1
|
Mokhtari A, Casale R, Salahuddin Z, Paquier Z, Guiot T, Woodruff HC, Lambin P, Van Laethem JL, Hendlisz A, Bali MA. Development of Clinical Radiomics-Based Models to Predict Survival Outcome in Pancreatic Ductal Adenocarcinoma: A Multicenter Retrospective Study. Diagnostics (Basel) 2024; 14:712. [PMID: 38611625 PMCID: PMC11011556 DOI: 10.3390/diagnostics14070712] [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: 02/17/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
PURPOSE This multicenter retrospective study aims to identify reliable clinical and radiomic features to build machine learning models that predict progression-free survival (PFS) and overall survival (OS) in pancreatic ductal adenocarcinoma (PDAC) patients. METHODS Between 2010 and 2020 pre-treatment contrast-enhanced CT scans of 287 pathology-confirmed PDAC patients from two sites of the Hopital Universitaire de Bruxelles (HUB) and from 47 hospitals within the HUB network were retrospectively analysed. Demographic, clinical, and survival data were also collected. Gross tumour volume (GTV) and non-tumoral pancreas (RPV) were semi-manually segmented and radiomics features were extracted. Patients from two HUB sites comprised the training dataset, while those from the remaining 47 hospitals of the HUB network constituted the testing dataset. A three-step method was used for feature selection. Based on the GradientBoostingSurvivalAnalysis classifier, different machine learning models were trained and tested to predict OS and PFS. Model performances were assessed using the C-index and Kaplan-Meier curves. SHAP analysis was applied to allow for post hoc interpretability. RESULTS A total of 107 radiomics features were extracted from each of the GTV and RPV. Fourteen subgroups of features were selected: clinical, GTV, RPV, clinical & GTV, clinical & GTV & RPV, GTV-volume and RPV-volume both for OS and PFS. Subsequently, 14 Gradient Boosting Survival Analysis models were trained and tested. In the testing dataset, the clinical & GTV model demonstrated the highest performance for OS (C-index: 0.72) among all other models, while for PFS, the clinical model exhibited a superior performance (C-index: 0.70). CONCLUSIONS An integrated approach, combining clinical and radiomics features, excels in predicting OS, whereas clinical features demonstrate strong performance in PFS prediction.
Collapse
Affiliation(s)
- Ayoub Mokhtari
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Roberto Casale
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Zohaib Salahuddin
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
| | - Zelda Paquier
- Medical Physics Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Thomas Guiot
- Medical Physics Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Henry C. Woodruff
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW—School for Oncology and Reproduction, Maastricht University Medical Centre+, 6229HX Maastricht, The Netherlands
| | - Philippe Lambin
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW—School for Oncology and Reproduction, Maastricht University Medical Centre+, 6229HX Maastricht, The Netherlands
| | - Jean-Luc Van Laethem
- Department of Gastroenterology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Alain Hendlisz
- Department of Gastroenterology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Maria Antonietta Bali
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|
2
|
Ahmed TM, Kawamoto S, Hruban RH, Fishman EK, Soyer P, Chu LC. A primer on artificial intelligence in pancreatic imaging. Diagn Interv Imaging 2023; 104:435-447. [PMID: 36967355 DOI: 10.1016/j.diii.2023.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 06/18/2023]
Abstract
Artificial Intelligence (AI) is set to transform medical imaging by leveraging the vast data contained in medical images. Deep learning and radiomics are the two main AI methods currently being applied within radiology. Deep learning uses a layered set of self-correcting algorithms to develop a mathematical model that best fits the data. Radiomics converts imaging data into mineable features such as signal intensity, shape, texture, and higher-order features. Both methods have the potential to improve disease detection, characterization, and prognostication. This article reviews the current status of artificial intelligence in pancreatic imaging and critically appraises the quality of existing evidence using the radiomics quality score.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ralph H Hruban
- Sol Goldman Pancreatic Research Center, Department of Pathology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Philippe Soyer
- Université Paris Cité, Faculté de Médecine, Department of Radiology, Hôpital Cochin-APHP, 75014, 75006, Paris, France, 7501475006
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
3
|
Shi C, Chen W, Davis R, Morse MA. Venous Invasion in Pancreatic Neuroendocrine Tumors Is Independently Associated With Disease-free Survival and Overall Survival. Am J Surg Pathol 2023; 47:678-685. [PMID: 37017316 DOI: 10.1097/pas.0000000000002038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
In this study, we evaluated venous invasion and its association with survival in patients with resected pancreatic neuroendocrine tumor (PanNET). Surgical Pathology Archives were searched for pancreatectomies performed for PanNET between October 1, 2005, and December 31, 2019. Hematoxylin and eosin (H&E)-stained slides were evaluated for venous invasion, and Movat's stain was performed in all cases with no venous invasion detected on H&E stains. Pathology reports and electronic medical records were also reviewed. Venous invasion was identified in 23 of 145 (15.9%) cases on H&E stains, and Movat's stain identified additional 34 cases with venous invasion (39.3% overall). Orphan arteries with adjacent well-defined tumor nodules or subtle hyalinizing nodules in hyalinizing tumors are highly specific for venous invasion. In stage I-III cases (n=122), venous invasion was associated with larger tumor size, higher World Health Organization (WHO) tumor grade, perineural invasion, extrapancreatic extension, lymph node metastasis, and liver metastasis ( P <0.05). In univariate analyses, tumor size, WHO grade, venous invasion, perineural invasion, T stage, and lymph node metastasis all correlated with disease-free survival; however, only venous invasion was associated with worse disease-free survival in multivariate analyses ( P <0.01). In all-stage cases, venous invasion was the only attributor associated with worse overall survival in multivariate analyses ( P =0.03). In summary, venous invasion in PanNET can be histologically subtle, and Movat's stain can greatly increase the detection rate. More importantly, enhanced venous invasion by Movat's stain correlates independently with disease-free survival in patients with stage I-III tumors and overall survival in all-stage patients.
Collapse
Affiliation(s)
| | | | | | - Michael A Morse
- Medicine, Medical Oncology, Duke University Medical Center, Durham, NC
| |
Collapse
|
4
|
Zeng C, He R, Dai Y, Lu X, Deng L, Zhu Q, Liu Y, Liu Q, Lu W, Wang Y, Jin J. Identification of TGF-β signaling-related molecular patterns, construction of a prognostic model, and prediction of immunotherapy response in gastric cancer. Front Pharmacol 2022; 13:1069204. [PMID: 36467074 PMCID: PMC9715605 DOI: 10.3389/fphar.2022.1069204] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/07/2022] [Indexed: 06/22/2024] Open
Abstract
Background: TGF-β signaling pathway plays an essential role in tumor progression and immune responses. However, the link between TGF-β signaling pathway-related genes (TSRGs) and clinical prognosis, tumor microenvironment (TME), and immunotherapy in gastric cancer is unclear. Methods: Transcriptome data and related clinical data of gastric cancer were downloaded from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, and 54 TSRGs were obtained from the Molecular Signatures Database (MSigDB). We systematically analyzed the expression profile characteristics of 54 TSRGs in 804 gastric cancer samples and examined the differences in prognosis, clinicopathological features, and TME among different molecular subtypes. Subsequently, TGF-β-related prognostic models were constructed using univariate and least absolute shrinkage and selection operator (LASSO) Cox regression analysis to quantify the degree of risk in each patient. Patients were divided into two high- and low-risk groups based on the median risk score. Finally, sensitivity to immune checkpoint inhibitors (ICIs) and anti-tumor agents was assessed in patients in high- and low-risk groups. Results: We identified two distinct TGF-β subgroups. Compared to TGF-β cluster B, TGF-β cluster A exhibits an immunosuppressive microenvironment with a shorter overall survival (OS). Then, a novel TGF-β-associated prognostic model, including SRPX2, SGCE, DES, MMP7, and KRT17, was constructed, and the risk score was demonstrated as an independent prognostic factor for gastric cancer patients. Further studies showed that gastric cancer patients in the low-risk group, characterized by higher tumor mutation burden (TMB), the proportion of high microsatellite instability (MSI-H), immunophenoscore (IPS), and lower tumor immune dysfunction and exclusion (TIDE) score, had a better prognosis, and linked to higher response rate to immunotherapy. In addition, the risk score and anti-tumor drug sensitivity were strongly correlated. Conclusion: These findings highlight the importance of TSRGs, deepen the understanding of tumor immune microenvironment, and guide individualized immunotherapy for gastric cancer patients.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Rong He
- Department of Medical Oncology, Shanghai Tenths People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuyang Dai
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaohuan Lu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Linghui Deng
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Zhu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Yu Liu
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Qian Liu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Wenbin Lu
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Yue Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianhua Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
5
|
Zhao S, Li J, Zhang H, Qi L, Du Y, Kogiso M, Braun FK, Xiao S, Huang Y, Li J, Teo WY, Lindsay H, Baxter P, Su JMF, Adesina A, Laczik M, Genevini P, Veillard AC, Schvartzman S, Berguet G, Ding SR, Du L, Stephan C, Yang J, Davies PJA, Lu X, Chintagumpala M, Parsons DW, Perlaky L, Xia YF, Man TK, Huang Y, Sun D, Li XN. Epigenetic Alterations of Repeated Relapses in Patient-matched Childhood Ependymomas. Nat Commun 2022; 13:6689. [PMID: 36335125 PMCID: PMC9637194 DOI: 10.1038/s41467-022-34514-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022] Open
Abstract
Recurrence is frequent in pediatric ependymoma (EPN). Our longitudinal integrated analysis of 30 patient-matched repeated relapses (3.67 ± 1.76 times) over 13 years (5.8 ± 3.8) reveals stable molecular subtypes (RELA and PFA) and convergent DNA methylation reprogramming during serial relapses accompanied by increased orthotopic patient derived xenograft (PDX) (13/27) formation in the late recurrences. A set of differentially methylated CpGs (DMCs) and DNA methylation regions (DMRs) are found to persist in primary and relapse tumors (potential driver DMCs) and are acquired exclusively in the relapses (potential booster DMCs). Integrating with RNAseq reveals differentially expressed genes regulated by potential driver DMRs (CACNA1H, SLC12A7, RARA in RELA and HSPB8, GMPR, ITGB4 in PFA) and potential booster DMRs (PLEKHG1 in RELA and NOTCH, EPHA2, SUFU, FOXJ1 in PFA tumors). DMCs predicators of relapse are also identified in the primary tumors. This study provides a high-resolution epigenetic roadmap of serial EPN relapses and 13 orthotopic PDX models to facilitate biological and preclinical studies.
Collapse
Affiliation(s)
- Sibo Zhao
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.413584.f0000 0004 0383 5679Jane and John Justin Neurosciences Center, Cook Children’s Medical Center, Fort Worth, TX 76104 USA ,grid.413584.f0000 0004 0383 5679Hematology and Oncology Center, Cook Children’s Medical Center, Fort Worth, TX 76104 USA
| | - Jia Li
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA ,grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA ,grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University; and Guangzhou Laboratory, Bioland, 510120 Guangzhou, Guangdong P. R. China
| | - Huiyuan Zhang
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Lin Qi
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Yuchen Du
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Mari Kogiso
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Frank K. Braun
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Sophie Xiao
- grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Yulun Huang
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.263761.70000 0001 0198 0694Department of Neurosurgery and Brain and Nerve Research Laboratory, the First Affiliated Hospital, and Department of Neurosurgery, Dushu Lake Hospital, Suzhou Medical College, Soochow University, 215007 Suzhou, P. R. China
| | - Jianfang Li
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Wan-Yee Teo
- grid.410724.40000 0004 0620 9745Humphrey Oei Institute of Cancer Research, National Cancer Center Singapore, Singapore, 169610 Singapore ,grid.428397.30000 0004 0385 0924Cancer and Stem Cell Biology Program, Duke-NUS Medical School Singapore, Singapore, Singapore ,grid.414963.d0000 0000 8958 3388KK Women’s & Children’s Hospital Singapore, Singapore, Singapore ,grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Holly Lindsay
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Patricia Baxter
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Jack M. F. Su
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Adekunle Adesina
- grid.39382.330000 0001 2160 926XDepartment of Pathology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Miklós Laczik
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Paola Genevini
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | | | - Sol Schvartzman
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Geoffrey Berguet
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Shi-Rong Ding
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Department of Radiation, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong P. R. China
| | - Liping Du
- grid.16753.360000 0001 2299 3507Clinical Cytogenetic Laboratory, Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Clifford Stephan
- grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA
| | - Jianhua Yang
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Peter J. A. Davies
- grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA
| | - Xinyan Lu
- grid.16753.360000 0001 2299 3507Clinical Cytogenetic Laboratory, Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Murali Chintagumpala
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Donald William Parsons
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Laszlo Perlaky
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Yun-Fei Xia
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Department of Radiation, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong P. R. China
| | - Tsz-Kwong Man
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Yun Huang
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Deqiang Sun
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Xiao-Nan Li
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
6
|
Jo JH, Jung DE, Lee HS, Park SB, Chung MJ, Park JY, Bang S, Park SW, Cho S, Song SY. A phase I/II study of ivaltinostat combined with gemcitabine and erlotinib in patients with untreated locally advanced or metastatic pancreatic adenocarcinoma. Int J Cancer 2022; 151:1565-1577. [PMID: 35657348 PMCID: PMC9545559 DOI: 10.1002/ijc.34144] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 12/03/2022]
Abstract
This phase I/II study evaluated the safety and efficacy of a new histone deacetylase (HDAC) inhibitor, ivaltinostat, in combination with gemcitabine and erlotinib for advanced pancreatic ductal adenocarcinoma (PDAC). Patients diagnosed with unresectable, histologically confirmed PDAC who had not undergone previous therapy were eligible. Phase I had a 3 + 3 dose escalation design to determine the maximum tolerable dose (MTD) of ivaltinostat (intravenously on days 1, 8 and 15) with gemcitabine (1000 mg/m2 intravenously on days 1, 8 and 15) and erlotinib (100 mg/day, orally) for a 28-day cycle. In phase II, patients received a six-cycle treatment with the MTD of ivaltinostat determined in phase I. The primary endpoint was the objective response rate (ORR). Secondary endpoints included overall survival (OS), disease control rate (DCR) and progression-free survival (PFS). The MTD of ivaltinostat for the phase II trial was determined to be 250 mg/m2 . In phase II, 24 patients were enrolled. The median OS and PFS were 8.6 (95% confidence interval [CI]: 5.3-11.2) and 5.3 months (95% CI: 3.7-5.8). Of the 16 patients evaluated for response, ORR and DCR were 25.0% and 93.8% with a median OS/PFS of 10.8 (95% CI: 8.3-16.7)/5.8 (95% CI: 4.6-6.7) months. Correlative studies showed that mutation burden detected by cfDNA and specific blood markers such as TIMP1, pro-MMP10, PECAM1, proMMP-2 and IGFBP1 were associated with clinical outcomes. Although the result of a small study, a combination of ivaltinostat, gemcitabine and erlotinib appeared to be a potential treatment option for advanced PDAC.
Collapse
Affiliation(s)
- Jung Hyun Jo
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Dawoon E. Jung
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Hee Seung Lee
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Soo Been Park
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Seung Woo Park
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Sangsook Cho
- CG PharmaceuticalsOrindaCaliforniaUSA
- CrystalGenomicsSeongnamsi, GyeonggidoSouth Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| |
Collapse
|
7
|
Ahn B, Jung JK, Jung H, Ryu YM, Kim YW, Song TJ, Park DH, Hwang DW, Cho H, Kim SY, Hong SM. Double Ki-67 and synaptophysin labeling in pancreatic neuroendocrine tumor biopsies. Pancreatology 2022; 22:427-434. [PMID: 35292233 DOI: 10.1016/j.pan.2022.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/21/2022] [Accepted: 03/06/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic neuroendocrine tumors (PanNETs) are frequently detected on endoscopic ultrasound-guided fine-needle aspiration biopsy (EUS-FNAB) specimens. The conventional methods for evaluating the Ki-67 labeling index (Ki67LI) in EUS-FNAB specimens are laborious, and their results are difficult to interpret. More practical and easy methods for evaluating the Ki67LI in PanNETs from EUS-FNAB specimens is increasing in need. METHODS We used double Ki-67 and synaptophysin (double Ki-Syn) antibody cocktail; Ki67LI, total Ki-67 positive cells, and total tumor cells were counted and compared with those detected on conventional single Ki-67 immunostaining (single Ki-67) of 96 PanNETs [Grade 1 (G1), 68 cases (71%); G2, 26 (27%); G3, 2 (2%)] from EUS-FNAB specimens. RESULTS The tumor grading between double Ki-Syn and single Ki-67 immunolabeling was highly concordant (correlation, 0.95; Fisher's exact test, P < 0.001). Seven EUS-FNAB specimens (7%) had discrepant results, of which 2 were removed through surgical resection and showed the same tumor grade as that detected on double Ki-Syn immunolabeling. Fifty-four specimens (56%) had higher Ki-67 positive tumor cell counts on single Ki-67 immunolabeling. Sixty-two specimens (65%) had higher total tumor cell counts on double Ki-Syn immunolabeling. The number of specimens with less than 500 total counted tumor cells were significantly reduced when double Ki-Syn immunolabeling was applied [P = 0.046; single Ki-67, 17 specimens (18%); double Ki-Syn, 9 specimens (9%)]. CONCLUSION Double Ki-Syn immunolabeling enables the accurate counting of the number of proliferating tumor cells without including inflammatory and contaminant epithelial cells compared with single Ki-67 immunolabeling in PanNETs from EUS-FNAB specimens.
Collapse
Affiliation(s)
- Bokyung Ahn
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Kying Jung
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - HaeSung Jung
- Department of Statistics, Korea University, Seoul, Republic of Korea
| | - Yeon-Mi Ryu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Yeon Wook Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Tae Jun Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Do Hyun Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dae Wook Hwang
- Department of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - HyungJun Cho
- Department of Statistics, Korea University, Seoul, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Hao W, Cui Y, Fan Y, Chen M, Yang G, Wang Y, Yang M, Li Z, Gong W, Yang Y, Gao C. Hybrid membrane-coated nanosuspensions for multi-modal anti-glioma therapy via drug and antigen delivery. J Nanobiotechnology 2021; 19:378. [PMID: 34801032 PMCID: PMC8606100 DOI: 10.1186/s12951-021-01110-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Glioma is one of the deadliest human cancers. Although many therapeutic strategies for glioma have been explored, these strategies are seldom used in the clinic. The challenges facing the treatment of glioma not only involve the development of chemotherapeutic drugs and immunotherapeutic agents, but also the lack of a powerful platform that could deliver these two moieties to the targeted sites. Herein, we developed chemoimmunotherapy delivery vehicles based on C6 cell membranes and DC membranes to create hybrid membrane-coated DTX nanosuspensions (DNS-[C6&DC]m). RESULTS Results demonstrated successful hybrid membrane fusion and nanosuspension functionalization, and DNS-[C6&DC]m could be used for different modes of anti-glioma therapy. For drug delivery, membrane coating could be applied to target the source cancer cells via a homotypic-targeting mechanism of the C6 cell membrane. For cancer immunotherapy, biomimetic nanosuspension enabled an immune response based on the professional antigen-presenting characteristic of the dendritic cell membrane (DCm), which carry the full array of cancer cell membrane antigens and facilitate the uptake of membrane-bound tumor antigens for efficient presentation and downstream immune n. CONCLUSION DNS-[C6&DC]m is a multifunctional biomimetic nano-drug delivery system with the potential to treat gliomas through tumor-targeted drug delivery combined with immunotherapy, thereby presenting a promising approach that may be utilized for multiple modes of cancer therapy.
Collapse
Affiliation(s)
- Wenyan Hao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yuexin Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yueyue Fan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Mengyu Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Guobao Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| |
Collapse
|
9
|
Wood LD, Ewald AJ. Organoids in cancer research: a review for pathologist-scientists. J Pathol 2021; 254:395-404. [PMID: 33886125 DOI: 10.1002/path.5684] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 12/20/2022]
Abstract
The use of three-dimensional (3D) culture models for cancer research has expanded greatly in recent years, with studies in almost every tumor type addressing a wide variety of research questions. Multiple distinct 3D culture approaches are now available, each with its own advantages and disadvantages, as well as most effective applications. In this review, we focus on one of these 3D culture models, organoids, in which multicellular units are isolated from primary or metastatic tumors and cultured in extracellular matrix gels. Organoids can be studied in acute cultures for short times after isolation, or passaged and biobanked for long-term use. We define this model system and describe some key studies in which organoid culture models were used to investigate cellular strategies and molecular mechanisms driving cancer initiation and progression, highlighting research questions for which this model is particularly well suited. In addition, as interest in implementing organoid systems continues to expand, we discuss key considerations in developing a new organoid research program. Our goal is to demonstrate the power and utility of organoid models and provide guidance for investigators who are considering implementation of these models in their own research programs. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Laura D Wood
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew J Ewald
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|