADC ratio (ICC, 0.774-0.878). The ADC ratio was significantly higher in the poor differentiation, T3-4 stage, lymph node metastasis (LNM)-positive, extranodal extension (ENE)-positive, tumor deposit (TD)-positive, and lymphovascular invasion (LVI)-positive groups than that in the well-moderate differentiation, T1-2 stage, LNM-negative, ENE-negative, TD-negative, and LVI-negative groups (p = 0.008, < 0.001, < 0.001, 0.001, < 0.001, and < 0.001, respectively). The area under the ROC curve (AUC) of the ADC ratio was the highest for assessing poor differentiation (0.700), T3-4 stage (0.707), LNM-positive (0.776), TD-positive (0.848), and LVI-positive (0.778). Both the ADC ratio (AUC = 0.677) and ADCpmean (AUC = 0.686) showed higher diagnostic performance for assessing ENE. CONCLUSION The ADC ratio could provide better predictive performance for assessing preoperative prognostic factors in resectable rectal cancer. KEY POINTS • Both the peritumor/tumor ADC ratio and ADCpmean are correlated with important prognostic factors of resectable rectal cancer. • Both peritumor ADC and peritumor/tumor ADC ratio had higher diagnostic performance than tumor ADC for assessment of prognostic factors in resectable rectal cancer. • Peritumor/tumor ADC ratio showed the most capability for the assessment of prognostic factors in resectable rectal cancer.
Collapse
|
19
|
Ideguchi M, Nishizaki T, Ikeda N, Fujii N, Ohno M, Shimabukuro T, Kimura T, Ikeda E, Suga K. Investigation of histological heterogeneity based on the discrepancy between the hyperintense area on T2-weighted images and the accumulation area on 11C-methionine PET in minimally enhancing glioma. INTERDISCIPLINARY NEUROSURGERY 2022. [DOI: 10.1016/j.inat.2021.101364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
20
|
Rabe E, Cioni D, Baglietto L, Fornili M, Gabelloni M, Neri E. Can the computed tomography texture analysis of colorectal liver metastases predict the response to first-line cytotoxic chemotherapy? World J Hepatol 2022; 14:244-259. [PMID: 35126852 PMCID: PMC8790398 DOI: 10.4254/wjh.v14.i1.244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/04/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Artificial intelligence in radiology has the potential to assist with the diagnosis, prognostication and therapeutic response prediction of various cancers. A few studies have reported that texture analysis can be helpful in predicting the response to chemotherapy for colorectal liver metastases, however, the results have varied. Necrotic metastases were not clearly excluded in these studies and in most studies the full range of texture analysis features were not evaluated. This study was designed to determine if the computed tomography (CT) texture analysis results of non-necrotic colorectal liver metastases differ from previous reports. A larger range of texture features were also evaluated to identify potential new biomarkers.
AIM To identify potential new imaging biomarkers with CT texture analysis which can predict the response to first-line cytotoxic chemotherapy in non-necrotic colorectal liver metastases (CRLMs).
METHODS Patients who presented with CRLMs from 2012 to 2020 were retrospectively selected on the institutional radiology information system of our private radiology practice. The inclusion criteria were non-necrotic CRLMs with a minimum size of 10 mm (diagnosed on archived 1.25 mm portal venous phase CT scans) which were treated with standard first-line cytotoxic chemotherapy (FOLFOX, FOLFIRI, FOLFOXIRI, CAPE-OX, CAPE-IRI or capecitabine). The final study cohort consisted of 29 patients. The treatment response of the CRLMs was classified according to the RECIST 1.1 criteria. By means of CT texture analysis, various first and second order texture features were extracted from a single non-necrotic target CRLM in each responding and non-responding patient. Associations between features and response to chemotherapy were assessed by logistic regression models. The prognostic accuracy of selected features was evaluated by using the area under the curve.
RESULTS There were 15 responders (partial response) and 14 non-responders (7 stable and 7 with progressive disease). The responders presented with a higher number of CRLMs (P = 0.05). In univariable analysis, eight texture features of the responding CRLMs were associated with treatment response, but due to strong correlations among some of the features, only two features, namely minimum histogram gradient intensity and long run low grey level emphasis, were included in the multiple analysis. The area under the receiver operating characteristic curve of the multiple model was 0.80 (95%CI: 0.64 to 0.96), with a sensitivity of 0.73 (95%CI: 0.48 to 0.89) and a specificity of 0.79 (95%CI: 0.52 to 0.92).
CONCLUSION Eight first and second order texture features, but particularly minimum histogram gradient intensity and long run low grey level emphasis are significantly correlated with treatment response in non-necrotic CRLMs.
Collapse
Affiliation(s)
- Etienne Rabe
- Academic Radiology, Master in Oncologic Imaging, Department of Translational Research, University of Pisa, Pisa 56126, Italy
- Bay Radiology-Cancercare Oncology Centre, Bay Radiology, Port Elizabeth 6001, Eastern Cape, South Africa
| | - Dania Cioni
- Academic Radiology, Master in Oncologic Imaging, Department of Translational Research, University of Pisa, Pisa 56126, Italy
| | - Laura Baglietto
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Marco Fornili
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Michela Gabelloni
- Academic Radiology, Master in Oncologic Imaging, Department of Translational Research, University of Pisa, Pisa 56126, Italy
| | - Emanuele Neri
- Academic Radiology, Master in Oncologic Imaging, Department of Translational Research, University of Pisa, Pisa 56126, Italy
| |
Collapse
|
21
|
Mahhengam N, Kazemnezhad K, Setia Budi H, Ansari MJ, Olegovich Bokov D, Suksatan W, Thangavelu L, Siahmansouri H. Targeted therapy of tumor microenvironment by gold nanoparticles as a new therapeutic approach. J Drug Target 2022; 30:494-510. [DOI: 10.1080/1061186x.2022.2032095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Negah Mahhengam
- Faculty of General Medicine, Belarusian State Medical University, Minsk, Belarus.
| | - Kimia Kazemnezhad
- Faculty of General Medicine, Belarusian State Medical University, Minsk, Belarus.
| | - Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University,Al-kharj, Saudi Arabia.
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, 119991, Russian Federation.
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand.
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India.
| | - Homayoon Siahmansouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Lisson CS, Lisson CG, Achilles S, Mezger MF, Wolf D, Schmidt SA, Thaiss WM, Bloehdorn J, Beer AJ, Stilgenbauer S, Beer M, Götz M. Longitudinal CT Imaging to Explore the Predictive Power of 3D Radiomic Tumour Heterogeneity in Precise Imaging of Mantle Cell Lymphoma (MCL). Cancers (Basel) 2022; 14:393. [PMID: 35053554 PMCID: PMC8773890 DOI: 10.3390/cancers14020393] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
The study's primary aim is to evaluate the predictive performance of CT-derived 3D radiomics for MCL risk stratification. The secondary objective is to search for radiomic features associated with sustained remission. Included were 70 patients: 31 MCL patients and 39 control subjects with normal axillary lymph nodes followed over five years. Radiomic analysis of all targets (n = 745) was performed and features selected using the Mann Whitney U test; the discriminative power of identifying "high-risk MCL" was evaluated by receiver operating characteristics (ROC). The four radiomic features, "Uniformity", "Entropy", "Skewness" and "Difference Entropy" showed predictive significance for relapse (p < 0.05)-in contrast to the routine size measurements, which showed no relevant difference. The best prognostication for relapse achieved the feature "Uniformity" (AUC-ROC-curve 0.87; optimal cut-off ≤0.0159 to predict relapse with 87% sensitivity, 65% specificity, 69% accuracy). Several radiomic features, including the parameter "Short Axis," were associated with sustained remission. CT-derived 3D radiomics improves the predictive estimation of MCL patients; in combination with the ability to identify potential radiomic features that are characteristic for sustained remission, it may assist physicians in the clinical management of MCL.
Collapse
Affiliation(s)
- Catharina Silvia Lisson
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Personalized Medicine (ZPM), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Christoph Gerhard Lisson
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Sherin Achilles
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Marc Fabian Mezger
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Visual Computing Group, Institute of Media Informatics, Ulm University, James-Franck-Ring, 89081 Ulm, Germany
| | - Daniel Wolf
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Visual Computing Group, Institute of Media Informatics, Ulm University, James-Franck-Ring, 89081 Ulm, Germany
| | - Stefan Andreas Schmidt
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Personalized Medicine (ZPM), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Wolfgang M Thaiss
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Johannes Bloehdorn
- Department of Internal Medicine III, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Ambros J Beer
- Center for Personalized Medicine (ZPM), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Translational Imaging "From Molecule to Man" (MoMan), Department of Internal Medicine II, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- i2SouI-Innovative Imaging in Surgical Oncology Ulm, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Comprehensive Cancer Center Ulm (CCCU), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Meinrad Beer
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Personalized Medicine (ZPM), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Translational Imaging "From Molecule to Man" (MoMan), Department of Internal Medicine II, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- i2SouI-Innovative Imaging in Surgical Oncology Ulm, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Michael Götz
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- German Cancer Research Center (DKFZ), Division Medical Image Computing, 69120 Heidelberg, Germany
| |
Collapse
|
23
|
Park HJ, Qin L, Bakouny Z, Krajewski KM, Van Allen EM, Choueiri TK, Shinagare AB. OUP accepted manuscript. Oncologist 2022; 27:389-397. [PMID: 35348767 PMCID: PMC9074990 DOI: 10.1093/oncolo/oyac034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 01/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Materials and Methods Results Conclusion
Collapse
Affiliation(s)
- Hyo Jung Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Lei Qin
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katherine M Krajewski
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Atul B Shinagare
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Corresponding author: Atul B. Shinagare, Department of Radiology, Brigham and Womens Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA. Tel.: +1 6176322988; Fax: +1 6175828574;
| |
Collapse
|
24
|
Response prediction of neoadjuvant chemoradiation therapy in locally advanced rectal cancer using CT-based fractal dimension analysis. Eur Radiol 2021; 32:2426-2436. [PMID: 34643781 DOI: 10.1007/s00330-021-08303-z] [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: 06/13/2021] [Revised: 08/10/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES There are individual variations in neo-adjuvant chemoradiation therapy (nCRT) in patients with locally advanced rectal cancer (LARC). No reliable modality currently exists that can predict the efficacy of nCRT. The purpose of this study is to assess if CT-based fractal dimension and filtration-histogram texture analysis can predict therapeutic response to nCRT in patients with LARC. METHODS In this retrospective study, 215 patients (average age: 57 years (18-87 years)) who received nCRT for LARC between June 2005 and December 2016 and underwent a staging diagnostic portal venous phase CT were identified. The patients were randomly divided into two datasets: a training set (n = 170), and a validation set (n = 45). Tumor heterogeneity was assessed on the CT images using fractal dimension (FD) and filtration-histogram texture analysis. In the training set, the patients with pCR and non-pCR were compared in univariate analysis. Logistic regression analysis was applied to identify the predictive value of efficacy of nCRT and receiver operating characteristic analysis determined optimal cutoff value. Subsequently, the most significant parameter was assessed in the validation set. RESULTS Out of the 215 patients evaluated, pCR was reached in 20.9% (n = 45/215) patients. In the training set, 7 out of 37 texture parameters showed significant difference comparing between the pCR and non-pCR groups and logistic multivariable regression analysis incorporating clinical and 7 texture parameters showed that only FD was associated with pCR (p = 0.001). The area under the curve of FD was 0.76. In the validation set, we applied FD for predicting pCR and sensitivity, specificity, and accuracy were 60%, 89%, and 82%, respectively. CONCLUSION FD on pretreatment CT is a promising parameter for predicting pCR to nCRT in patients with LARC and could be used to help make treatment decisions. KEY POINTS • Fractal dimension analysis on pretreatment CT was associated with response to neo-adjuvant chemoradiation in patients with locally advanced rectal cancer. • Fractal dimension is a promising biomarker for predicting pCR to nCRT and may potentially select patients for individualized therapy.
Collapse
|
25
|
Huang Y, Fan J, Li Y, Fu S, Chen Y, Wu J. Imaging of Tumor Hypoxia With Radionuclide-Labeled Tracers for PET. Front Oncol 2021; 11:731503. [PMID: 34557414 PMCID: PMC8454408 DOI: 10.3389/fonc.2021.731503] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/19/2021] [Indexed: 01/27/2023] Open
Abstract
The hypoxic state in a solid tumor refers to the internal hypoxic environment that appears as the tumor volume increases (the maximum radius exceeds 180-200 microns). This state can promote angiogenesis, destroy the balance of the cell’s internal environment, and lead to resistance to radiotherapy and chemotherapy, as well as poor prognostic factors such as metastasis and recurrence. Therefore, accurate quantification, mapping, and monitoring of hypoxia, targeted therapy, and improvement of tumor hypoxia are of great significance for tumor treatment and improving patient survival. Despite many years of development, PET-based hypoxia imaging is still the most widely used evaluation method. This article provides a comprehensive overview of tumor hypoxia imaging using radionuclide-labeled PET tracers. We introduced the mechanism of tumor hypoxia and the reasons leading to the poor prognosis, and more comprehensively included the past, recent and ongoing studies of PET radiotracers for tumor hypoxia imaging. At the same time, the advantages and disadvantages of mainstream methods for detecting tumor hypoxia are summarized.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Junying Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Oncology, Academician (Expert) Workstation of Sichuan Province, Luzhou, China
| | - Yue Chen
- Department of Oncology, Academician (Expert) Workstation of Sichuan Province, Luzhou, China.,Nuclear Medicine and Molecular Imaging key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Oncology, Academician (Expert) Workstation of Sichuan Province, Luzhou, China
| |
Collapse
|
26
|
Xie T, Zhao Q, Fu C, Grimm R, Gu Y, Peng W. Improved value of whole-lesion histogram analysis on DCE parametric maps for diagnosing small breast cancer (≤ 1 cm). Eur Radiol 2021; 32:1634-1643. [PMID: 34505195 DOI: 10.1007/s00330-021-08244-7] [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: 02/23/2021] [Revised: 07/21/2021] [Accepted: 08/03/2021] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To determine if whole-lesion histogram analysis on dynamic contrast-enhanced (DCE) parametric maps help to improve the diagnostic accuracy of small suspicious breast lesions (≤ 1 cm). METHODS This retrospective study included 99 female patients with 114 lesions (40 malignant and 74 benign lesions) suspicious on magnetic resonance imaging (MRI).Two radiologists reviewed all lesions and descripted the morphologic and kinetic characteristics according to BI-RADS by consensus. Whole lesions were segmented on DCE parametric maps (washin and washout), and quantitative histogram features were extracted. Univariate analysis and multivariate logistic regression analysis with forward stepwise covariate selection were performed to identify significant variables. Diagnostic performance was assessed and compared with that of qualitative BI-RADS assessment and quantitative histogram analysis by ROC analysis. RESULTS For malignancy defined as a washout or plateau pattern, the qualitative kinetic pattern showed a significant difference between the two groups (p = 0.023), yielding an AUC of 0.603 (95% confidence interval [CI]: 0.507, 0.694). The mean and median of washout were independent quantitative predictors of malignancy (p = 0.002, 0.010), achieving an AUC of 0.796 (95% CI: 0. 709, 0.865). The AUC of the quantitative model was better than that of the qualitative model (p < 0.001). CONCLUSIONS Compared with the qualitative BI-RADS assessment, quantitative whole-lesion histogram analysis on DCE parametric maps was better to discriminate between small benign and malignant breast lesions (≤ 1 cm) initially defined as suspicious on DCE-MRI. KEY POINTS • For malignancy defined as a washout or plateau, the kinetic pattern may provide information to diagnose small breast cancer. • The mean and median of washout map were significantly lower for small malignant breast lesions than for benign lesions. • Quantitative histogram analysis on MRI parametric maps improves diagnostic accuracy for small breast cancer, which may obviate unnecessary biopsy.
Collapse
Affiliation(s)
- Tianwen Xie
- Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Qiufeng Zhao
- Department of Radiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Caixia Fu
- MR Applications Development, Siemens Shenzhen Magnetic Resonance Ltd, Shenzhen, People's Republic of China
| | - Robert Grimm
- MR Application Predevelopment, Siemens Healthcare, Erlangen, Germany
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Weijun Peng
- Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
27
|
Asadian S, Rezaeian N, Hosseini L, Toloueitabar Y, Hemmati Komasi MM. The role of cardiac CT and MRI in the diagnosis and management of primary cardiac lymphoma: A comprehensive review. Trends Cardiovasc Med 2021; 32:408-420. [PMID: 34454052 DOI: 10.1016/j.tcm.2021.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/05/2021] [Accepted: 08/20/2021] [Indexed: 12/18/2022]
Abstract
Primary cardiac tumors comprise a distinct category of disorders that result in significant cardiac complications. Primary cardiac lymphomas (PCLs) constitute the second most frequent primary malignancy involving the heart. Without treatment, survival may be limited to just a few months; however, a timely therapeutic schedule may prolong the five-year survival. Accordingly, robust diagnostic modalities are essential to improve prognosis. We herein review the literature available in PubMed, MEDLINE, Cochrane, Google Scholar and Scopus databases. Our review demonstrated that cardiac computed tomography (CT) and magnetic resonance imaging (MRI) employ multiple advanced sequences for tumor characterization with or without a contrast agent. These methods assist not only in differentiating PCLs from other cardiac masses such as cardiac thrombi but also in defining the extent of PCLs and conducting a safe biopsy. Cardiac magnetic resonance (CMR) and CT imaging provide essential knowledge regarding PCLs and cardiotoxicity induced by therapeutic regimens. The application of these robust imaging modalities aids in the early diagnosis of PCLs, accelerates the initiation of the treatment program, and improves patient outcomes significantly. Also presented is our introduction into novel techniques and the feasibility of their use to diagnose and treat cardiac masses, particularly PCLs. It should be mentioned that the paramount role of FDG-PET was not the focus of this paper.
Collapse
Affiliation(s)
- Sanaz Asadian
- Rajaie Cardiovascular Medical and Research Center, Tehran, Iran
| | - Nahid Rezaeian
- Rajaie Cardiovascular Medical and Research Center, Tehran, Iran.
| | - Leila Hosseini
- Rajaie Cardiovascular Medical and Research Center, Tehran, Iran
| | | | | |
Collapse
|
28
|
Wang J, Zhou M, Chen F, Liu X, Gao J, Wang W, Wang H, Yu H. Stimuli-Sheddable Nanomedicine Overcoming Pathophysiological Barriers for Potentiating Immunotherapy of Cancer. J Biomed Nanotechnol 2021; 17:1486-1509. [PMID: 34544528 DOI: 10.1166/jbn.2021.3134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Immunotherapy displays potent potential for clinical cancer management by activating the protective immune response; however, the microenvironment of the immunosuppressive tumor restricts the efficiency of immunotherapies. Along with the complex pathophysiological barrier of the solid tumors, successful immunotherapeutic delivery remains a formidable challenge for conventional nanomedicine. Stimuli-sheddable nano vectors may facilitate the delivery of cargoes to tumors with minimal premature cargo leakage in blood circulation while enhancing the tumor penetration of nanomedicines by deshielding the polyethylene glycol (PEG) corona upon endogenous activity such as acidity, enzymes and glutathione, or external stimuli, such as laser irradiation. Throughout this study, researchers overviewed the recent advances of nanomedicine-based cancer immunotherapy using the stimuli-responsive deshielding nano vectors, which allowed researchers to integrate multiple therapeutic regimens for inducing immunogenic cell death. This aided in blocking the immune checkpoints, repolarizing the macrophages, and regulating the kynurenine metabolism. Furthermore, researchers discussed the critical issues in the development of stimuli-sheddable nanoimmunodulators, primarily aimed at speeding up their clinical translation. Finally, researchers provided novel perspectives for improving cancer management with the stimuli-sheddable nanomedicine.
Collapse
Affiliation(s)
- Jiaxin Wang
- College of Chemistry and Chemical Engineering, Inner Magnolia University, Huhhot, 010021, China
| | - Mengxue Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Fangmin Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Jin Gao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Hui Wang
- College of Chemistry and Chemical Engineering, Inner Magnolia University, Huhhot, 010021, China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
29
|
Song SE, Seo BK, Cho KR, Woo OH, Ganeshan B, Kim ES, Cha J. Prediction of Inflammatory Breast Cancer Survival Outcomes Using Computed Tomography-Based Texture Analysis. Front Bioeng Biotechnol 2021; 9:695305. [PMID: 34354986 PMCID: PMC8329959 DOI: 10.3389/fbioe.2021.695305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/18/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Although inflammatory breast cancer (IBC) has poor overall survival (OS), there is little information about using imaging features for predicting the prognosis. Computed tomography (CT)-based texture analysis, a non-invasive technique to quantify tumor heterogeneity, could be a potentially useful imaging biomarker. The aim of the article was to investigate the usefulness of chest CT-based texture analysis to predict OS in IBC patients. Methods: Of the 3,130 patients with primary breast cancers between 2006 and 2016, 104 patients (3.3%) with IBC were identified. Among them, 98 patients who underwent pre-treatment contrast-enhanced chest CT scans, got treatment in our institution, and had a follow-up period of more than 2 years were finally included for CT-based texture analysis. Texture analysis was performed on CT images of 98 patients, using commercially available software by two breast radiologists. Histogram-based textural features, such as quantification of variation in CT attenuation (mean, standard deviation, mean of positive pixels [MPP], entropy, skewness, and kurtosis), were recorded. To dichotomize textural features for survival analysis, receiver operating characteristic curve analysis was used to determine cutoff points. Clinicopathologic variables, such as age, node stage, metastasis stage at the time of diagnosis, hormonal receptor positivity, human epidermal growth factor receptor 2 positivity, and molecular subtype, were assessed. A Cox proportional hazards model was used to determine the association of textural features and clinicopathologic variables with OS. Results: During a mean follow-up period of 47.9 months, 41 of 98 patients (41.8%) died, with a median OS of 20.0 months. The textural features of lower mean attenuation, standard deviation, MPP, and entropy on CT images were significantly associated with worse OS, as was the M1 stage among clinicopathologic variables (all P-values < 0.05). In multivariate analysis, lower mean attenuation (hazard ratio [HR], 3.26; P = 0.003), lower MPP (HR, 3.03; P = 0.002), and lower entropy (HR, 2.70; P = 0.009) on chest CT images were significant factors independent from the M1 stage for predicting worse OS. Conclusions: Lower mean attenuation, MPP, and entropy on chest CT images predicted worse OS in patients with IBC, suggesting that CT-based texture analysis provides additional predictors for OS.
Collapse
Affiliation(s)
- Sung Eun Song
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Bo Kyoung Seo
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, South Korea
| | - Kyu Ran Cho
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Ok Hee Woo
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Balaji Ganeshan
- Institute of Nuclear Medicine, University College London Hospitals NHS Trust, London, United Kingdom
| | - Eun Sil Kim
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, South Korea
| | - Jaehyung Cha
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, South Korea
| |
Collapse
|
30
|
Park NJY, Jeong JY, Park JY, Kim HJ, Park CS, Lee J, Park HY, Jung JH, Kim WW, Chae YS, Lee SJ, Kim WH. Peritumoral edema in breast cancer at preoperative MRI: an interpretative study with histopathological review toward understanding tumor microenvironment. Sci Rep 2021; 11:12992. [PMID: 34155253 PMCID: PMC8217499 DOI: 10.1038/s41598-021-92283-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Peritumoral edema (PE) of breast cancer at T2-weighted MR images is considered a poor prognostic sign and may represent the microenvironment surrounding the tumor; however, its histopathological mechanism remains unclear. The purpose of the study was to identify and describe detailed histopathological characteristics associated with PE at preoperative breast MRI in breast cancer patients. This retrospective study included breast cancer patients who had undergone preoperative MRI and surgery between January 2011 and December 2012. Two radiologists determined the presence of PE in consensus based on the signal intensity surrounding the tumor at T2-weighted images. The following detailed histopathological characteristics were reviewed by two breast pathologists using four-tiered grades; lymphovascular invasion, vessel ectasia, stromal fibrosis, growth pattern, and tumor budding. Tumor necrosis and tumor infiltrating lymphocytes were assessed using a percent scale. Baseline clinicopathological characteristics, including age and histologic grade, were collected. The associations between detailed histopathologic characteristics and PE were examined using multivariable logistic regression with odds ratio (OR) calculation. A total of 136 women (median age, 49 ± 9 years) were assessed; among them 34 (25.0%) had PE. After adjustment of baseline clinicopathological characteristics that were significantly associated with PE (age, T stage, N stage, histologic grade, and subtype, all Ps < 0.05), lymphovascular invasion (P = 0.009), vessel ectasia (P = 0.021), stromal fibrosis (P = 0.024), growth pattern (P = 0.036), and tumor necrosis (P < 0.001) were also associated with PE. In comparison with patients without PE, patients with PE were more likely to have a higher degree of lymphovascular invasion (OR, 2.9), vessel ectasia (OR, 3.3), stromal fibrosis (OR, 2.5), lesser degree of infiltrative growth pattern (OR, 0.4), and higher portion of tumor necrosis (OR, 1.4). PE of breast cancer at MRI is associated with detailed histopathological characteristics of lymphovascular invasion, vessel ectasia, stromal fibrosis, growth pattern, and tumor necrosis, suggesting a relevance for tumor microenvironment.
Collapse
Affiliation(s)
- Nora Jee-Young Park
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Ji Young Park
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Hye Jung Kim
- Department of Radiology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Chan Sub Park
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Jeeyeon Lee
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Ho Yong Park
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Jin Hyang Jung
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Wan Wook Kim
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Yee Soo Chae
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Soo Jung Lee
- Department of Oncology/Hematology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea
| | - Won Hwa Kim
- Department of Radiology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, South Korea.
| |
Collapse
|
31
|
Frank V, Shariati S, Budai BK, Fejér B, Tóth A, Orbán V, Bérczi V, Kaposi PN. CT texture analysis of abdominal lesions – Part II: Tumors of the Kidney and Pancreas. IMAGING 2021. [DOI: 10.1556/1647.2021.00020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
AbstractIt has been proven in a few early studies that radiomic analysis offers a promising opportunity to detect or differentiate between organ lesions based on their unique texture parameters. Recently, the utilization of CT texture analysis (CTTA) has been receiving significant attention, especially for response evaluation and prognostication of different oncological diagnoses. In this review article, we discuss the unique ability of radiomics and its subfield CTTA to diagnose lesions in the pancreas and kidney. We review studies in which CTTA was used for the classification of histology grades in pancreas and kidney tumors. We also review the role of radiogenomics in the prediction of the molecular and genetic subtypes of pancreatic tumors. Furthermore, we provide a short report on recent advancements of radiomic analysis in predicting prognosis and survival of patients with pancreatic and renal cancers.
Collapse
Affiliation(s)
- Veronica Frank
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Sonaz Shariati
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Bettina Katalin Budai
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Bence Fejér
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Ambrus Tóth
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Vince Orbán
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Viktor Bérczi
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Pál Novák Kaposi
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| |
Collapse
|
32
|
Khorrami M, Bera K, Thawani R, Rajiah P, Gupta A, Fu P, Linden P, Pennell N, Jacono F, Gilkeson RC, Velcheti V, Madabhushi A. Distinguishing granulomas from adenocarcinomas by integrating stable and discriminating radiomic features on non-contrast computed tomography scans. Eur J Cancer 2021; 148:146-158. [PMID: 33743483 PMCID: PMC8087632 DOI: 10.1016/j.ejca.2021.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To identify stable and discriminating radiomic features on non-contrast CT scans to develop more generalisable radiomic classifiers for distinguishing granulomas from adenocarcinomas. METHODS In total, 412 patients with adenocarcinomas and granulomas from three institutions were retrospectively included. Segmentations of the lung nodules were performed manually by an expert radiologist in a 2D axial view. Radiomic features were extracted from intra- and perinodular regions. A total of 145 patients were used as part of the training set (Str), whereas 205 patients were used as part of test set I (Ste1) and 62 patients were used as part of independent test set II (Ste2). To mitigate the variation of CT acquisition parameters, we defined 'stable' radiomic features as those for which the feature expression remains relatively unchanged between different sites, as assessed using a Wilcoxon rank-sum test. These stable features were used to develop more generalisable radiomic classifiers that were more resilient to variations in lung CT scans. Features were ranked based on two criteria, firstly based on discriminability (i.e. maximising AUC) alone and subsequently based on maximising both feature stability and discriminability. Different machine-learning classifiers (Linear discriminant analysis, Quadratic discriminant analysis, Support vector machines and random forest) were trained with features selected using the two different criteria and then compared on the two independent test sets for distinguishing granulomas from adenocarcinomas, in terms of area under the receiver operating characteristic curve. RESULTS In the test sets, classifiers constructed using the criteria involving maximising feature stability and discriminability simultaneously achieved higher AUC compared with the discriminating alone criteria (Ste1 [n = 205]: maximum AUCs of 0.85versus . 0.80; p-value = 0.047 and Ste2 [n = 62]: maximum AUCs of 0.87 versus. 0.79; p-value = 0.021). These differences held for features extracted from scans with <3 mm slice thickness (AUC = 0.88 versus. 0.80; p-value = 0.039, n = 100) and for the ≥3 mm cases (AUC = 0.81 versus. 0.76; p-value = 0.034, n = 105). In both experiments, shape and peritumoural texture features had a higher stability compared with intratumoural texture features. CONCLUSIONS Our study suggests that explicitly accounting for both stability and discriminability results in more generalisable radiomic classifiers to distinguish adenocarcinomas from granulomas on non-contrast CT scans. Our results also showed that peritumoural texture and shape features were less affected by the scanner parameters compared with intratumoural texture features; however, they were also less discriminating compared with intratumoural features.
Collapse
Affiliation(s)
- Mohammadhadi Khorrami
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Rajat Thawani
- OHSU Knight Cancer Institute, Oregon Health & Science University, Oregon, USA
| | - Prabhakar Rajiah
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Amit Gupta
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, CWRU, Cleveland, OH, USA
| | - Philip Linden
- Thoracic and Esophageal Surgery Department, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Nathan Pennell
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Frank Jacono
- Pulmonary Section, Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Robert C Gilkeson
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA; Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA.
| |
Collapse
|
33
|
Ma M, Liang J, Zhang D, Xu X, Cheng Q, Xiao Z, Shi C, Luo L. Monitoring Treatment Efficacy of Antiangiogenic Therapy Combined With Hypoxia-Activated Prodrugs Online Using Functional MRI. Front Oncol 2021; 11:672047. [PMID: 33996599 PMCID: PMC8120295 DOI: 10.3389/fonc.2021.672047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/06/2021] [Indexed: 01/12/2023] Open
Abstract
Objective This study aimed to investigate the effectiveness of intravoxel incoherent motion (IVIM) diffusion-weighted imaging (DWI) and blood oxygen level-dependent (BOLD) magnetic resonance imaging (MRI) in monitoring tumor responses to antiangiogenic therapy combined with hypoxia-activated prodrugs (HAPs). Materials and methods Establishing colon cancer xenograft model by subcutaneously injecting the HCT116 cell line into BALB/C nude mice. Twenty-four tumor-bearing mice were randomly divided into four groups and injected with bevacizumab combined with TH-302 (A), bevacizumab (B), TH-302 (C), or saline (D) on days 1, 4, 7, 10 and 13. Functional MRI was performed before and at 3, 6, 9, 12 and 15 days after treatment. Pathologic examinations, including HE staining, HIF-1α and CD31 immunohistochemical staining, and TUNEL and Ki-67 immunofluorescent staining, were performed after the last scan. Results At the end of the study, Group A showed the lowest tumor volume, followed by Groups B, C, and D (F=120.652, P<0.001). For pathologic examinations, Group A showed the lowest percentage of CD31 staining (F=73.211, P<0.001) and Ki-67 staining (F=231.170, P<0.001), as well as the highest percentage of TUNEL staining (F=74.012, P<0.001). Moreover, the D* and f values exhibited positive correlations with CD31 (r=0.868, P<0.001, and r=0.698, P=0.012, respectively). R2* values was positively correlated with HIF-1α (r=0.776, P=0.003). D values were positively correlated with TUNEL (r=0.737, P=0.006) and negatively correlated with Ki-67 (r=0.912, P<0.001). The standard ADC values were positive correlated with TUNEL (r=0.672, P=0.017) and negative correlated with Ki-67 (r=0.873, P<0.001). Conclusion Anti-angiogenic agents combined with HAP can inhibit tumor growth effectively. In addition, IVIM-DWI and BOLD-MRI can be used to monitor the tumor microenvironment, including perfusion, hypoxia, cell apoptosis and proliferation, in a noninvasive manner.
Collapse
Affiliation(s)
- Mengjie Ma
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jianye Liang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Dong Zhang
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xi Xu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qingqing Cheng
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zeyu Xiao
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Changzheng Shi
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liangping Luo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
34
|
Thiruthaneeswaran N, Bibby BAS, Yang L, Hoskin PJ, Bristow RG, Choudhury A, West C. Lost in application: Measuring hypoxia for radiotherapy optimisation. Eur J Cancer 2021; 148:260-276. [PMID: 33756422 DOI: 10.1016/j.ejca.2021.01.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022]
Abstract
The history of radiotherapy is intertwined with research on hypoxia. There is level 1a evidence that giving hypoxia-targeting treatments with radiotherapy improves locoregional control and survival without compromising late side-effects. Despite coming in and out of vogue over decades, there is now an established role for hypoxia in driving molecular alterations promoting tumour progression and metastases. While tumour genomic complexity and immune profiling offer promise, there is a stronger evidence base for personalising radiotherapy based on hypoxia status. Despite this, there is only one phase III trial targeting hypoxia modification with full transcriptomic data available. There are no biomarkers in routine use for patients undergoing radiotherapy to aid management decisions, and a roadmap is needed to ensure consistency and provide a benchmark for progression to application. Gene expression signatures address past limitations of hypoxia biomarkers and could progress biologically optimised radiotherapy. Here, we review recent developments in generating hypoxia gene expression signatures and highlight progress addressing the challenges that must be overcome to pave the way for their clinical application.
Collapse
Affiliation(s)
- Niluja Thiruthaneeswaran
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Becky A S Bibby
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Lingjang Yang
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Mount Vernon Cancer Centre, Northwood, UK
| | - Robert G Bristow
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
35
|
Abstract
Viral infections lead to the death of more than a million people each year around the world, both directly and indirectly. Viruses interfere with many cell functions, particularly critical pathways for cell death, by affecting various intracellular mediators. MicroRNAs (miRNAs) are a major example of these mediators because they are involved in many (if not most) cellular mechanisms. Virus-regulated miRNAs have been implicated in three cell death pathways, namely, apoptosis, autophagy, and anoikis. Several molecules (e.g., BECN1 and B cell lymphoma 2 [BCL2] family members) are involved in both apoptosis and autophagy, while activation of anoikis leads to cell death similar to apoptosis. These mechanistic similarities suggest that common regulators, including some miRNAs (e.g., miR-21 and miR-192), are involved in different cell death pathways. Because the balance between cell proliferation and cell death is pivotal to the homeostasis of the human body, miRNAs that regulate cell death pathways have drawn much attention from researchers. miR-21 is regulated by several viruses and can affect both apoptosis and anoikis via modulating various targets, such as PDCD4, PTEN, interleukin (IL)-12, Maspin, and Fas-L. miR-34 can be downregulated by viral infection and has different effects on apoptosis, depending on the type of virus and/or host cell. The present review summarizes the existing knowledge on virus-regulated miRNAs involved in the modulation of cell death pathways. Understanding the mechanisms for virus-mediated regulation of cell death pathways could provide valuable information to improve the diagnosis and treatment of many viral diseases.
Collapse
|
36
|
Hypoxia-Driven Effects in Cancer: Characterization, Mechanisms, and Therapeutic Implications. Cells 2021; 10:cells10030678. [PMID: 33808542 PMCID: PMC8003323 DOI: 10.3390/cells10030678] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Hypoxia, a common feature of solid tumors, greatly hinders the efficacy of conventional cancer treatments such as chemo-, radio-, and immunotherapy. The depletion of oxygen in proliferating and advanced tumors causes an array of genetic, transcriptional, and metabolic adaptations that promote survival, metastasis, and a clinically malignant phenotype. At the nexus of these interconnected pathways are hypoxia-inducible factors (HIFs) which orchestrate transcriptional responses under hypoxia. The following review summarizes current literature regarding effects of hypoxia on DNA repair, metastasis, epithelial-to-mesenchymal transition, the cancer stem cell phenotype, and therapy resistance. We also discuss mechanisms and pathways, such as HIF signaling, mitochondrial dynamics, exosomes, and the unfolded protein response, that contribute to hypoxia-induced phenotypic changes. Finally, novel therapeutics that target the hypoxic tumor microenvironment or interfere with hypoxia-induced pathways are reviewed.
Collapse
|
37
|
Łukasik P, Załuski M, Gutowska I. Cyclin-Dependent Kinases (CDK) and Their Role in Diseases Development-Review. Int J Mol Sci 2021; 22:ijms22062935. [PMID: 33805800 PMCID: PMC7998717 DOI: 10.3390/ijms22062935] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) are involved in many crucial processes, such as cell cycle and transcription, as well as communication, metabolism, and apoptosis. The kinases are organized in a pathway to ensure that, during cell division, each cell accurately replicates its DNA, and ensure its segregation equally between the two daughter cells. Deregulation of any of the stages of the cell cycle or transcription leads to apoptosis but, if uncorrected, can result in a series of diseases, such as cancer, neurodegenerative diseases (Alzheimer’s or Parkinson’s disease), and stroke. This review presents the current state of knowledge about the characteristics of cyclin-dependent kinases as potential pharmacological targets.
Collapse
Affiliation(s)
- Paweł Łukasik
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Michał Załuski
- Department of Pharmaceutical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
- Correspondence:
| |
Collapse
|
38
|
Zhang T, Zhang Y, Liu X, Xu H, Chen C, Zhou X, Liu Y, Ma X. Application of Radiomics Analysis Based on CT Combined With Machine Learning in Diagnostic of Pancreatic Neuroendocrine Tumors Patient's Pathological Grades. Front Oncol 2021; 10:521831. [PMID: 33643890 PMCID: PMC7905094 DOI: 10.3389/fonc.2020.521831] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/11/2020] [Indexed: 02/05/2023] Open
Abstract
Purpose To evaluate the value of multiple machine learning methods in classifying pathological grades (G1,G2, and G3), and to provide the best machine learning method for the identification of pathological grades of pancreatic neuroendocrine tumors (PNETs) based on radiomics. Materials and Methods A retrospective study was conducted on 82 patients with Pancreatic Neuroendocrine tumors. All patients had definite pathological diagnosis and grading results. Using Lifex software to extract the radiomics features from CT images manually. The sensitivity, specificity, area under the curve (AUC) and accuracy were used to evaluate the performance of the classification model. Result Our analysis shows that the CT based radiomics features combined with multi algorithm machine learning method has a strong ability to identify the pathological grades of pancreatic neuroendocrine tumors. DC + AdaBoost, DC + GBDT, and Xgboost+RF were very valuable for the differential diagnosis of three pathological grades of PNET. They showed a strong ability to identify the pathological grade of pancreatic neuroendocrine tumors. The validation set AUC of DC + AdaBoost is 0.82 (G1 vs G2), 0.70 (G2 vs G3), and 0.85 (G1 vs G3), respectively. Conclusion In conclusion, based on enhanced CT radiomics features could differentiate between different pathological grades of pancreatic neuroendocrine tumors. Feature selection method Distance Correlation + classifier method Adaptive Boosting show a good application prospect.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - YueHua Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xinglong Liu
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hanyue Xu
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chaoyue Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuan Zhou
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yichun Liu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Yang Y, Gu Z, Tang J, Zhang M, Yang Y, Song H, Yu C. MnO 2 Nanoflowers Induce Immunogenic Cell Death under Nutrient Deprivation: Enabling an Orchestrated Cancer Starvation-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002667. [PMID: 33643794 PMCID: PMC7887587 DOI: 10.1002/advs.202002667] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/11/2020] [Indexed: 05/24/2023]
Abstract
MnO2 nanoparticles have been widely employed in cancer immunotherapy, playing a subsidiary role in assisting immunostimulatory drugs by improving their pharmacokinetics and/or creating a favorable microenvironment. Here, the stereotype of the subsidiary role of MnO2 nanoparticles in cancer immunotherapy is challenged. This study unravels an intrinsic immunomodulatory property of MnO2 nanoparticles as a unique nutrient-responsive immunogenic cell death (ICD) inducer, capable of directly modulating immunosurveillance toward tumor cells. MnO2 nanoflowers (MNFs) constructed via a one pot self-assembly approach selectively induce ICD to nutrient-deprived but not nutrient-replete cancer cells, which is confirmed by the upregulated damage associated molecular patterns in vitro and a prophylactic vaccination in vivo. The underlying mechanism of the MNFs-mediated selective ICD induction is likely associated with the concurrently upregulated oxidative stress and autophagy. Built on their unique immunomodulatory properties, an innovative nanomaterials orchestrated cancer starvation-immunotherapy is successfully developed, which is realized by the in situ vaccination with MNFs and vascular disrupting agents that cut off intratumoral nutrient supply, eliciting potent efficacy for suppressing local and distant tumors. These findings open up a new avenue toward biomedical applications of MnO2 materials, enabling an innovative therapeutics paradigm with great clinical significance.
Collapse
Affiliation(s)
- Yannan Yang
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaBrisbaneQLD4072Australia
| | - Zhengying Gu
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaBrisbaneQLD4072Australia
| | - Jie Tang
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaBrisbaneQLD4072Australia
| | - Min Zhang
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaBrisbaneQLD4072Australia
- School of Chemistry and Molecular EngineeringEast China Normal UniversityShanghai200241P. R. China
| | - Yang Yang
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaBrisbaneQLD4072Australia
| | - Hao Song
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaBrisbaneQLD4072Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaBrisbaneQLD4072Australia
- School of Chemistry and Molecular EngineeringEast China Normal UniversityShanghai200241P. R. China
| |
Collapse
|
40
|
Chuang CH, Dorsch M, Dujardin P, Silas S, Ueffing K, Hölken JM, Yang D, Winslow MM, Grüner BM. Altered Mitochondria Functionality Defines a Metastatic Cell State in Lung Cancer and Creates an Exploitable Vulnerability. Cancer Res 2021; 81:567-579. [PMID: 33239425 PMCID: PMC8137518 DOI: 10.1158/0008-5472.can-20-1865] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/05/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
Lung cancer is a prevalent and lethal cancer type that leads to more deaths than the next four major cancer types combined. Metastatic cancer spread is responsible for most cancer-related deaths but the cellular changes that enable cancer cells to leave the primary tumor and establish inoperable and lethal metastases remain poorly understood. To uncover genes that are specifically required to sustain metastasis survival or growth, we performed a genome-scale pooled lentiviral-shRNA library screen in cells that represent nonmetastatic and metastatic states of lung adenocarcinoma. Mitochondrial ribosome and mitochondria-associated genes were identified as top gene sets associated with metastasis-specific lethality. Metastasis-derived cell lines in vitro and metastases analyzed ex vivo from an autochthonous lung cancer mouse model had lower mitochondrial membrane potential and reduced mitochondrial functionality than nonmetastatic primary tumors. Electron microscopy of metastases uncovered irregular mitochondria with bridging and loss of normal membrane structure. Consistent with these findings, compounds that inhibit mitochondrial translation or replication had a greater effect on the growth of metastasis-derived cells. Finally, mice with established tumors developed fewer metastases upon treatment with phenformin in vivo. These results suggest that the metastatic cell state in lung adenocarcinoma is associated with a specifically altered mitochondrial functionality that can be therapeutically exploited. SIGNIFICANCE: This study characterizes altered mitochondria functionality of the metastatic cell state in lung cancer and opens new avenues for metastasis-specific therapeutic targeting.
Collapse
Affiliation(s)
- Chen-Hua Chuang
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Madeleine Dorsch
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University of Duisburg-Essen, Essen, Germany
| | - Philip Dujardin
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University of Duisburg-Essen, Essen, Germany
| | - Sukrit Silas
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California
| | - Kristina Ueffing
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University of Duisburg-Essen, Essen, Germany
| | - Johanna M Hölken
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University of Duisburg-Essen, Essen, Germany
| | - Dian Yang
- Cancer Biology Program, Stanford University School of Medicine, Stanford, California
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, California
- Cancer Biology Program, Stanford University School of Medicine, Stanford, California
- Department of Pathology, Stanford University School of Medicine, Stanford, California
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Barbara M Grüner
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University of Duisburg-Essen, Essen, Germany.
- German Cancer Consortium (DKTK) partner site Essen, Essen, Germany
| |
Collapse
|
41
|
Yu Y, Xiang K, Xu M, Li Y, Cui J, Zhang L, Tang X, Zhu X, Qian L, Zhang M, Yang Y, Yu Q, Shen Y, Gan Z. Prodrug Nanomedicine Inhibits Chemotherapy-Induced Proliferative Burst by Altering the Deleterious Intercellular Communication. ACS NANO 2021; 15:781-796. [PMID: 33410660 DOI: 10.1021/acsnano.0c07113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chemotherapy is one of the most commonly used clinical antitumor strategies. However, the therapy-induced proliferative burst, which always accompanies drug resistance and metastasis, has become a major obstacle during treatment. Except for some endogenous cellular or genetic mechanisms and some microenvironmental selection pressures, the intercellular connections in the tumor microenvironment (TME) are also thought to be the driving force for the acquired drug resistance and proliferative burst. Even though some pathway inhibitors or cell exempting strategies could be applied to partially avoid these unwanted communications, the complexity of the TME and the limited knowledge about those unknown detrimental connections might greatly compromise the efforts. Therefore, a more broad-spectrum strategy is urgently needed to relieve the drug-induced burst proliferation during various treatments. In this article, based on the possible discrepancies in metabolic activity between cells with different growth rates, several ester-bond-based prodrugs were synthesized. After screening, 7-ethyl-10-hyodroxycamptothecin-based prodrug nanoparticles were found to efficiently overcome the paclitaxel resistance, to selectively act on the malignantly proliferated drug-resistant cells and, furthermore, to greatly diminish the proliferative effect of common cytotoxic agents by blocking the detrimental intercellular connections. With the discriminating ability against malignant proliferating cells, the as-prepared prodrug nanomedicine exhibited significant anticancer efficacy against both drug-sensitive and drug-resistant tumor models, either by itself or by combining with highly potent nonselective chemotherapeutics. This work provides a different perspective and a possible solution for the treatment of therapy-induced burst proliferation.
Collapse
Affiliation(s)
- Yanting Yu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Keqi Xiang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mingzhi Xu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yuqiang Li
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiajunzi Cui
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lanqiong Zhang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaohu Tang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xianqi Zhu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lili Qian
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Meng Zhang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yan Yang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qingsong Yu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zhihua Gan
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
42
|
Weissinger M, Vogel J, Kupferschläger J, Dittmann H, Castaneda Vega SG, Grosse U, Artzner C, Nikolaou K, la Fougere C, Grözinger G. Correlation of C-arm CT acquired parenchymal blood volume (PBV) with 99mTc-macroaggregated albumin (MAA) SPECT/CT for radioembolization work-up. PLoS One 2020; 15:e0244235. [PMID: 33378338 PMCID: PMC7773241 DOI: 10.1371/journal.pone.0244235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/05/2020] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE SPECT/CT with 99mTc-macroaggregated albumin (MAA) is generally used for diagnostic work-up prior to transarterial radioembolization (TARE) to exclude shunts and to provide additional information for treatment stratification and dose calculation. C-arm CT is used for determination of lobular vascular supply and assessment of parenchymal blood volume (PBV). Aim of this study was to correlate MAA-uptake and PBV-maps in hepatocellular carcinoma (HCC) and hepatic metastases of the colorectal carcinoma (CRC). MATERIALS AND METHODS 34 patients underwent a PBV C-arm CT immediately followed by 99mTc-MAA injection and a SPECT/CT acquisition after 1 h uptake. MAA-uptake and PBV-maps were visually assessed and semi-quantitatively analyzed (MAA-tumor/liver-parenchyma = MAA-TBR or PBV in ml/100ml). In case of a poor match, tumors were additionally correlated with post-TARE 90Y-Bremsstrahlung-SPECT/CT as a reference. RESULTS 102 HCC or CRC metastases were analyzed. HCC presented with significantly higher MAA-TBR (7.6 vs. 3.9, p<0.05) compared to CRC. Tumors showed strong intra- and inter-individual dissimilarities between TBR and PBV with a weak correlations for capsular HCCs (r = 0.45, p<0.05) and no correlation for CRC. The demarcation of lesions was slightly better for both HCC and CRC in PBV-maps compared to MAA-SPECT/CT (exact match: 52%/50%; same intensity/homogeneity: 38%/39%; insufficient 10%/11%). MAA-SPECT/CT revealed a better visual correlation with post-therapeutic 90Y-Bremsstrahlung-SPECT/CT. CONCLUSION The acquisition of PBV can improve the detectability of small intrahepatic tumors and correlates with the MAA-Uptake in HCC. The results indicate that 99mTc-MAA-SPECT/CT remains to be the superior method for the prediction of post-therapeutic 90Y-particle distribution, especially in CRC. However, intra-procedural PBV acquisition has the potential to become an additional factor for TARE planning, in addition to improving the determination of segment and tumor blood supply, which has been demonstrated previously.
Collapse
Affiliation(s)
- Matthias Weissinger
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
| | - Jonas Vogel
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | - Jürgen Kupferschläger
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
| | - Helmut Dittmann
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
| | - Salvador Guillermo Castaneda Vega
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
- Department for Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tuebingen, Tuebingen, Germany
| | - Ulrich Grosse
- Department of Diagnostic and Interventional Radiology, Kantonsspital Frauenfeld, Frauenfeld, Switzerland
| | - Christoph Artzner
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany
- iFIT-Cluster of Excellence, Eberhard Karls University Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tuebingen, Tuebingen, Germany
| | - Christian la Fougere
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
- iFIT-Cluster of Excellence, Eberhard Karls University Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tuebingen, Tuebingen, Germany
- * E-mail:
| | - Gerd Grözinger
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
43
|
Ni XQ, Yin HK, Fan GH, Shi D, Xu L, Jin D. Differentiation of pulmonary sclerosing pneumocytoma from solid malignant pulmonary nodules by radiomic analysis on multiphasic CT. J Appl Clin Med Phys 2020; 22:158-164. [PMID: 33369106 PMCID: PMC7882110 DOI: 10.1002/acm2.13154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/11/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Purpose To investigate the diagnostic value and feasibility of radiomics‐based texture analysis in differentiating pulmonary sclerosing pneumocytoma (PSP) from solid malignant pulmonary nodules (SMPN) on single‐ and three‐phase computed tomography (CT) images. Materials and Methods A total of 25 PSP patients and 35 SMPN patients with pathologically confirmed results were retrospectively included in this study. For each patient, the tumor regions were manually labeled in images acquired at the noncontrast phase (NCP), arterial phase (AP), and venous phase (VP). The least absolute shrinkage and selection operator (LASSO) method was used to select the most useful predictive features extracted from the CT images. The predictive models that discriminate PSP from SMPN based on single‐phase CT images (NCP, AP, and VP) or three‐phase CT images (Combined model) were developed and validated through fivefold cross‐validation using a logistic regression classifier. Model performance was evaluated using receiver operating characteristic (ROC) analysis. The predictive performance was also compared between the Combined model and human readers. Results Four, five, and five features were selected from NCP, AP, and VP CT images for the development of radiomic models, respectively. The NCP, AP, and VP models exhibited areas under the curve (AUCs) of 0.748 (95% confidence interval [CI], 0.620–0.852), 0.749 (95% CI, 0.620–0.852), and 0.790 (95% CI, 0.665–0.884) in the validation dataset, respectively. The Combined model based on three‐phase CT images outperformed the NCP, AP, and VP models (all p < 0.05), yielding an AUC of 0.882 (95% CI, 0.773–0.951) in the validation dataset. The Combined model displayed noninferior performance compared to two senior radiologists; however, it outperformed two junior radiologists (p = 0.004 and 0.001, respectively). Conclusion The Combined model based on radiomic features extracted from three‐phase CT images achieved radiologist‐level performance and could be used as promising noninvasive tool to differentiate PSP from SMPN.
Collapse
Affiliation(s)
- Xiao-Qiong Ni
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong-Kun Yin
- Beijing Infervision Technology Co.,Ltd, Beijing, China
| | - Guo-Hua Fan
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dai Shi
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Xu
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dan Jin
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
44
|
Contrast-Enhanced CT-based Textural Parameters as Potential Prognostic Factors of Survival for Colorectal Cancer Patients Receiving Targeted Therapy. Mol Imaging Biol 2020; 23:427-435. [PMID: 33108800 DOI: 10.1007/s11307-020-01552-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 09/07/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE This study was designed to estimate the clinical significance of the contrast-enhanced computed tomography (CT) textural features for prediction of survival in colorectal cancer (CRC) patients receiving targeted therapy (bevacizumab and cetuximab). PROCEDURES The LifeX software was used to extract the textural parameters of the tumor lesions in the contrast-enhanced CT. We used the least absolute shrinkage and selection operator (LASSO) Cox regression and random forest method to screen the non-redundant radiomic features and constructed the CT imaging score. Univariate and multivariate analyses through the Cox proportional hazards model were performed to assess the prognostic clinical factor. Based on the result of multivariate analysis and CT imaging score, combined nomogram model was constructed to predict the overall survival (OS) of patients. Decision curves analysis was employed to evaluate the performance of the combined model and clinical model. RESULTS After comparative analysis of the area under curve of the receiver operating characteristic (ROC) curve, we chose the result of random forest model as CT imaging score. Considering the clinical practice and the result of analysis, age, surgery, and lactate dehydrogenase (LDH) level have been introduced into clinical model. Based on the result of analysis and the CT imaging score, we constructed the nomogram combined model. C-index and calibration curve verified the goodness of fit and discrimination of the combined model. Decision curve analysis (DCA) demonstrated that the combined model showed the better net benefit for a 3-year OS than clinical model. CONCLUSIONS In conclusion, the study provides preliminary evidences that several radiomic parameters of tumor lesions derived from CT images were prognostic factors and predictive markers for CRC patients who are candidates for targeted therapy (bevacizumab and cetuximab).
Collapse
|
45
|
Li M, Zhu YZ, Zhang YC, Yue YF, Yu HP, Song B. Radiomics of rectal cancer for predicting distant metastasis and overall survival. World J Gastroenterol 2020; 26:5008-5021. [PMID: 32952346 PMCID: PMC7476170 DOI: 10.3748/wjg.v26.i33.5008] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/16/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Rectal cancer (RC) patient stratification by different factors may yield variable results. Therefore, more efficient prognostic biomarkers are needed for improved risk stratification, personalized treatment, and prognostication of RC patients.
AIM To build a novel model for predicting the presence of distant metastases and 3-year overall survival (OS) in RC patients.
METHODS This was a retrospective analysis of 148 patients (76 males and 72 females) with RC treated with curative resection, without neoadjuvant or postoperative chemoradiotherapy, between October 2012 and December 2015. These patients were allocated to a training or validation set, with a ratio of 7:3. Radiomic features were extracted from portal venous phase computed tomography (CT) images of RC. The least absolute shrinkage and selection operator regression analysis was used for feature selection. Multivariate logistic regression analysis was used to develop the radiomics signature (Rad-score) and the clinicoradiologic risk model (the combined model). Receiver operating characteristic curves were constructed to evaluate the diagnostic performance of the models for predicting distant metastasis of RC. The association of the combined model with 3-year OS was investigated by Kaplan-Meier survival analysis.
RESULTS A total of 51 (34.5%) patients had distant metastases, while 26 (17.6%) patients died, and 122 (82.4%) patients lived at least 3 years post-surgery. The values of both the Rad-score (consisted of three selected features) and the combined model were significantly different between the distant metastasis group and the non-metastasis group (0.46 ± 0.21 vs 0.32 ± 0.24 for the Rad-score, and 0.60 ± 0.23 vs 0.28 ± 0.26 for the combined model; P < 0.001 for both models). Predictors contained in the combined model included the Rad-score, pathological N-stage, and T-stage. The addition of histologic grade to the model failed to show incremental prognostic value. The combined model showed good discrimination, with areas under the curve of 0.842 and 0.802 for the training set and validation set, respectively. For the survival analysis, the combined model was associated with an improved OS in the whole cohort and the respective subgroups.
CONCLUSION This study presents a clinicoradiologic risk model, visualized in a nomogram, that can be used to facilitate individualized prediction of distant metastasis and 3-year OS in patients with RC.
Collapse
Affiliation(s)
- Mou Li
- Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yu-Zhou Zhu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yong-Chang Zhang
- Department of Radiology, Chengdu Seventh People’s Hospital, Chengdu 610213, Sichuan Province, China
| | - Yu-Feng Yue
- Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hao-Peng Yu
- Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bin Song
- Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
46
|
Interaction between Immunotherapy and Antiangiogenic Therapy for Cancer. Molecules 2020; 25:molecules25173900. [PMID: 32859106 PMCID: PMC7504110 DOI: 10.3390/molecules25173900] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022] Open
Abstract
Although immunotherapy has led to durable responses in diverse cancers, unfortunately, there has been limited efficacy and clinical response rates due to primary or acquired resistance to immunotherapy. To maximize the potential of immunotherapy, combination therapy with antiangiogenic drugs seems to be promising. Some phase III trials showed superiority for survival with the combination of immunotherapy and antiangiogenic therapy. In this study, we describe a synergistic mechanism of immunotherapy and antiangiogenic therapy and summarize current clinical trials of these combinations.
Collapse
|
47
|
Chen L, Wang H, Zeng H, Zhang Y, Ma X. Evaluation of CT-based radiomics signature and nomogram as prognostic markers in patients with laryngeal squamous cell carcinoma. Cancer Imaging 2020; 20:28. [PMID: 32321585 PMCID: PMC7178759 DOI: 10.1186/s40644-020-00310-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/15/2020] [Indexed: 02/05/2023] Open
Abstract
Background The aim of this study was to evaluate the prognostic value of radiomics signature and nomogram based on contrast-enhanced computed tomography (CT) in patients after surgical resection of laryngeal squamous cell carcinoma (LSCC). Methods All patients (n = 136) were divided into the training cohort (n = 96) and validation cohort (n = 40). The LASSO regression method was performed to construct radiomics signature from CT texture features. Then a radiomics nomogram incorporating the radiomics signature and clinicopathologic factors was established to predict overall survival (OS). The validation of nomogram was evaluated by calibration curve, concordance index (C-index) and decision curve. Results Based on three selected texture features, the radiomics signature showed high C-indexes of 0.782 (95%CI: 0.656–0.909) and 0.752 (95%CI, 0.614–0.891) in the two cohorts. The radiomics nomogram had significantly better discrimination capability than cancer staging in the training cohort (C-index, 0.817 vs. 0.682; P = 0.009) and validation cohort (C-index, 0.913 vs. 0.699; P = 0.019), as well as a good agreement between predicted and actual survival in calibration curves. Decision curve analysis also suggested improved clinical utility of radiomics nomogram. Conclusions Radiomics signature and nomogram showed favorable prediction accuracy for OS, which might facilitate the individualized risk stratification and clinical decision-making in LSCC patients.
Collapse
Affiliation(s)
- Linyan Chen
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center, No.37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Haiyang Wang
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hao Zeng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center, No.37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Yi Zhang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center, No.37, Guoxue Alley, Chengdu, 610041, People's Republic of China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center, No.37, Guoxue Alley, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
48
|
Ye J, Ling J, Lv Y, Chen J, Cai J, Chen M. Pulmonary adenocarcinoma appearing as ground-glass opacity nodules identified using non-enhanced and contrast-enhanced CT texture analysis: A retrospective analysis. Exp Ther Med 2020; 19:2483-2490. [PMID: 32256725 PMCID: PMC7086215 DOI: 10.3892/etm.2020.8511] [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: 07/29/2019] [Accepted: 12/04/2019] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the ability of CT-based texture analysis to differentiate invasive adenocarcinoma (IA) from pre-invasive lesions (PIL) or minimally IA (MIA) appearing as ground-glass opacity (GGO) nodules, and to further compare the performance of non-enhanced CT (NECT) images with that of contrast-enhanced CT (CECT) images. A total of 77 patients with GGO nodules and surgically confirmed pulmonary adenocarcinoma were included in the present retrospective study. Each GGO nodule was manually segmented and its texture features were extracted from NECT and CECT images using in-house developed software coded in MATLAB (MathWorks). The independent-samples t-test was used to select the texture features with statistically significant differences between IA and MIA/PIL. Multivariate logistic regression and receiver operating characteristics (ROC) curve analyses were performed to identify predictive features. Of the 77 GGO nodules, 12 were atypical adenomatous hyperplasia or adenocarcinoma in situ (15.6%), 36 were MIA (46.8%) and 29 were IA (37.7%). IA and MIA/PIL exhibited significant differences in most histogram features and gray-level co-occurrence matrix features (P<0.05). Multivariate logistic regression and ROC curve analyses revealed that smaller energy and higher entropy were significant differentiators of IA from MIA and PIL, irrespective of whether NECT images [area under the curve (AUC): 0.839, 0.859] or CECT images (AUC: 0.818, 0.820) are used. Texture analysis of CT images, regardless of whether NECT or CECT is used, has the potential to distinguish IA from PIL or MIA, particularly the parameters of energy and entropy. Furthermore, NECT images were simpler to obtain and no contrast agent was required; thus, analysis with NECT may be a preferred choice.
Collapse
Affiliation(s)
- Jing Ye
- Department of Medical Imaging, Yangzhou University Clinical College Subei People's Hospital, Yangzhou, Jiangsu 225002, P.R. China
| | - Jun Ling
- Department of Medical Imaging, Yangzhou University Clinical College Subei People's Hospital, Yangzhou, Jiangsu 225002, P.R. China
| | - Yan Lv
- Department of Medical Imaging, Yangzhou University Clinical College Subei People's Hospital, Yangzhou, Jiangsu 225002, P.R. China
| | - Juan Chen
- Department of Medical Imaging, Yangzhou University Clinical College Subei People's Hospital, Yangzhou, Jiangsu 225002, P.R. China
| | - Junhui Cai
- Department of Medical Imaging, Yangzhou University Clinical College Subei People's Hospital, Yangzhou, Jiangsu 225002, P.R. China
| | - Mingxiang Chen
- Department of Medical Imaging, Yangzhou University Clinical College Subei People's Hospital, Yangzhou, Jiangsu 225002, P.R. China
| |
Collapse
|
49
|
Oh J, Lee JM, Park J, Joo I, Yoon JH, Lee DH, Ganeshan B, Han JK. Hepatocellular Carcinoma: Texture Analysis of Preoperative Computed Tomography Images Can Provide Markers of Tumor Grade and Disease-Free Survival. Korean J Radiol 2020; 20:569-579. [PMID: 30887739 PMCID: PMC6424831 DOI: 10.3348/kjr.2018.0501] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 09/29/2018] [Indexed: 11/25/2022] Open
Affiliation(s)
- Jiseon Oh
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Min Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea.
| | - Junghoan Park
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Ijin Joo
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Hee Yoon
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ho Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Balaji Ganeshan
- Institute of Nuclear Medicine, University College London, London, UK
| | - Joon Koo Han
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
50
|
Xu Y, Xu Q, Ma Y, Duan J, Zhang H, Liu T, Li L, Sun H, Shi K, Xie S, Wang W. Characterizing MRI features of rectal cancers with different KRAS status. BMC Cancer 2019; 19:1111. [PMID: 31727020 PMCID: PMC6857233 DOI: 10.1186/s12885-019-6341-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND To investigate whether MRI findings, including texture analysis, can differentiate KRAS mutation status in rectal cancer. METHODS Totally, 158 patients with pathologically proved rectal cancers and preoperative pelvic MRI examinations were enrolled. Patients were stratified into two groups: KRAS wild-type group (KRASwt group) and KRAS mutation group (KRASmt group) according to genomic DNA extraction analysis. MRI findings of rectal cancers (including texture features) and relevant clinical characteristics were statistically evaluated to identify the differences between the two groups. The independent samples t test or Mann-Whitney U test were used for continuous variables. The differences of the remaining categorical polytomous variables were analyzed using the Chi-square test or Fisher exact test. A receiver operating characteristic (ROC) curve analysis was performed to evaluate the discriminatory power of MRI features. The area under the ROC curve (AUC) and the optimal cut-off values were calculated using histopathology diagnosis as a reference; meanwhile, sensitivity and specificity were determined. RESULTS Mean values of six texture parameters (Mean, Variance, Skewness, Entropy, gray-level nonuniformity, run-length nonuniformity) were significantly higher in KRASmt group compared to KRASwt group (p < 0.0001, respectively). The AUC values of texture features ranged from 0.703~0.813. In addition, higher T stage and lower ADC values were observed in the KRASmt group compared to KRASwt group (t = 7.086, p = 0.029; t = - 2.708, p = 0.008). CONCLUSION The MRI findings of rectal cancer, especially texture features, showed an encouraging value for identifying KRAS status.
Collapse
Affiliation(s)
- Yanyan Xu
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Qiaoyu Xu
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Yanhui Ma
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Jianghui Duan
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Haibo Zhang
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Tongxi Liu
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Lu Li
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Hongliang Sun
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China.
| | - Kaining Shi
- Philips Healthcare, Beijing, 100001, People's Republic of China
| | - Sheng Xie
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Wu Wang
- Department of Radiology, China-Japan Friendship Hospital, No.2 Yinghua East Street, Chaoyang District, Beijing, 100029, People's Republic of China
| |
Collapse
|