1
|
Han S, Zhang T, Deng W, Han S, Wu H, Jiang B, Xie W, Chen Y, Deng T, Wen X, Liu N, Fan J. Deep learning progressive distill for predicting clinical response to conversion therapy from preoperative CT images of advanced gastric cancer patients. Sci Rep 2025; 15:17092. [PMID: 40379665 PMCID: PMC12084415 DOI: 10.1038/s41598-025-01063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Identifying patients suitable for conversion therapy through early non-invasive screening is crucial for tailoring treatment in advanced gastric cancer (AGC). This study aimed to develop and validate a deep learning method, utilizing preoperative computed tomography (CT) images, to predict the response to conversion therapy in AGC patients. This retrospective study involved 140 patients. We utilized Progressive Distill (PD) methodology to construct a deep learning model for predicting clinical response to conversion therapy based on preoperative CT images. Patients in the training set (n = 112) and in the test set (n = 28) were sourced from The First Affiliated Hospital of Wenzhou Medical University between September 2017 and November 2023. Our PD models' performance was compared with baseline models and those utilizing Knowledge Distillation (KD), with evaluation metrics including accuracy, sensitivity, specificity, receiver operating characteristic curves, areas under the receiver operating characteristic curve (AUCs), and heat maps. The PD model exhibited the best performance, demonstrating robust discrimination of clinical response to conversion therapy with an AUC of 0.99 and accuracy of 99.11% in the training set, and 0.87 AUC and 85.71% accuracy in the test set. Sensitivity and specificity were 97.44% and 100% respectively in the training set, 85.71% and 85.71% each in the test set, suggesting absence of discernible bias. The deep learning model of PD method accurately predicts clinical response to conversion therapy in AGC patients. Further investigation is warranted to assess its clinical utility alongside clinicopathological parameters.
Collapse
Grants
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
- 2023D007, 2023D015, 2023D033, 2023D034, 2023D035 Municipal Government of Quzhou
Collapse
Affiliation(s)
- Saiyi Han
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China
| | - Tong Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China
- Yangtze Delta Region Institute(Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Wenzhuo Deng
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China
- Yangtze Delta Region Institute(Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Shaoliang Han
- Department of The Gastrointestinal Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Honghao Wu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China
- Yangtze Delta Region Institute(Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Beier Jiang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China
| | - Weidong Xie
- Department of The Gastrointestinal Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Yide Chen
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China
- Yangtze Delta Region Institute(Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Tao Deng
- Department of The Gastrointestinal Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xuewen Wen
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China
- Yangtze Delta Region Institute(Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Nianbo Liu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China
- Yangtze Delta Region Institute(Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan, PR China
| | - Jianping Fan
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, PR China.
| |
Collapse
|
2
|
Chen H, Gou L, Fang Z, Dou Q, Chen H, Chen C, Qiu Y, Zhang J, Ning C, Hu Y, Deng H, Yu J, Li G. Artificial intelligence assisted real-time recognition of intra-abdominal metastasis during laparoscopic gastric cancer surgery. NPJ Digit Med 2025; 8:9. [PMID: 39757250 DOI: 10.1038/s41746-024-01372-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Laparoscopic exploration (LE) is crucial for diagnosing intra-abdominal metastasis (IAM) in advanced gastric cancer (GC). However, overlooking single, tiny, and occult IAM lesions during LE can severely affect the treatment and prognosis due to surgeons' visual misinterpretations. To address this, we developed the artificial intelligence laparoscopic exploration system (AiLES) to recognize IAM lesions with various metastatic extents and locations. The AiLES was developed based on a dataset consisting of 5111 frames from 100 videos, using 4130 frames for model development and 981 frames for evaluation. The AiLES achieved a Dice score of 0.76 and a recognition speed of 11 frames per second, demonstrating robust performance in different metastatic extents (0.74-0.76) and locations (0.63-0.90). Furthermore, AiLES performed comparably to novice surgeons in IAM recognition and excelled in recognizing tiny and occult lesions. Our results demonstrate that the implementation of AiLES could enhance accurate tumor staging and assist individualized treatment decisions.
Collapse
Affiliation(s)
- Hao Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Longfei Gou
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwen Fang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Qi Dou
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Haobin Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chang Chen
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yuqing Qiu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Jinglin Zhang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Chenglin Ning
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Yanfeng Hu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haijun Deng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jiang Yu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
3
|
Han S, Han S, Qian J, Guo M, Fan J. Analysis of the therapeutic effect and influencing factors on unresectable gastric cancer treated with conversion therapy. Front Oncol 2024; 14:1435398. [PMID: 39540147 PMCID: PMC11557377 DOI: 10.3389/fonc.2024.1435398] [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: 05/20/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Background Gastric cancer (GC) is one of the leading causes of cancer-related death in China, and with the extensive development of conversion therapy, the treatment of advanced unresectable gastric cancer (AUGC) patients has ushered in a new dawn. This study aimed to analyze the efficacy of conversion therapy in AUGC patients and explored the relevant factors affecting the efficacy. Method We collected information from GC patients who received conversion therapy from this center and designed a retrospective study. Results We collected relevant clinical data from 160 patients with AUGC. A total of 120 patients who underwent routine R0 resection were identified as conversion cases. A total of 25 patients (15.6%) achieved pCR, 92 patients (57.5%) achieved objective response rate (ORR), 140 patients (87.5%) achieved disease control rate (DCR), and 20 cases (12.5%) observed tumor progression. There were 86 patients who achieved pathological downgrading, with a total downgrading rate of 53.8%. Among the 160 patients, 37 patients (23.1%) had postoperative complications of varying degrees. A total of 72 patients (45.0%) had tumor recurrence/progression at the end of follow-up. The last chemotherapy and surgery (CST) (OR = 1.046, 95% CI 1.013-1.081, p = 0.006), tumor invasion (OR = 32.096, 95% CI 5.091-202.349, p < 0.001), and distant metastasis (OR = 7.050, 95% CI 1.888-26.323, p = 0.004) were independent factors influencing the efficacy of conversion therapy. Conclusion Conversion therapy may have a good therapeutic efficacy for AUGC, and some clinical factors affect the efficacy response.
Collapse
Affiliation(s)
- Saiyi Han
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Shaoliang Han
- Department of The Gastrointestinal Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jun Qian
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Mengfu Guo
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Jianping Fan
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| |
Collapse
|
4
|
Wang J, Liang Y, Xue A, Xiao J, Zhao X, Cao S, Li P, Dong J, Li Y, Xu Z, Yang L. Intratumoral CXCL13 + CD160 + CD8 + T cells promote the formation of tertiary lymphoid structures to enhance the efficacy of immunotherapy in advanced gastric cancer. J Immunother Cancer 2024; 12:e009603. [PMID: 39244216 PMCID: PMC11381742 DOI: 10.1136/jitc-2024-009603] [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] [Accepted: 08/19/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Stage IV gastric cancer is a highly heterogeneous and lethal tumor with few therapeutic strategies. The combination of programmed cell death protein 1 inhibitors and chemotherapy is currently the standard frontline treatment regimen for advanced gastric cancer. Nevertheless, it remains a great challenge to screen the beneficiaries of immunochemotherapy and expand indications for this treatment regimen. METHODS We conducted a pathological assessment to ascertain the importance of tertiary lymphoid structures based on the tissue samples collected from patients with stage IV gastric cancer (n=15) both prior to and following immunochemotherapy treatment. Additionally, we used spatial (n=10) and single-cell transcriptional analysis (n=97) to investigate the key regulators of tertiary lymphoid structures (TLSs). Multiplex immunofluorescence and image analysis (n=34) were performed to explore the association between tumor-infiltrating CXCL13+ CD160+ CD8+ T cells and TLSs. The relationship between CXCL13+ CD160+ CD8+ T cells and the responsiveness to immunotherapy was also evaluated by multiplex immunofluorescence and image analysis approaches (n=15). Furthermore, we explored the intrinsic characteristics of CXCL13+ CD160+ CD8+ T cells through various experimental techniques, including quantitative reverse transcription-PCR, western blot, and flow cytometry. RESULTS We found that responders exhibited higher levels of TLSs and CXCL13+ CD160+ CD8+ T cells in biopsy tissues prior to immunochemotherapy compared with non-responders. Following conversion therapy, responders also had a higher percentage of mature TLSs and a higher number of CXCL13+ CD160+ CD8+ T cells in surgical resections. Moreover, we discovered that vitamin B6 in CD160+ CD8+ T cells could reduce the ubiquitination modification of HIF-1α by MDM2, thereby attenuating the degradation of HIF-1α. Consequently, this led to the transcriptional upregulation of CXCL13 expression, facilitating the recruitment of CXCR5+ B cells and the formation of TLSs. CONCLUSION The number and maturity of TLSs, along with the extent of CXCL13+ CD160+ CD8+ T-cell infiltration, might function as potential indicators for assessing the effectiveness of immunotherapy in treating gastric malignancies. Furthermore, our research suggests that vitamin B6 could enhance the secretion of CXCL13 by CD160+ CD8+ T cells by reducing the degradation of HIF-1α. Additionally, we demonstrate that vitamin B6 supplementation or targeting pyridoxal kinase could substantially improve the efficacy of immunotherapies for gastric cancer.
Collapse
Affiliation(s)
- Jiawei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuan Liang
- Southeast University, Nanjing, Jiangsu, China
| | - Ao Xue
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Xiao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinyu Zhao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuqing Cao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pengyu Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiacheng Dong
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuan Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Meng W, Pan L, Huang L, Li Q, Sun Y. Applications of image-guided locoregional transarterial chemotherapy in patients with inoperable colorectal cancer: a review. Front Oncol 2024; 14:1464242. [PMID: 39246324 PMCID: PMC11377196 DOI: 10.3389/fonc.2024.1464242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
With the development of comprehensive treatment, locoregional transarterial chemotherapy has become an alternative conversion therapy, palliative therapy, and neoadjuvant therapy for many solid malignant tumors. Locoregional transarterial chemotherapy, which is most frequently used for treating liver cancer, has the characteristics of high regional efficacy and few systemic adverse reactions. In recent years, the number of relevant reports of locoregional chemotherapy for treating initially inoperable colorectal cancer (CRC), including non-metastatic and metastatic CRC, has gradually increased. However, the specific treatment options for such locoregional therapy are not the same, and its indications, medication regimens and combined treatments have not reached any consensus. In this review, the application status of locoregional transarterial chemotherapy in primary and metastatic CRC patients has been reviewed and summarized to provide a reference for future clinical work and scientific research.
Collapse
Affiliation(s)
- Wenjun Meng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Lu Pan
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing, Sichuan University, Chengdu, China
| | - Li Huang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing, Sichuan University, Chengdu, China
| | - Qing Li
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Sun
- Department of Oncology and Hematology, Air Force Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
6
|
Zhu J, Wang Y, Xu B, Pu W, He P, Zhang J, Wang H, Chen H. Photodynamic therapy may salvage chemotherapy failure in gastric cancer: A case report and a literature review. Photodiagnosis Photodyn Ther 2024; 46:104038. [PMID: 38447816 DOI: 10.1016/j.pdpdt.2024.104038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/29/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
Given the highly heterogeneous characteristics of advanced gastric cancer (GC), most patients must receive neoadjuvant therapy or conversion therapy consisting of chemotherapy to decrease tumor grade and improve the likelihood of complete resection. Drug resistance, however, always leads to an aborted conversion therapy and inevitable death. When meet drug resistance, alternative drug regimens will be applied with immunotherapy or targeted therapy, whose clinical efficacy remains limited when new drug resistance or severer liver and kidney toxicity emerge. Photodynamic therapy (PDT), a novel treatment, has demonstrated remarkable therapeutic efficacy in different stages of GC. However, no report has been reported so far on the clinical application of photodynamic therapy in conversion therapy after drug resistance. Here we report a case of middle-aged patient with advanced GC, who experienced failure of conversion therapy consisted of multi-line chemotherapy along with immunotherapy. Ultimate success was achieved through a comprehensive conversion therapy of PDT, chemotherapy, immunotherapy, and targeted therapy. Subsequently, the patient underwent robotic-assisted radical gastrectomy while the surgical specimen showed no tumor cell exists. The patient underwent 3 cycles of systemic adjuvant therapy following surgical intervention. Presently, the patient remains 17 months in a satisfactory state of health.
Collapse
Affiliation(s)
- Jingyu Zhu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yunpeng Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Bo Xu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Weigao Pu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Puyi He
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Jing Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Haiyun Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Hao Chen
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Department of Surgical Oncology, The Second Hospital of Lanzhou University, Lanzhou 730030, China; Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou 730030, China.
| |
Collapse
|
7
|
Liang H, Yan X, Li Z, Chen X, Qiu Y, Li F, Wang M, Huang Z, Huang K, Xie Q, Zhang H, Zhong R, Zhao Z, Zou Y, Yu J, Hu Y, Liu H, Li G, Zhao L. Clinical outcomes of conversion surgery following immune checkpoint inhibitors and chemotherapy in stage IV gastric cancer. Int J Surg 2023; 109:4162-4172. [PMID: 37720943 PMCID: PMC10720795 DOI: 10.1097/js9.0000000000000738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND The clinical benefit of conversion surgery following immunochemotherapy in patients with stage IV gastric cancer (GC) remains uncertain. This study aims to clarify the clinical outcomes of conversion surgery for such patients. METHODS This retrospective cohort study enroled consecutive patients with stage IV GC treated with a combination of immune checkpoint inhibitors and chemotherapy and/or anti-human epidermal growth factor receptor-2 targeted therapy as first-line therapy. Cumulative survival curves were estimated using Kaplan-Meier method. Logistic regression and Cox regression analyses were conducted to identify factors associated with conversion surgery and survival, respectively. RESULTS Among the 136 patients included in the study. The disease control rate was 72.1% (98/136), with objective response rate in 58.8% (80/136) and complete response rate in 5.9% (8/136). Among 98 patients with disease control, 56 patients underwent palliative immunochemotherapy with median progression-free survival (PFS) and overall survival at 9.2 and 16.2 months, respectively; the remaining 42 patients underwent conversion surgery, yielding an unreached median PFS over a 19.0-month median follow-up, accompanied by 1-year overall survival and PFS rates of 96.6% and 89.1%, respectively. The R0 resection rate reached 90.5% (38/42). 7 out of 42 patients achieved pathological complete response, of whom three patients demonstrated human epidermal growth factor receptor-2 positivity. No serious complications leading to death were observed during the perioperative period. Multivariate analysis indicated that programmed death ligand 1 combined positive score greater than or equal to 5 (odds ratio, 0.22; 95% CI, 0.08-0.57; P =0.002) favored successful conversion surgery, while signet ring cell carcinoma (hazard ratio, 6.29; 95% CI, 1.56-25.36; P =0.010) was the poor prognostic factor associated with survival in patients who underwent conversion surgery. CONCLUSIONS Conversion surgery holds the potential for significant survival benefits in stage IV GC patients who have achieved a favourable clinical response to immunochemotherapy. Individuals with signet ring cell carcinoma may experience increased post-conversion surgery recurrence.
Collapse
Affiliation(s)
- Huayuan Liang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Xiao Yan
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Zhiwei Li
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Xinhua Chen
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Yaopeng Qiu
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Fengping Li
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Minghao Wang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Zhicheng Huang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Kaihua Huang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Qing Xie
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Huimin Zhang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Rou Zhong
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Zhuoyang Zhao
- Department of Pathology, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuan Zou
- Department of Pathology, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiang Yu
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
- Multidisciplinary Team of Gastric Tumor , Nanfang Hospital, Southern Medical University
| | - Yanfeng Hu
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
- Multidisciplinary Team of Gastric Tumor , Nanfang Hospital, Southern Medical University
| | - Hao Liu
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
- Multidisciplinary Team of Gastric Tumor , Nanfang Hospital, Southern Medical University
| | - Guoxin Li
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
- Multidisciplinary Team of Gastric Tumor , Nanfang Hospital, Southern Medical University
| | - Liying Zhao
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
- Multidisciplinary Team of Gastric Tumor , Nanfang Hospital, Southern Medical University
| |
Collapse
|