1
|
Miao H, Xu C, Gao W, Zhong L, Li H, Wen Z, Ren Q, Chen Y. PYGB targeted by androgen receptor contributes to tumor progression and metabolic reprogramming in esophageal squamous carcinoma. Cell Signal 2024; 124:111481. [PMID: 39442902 DOI: 10.1016/j.cellsig.2024.111481] [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: 07/18/2024] [Revised: 10/07/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND The incidence and mortality rates of esophageal squamous cell carcinoma (ESCC) are conspicuously augmented in men in contrast to women. The androgen receptor (AR), prevalently associated with the manifestation of male characteristics, is regarded as a pivotal determinant in tumor progression. Nevertheless, its exact role in ESCC remains insufficiently delineated. METHODS In this study, we probed the expression levels of AR and glucose metabolism enzymes in ESCC tissues by means of immunohistochemistry. We exploited chromatin immunoprecipitation and dual luciferase reporter assays to delve into the transcriptional regulatory interrelationships between AR and these enzymes. A gamut of molecular techniques-including multi-omics sequencing, colony formation assays, cell counting kit 8 (CCK8), 5-Ethynyl-2'-deoxyuridine (EdU) incorporation assays, wound-healing assays, transwell migration assays, extracellular acidification rate (ECAR) measurements, lipid droplet fluorescence imaging, and xenograft models-were enlisted to illuminate the functions of these enzymes within ESCC cells. RESULTS Our discoveries manifested that AR expression was strikingly higher in male ESCC tissues than in their female counterparts. Significantly, we discerned that glycogen phosphorylase B (PYGB), a cardinal enzyme implicated in glucose metabolism, demonstrated not only a positive correlation with AR expression but also an association with adverse prognostic outcomes for ESCC patients. Moreover, AR directly binds to the promoter region of the PYGB gene, thereby potentiating its transcriptional activity. This upregulation of PYGB was ascertained to facilitate proliferation, invasion, and metastasis among ESCC cells while intensifying glycolysis and modifying lipid metabolism pathways within these cells. In animal models employing nude mice, elevated PYGB levels were witnessed to expedite subcutaneous tumor growth as well as lung metastasis. CONCLUSIONS Collectively, our study establishes PYGB as a direct target of AR that assumes an indispensable role in both tumor progression and metabolic reprogramming affiliated with ESCC, thus paving novel avenues for therapeutic strategies centered on metabolic intercessions.
Collapse
Affiliation(s)
- Huikai Miao
- Institute of Pharmaceutical Research, Shandong Key Laboratory of Digital Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chunmei Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wuyou Gao
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Leqi Zhong
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongmu Li
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhesheng Wen
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiannan Ren
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Youfang Chen
- Department of Thoracic Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
2
|
Nogueira-Lima E, Alves T, Etchebehere E. 18F-Fluoride PET/CT-Updates. Semin Nucl Med 2024:S0001-2998(24)00083-7. [PMID: 39393951 DOI: 10.1053/j.semnuclmed.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
Sodium Fluoride-18 production started in the 1940s and was described clinically for the first time in 1962 as a bone-imaging agent. However, its use became dormant with the development of conventional bone scintigraphy, especially due to its low cost. Conventional bone scintigraphy has been the most utilized Nuclear Medicine technique for identifying osteoblastic bone metastases, especially in prostate and breast cancers for decades and is also employed to identify benign bone disease, especially in the orthopedic setting. While bone scintigraphy is highly sensitive, it lacks adequate specificity. With the advent of high-quality 3D Whole-Body Positron Emission Tomography combined with computed tomography (PET/CT), images, Sodium Fluoride-18 imaging with PET/CT (Fluoride PET/CT) re-emerged. This PET/CT bone-imaging agent provides higher sensitivity and specificity to detect bone lesions in both the oncological scenario as well as to identify benign bone and joint disorders. PET/CT bone-imaging provides a precise view of the bone metabolism remodeling processes at a molecular level, throughout the skeleton, and combines anatomical information, enhancing diagnostic specificity and accuracy. This article review will explore the updates on clinical applications of Fluoride PET/CT in oncology and benign conditions encompassing orthopedic, inflammatory and cardiovascular conditions and treatment response assessment.
Collapse
Affiliation(s)
- Ellen Nogueira-Lima
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Thiago Alves
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Elba Etchebehere
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
3
|
Sun X, Niwa T, Kazama T, Okazaki T, Koyanagi K, Kumaki N, Hashimoto J, Ozawa S. Preoperative dual-energy computed tomography and positron-emission tomography evaluation of lymph node metastasis in esophageal squamous cell carcinoma. PLoS One 2024; 19:e0309653. [PMID: 39302928 DOI: 10.1371/journal.pone.0309653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 08/15/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE To investigate the detectability of lymph node metastasis in patients with esophageal squamous cell carcinoma using a combination of dual-energy computed tomography (CT) and positron-emission tomography (PET) parameters. METHODS We analyzed dual-energy CT and PET preoperative data in 27 consecutive patients with esophageal squamous cell carcinoma (23 men, 4 women; mean age, 73.7 years). We selected lymph nodes with a short-axis diameter of ≥5 mm and measured CT values, iodine concentrations, fat fractions, long- and short-axis diameters, and ratio of long- and short-axis diameters. We performed visual assessment of lymph node characteristics based on dual-energy CT and determined the maximum standardized uptake value via PET. The measured values were postoperatively compared between pathologically confirmed metastatic and nonmetastatic lymph nodes. Stepwise logistic regression analysis was performed to determine factors associated with lymph node metastasis. Diagnostic accuracy was assessed via receiver operating characteristic curve analysis. RESULTS Overall, 18 metastatic and 37 nonmetastatic lymph nodes were detected. CT values, iodine concentrations, fat fractions, and the maximum standardized uptake values differed significantly between metastatic and nonmetastatic lymph nodes (p < 0.05). Stepwise logistic regression showed that iodine concentration and the maximum standardized uptake value were significant predictors of metastatic lymph nodes. The areas under the curve of iodine concentrations and maximum standardized uptake values were 0.809 and 0.833, respectively. The area under the curve of the combined parameters was 0.884, with 83.3% sensitivity and 86.5% specificity. CONCLUSION Combined dual-energy CT and PET parameters improved the diagnosis of lymph node metastasis in patients with esophageal cancer.
Collapse
Affiliation(s)
- Xuyang Sun
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Japan
| | - Tetsu Niwa
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Japan
| | - Toshiki Kazama
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Japan
| | - Takashi Okazaki
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Japan
| | - Kazuo Koyanagi
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Nobue Kumaki
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Jun Hashimoto
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Japan
| | - Soji Ozawa
- Department of Surgery, Tamakyuryo Hospital, Machida, Japan
| |
Collapse
|
4
|
Wang R, Liu S, Chen B, Li Q, Cheng X, Zhu Y, Zhang L, Hu Y, Liu M, Hu Y, Xi M. Prognostic significance of PET/CT and its association with immuno-genomic profiling in oesophageal squamous cell carcinoma treated with immunotherapy plus chemoradiotherapy: results from a phase II study. Br J Cancer 2024; 131:709-717. [PMID: 38937623 PMCID: PMC11333745 DOI: 10.1038/s41416-024-02779-4] [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: 12/19/2023] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND A phase II trial (EC-CRT-001) demonstrated the promising efficacy of combining toripalimab (an anti-PD-1 antibody) with definitive chemoradiotherapy (CRT) for locally advanced oesophageal squamous cell carcinoma (ESCC). Biomarkers are key to identifying patients who may benefit from this therapeutic approach. METHODS Of the 42 patients with ESCC who received toripalimab combined with definitive CRT, 37 were included in this analysis. Baseline assessments included PET/CT metabolic parameters (SUVmax, SUVmean, SUVpeak, MTV, and TLG), RNA sequencing of tumour biopsies to quantify the tissue mutational burden (TMB), and multiplex immunofluorescence staining to estimate immune cell infiltration in the tumour microenvironment (TME). Frozen neoplastic samples were procured for RNA sequencing to further explore the immune-related TME. RESULTS Among the 37 patients, high baseline SUVmax (≥12.0; OR = 6.5, 95% CI 1.4-48.2, p = 0.032) and TLG (≥121.8; OR = 6.8, 95% CI 1.6-33.5, p = 0.012) were significantly correlated with lower complete response rates. All five PET/CT parameters were notably associated with overall survival; only SUVmax and TLG were associated with a significantly worse progression-free survival. A trend towards an inverse correlation was observed between SUVmax and TMB (R = -0.33, p = 0.062). PD-1 + CD8 + T cell infiltration was negatively correlated with MTV (R = -0.355, p = 0.034) and TLG (R = -0.385, p = 0.021). Moreover, RNA sequencing revealed that the high TLG subgroup exhibited low immune cell infiltration, indicating an immunosuppressive landscape. CONCLUSIONS High baseline SUVmax and TLG might predict poorer treatment response and worse survival in patients with ESCC undergoing immunotherapy combined with CRT. In addition, high PET/CT metabolic parameters, particularly TLG, were correlated with an immunosuppressive TME, which warrants further exploration.
Collapse
Affiliation(s)
- Ruixi Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shiliang Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Baoqing Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiaoqiao Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xingyuan Cheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yujia Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yonghong Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengzhong Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yingying Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mian Xi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, China.
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
5
|
Zhang S, Li K, Sun Y, Wan Y, Ao Y, Zhong Y, Liang M, Wang L, Chen X, Pei X, Hu Y, Chen D, Li M, Shan H. Deep Learning for Automatic Gross Tumor Volumes Contouring in Esophageal Cancer Based on Contrast-Enhanced Computed Tomography Images: A Multi-Institutional Study. Int J Radiat Oncol Biol Phys 2024; 119:1590-1600. [PMID: 38432286 DOI: 10.1016/j.ijrobp.2024.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/02/2024] [Accepted: 02/18/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE To develop and externally validate an automatic artificial intelligence (AI) tool for delineating gross tumor volume (GTV) in patients with esophageal squamous cell carcinoma (ESCC), which can assist in neo-adjuvant or radical radiation therapy treatment planning. METHODS AND MATERIALS In this multi-institutional study, contrast-enhanced CT images from 580 eligible ESCC patients were retrospectively collected. The GTV contours delineated by 2 experts via consensus were used as ground truth. A 3-dimensional deep learning model was developed for GTV contouring in the training cohort and internally and externally validated in 3 validation cohorts. The AI tool was compared against 12 board-certified experts in 25 patients randomly selected from the external validation cohort to evaluate its assistance in improving contouring performance and reducing variation. Contouring performance was measured using dice similarity coefficient (DSC) and average surface distance. Additionally, our previously established radiomics model for predicting pathologic complete response was used to compare AI-generated and ground truth contours, to assess the potential of the AI contouring tool in radiomics analysis. RESULTS The AI tool demonstrated good GTV contouring performance in multicenter validation cohorts, with median DSC values of 0.865, 0.876, and 0.866 and median average surface distance values of 0.939, 0.789, and 0.875 mm, respectively. Furthermore, the AI tool significantly improved contouring performance for half of 12 board-certified experts (DSC values, 0.794-0.835 vs 0.856-0.881, P = .003-0.048), reduced the intra- and interobserver variations by 37.4% and 55.2%, respectively, and saved contouring time by 77.6%. In the radiomics analysis, 88.7% of radiomic features from ground truth and AI-generated contours demonstrated stable reproducibility, and similar pathologic complete response prediction performance for these contours (P = .430) was observed. CONCLUSIONS Our AI contouring tool can improve GTV contouring performance and facilitate radiomics analysis in ESCC patients, which indicates its potential for GTV contouring during radiation therapy treatment planning and radiomics studies.
Collapse
Affiliation(s)
- Shuaitong Zhang
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Kunwei Li
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yuchen Sun
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Yun Wan
- Department of Radiology, Xinyi City People's Hospital, Xinyi, Guangdong, China
| | - Yong Ao
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in South China, Guangdong Esophageal Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Yinghua Zhong
- Department of Radiology, The Third People's Hospital of Zhuhai, Zhuhai, Guangdong, China
| | - Mingzhu Liang
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Lizhu Wang
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Xiangmeng Chen
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Xiaofeng Pei
- Department of Radiation Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yi Hu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in South China, Guangdong Esophageal Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.
| | - Duanduan Chen
- School of Medical Technology, Beijing Institute of Technology, Beijing, China.
| | - Man Li
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China.
| | - Hong Shan
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China.
| |
Collapse
|
6
|
Liu J, Sui C, Bian H, Li Y, Wang Z, Fu J, Qi L, Chen K, Xu W, Li X. Radiomics based on 18F-FDG PET/CT for prediction of pathological complete response to neoadjuvant therapy in non-small cell lung cancer. Front Oncol 2024; 14:1425837. [PMID: 39132503 PMCID: PMC11310012 DOI: 10.3389/fonc.2024.1425837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose This study aimed to establish and evaluate the value of integrated models involving 18F-FDG PET/CT-based radiomics and clinicopathological information in the prediction of pathological complete response (pCR) to neoadjuvant therapy (NAT) for non-small cell lung cancer (NSCLC). Methods A total of 106 eligible NSCLC patients were included in the study. After volume of interest (VOI) segmentation, 2,016 PET-based and 2,016 CT-based radiomic features were extracted. To select an optimal machine learning model, a total of 25 models were constructed based on five sets of machine learning classifiers combined with five sets of predictive feature resources, including PET-based alone radiomics, CT-based alone radiomics, PET/CT-based radiomics, clinicopathological features, and PET/CT-based radiomics integrated with clinicopathological features. Area under the curves (AUCs) of receiver operator characteristic (ROC) curves were used as the main outcome to assess the model performance. Results The hybrid PET/CT-derived radiomic model outperformed PET-alone and CT-alone radiomic models in the prediction of pCR to NAT. Moreover, addition of clinicopathological information further enhanced the predictive performance of PET/CT-derived radiomic model. Ultimately, the support vector machine (SVM)-based PET/CT radiomics combined clinicopathological information presented an optimal predictive efficacy with an AUC of 0.925 (95% CI 0.869-0.981) in the training cohort and an AUC of 0.863 (95% CI 0.740-0.985) in the test cohort. The developed nomogram involving radiomics and pathological type was suggested as a convenient tool to enable clinical application. Conclusions The 18F-FDG PET/CT-based SVM radiomics integrated with clinicopathological information was an optimal model to non-invasively predict pCR to NAC for NSCLC.
Collapse
Affiliation(s)
- Jianjing Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Haiman Bian
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yue Li
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Jie Fu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Lisha Qi
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Kun Chen
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
7
|
Shi YJ, Yan S, Yang X, Guan Z, Li XT, Wang LL, Dai L, Sun YS. Early Contrast-Enhanced MR for Diagnosing Complete Tumor Response of Locally Advanced Esophageal Squamous Cell Carcinoma After Neoadjuvant Therapy: A Retrospective Comparative Study. Ann Surg Oncol 2024; 31:4271-4280. [PMID: 38453768 DOI: 10.1245/s10434-024-15123-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/14/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND This study assessed the performance of early contrast-enhanced magnetic resonance (ECE-MR) in the detecting of complete tumor response (ypT0) in patients with esophageal squamous cell carcinoma following neoadjuvant therapy. PATIENTS AND METHODS Preoperative MR images of consecutive patients who underwent neoadjuvant therapy and surgical resection were reviewed retrospectively. The accuracy of ECE-MR and T2WI+DWI was evaluated by comparing the findings with pathological results. Receiver operating characteristic curve analysis was used to assess the diagnostic performance, and DeLong method was applied to compare the areas under the curves (AUC). Chi-squared analysis was conducted to explore the difference in pathological changes. RESULTS A total of 198 patients (mean age 62.6 ± 7.8 years, 166 men) with 201 lesions were included. The AUC of ECE-MR was 0.85 (95% CI 0.79-0.90) for diagnosing ypT1-4, which was significantly higher than that of T2WI+DWI (AUC 0.69, 95% CI 0.63-0.76, p < 0.001). The diagnostic performance of both T2WI+DWI and ECE-MR improved with increasing tumor stage. The AUCs of ECE-MRI were higher in ypT1 and ypT2 tumors than T2WI+DWI. Degree 2-3 tumor-infiltrating lymphocytes and neutrophils were commonly seen in ypT0 tumors misdiagnosed by ECE-MR. CONCLUSIONS Visual evaluation of ECE-MR is a promising diagnostic protocol for the detection of complete tumor response, especially for differentiation with early stage tumors. The accurate diagnosis of complete tumor response after neoadjuvant therapy using imaging modalities is of important significance for clinical decision-making for patients with esophageal squamous cell carcinoma. It is hoped that early contrast-enhanced MR will provide supportive advice for the development of individualized treatment options for patients.
Collapse
Affiliation(s)
- Yan-Jie Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Hai Dian District, Beijing, China
| | - Shuo Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Hai Dian District, Beijing, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and Institute, Hai Dian District, Beijing, China
| | - Zhen Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Hai Dian District, Beijing, China
| | - Xiao-Ting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Hai Dian District, Beijing, China
| | - Lin-Lin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Hai Dian District, Beijing, China
| | - Liang Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Hai-Dian District, Beijing, China.
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Hai Dian District, Beijing, China.
| |
Collapse
|
8
|
Ruan Y, Ma Y, Ma M, Liu C, Su D, Guan X, Yang R, Wang H, Li T, Zhou Y, Ma J, Zhang Y. Dynamic radiological features predict pathological response after neoadjuvant immunochemotherapy in esophageal squamous cell carcinoma. J Transl Med 2024; 22:471. [PMID: 38762454 PMCID: PMC11102630 DOI: 10.1186/s12967-024-05291-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/09/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND Neoadjuvant immunochemotherapy (NICT) plus esophagectomy has emerged as a promising treatment option for locally advanced esophageal squamous cell carcinoma (LA-ESCC). Pathologic complete response (pCR) is a key indicator associated with great efficacy and overall survival (OS). However, there are insufficient indicators for the reliable assessment of pCR. METHODS 192 patients with LA-ESCC treated with NICT from December 2019 to October 2023 were recruited. According to pCR status, patients were categorized into pCR group (22.92%) and non-pCR group (77.08%). Radiological features of pretreatment and preoperative CT images were extracted. Logistic and COX regressions were trained to predict pathological response and prognosis, respectively. RESULTS Four of the selected radiological features were combined to construct an ESCC preoperative imaging score (ECPI-Score). Logistic models revealed independent associations of ECPI-Score and vascular sign with pCR, with AUC of 0.918 in the training set and 0.862 in the validation set, respectively. After grouping by ECPI-Score, a higher proportion of pCR was observed among the high-ECPI group and negative vascular sign. Kaplan Meier analysis demonstrated that recurrence-free survival (RFS) with negative vascular sign was significantly better than those with positive (P = 0.038), but not for OS (P = 0.310). CONCLUSIONS This study demonstrates dynamic radiological features are independent predictors of pCR for LA-ESCC treated with NICT. It will guide clinicians to make accurate treatment plans.
Collapse
Affiliation(s)
- Yuli Ruan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Yue Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Ming Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China
| | - Dan Su
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Xin Guan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Rui Yang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China
| | - Hong Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Tianqin Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Yang Zhou
- Department of Radiology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China.
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China.
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang, 150001, People's Republic of China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin, China.
| |
Collapse
|
9
|
Tan R, Sui C, Wang C, Zhu T. MRI-based intratumoral and peritumoral radiomics for preoperative prediction of glioma grade: a multicenter study. Front Oncol 2024; 14:1401977. [PMID: 38803534 PMCID: PMC11128562 DOI: 10.3389/fonc.2024.1401977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Background Accurate preoperative prediction of glioma is crucial for developing individualized treatment decisions and assessing prognosis. In this study, we aimed to establish and evaluate the value of integrated models by incorporating the intratumoral and peritumoral features from conventional MRI and clinical characteristics in the prediction of glioma grade. Methods A total of 213 glioma patients from two centers were included in the retrospective analysis, among which, 132 patients were classified as the training cohort and internal validation set, and the remaining 81 patients were zoned as the independent external testing cohort. A total of 7728 features were extracted from MRI sequences and various volumes of interest (VOIs). After feature selection, 30 radiomic models depended on five sets of machine learning classifiers, different MRI sequences, and four different combinations of predictive feature sources, including features from the intratumoral region only, features from the peritumoral edema region only, features from the fusion area including intratumoral and peritumoral edema region (VOI-fusion), and features from the intratumoral region with the addition of features from peritumoral edema region (feature-fusion), were established to select the optimal model. A nomogram based on the clinical parameter and optimal radiomic model was constructed for predicting glioma grade in clinical practice. Results The intratumoral radiomic models based on contrast-enhanced T1-weighted and T2-flair sequences outperformed those based on a single MRI sequence. Moreover, the internal validation and independent external test underscored that the XGBoost machine learning classifier, incorporating features extracted from VOI-fusion, showed superior predictive efficiency in differentiating between low-grade gliomas (LGG) and high-grade gliomas (HGG), with an AUC of 0.805 in the external test. The radiomic models of VOI-fusion yielded higher prediction efficiency than those of feature-fusion. Additionally, the developed nomogram presented an optimal predictive efficacy with an AUC of 0.825 in the testing cohort. Conclusion This study systematically investigated the effect of intratumoral and peritumoral radiomics to predict glioma grading with conventional MRI. The optimal model was the XGBoost classifier coupled radiomic model based on VOI-fusion. The radiomic models that depended on VOI-fusion outperformed those that depended on feature-fusion, suggesting that peritumoral features should be rationally utilized in radiomic studies.
Collapse
Affiliation(s)
- Rui Tan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Chao Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People’s Hospital), Shandong, China
| | - Tao Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
10
|
Ma J, Wang X, Tang M, Zhang C. Preoperative prediction of pancreatic neuroendocrine tumor grade based on 68Ga-DOTATATE PET/CT. Endocrine 2024; 83:502-510. [PMID: 37715934 PMCID: PMC10850018 DOI: 10.1007/s12020-023-03515-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 08/29/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE To establish a prediction model for preoperatively predicting grade 1 and grade 2/3 tumors in patients with pancreatic neuroendocrine tumors (PNETs) based on 68Ga-DOTATATE PET/CT. METHODS Clinical data of 41 patients with PNETs were included in this study. According to the pathological results, they were divided into grade 1 and grade 2/3. 68Ga-DOTATATE PET/CT images were collected within one month before surgery. The clinical risk factors and significant radiological features were filtered, and a clinical predictive model based on these clinical and radiological features was established. 3D slicer was used to extracted 107 radiomic features from the region of interest (ROI) of 68Ga-dotata PET/CT images. The Pearson correlation coefficient (PCC), recursive feature elimination (REF) based five-fold cross validation were adopted for the radiomic feature selection, and a radiomic score was computed subsequently. The comprehensive model combining the clinical risk factors and the rad-score was established as well as the nomogram. The performance of above clinical model and comprehensive model were evaluated and compared. RESULTS Adjacent organ invasion, N staging, and M staging were the risk factors for PNET grading (p < 0.05). 12 optimal radiomic features (3 PET radiomic features, 9 CT radiomic features) were screen out. The clinical predictive model achieved an area under the curve (AUC) of 0.785. The comprehensive model has better predictive performance (AUC = 0.953). CONCLUSION We proposed a comprehensive nomogram model based on 68Ga-DOTATATE PET/CT to predict grade 1 and grade 2/3 of PNETs and assist personalized clinical diagnosis and treatment plans for patients with PNETs.
Collapse
Affiliation(s)
- Jiao Ma
- Department of Nuclear Medicine, The Affilliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, PR China
| | - Xiaoyong Wang
- Department of Radiology, The Affilliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, PR China
| | - Mingsong Tang
- Department of Radiology, The Affilliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, PR China
| | - Chunyin Zhang
- Department of Nuclear Medicine, The Affilliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, PR China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, PR China.
- Academician (expert) Workstation of Sichuan Province, Luzhou, 646000, Sichuan, PR China.
| |
Collapse
|
11
|
Miao Y, Feng R, Yu T, Guo R, Zhang M, Wang Y, Hai W, Shangguan C, Zhu Z, Li B. Value of 68Ga-FAPI-04 and 18F-FDG PET/CT in Early Prediction of Pathologic Response to Neoadjuvant Chemotherapy in Locally Advanced Gastric Cancer. J Nucl Med 2024; 65:213-220. [PMID: 38164574 DOI: 10.2967/jnumed.123.266403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/07/2023] [Indexed: 01/03/2024] Open
Abstract
This prospective study investigated whether PET parameters from 18F-FDG and 68Ga-fibroblast activation protein inhibitor (FAPI)-04 PET/CT can predict a pathologic response to neoadjuvant chemotherapy (NAC) early in patients with locally advanced gastric cancer (LAGC). Methods: The study included 28 patients with LAGC who underwent 18F-FDG PET/CT and 68Ga-FAPI-04 PET/CT at baseline and after 1 cycle of NAC. PET parameters including SUV and tumor-to-background ratio (TBR), as well as the change rate of SUV and TBR, were recorded. Patients were classified as major or minor pathologic responders according to postoperative pathology findings. We compared the PET parameters between the 2 pathologic response groups and different treatment regimens and analyzed their predictive performance for tumor pathologic response. Results: Major pathologic responders had significantly lower 68Ga-FAPI change rates (percentage SUVmax [%SUVmax], percentage SUVpeak [%SUVpeak], and percentage TBR [%TBR]) than minor pathologic responders. Among the PET parameters, 68Ga-FAPI %SUVmax (area under the curve, 0.856; P = 0.009), %SUVpeak (area under the curve, 0.811; P = 0.022), and %TBR (area under the curve, 0.864; P = 0.007) were significant parameters for early prediction of pathologic response to NAC in LAGC; they had the same predictive accuracy of 89.29%, with the thresholds of decrease to at least 52.43%, 60.46%, and 52.96%, respectively. In addition, 68Ga-FAPI %SUVmax and %TBR showed significant differences between the different treatment regimens. Conclusion: In this preliminary study, 68Ga-FAPI-04 PET change rate parameters were preferable to 18F-FDG in predicting pathologic response to NAC at an early stage in LAGC. 68Ga-FAPI %SUVmax and %TBR may be better predictors of therapeutic response between different treatment regimens. These findings may help optimize the treatment for patients with LAGC.
Collapse
Affiliation(s)
- Ying Miao
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runhua Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Teng Yu
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Wang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wangxi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengfang Shangguan
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; and
| | - Zhenggang Zhu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China;
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China;
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, China
| |
Collapse
|
12
|
Song XY, Liu J, Li HX, Cai XW, Li ZG, Su YC, Li Y, Dong XH, Yu W, Fu XL. Enhancing Prediction for Tumor Pathologic Response to Neoadjuvant Immunochemotherapy in Locally Advanced Esophageal Cancer by Dynamic Parameters from Clinical Assessments. Cancers (Basel) 2023; 15:4377. [PMID: 37686655 PMCID: PMC10486879 DOI: 10.3390/cancers15174377] [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: 07/17/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
To develop accurate and accessible prediction methods for assessing pathologic response following NICT prior to surgery, we conducted a retrospective study including 137 patients with esophageal squamous cell carcinoma (ESCC) who underwent surgery after two cycles of NICT between January 2019 and March 2022 at our center. We collected clinical parameters to evaluate the dynamic changes in the primary tumor. Univariate and multivariate analyses were performed to determine the correlations between these parameters and the pathologic response of the primary tumor. Subsequently, we constructed prediction models for pCR and MPR using multivariate logistic regression. The MPR prediction Model 2 was internally validated using bootstrapping and externally validated using an independent cohort from our center. The univariate logistic analysis revealed significant differences in clinical parameters reflecting tumor regression among patients with varying pathologic responses. The clinical models based on these assessments demonstrated excellent predictive performance, with the training cohort achieving a C-index of 0.879 for pCR and 0.912 for MPR, while the testing cohort also achieved a C-index of 0.912 for MPR. Notably, the MPR prediction Model 2, with a threshold cut-off of 0.74, exhibited 92.7% specificity and greater than 70% sensitivity, indicating a low rate of underestimating residual tumors. In conclusion, our study demonstrated the high accuracy of clinical assessment-based models in pathologic response prediction, aiding in decision-making regarding organ preservation and radiotherapy adjustments after induction immunochemotherapy.
Collapse
Affiliation(s)
- Xin-Yun Song
- Department of Radiation Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (X.-Y.S.)
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jun Liu
- Department of Radiation Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (X.-Y.S.)
| | - Hong-Xuan Li
- Department of Radiation Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (X.-Y.S.)
| | - Xu-Wei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (X.-Y.S.)
| | - Zhi-Gang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yu-Chen Su
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yue Li
- Department of Radiation Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (X.-Y.S.)
| | - Xiao-Huan Dong
- Department of Radiation Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (X.-Y.S.)
| | - Wen Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (X.-Y.S.)
| | - Xiao-Long Fu
- Department of Radiation Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (X.-Y.S.)
| |
Collapse
|
13
|
Wang S, Di S, Lu J, Xie S, Yu Z, Liang Y, Gong T. 18 F-FDG PET/CT predicts the role of neoadjuvant immunochemotherapy in the pathological response of esophageal squamous cell carcinoma. Thorac Cancer 2023; 14:2338-2349. [PMID: 37424279 PMCID: PMC10447171 DOI: 10.1111/1759-7714.15024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023] Open
Abstract
BACKGROUND This study aimed to investigate the predictive value of 18 F-FDG PET/CT for pathological response after neoadjuvant immunochemotherapy (NICT) in patients with esophageal squamous cell carcinoma (ESCC). METHODS The clinical data of 54 patients with ESCC who underwent two cycles of NICT followed by surgery were retrospectively analyzed. NICT consisted of PD-1 blockade therapy combined with chemotherapy. 18 F-FDG PET/CT scans were performed before and after NICT. The pathological results after surgery were used to assess the degree of pathological response. The scan parameters of 18 F-FDG PET/CT and their changes before and after NICT were compared with the pathological response. RESULTS Among the 54 patients, 10 (18.5%) achieved complete pathological response (pCR) and 21 (38.9%) achieved major pathological response (MPR). The post-NICT scan parameters and their changes were significantly associated with the pathological response. In addition, the values of the changes in the scanned parameters before and after treatment can further predict the pathological response of the patient. CONCLUSION 18 F-FDG PET/CT is a useful tool to evaluate the efficacy of NICT and predict pathological response in patients with ESCC. The post-NICT scan parameters and their changes can help identify patients who are likely to achieve pCR or MPR.
Collapse
Affiliation(s)
- Shuohua Wang
- Department of Thoracic SurgeryNavy Clinical College, Anhui Medical UniversityHefeiChina
- Department of Thoracic SurgeryThe Fifth School of Clinical Medicine, Anhui Medical UniversityHefeiChina
| | - Shouyin Di
- Department of Thoracic SurgeryThe Sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Jing Lu
- Department of Thoracic SurgeryThe Sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Shun Xie
- Department of Thoracic SurgeryNavy Clinical College, Anhui Medical UniversityHefeiChina
- Department of Thoracic SurgeryThe Fifth School of Clinical Medicine, Anhui Medical UniversityHefeiChina
| | - Zhenyang Yu
- Department of PathologyThe Sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Yingkui Liang
- Department of Nuclear MedicineThe Sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Taiqian Gong
- Department of Thoracic SurgeryNavy Clinical College, Anhui Medical UniversityHefeiChina
- Department of Thoracic SurgeryThe Fifth School of Clinical Medicine, Anhui Medical UniversityHefeiChina
| |
Collapse
|
14
|
Wang H, Song C, Zhao X, Deng W, Dong J, Shen W. Evaluation of neoadjuvant immunotherapy and traditional neoadjuvant therapy for resectable esophageal cancer: a systematic review and single-arm and network meta-analysis. Front Immunol 2023; 14:1170569. [PMID: 37251393 PMCID: PMC10213267 DOI: 10.3389/fimmu.2023.1170569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Objective This systematic review and meta-analysis aimed to investigate the role of neoadjuvant immunochemotherapy with or without radiotherapy [NIC(R)T] compared to traditional neoadjuvant therapies, without immunotherapy [NC(R)T]. Summary background data NCRT followed by surgical resection is recommended for patients with early-stage esophageal cancer. However, it is uncertain whether adding immunotherapy to preoperative neoadjuvant therapy would improve patient outcomes when radical surgery is performed following neoadjuvant therapy. Methods We searched PubMed, Web of Science, Embase, and Cochrane Central databases, as well as international conference abstracts. Outcomes included R0, pathological complete response (pCR), major pathological response (mPR), overall survival (OS) and disease-free survival (DFS) rates. Results We included data from 5,034 patients from 86 studies published between 2019 and 2022. We found no significant differences between NICRT and NCRT in pCR or mPR rates. Both were better than NICT, with NCT showing the lowest response rate. Neoadjuvant immunotherapy has a significant advantage over traditional neoadjuvant therapy in terms of 1-year OS and DFS, with NICT having better outcomes than any of the other three treatments. There were no significant differences among the four neoadjuvant treatments in terms of R0 rates. Conclusions Among the four neoadjuvant treatment modalities, NICRT and NCRT had the highest pCR and mPR rates. There were no significant differences in the R0 rates among the four treatments. Adding immunotherapy to neoadjuvant therapy improved 1-year OS and DFS, with NICT having the highest rates compared to the other three modalities. Systematic Review Registration https://inplasy.com/inplasy-2022-12-0060/, identifier INPLASY2022120060.
Collapse
Affiliation(s)
| | | | | | | | | | - Wenbin Shen
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
15
|
Gao L, Hong ZN, Wu L, Yang Y, Kang M. Residual tumor model in esophageal squamous cell carcinoma after neoadjuvant immunochemotherapy: Frequently involves the mucosa and/or submucosa. Front Immunol 2022; 13:1008681. [PMID: 36569913 PMCID: PMC9780370 DOI: 10.3389/fimmu.2022.1008681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/31/2022] [Indexed: 12/13/2022] Open
Abstract
Objectives The efficacy and safety of neoadjuvant immunochemotherapy (nICT) are widely explored in locally advanced esophageal squamous cell carcinoma (ESCC). Whether the "wait-and-see" strategy is applicable in ESCC after nICT is still lacking a theoretical basis. This study aimed to preliminarily explore the distribution of residual tumors and the regression pattern of ESCC after nICT. Methods Patients undergoing radical esophagectomy after nICT in Fujian Medical University Union Hospital between January 2020 and March 2022 were identified. The resection specimens were re-evaluated by one experienced pathologist. The pathological response was assessed by tumor regression grade (TRG) (modified Ryan scheme). The TRG grade was divided into grades 0 (pathological complete response), 1, 2, and 3. The pathological stage was evaluated in the Eighth Edition AJCC. In the non-pCR group, the residual model was divided into four types: Type I, regression towards the lumen; type II, regression towards the invasive front; type III, concentric regression; and type IV, scattered regression. Results A total of 95 consecutive patients were included for analysis. Seventy-six (80.0%) of 95 patients were in non-pCR (pathological complete response), and nine patients (9/76, 11.84%) had isolated residual tumors in lymph nodes. There was no significant difference in baseline characteristics between the pCR group and the non-pCR group (p > 0.05). The overall distribution of TRG for all esophageal wall layers was TRG 0 = 28 (28/95, 29.5%), TRG 1 = 17 (17/95, 17.9%), TRG 2 = 18 (18.9%, 18/95), and TRG 3 = 32 (32/95, 33.7%). In 67 patients with residual tumors in the esophageal wall (TRG ≧1), 63 (63/67, 94.0%) had residual tumor cells in the mucosa and/or submucosa, and four had isolated residual tumors in the muscle layer (4/67, 6.0%). Further analysis showed eight (8/67, 11.9%) patients with submucosal involvement but without mucosal involvement. The distribution of regression patterns was type I (n = 35, 52.2%), type II (n = 3, 4.5%), type III (n = 8, 11.9%), and type IV (n = 21, 31.3%). Conclusions The mucosa and/or submucosa are frequently involved in residual malignancy, and the frequent regression models are regression toward the lumen and random regression. There is an opportunity to carefully test the residual tumors in a subgroup of the population with ESCC following nICT. However, some patients had residual tumors only in the muscle layer or lymph nodes. The clinical application of the wait-and-see strategy in ESCC after nICT should be explored using an appropriate evaluation protocol.
Collapse
Affiliation(s)
- Lei Gao
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China,Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Zhi-Nuan Hong
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China,Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Long Wu
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yinghong Yang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China,*Correspondence: Mingqiang Kang, ; Yinghong Yang,
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China,Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China,*Correspondence: Mingqiang Kang, ; Yinghong Yang,
| |
Collapse
|