1
|
Lewis KA, Diggs LP, Badgwell BD. Educational Review: Updates on Therapeutic Strategies for Gastric Cancer with Peritoneal Metastasis. Ann Surg Oncol 2025; 32:3672-3687. [PMID: 40016614 DOI: 10.1245/s10434-025-17069-3] [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: 01/28/2025] [Accepted: 02/09/2025] [Indexed: 03/01/2025]
Abstract
Gastric cancer (GC) commonly presents in advanced stages with metastatic spread to the peritoneal cavity, and outcomes associated with gastric cancer with peritoneal metastasis (GCPM) continue to carry a dismal prognosis. Persistent challenges in the detection of peritoneal metastasis (PM) have resulted in a relative paucity of high-quality data to inform management decisions. Several consensus groups have published recommendations to guide management, including most recently the National Comprehensive Cancer Network guidelines, which now include cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) as a potential treatment modality in select patients with GCPM. Multiple clinical trials have investigated the use of CRS/HIPEC and other peritoneal-directed therapies, such as intraperitoneal chemotherapy (IPC) and pressurized intraperitoneal aerosolized chemotherapy (PIPAC). As high-volume centers work to incorporate such therapies into their practice, ongoing clinical trials are aimed at understanding their efficacy. Recent findings have improved understanding of the mechanisms and pathophysiology underlying GCPM while the discovery of novel targets offers potential for drug development and therapeutic strategies to overcome treatment resistance. This review highlights recent advancements and addresses the persistent challenges in managing GCPM while also offering a comprehensive summary of current guidelines and treatment strategies.
Collapse
Affiliation(s)
- Kever A Lewis
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Laurence P Diggs
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Sigler GI, Murtha J, Varley PR. Diagnostic Advances and Novel Therapeutics in Peritoneal Metastasis. Surg Oncol Clin N Am 2025; 34:173-194. [PMID: 40015798 DOI: 10.1016/j.soc.2024.12.005] [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] [Indexed: 03/01/2025]
Abstract
Appropriate assessment of disease burden in patients with peritoneal surface malignancy (PSM) is critical for treatment decision-making, and conventional cross-sectional imaging (computed tomography and/or MRI) often underestimates burden of disease. Advances in imaging for PSM include novel functional imaging modalities that target cells unique to the tumor microenvironment. Novel alternative methods of diagnosis and disease monitoring are also potentially applicable to management of PSM. These include forms of "liquid biopsy" targeting circulating tumor DNA. Novel regional therapies include both new therapeutic agents (immune-based and nanoparticle-based), as well as new methods of delivery such as pressurized intraperitoneal aerosolized chemotherapy.
Collapse
Affiliation(s)
- Gregory I Sigler
- Division of Surgical Oncology, Department of General Surgery, Complex General Surgical Oncology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Jacqueline Murtha
- Department of General Surgery, General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Patrick R Varley
- Division of Surgical Oncology, Department of General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA; William S. Middleton Memorial Veterans Affairs Hospital, Madison, WI, USA.
| |
Collapse
|
3
|
Zhang F, Ding Z, Lian Y, Yang X, Hu P, Liu Y, Xu L, Li Z, Qiu H. Prophylactic antibiotic use is associated with better clinical outcomes in gastric cancer patients receiving immunotherapy. Oncologist 2025; 30:oyae362. [PMID: 40036772 PMCID: PMC11879193 DOI: 10.1093/oncolo/oyae362] [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: 04/26/2024] [Accepted: 11/03/2024] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The relationship between antibiotic treatment and immunotherapy efficacy is complex. METHODS This study was a single-center study. History of antibiotic use in gastric cancer (GC) patients within 1 or 3 months prior to immunotherapy was collected. Patients were categorized into 3 groups according to whether they had used antibiotics prior to immunotherapy: none, prophylactic use, and infection. RESULTS A total of 252 GC patients received immunotherapy, of which 38.5% (97/252) received antibiotic treatment within 1 month before immunotherapy (prophylactic use in 72.2% of patients) and 48.8% (123/252) received antibiotic treatment within 3 months before immunotherapy (prophylactic use in 74.8% of patients). The prophylactic use of antibiotic within 1 month prior to immunotherapy significantly improved overall survival (OS) compared with patients who received anti-infective therapy and had no history of antibiotic use (prophylactic vs infection: OS, 22.6 vs 9.7 m, HR, 0.53, 95% CI, 0.27-1.07; prophylactic vs none: OS, 22.6 vs 14.7 m, HR, 0.57; 95% CI, 0.39-0.83). The use of antibiotics in infected patients did not increase the risk of death in patients compared with those who did not use antibiotics. Prophylactic antibiotic use within 1 month before immunotherapy is an independent prognostic factor for OS. CONCLUSIONS Prophylactic use of antibiotics is associated with better prognosis in GC patients receiving immunotherapy. Therefore, there is no necessity to delay the use of immune checkpoint inhibitors in this group of patients.
Collapse
Affiliation(s)
- Fangyuan Zhang
- Department of Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong, University of Science and Technology, Wuhan 430000, Hubei, China
| | - Zixuan Ding
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yongping Lian
- Department of Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong, University of Science and Technology, Wuhan 430000, Hubei, China
| | - Xiao Yang
- Department of Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong, University of Science and Technology, Wuhan 430000, Hubei, China
| | - Pengbo Hu
- Department of Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong, University of Science and Technology, Wuhan 430000, Hubei, China
| | - Yongqing Liu
- Department of Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong, University of Science and Technology, Wuhan 430000, Hubei, China
| | - Liang Xu
- Department of Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong, University of Science and Technology, Wuhan 430000, Hubei, China
| | - Zhou Li
- Department of Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong, University of Science and Technology, Wuhan 430000, Hubei, China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong, University of Science and Technology, Wuhan 430000, Hubei, China
| |
Collapse
|
4
|
Mela E, Theodorou AP, Kimpizi D, Konstantinou K, Belimezakis N, Schizas D, Theodorou D, Triantafyllou T. Emerging Trends in the Management of Gastric Malignancy with Peritoneal Dissemination: Same Disease, Heterogeneous Prognosis. Cancers (Basel) 2025; 17:117. [PMID: 39796744 PMCID: PMC11719794 DOI: 10.3390/cancers17010117] [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: 11/29/2024] [Revised: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Gastric cancer is a significant global contributor to cancer-related mortality. Stage IV gastric cancer represents a significant percentage of patients in Western countries, with peritoneal dissemination being the most prevalent site. Peritoneal disease comprises two distinct entities, macroscopic (P1) and microscopic (P0CY1), which are associated with poor long-term survival rates. Although the present standard of treatment is palliative chemotherapy, a global controversy has arisen concerning specific patients with limited disease burden or conversion to negative lavage cytology following chemotherapy. Available approaches include systemic or intraperitoneal chemotherapy, upfront gastrectomy, and conversion surgery. This review consolidated the current evidence regarding multimodal management, indicating prolonged survival for this distinct subgroup of patients. Considering the complexity of peritoneal metastases, the potential of the multimodal approach unveils promising prospects for identifying the optimal treatment for this particular subset of stage IV patients and thus enhancing their survival outcomes.
Collapse
Affiliation(s)
- Evgenia Mela
- First Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Hippocration General Hospital, 11527 Athens, Greece; (A.P.T.); (D.K.); (K.K.); (N.B.); (D.T.); (T.T.)
| | - Andreas Panagiotis Theodorou
- First Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Hippocration General Hospital, 11527 Athens, Greece; (A.P.T.); (D.K.); (K.K.); (N.B.); (D.T.); (T.T.)
| | - Despina Kimpizi
- First Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Hippocration General Hospital, 11527 Athens, Greece; (A.P.T.); (D.K.); (K.K.); (N.B.); (D.T.); (T.T.)
| | - Kyriaki Konstantinou
- First Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Hippocration General Hospital, 11527 Athens, Greece; (A.P.T.); (D.K.); (K.K.); (N.B.); (D.T.); (T.T.)
| | - Nektarios Belimezakis
- First Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Hippocration General Hospital, 11527 Athens, Greece; (A.P.T.); (D.K.); (K.K.); (N.B.); (D.T.); (T.T.)
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 11527 Athens, Greece;
| | - Dimitrios Theodorou
- First Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Hippocration General Hospital, 11527 Athens, Greece; (A.P.T.); (D.K.); (K.K.); (N.B.); (D.T.); (T.T.)
| | - Tania Triantafyllou
- First Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Hippocration General Hospital, 11527 Athens, Greece; (A.P.T.); (D.K.); (K.K.); (N.B.); (D.T.); (T.T.)
| |
Collapse
|
5
|
Zhu ZN, Feng QX, Li Q, Xu WY, Liu XS. Machine learning-based CT radiomics approach for predicting occult peritoneal metastasis in advanced gastric cancer preoperatively. Clin Radiol 2025; 80:106727. [PMID: 39571365 DOI: 10.1016/j.crad.2024.10.008] [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: 02/20/2024] [Revised: 08/29/2024] [Accepted: 10/14/2024] [Indexed: 01/18/2025]
Abstract
AIM To develop a machine learning-based CT radiomics model to preoperatively diagnose occult peritoneal metastasis (OPM) in advanced gastric cancer (AGC) patients. MATERIALS AND METHODS A total of 177 AGC patients were retrospectively analyzed. Four regions of interest (ROIs) along the largest area of tumor (core ROI) and corresponding tumor mesenteric fat space (peri ROI) were manually delineated on the arterial (A-core and A-peri) and venous phase (V-core and V-peri) of CT images. A total of 1316 radiomics features were extracted from each ROI. Then, ten machine learning classification algorithms were used to develop the model. An integrated radiomics nomogram was established to predict OPM individually. RESULTS For the radiomics of tumor mesenteric fat space, the AUCs of A-peri in training and test sets were 0.881 and 0.800, respectively. And the AUCs of V-peri were 0.838 and 0.815, respectively. In terms of primary tumor' s radiomics signature, the AUCs of A-core in training and test sets were 0.862 and 0.691, respectively. The AUCs of V-core were 0.831 and 0.620. Integrated radiomics model showed the highest AUC value when it compared to each single radiomics score in the training (0.943 vs 0.831-0.881) and test set (0.835 vs 0.620-0.815). Radiomics nomogram demonstrated good diagnostic accuracy with a C-index of 0.948. CONCLUSION Both the radiomics of tumor mesenteric fat space and primary tumor were associated with OPM. A CT radiomics nomogram had a relatively good predictive performance for detecting OPM in patients with AGC.
Collapse
Affiliation(s)
- Z-N Zhu
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| | - Q-X Feng
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| | - Q Li
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| | - W-Y Xu
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| | - X-S Liu
- Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210000, China.
| |
Collapse
|
6
|
Kim IH, Kang SJ, Choi W, Seo AN, Eom BW, Kang B, Kim BJ, Min BH, Tae CH, Choi CI, Lee CK, An HJ, Byun HK, Im HS, Kim HD, Cho JH, Pak K, Kim JJ, Bae JS, Yu JI, Lee JW, Choi J, Kim JH, Choi M, Jung MR, Seo N, Eom SS, Ahn S, Kim SJ, Lee SH, Lim SH, Kim TH, Han HS. Korean Practice Guidelines for Gastric Cancer 2024: An Evidence-based, Multidisciplinary Approach (Update of 2022 Guideline). J Gastric Cancer 2025; 25:5-114. [PMID: 39822170 PMCID: PMC11739648 DOI: 10.5230/jgc.2025.25.e11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025] Open
Abstract
Gastric cancer is one of the most common cancers in both Korea and worldwide. Since 2004, the Korean Practice Guidelines for Gastric Cancer have been regularly updated, with the 4th edition published in 2022. The 4th edition was the result of a collaborative work by an interdisciplinary team, including experts in gastric surgery, gastroenterology, endoscopy, medical oncology, abdominal radiology, pathology, nuclear medicine, radiation oncology, and guideline development methodology. The current guideline is the 5th version, an updated version of the 4th edition. In this guideline, 6 key questions (KQs) were updated or proposed after a collaborative review by the working group, and 7 statements were developed, or revised, or discussed based on a systematic review using the MEDLINE, Embase, Cochrane Library, and KoreaMed database. Over the past 2 years, there have been significant changes in systemic treatment, leading to major updates and revisions focused on this area. Additionally, minor modifications have been made in other sections, incorporating recent research findings. The level of evidence and grading of recommendations were categorized according to the Grading of Recommendations, Assessment, Development and Evaluation system. Key factors for recommendation included the level of evidence, benefit, harm, and clinical applicability. The working group reviewed and discussed the recommendations to reach a consensus. The structure of this guideline remains similar to the 2022 version. Earlier sections cover general considerations, such as screening, diagnosis, and staging of endoscopy, pathology, radiology, and nuclear medicine. In the latter sections, statements are provided for each KQ based on clinical evidence, with flowcharts supporting these statements through meta-analysis and references. This multidisciplinary, evidence-based gastric cancer guideline aims to support clinicians in providing optimal care for gastric cancer patients.
Collapse
Affiliation(s)
- In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Joo Kang
- Department of Internal Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Wonyoung Choi
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Bang Wool Eom
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Beodeul Kang
- Division of Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Bum Jun Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Anyang, Korea
| | - Byung-Hoon Min
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chung Hyun Tae
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Chang In Choi
- Department of Surgery, Pusan National University Hospital, Busan, Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Ho Jung An
- Division of Oncology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Hwa Kyung Byun
- Department of Radiation Oncology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Hyeon-Su Im
- Department of Hematology and Oncology, Ulsan University Hospital, Ulsan University College of Medicine, Ulsan, Korea
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jang Ho Cho
- Division of Medical Oncology, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Kyoungjune Pak
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Jae-Joon Kim
- Division of Hematology and Oncology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jae Seok Bae
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Korea
| | - Jeong Il Yu
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul, Korea
| | - Jeong Won Lee
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jungyoon Choi
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Jwa Hoon Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Miyoung Choi
- National Evidence-based Healthcare Collaborating Agency (NECA), Seoul, Korea
| | - Mi Ran Jung
- Department of Surgery, Chonnam National University Medical School, Gwangju, Korea
| | - Nieun Seo
- Department of Radiology, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Soo Eom
- Department of Surgery, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Soomin Ahn
- Department of Pathology and Translational Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Jin Kim
- Department of Radiology, National Cancer Center, Goyang, Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Hee Lim
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Seoul, Korea
| | - Tae-Han Kim
- Department of Surgery, Gyeongsang National University Changwon Hospital, Changwon, Korea.
| | - Hye Sook Han
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea.
| |
Collapse
|
7
|
Cai XX, Chen GM, Zheng ZQ, Yin YX, Wang S, Qiao L, Chen XJ, Zhao BW, Duan JL, Liang CC, Zhang RP, Wei CZ, Zhang FY, Huang BW, Liu ZX, Zhou ZW, Xie D, Cai MY, Yuan SQ, Li YF, Nie RC. Transcriptional landscape and predictive potential of long noncoding RNAs in peritoneal recurrence of gastric cancer. Mol Cancer 2024; 23:284. [PMID: 39736670 DOI: 10.1186/s12943-024-02196-4] [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: 07/16/2024] [Accepted: 12/08/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) play a critical role in gastric cancer (GC) progression and metastasis. However, research comprehensively exploring tissue-derived lncRNAs for predicting peritoneal recurrence in patients with GC remains limited. This study aims to investigate the transcriptional landscape of lncRNAs in GC with peritoneal metastasis (PM) and to develop an integrated lncRNA-based score to predict peritoneal recurrence in patients with GC after radical gastrectomy. METHODS We analyzed the transcriptome profile of lncRNAs in paired peritoneal, primary gastric tumor, and normal tissue specimens from 12 patients with GC in the Sun Yat-sen University Cancer Center (SYSUCC) discovery cohort. Key lncRNAs were identified via interactive analysis with the TCGA database and SYSUCC validation cohort. A score model was constructed using the LASSO regression model and nomogram COX regression and evaluated using receiver operating characteristic curves. The role of lncRNAs in the PM of GC was then examined through wound healing, Transwell, 3D multicellular tumor spheroid invasion, and peritoneal cavity xenograft tumorigenicity assays in mice. RESULT Five essential lncRNAs were screened and incorporated into the PM risk score to predict peritoneal recurrence-free survival (pRFS). We developed a comprehensive, integrated nomogram score, including the PM risk score, pT, pN, and tumor size, which could effectively predict the 5-year pRFS with an Area under the curve of 0.79 (95% CI: 0.71-0.88). Subsequently, we demonstrated that one of these lncRNAs, CASC15, promoted the invasion and migration of GC cells in vitro and facilitated the PM of GC cells in vivo, initially verifying that lncRNAs contribute to the PM of GC. Mechanistic analysis demonstrated that CASC15 promoted EMT and metastasis by activating the JNK and p38 pathways. CONCLUSION A lncRNA-based integrated score was constructed in this study to predict peritoneal recurrence in patients clinically.
Collapse
Affiliation(s)
- Xiao-Xia Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Guo-Ming Chen
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Zi-Qi Zheng
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yi-Xin Yin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Shuang Wang
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Li Qiao
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510060, P. R. China
| | - Xiao-Jiang Chen
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Bai-Wei Zhao
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jin-Ling Duan
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Cheng-Cai Liang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ruo-Peng Zhang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Cheng-Zhi Wei
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Fei-Yang Zhang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Bo-Wen Huang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Mu-Yan Cai
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Yuan-Fang Li
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Run-Cong Nie
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
8
|
van der Sluis K, Vollebergh MA, Kodach LL, van Dieren JM, de Hingh IHJT, Wijnhoven BPL, Verhoeven RHA, Sandick JWV. The clinical implications of staging laparoscopy in the diagnostic workup of gastric cancer patients: A population based study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108569. [PMID: 39134081 DOI: 10.1016/j.ejso.2024.108569] [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: 04/10/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Since 2016, staging laparoscopy has been implemented in the diagnostic workup of patients with gastric cancer. Staging laparoscopy aims to detect incurable disease (peritoneal metastases and irresectable tumors) and to prevent futile laparotomies. METHODS In this population-based nationwide study, we sought patient- and tumor characteristics associated with undergoing a staging laparoscopy. Additionally, we analyzed the prevalence of synchronous peritoneal metastases, the outcome of the staging laparoscopy and its clinical impact on treatment decisions. All patients diagnosed with non-cardia gastric cancer from the Netherlands Cancer Registry between 2016 and 2021 were included. RESULTS Alongside tumor characteristics, patient characteristics such as younger age, absence of comorbidities and lower WHO performance status were associated with performing a staging laparoscopy. In the study period, an increase in the proportion of patients who underwent a staging laparoscopy was observed, from 19.6% in 2016 to 32.3% in 2021 (p-value<0.001). In the same period, the prevalence of synchronous peritoneal metastases increased from 25% to 31%. In 37.6% of the patients who had the outcome of their staging laparoscopy reported, had incurable disease diagnosed during staging laparoscopy. Significantly less of these patients were treated with triplet regimens as compared to patients with a negative staging laparoscopy (18.5 vs. 76.3%; p-value<0.001). CONCLUSION The implementation of staging laparoscopy in gastric cancer patients paralleled the increase in diagnosis of incurable disease and a decrease in the application of triplet systemic therapies in these patients.
Collapse
Affiliation(s)
- K van der Sluis
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands.
| | - M A Vollebergh
- The Netherlands Cancer Institute, Department of Gastrointestinal Oncology, Amsterdam, the Netherlands
| | - L L Kodach
- The Netherlands Cancer Institute, Department of Pathology, Amsterdam, the Netherlands
| | - J M van Dieren
- The Netherlands Cancer Institute, Department of Gastrointestinal Oncology, Amsterdam, the Netherlands
| | - I H J T de Hingh
- Catharina Hospital, Department of Surgery, Eindhoven, the Netherlands; Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands; Netherlands Comprehensive Cancer Organization (IKNL), Department of Research & Development, Utrecht, the Netherlands
| | - B P L Wijnhoven
- Erasmus Medical Centre, Department of Surgery, Rotterdam, the Netherlands
| | - R H A Verhoeven
- Netherlands Comprehensive Cancer Organization (IKNL), Department of Research & Development, Utrecht, the Netherlands; Amsterdam UMC Location University of Amsterdam, Medical Oncology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - J W van Sandick
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Zhu ZN, Feng QX, Li Q, Xu WY, Liu XS. Utility of Combined Use of Imaging Features From Abdominopelvic CT and CA 125 to Identify Presence of CT Occult Peritoneal Metastases in Advanced Gastric Cancer. J Comput Assist Tomogr 2024; 48:734-742. [PMID: 38595104 DOI: 10.1097/rct.0000000000001600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
OBJECTIVE The purpose of this study is to identify the presence of occult peritoneal metastasis (OPM) in patients with advanced gastric cancer (AGC) by using clinical characteristics and abdominopelvic computed tomography (CT) features. METHODS This retrospective study included 66 patients with OPM and 111 patients without peritoneal metastasis (non-PM [NPM]) who underwent preoperative contrast-enhanced CT between January 2020 and December 2021. Occult PMs means PMs that are missed by CT but later diagnosed by laparoscopy or laparotomy. Patients with NPM means patients have neither PM nor other distant metastases, indicating there is no evidence of distant metastases in patients with AGC. Patients' clinical characteristics and CT features such as tumor marker, Borrmann IV, enhancement patterns, and pelvic ascites were observed by 2 experienced radiologists. Computed tomography features and clinical characteristics were combined to construct an indicator for identifying the presence of OPM in patients with AGC based on a logistic regression model. Receiver operating characteristic curves and the area under the receiver operating characteristic curve (AUC) were generated to assess the diagnostic performance of the combined indicator. RESULTS Four independent predictors (Borrmann IV, pelvic ascites, carbohydrate antigen 125, and normalized arterial CT value) differed significantly between OPM and NPM and performed outstandingly in distinguishing patients with OPM from those without PM (AUC = 0.643-0.696). The combined indicator showed a higher AUC value than the independent risk factors (0.820 vs 0.643-0.696). CONCLUSIONS The combined indicator based on abdominopelvic CT features and carbohydrate antigen 125 may assist clinicians in identifying the presence of CT OPMs in patients with AGC.
Collapse
Affiliation(s)
- Zhen-Ning Zhu
- From the Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | | | | | | | | |
Collapse
|
10
|
Lan YY, Han J, Liu YY, Lan L. Construction of a predictive model for gastric cancer neuroaggression and clinical validation analysis: A single-center retrospective study. World J Gastrointest Surg 2024; 16:2602-2611. [PMID: 39220072 PMCID: PMC11362950 DOI: 10.4240/wjgs.v16.i8.2602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/08/2024] [Accepted: 06/27/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND This study investigated the construction and clinical validation of a predictive model for neuroaggression in patients with gastric cancer. Gastric cancer is one of the most common malignant tumors in the world, and neuroinvasion is the key factor affecting the prognosis of patients. However, there is a lack of systematic analysis on the construction and clinical application of its prediction model. This study adopted a single-center retrospective study method, collected a large amount of clinical data, and applied statistics and machine learning technology to build and verify an effective prediction model for neuroaggression, with a view to providing scientific basis for clinical treatment decisions and improving the treatment effect and survival rate of patients with gastric cancer. AIM To investigate the value of a model based on clinical data, spectral computed tomography (CT) parameters and image omics characteristics for the preoperative prediction of nerve invasion in patients with gastric cancer. METHODS A retrospective analysis was performed on 80 gastric cancer patients who underwent preoperative energy spectrum CT at our hospital between January 2022 and August 2023, these patients were divided into a positive group and a negative group according to their pathological results. Clinicopathological data were collected, the energy spectrum parameters of primary gastric cancer lesions were measured, and single factor analysis was performed. A total of 214 image omics features were extracted from two-phase mixed energy images, and the features were screened by single factor analysis and a support vector machine. The variables with statistically significant differences were included in logistic regression analysis to construct a prediction model, and the performance of the model was evaluated using the subject working characteristic curve. RESULTS There were statistically significant differences in sex, carbohydrate antigen 199 expression, tumor thickness, Lauren classification and Borrmann classification between the two groups (all P < 0.05). Among the energy spectrum parameters, there were statistically significant differences in the single energy values (CT60-CT110 keV) at the arterial stage between the two groups (all P < 0.05) and statistically significant differences in CT values, iodide group values, standardized iodide group values and single energy values except CT80 keV at the portal vein stage between the two groups (all P < 0.05). The support vector machine model with the largest area under the curve was selected by image omics analysis, and its area under the curve, sensitivity, specificity, accuracy, P value and parameters were 0.843, 0.923, 0.714, 0.925, < 0.001, and c:g 2.64:10.56, respectively. Finally, based on the logistic regression algorithm, a clinical model, an energy spectrum CT model, an imaging model, a clinical + energy spectrum model, a clinical + imaging model, an energy spectrum + imaging model, and a clinical + energy spectrum + imaging model were established, among which the clinical + energy spectrum + imaging model had the best efficacy in diagnosing gastric cancer nerve invasion. The area under the curve, optimal threshold, Youden index, sensitivity and specificity were 0.927 (95%CI: 0.850-1.000), 0.879, 0.778, 0.778, and 1.000, respectively. CONCLUSION The combined model based on clinical features, spectral CT parameters and imaging data has good value for the preoperative prediction of gastric cancer neuroinvasion.
Collapse
Affiliation(s)
- Yu-Yin Lan
- Department of Stomatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Jing Han
- Department of Biobank, Zhejiang Cancer Hospital, Hangzhou 310005, Zhejiang Province, China
| | - Yan-Yan Liu
- Department of General Surgery, Peking University First Hospital, Beijing 100034, China
| | - Lei Lan
- Department of Oncology, Zhejiang Hospital, Hangzhou 310013, Zhejiang Province, China
| |
Collapse
|
11
|
Wang J, Liang JC, Lin FT, Ma J. Energy spectrum computed tomography multi-parameter imaging in preoperative assessment of vascular and neuroinvasive status in gastric cancer. World J Gastrointest Surg 2024; 16:2511-2520. [PMID: 39220074 PMCID: PMC11362936 DOI: 10.4240/wjgs.v16.i8.2511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Vascular and nerve infiltration are important indicators for the progression and prognosis of gastric cancer (GC), but traditional imaging methods have some limitations in preoperative evaluation. In recent years, energy spectrum computed tomography (CT) multiparameter imaging technology has been gradually applied in clinical practice because of its advantages in tissue contrast and lesion detail display. AIM To explore and analyze the value of multiparameter energy spectrum CT imaging in the preoperative assessment of vascular invasion (LVI) and nerve invasion (PNI) in GC patients. METHODS Data from 62 patients with GC confirmed by pathology and accompanied by energy spectrum CT scanning at our hospital between September 2022 and September 2023, including 46 males and 16 females aged 36-71 (57.5 ± 9.1) years, were retrospectively collected. The patients were divided into a positive group (42 patients) and a negative group (20 patients) according to the presence of LVI/PNI. The CT values (CT40 keV, CT70 keV), iodine concentration (IC), and normalized IC (NIC) of lesions in the upper energy spectrum CT images of the arterial phase, venous phase, and delayed phase 40 and 70 keV were measured, and the slopes of the energy spectrum curves [K (40-70)] from 40 to 70 keV were calculated. Arterial phase combined parameter, venous phase combined parameters (VP-ALLs), and delayed phase association parameters were calculated for patients with late-stage disease. The differences in the energy spectrum parameters between the positive and negative groups were compared, receiver operating characteristic (ROC) curves were plotted, and the area under the curve (AUC), sensitivity, specificity, and optimal threshold were calculated to measure the diagnostic efficiency of each parameter. RESULTS In the delayed phase, the CT40 keV, CT70 keV, K (40-70), IC, NIC, and CT70 keV and the NIC in the upper arterial and venous phases of energy spectrum CT were greater in the LVI/PNI-positive group than in the LVI-negative group. The representative parameters for the arterial phase NIC were 0.14 ± 0.04 in the positive group and 0.12 ± 0.04 in the negative group. The venous phase NIC was 0.5 (0.5, 0.6) in the positive group and 0.4 (0.4, 0.5) in the negative group. Last, for the delayed phase NIC, it was 0.6 ± 0.1 in the positive group and 0.5 ± 0.1 in the negative group (all P values are less than 0.05). ROC curve analysis demonstrated that the diagnostic efficacy of each parameter during the venous stage was superior to that during the arterial and delayed stages. Furthermore, the diagnostic efficacy of the combined parameter throughout all three stages was superior to that of any single parameter. The AUC, sensitivity, and specificity of the optimal parameter, VP-ALL, were 0.931 (95% confidence interval: 0.872-0.990), 80.95%, and 95.00%, respectively. CONCLUSION When assessing the condition of LVI and PNI (perineural invasion) in patients with GC prior to surgery, the ability to diagnose these conditions using venous stage parameters was superior to that using arterial stage and delayed stage parameters. Furthermore, the diagnostic accuracy of using a combination of parameters was better than that of using individual parameters alone.
Collapse
Affiliation(s)
- Jing Wang
- Department of Radiology, Pingluo County People's Hospital, Shizuishan 753400, Ningxia Hui Autonomous Region, China
| | - Jian-Cheng Liang
- Department of Radiology, Pingluo County People's Hospital, Shizuishan 753400, Ningxia Hui Autonomous Region, China
| | - Fa-Te Lin
- Department of Gastrointestinal Surgery, Jiangsu Provincial People's Hospital, Nanjing 210029, Jiangsu Province, China
| | - Jun Ma
- Department of Radiology, Pingluo County People's Hospital, Shizuishan 753400, Ningxia Hui Autonomous Region, China
| |
Collapse
|
12
|
van der Sluis K, Guchelaar NAD, Triemstra L, Mathijssen RHJ, Ruurda JP, Wijnhoven BPL, van Sandick JW. Staging laparoscopy in gastric cancer patients: From a Dutch nationwide Delphi consensus towards a standardized protocol. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108278. [PMID: 38531232 DOI: 10.1016/j.ejso.2024.108278] [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: 01/23/2024] [Revised: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Staging laparoscopy is a common diagnostic tool in gastric cancer, but its performance varies widely. The aim of this study was to gain Dutch nationwide consensus regarding the indications for and execution of staging laparoscopy in patients with gastric cancer. METHODS All surgeons in the Netherlands specialized in gastric cancer surgery (n = 52) were asked to participate in a Delphi consensus study. The study involved an initial questionnaire with a 3-point Likert scale, an online consensus meeting, and a second questionnaire using a 2-point Likert scale (agree/disagree). Consensus was defined as 70% or more agreement among participants. RESULTS In total, 45 experts completed both questionnaires (87% response rate). Consensus was reached on the indication to perform staging laparoscopy in cT3-4 or cN + or diffuse-type gastric cancer, including Siewert type III oesophagogastric junctional cancer. The experts agreed that if preoperative scans suggest infiltration of surrounding organs (cT4), the tumour's resectability should explicitly be investigated. Consensus was also reached for a systematic peritoneal cavity inspection according to Sugarbaker's Peritoneal Cancer Index (PCI) score. All regions should be inspected routinely, although the omental bursa may be inspected on indication. Aspiration of ascites or peritoneal washing should be performed for cytology. The experts agreed that restaging laparoscopy should be performed before resection in case of progressive disease on preoperative imaging. Without progression, global inspection was considered sufficient. CONCLUSIONS The results of this Dutch nationwide Delphi consensus study exposed the variability of performing staging laparoscopy in patients with gastric cancer and provided the concept for a standardized protocol.
Collapse
Affiliation(s)
| | - Niels A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Lianne Triemstra
- Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | | |
Collapse
|
13
|
Zhang S, Hu Q, Chen X, Zhou N, Huang Q, Tan S, Su M, Gou H. 68Ga-FAPI-04 positron emission tomography/CT and laparoscopy for the diagnosis of occult peritoneal metastasis in newly diagnosed locally advanced gastric cancer: study protocol of a single-centre prospective cohort study. BMJ Open 2024; 14:e075680. [PMID: 38643004 PMCID: PMC11033661 DOI: 10.1136/bmjopen-2023-075680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 03/22/2024] [Indexed: 04/22/2024] Open
Abstract
INTRODUCTION Accurate baseline clinical staging is critical to inform treatment decision-making for patients with gastric cancers. Peritoneal metastasis (PM) is the most common form of metastasis in gastric cancer and mainly diagnosed by diagnostic laparoscopy and peritoneal lavage evaluation. However, diagnostic laparoscopy is invasive and less cost-effective. It is urgent to develop a safe, fast and non-invasive functional imaging method to verify the peritoneal metastasis of gastric cancer. The aim of our study was to evaluate the proportion of patients in whom 68Ga-FAPI-04 positron emission tomography/CT (PET/CT) led to a change in treatment strategy and to assess the diagnostic accuracy of 68Ga-FAPI-04 PET/CT for the detection of occult peritoneal metastasis compared with laparoscopic exploration. METHODS AND ANALYSIS In this single-centre, prospective diagnostic test accuracy study, a total of 48 patients with locally advanced gastric or gastro-oesophageal junction adenocarcinoma (cT4a-b, N0-3, M0, based on CT images) who are considering radical tumour surgery will be recruited. All participants will undergo 68Ga-FAPI-04 PET/CT before the initiation of laparoscopic exploration. The primary outcome is the proportion of patients with occult peritoneal metastatic lesions detected by 68Ga-FAPI-04 PET/CT, leading to a change in therapy strategy. The secondary outcomes include the diagnostic performance of 68Ga-FAPI-04 PET/CT for occult peritoneal metastasis, including sensitivity, specificity, accuracy, positive predictive value and negative predictive value. ETHICS AND DISSEMINATION The study protocol was approved by the Ethics Committee of West China Hospital, Sichuan University (2022-1484). Study results will be presented at public and scientific conferences and in peer-reviewed journals. TRIAL REGISTRATION NUMBER ChiCTR2300067591.
Collapse
Affiliation(s)
- Shunyu Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Qiancheng Hu
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Xinchuan Chen
- Department of Hematology, Sichuan University, Chengdu, Sichuan, China
| | - Nan Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Qiyue Huang
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Sirui Tan
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Minggang Su
- Department of Nuclear Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Wei GX, Zhou YW, Li ZP, Qiu M. Application of artificial intelligence in the diagnosis, treatment, and recurrence prediction of peritoneal carcinomatosis. Heliyon 2024; 10:e29249. [PMID: 38601686 PMCID: PMC11004411 DOI: 10.1016/j.heliyon.2024.e29249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
Peritoneal carcinomatosis (PC) is a type of secondary cancer which is not sensitive to conventional intravenous chemotherapy. Treatment strategies for PC are usually palliative rather than curative. Recently, artificial intelligence (AI) has been widely used in the medical field, making the early diagnosis, individualized treatment, and accurate prognostic evaluation of various cancers, including mediastinal malignancies, colorectal cancer, lung cancer more feasible. As a branch of computer science, AI specializes in image recognition, speech recognition, automatic large-scale data extraction and output. AI technologies have also made breakthrough progress in the field of peritoneal carcinomatosis (PC) based on its powerful learning capacity and efficient computational power. AI has been successfully applied in various approaches in PC diagnosis, including imaging, blood tests, proteomics, and pathological diagnosis. Due to the automatic extraction function of the convolutional neural network and the learning model based on machine learning algorithms, AI-assisted diagnosis types are associated with a higher accuracy rate compared to conventional diagnosis methods. In addition, AI is also used in the treatment of peritoneal cancer, including surgical resection, intraperitoneal chemotherapy, systemic chemotherapy, which significantly improves the survival of patients with PC. In particular, the recurrence prediction and emotion evaluation of PC patients are also combined with AI technology, further improving the quality of life of patients. Here we have comprehensively reviewed and summarized the latest developments in the application of AI in PC, helping oncologists to comprehensively diagnose PC and provide more precise treatment strategies for patients with PC.
Collapse
Affiliation(s)
- Gui-Xia Wei
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yu-Wen Zhou
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Zhi-Ping Li
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Meng Qiu
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Radomski SN, Dunworth M, West JJ, Greer JB, Johnston FM, Ewald AJ. Intra- and Interpatient Drug Response Heterogeneity Exist in Patients Undergoing Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for Nongynecologic Cancers. Ann Surg Oncol 2024; 31:1996-2007. [PMID: 38175427 DOI: 10.1245/s10434-023-14696-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Select patients with peritoneal metastases are treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC). We assayed for intra- and interpatient drug response heterogeneity through testing of patient-derived tumor organoids (PDTOs). METHODS PDTOs were generated from CRS/HIPEC patients from December 2021 to September 2022 and subjected to an in vitro HIPEC drug screen. Drug response was assessed with a cell viability assay and cleaved caspase-3 staining. RESULTS A total of 31 patients were consented for tissue collection. Viable tissue was harvested from 23, and PDTO generation was successful in 13 (56%). PDTOs were analyzed from six appendiceal, three colorectal, two small bowel, one gastric, and one adrenal tumor. Drug screen results were generated in as few as 7 days (62%), with an average time of 12 days. Most patients received mitomycin-C (MMC) intraoperatively (n = 9); however, in only three cases was this agent considered the optimal choice in vitro. Three sets of PDTOs were resistant (defined as > 50% PDTO viability) to all agents tested and two were pan-sensitive (defined as 3 or more agents with < 50% PDTO viability). In three patients, organoids were generated from multiple metastatic sites and intrapatient drug response heterogeneity was observed. CONCLUSIONS Both intra- and interpatient drug response heterogeneity exist in patients undergoing CRS/HIPEC for nongynecologic abdominal cancers. Caution must be used when interpreting patient response to chemotherapeutic agents based on a single site of testing in those with metastatic disease.
Collapse
Affiliation(s)
- Shannon N Radomski
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew Dunworth
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Junior J West
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan B Greer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fabian M Johnston
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Andrew J Ewald
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
- Giovanis Institute for Translational Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
16
|
Xie J, Xue B, Bian S, Ji X, Lin J, Zheng X, Tang K. A radiomics nomogram based on 18 F-FDG PET/CT and clinical risk factors for the prediction of peritoneal metastasis in gastric cancer. Nucl Med Commun 2023; 44:977-987. [PMID: 37578301 DOI: 10.1097/mnm.0000000000001742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
PURPOSE Peritoneal metastasis (PM) is usually considered an incurable factor of gastric cancer (GC) and not fit for surgery. The aim of this study is to develop and validate an 18 F-FDG PET/CT-derived radiomics model combining with clinical risk factors for predicting PM of GC. METHOD In this retrospective study, 410 GC patients (PM - = 281, PM + = 129) who underwent preoperative 18 F-FDG PET/CT images from January 2015 to October 2021 were analyzed. The patients were randomly divided into a training cohort (n = 288) and a validation cohort (n = 122). The maximum relevance and minimum redundancy (mRMR) and the least shrinkage and selection operator method were applied to select feature. Multivariable logistic regression analysis was preformed to develop the predicting model. Discrimination, calibration, and clinical usefulness were used to evaluate the performance of the nomogram. RESULT Fourteen radiomics feature parameters were selected to construct radiomics model. The area under the curve (AUC) of the radiomics model were 0.86 [95% confidence interval (CI), 0.81-0.90] in the training cohort and 0.85 (95% CI, 0.78-0.92) in the validation cohort. After multivariable logistic regression, peritoneal effusion, mean standardized uptake value (SUVmean), carbohydrate antigen 125 (CA125) and radiomics signature showed statistically significant differences between different PM status patients( P < 0.05). They were chosen to construct the comprehensive predicting model which showed a performance with an AUC of 0.92 (95% CI, 0.89-0.95) in the training cohort and 0.92 (95% CI, 0.86-0.98) in the validation cohort, respectively. CONCLUSION The nomogram based on 18 F-FDG PET/CT radiomics features and clinical risk factors can be potentially applied in individualized treatment strategy-making for GC patients before the surgery.
Collapse
Affiliation(s)
- Jiageng Xie
- Departments of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Beihui Xue
- Departments of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuying Bian
- Departments of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaowei Ji
- Departments of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Lin
- Departments of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangwu Zheng
- Departments of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kun Tang
- Departments of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
17
|
Ho SYA, Tay KV. Systematic review of diagnostic tools for peritoneal metastasis in gastric cancer-staging laparoscopy and its alternatives. World J Gastrointest Surg 2023; 15:2280-2293. [PMID: 37969710 PMCID: PMC10642463 DOI: 10.4240/wjgs.v15.i10.2280] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Gastric cancer is one of the leading causes of cancer burden and mortality, often resulting in peritoneal metastasis in advanced stages with negative survival outcomes. Staging laparoscopy has become standard practice for suspected cases before a definitive gastrectomy or palliation. This systematic review aims to compare the efficacy of other diagnostic modalities instead of staging laparoscopy as the alternatives are able to reduce cost and invasive staging procedures. Recently, a radiomic model based on computed tomography and positron emission tomography (PET) has also emerged as another method to predict peritoneal metastasis. AIM To determine if the efficacy of computed tomography, magnetic resonance imaging and PET is comparable with staging laparoscopy. METHODS Articles comparing computed tomography, PET, magnetic resonance imaging, and radiomic models based on computed tomography and PET to staging laparoscopies were filtered out from the Cochrane Library, EMBASE, PubMed, Web of Science, and Reference Citations Analysis (https://www.referencecitationanalysis.com/). In the search for studies comparing computed tomography (CT) to staging laparoscopy, five retrospective studies and three prospective studies were found. Similarly, five retrospective studies and two prospective studies were also included for papers comparing CT to PET scans. Only one retrospective study and one prospective study were found to be suitable for papers comparing CT to magnetic resonance imaging scans. RESULTS Staging laparoscopy outperformed computed tomography in all measured aspects, namely sensitivity, specificity, positive predictive value and negative predictive value. Magnetic resonance imaging and PET produced mixed results, with the former shown to be only marginally better than computed tomography. CT performed slightly better than PET in most measured domains, except in specificity and true negative rates. We speculate that this may be due to the limited F-fluorodeoxyglucose uptake in small peritoneal metastases and in linitis plastica. Radiomic modelling, in its current state, shows promise as an alternative for predicting peritoneal metastases. With further research, deep learning and radiomic modelling can be refined and potentially applied as a preoperative diagnostic tool to reduce the need for invasive staging laparoscopy. CONCLUSION Staging laparoscopy was superior in all measured aspects. However, associated risks and costs must be considered. Refinements in radiomic modelling are necessary to establish it as a reliable screening technique.
Collapse
Affiliation(s)
| | - Kon Voi Tay
- Upper GI and Bariatric Division, General Surgery, Tan Tock Seng Hospital, Singapore 308433, Singapore
- Upper GI and Bariatric Division, General Surgery, Woodlands Health, Singapore 768024, Singapore
| |
Collapse
|
18
|
Gao H, Ji K, Bao L, Chen H, Lin C, Feng M, Tao L, Wang M. Establishment and verification of prediction model of occult peritoneal metastasis in advanced gastric cancer. World J Surg Oncol 2023; 21:320. [PMID: 37833730 PMCID: PMC10571475 DOI: 10.1186/s12957-023-03188-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/17/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND To investigate the risk factors associated with the development of occult peritoneal metastasis in advanced gastric cancer, and establish and externally validate a nomogram for predicting the occurrence of occult peritoneal metastasis in patients with advanced gastric cancer. METHODS A total of 111 patients with advanced gastric cancer who underwent laparoscopic exploration or peritoneal lavage cytology examination at the Affiliated Drum Tower Hospital of Nanjing University Medical School from August 2014 to December 2021 were retrospectively analyzed. The patients diagnosed between 2019 and 2021 were assigned to the training set (n = 64), while those diagnosed between 2014 and 2016 constituted the external validation set (n = 47). In the training set, patients were classified into two groups based on preoperative imaging and postoperative pathological data: the occult peritoneal metastasis group (OPMG) and the peritoneal metastasis negative group (PMNG). In the validation set, patients were classified into the occult peritoneal metastasis group (CY1P0, OPMG) and the peritoneal metastasis negative group (CY0P0, PMNG) based on peritoneal lavage cytology results. A nomogram was constructed using univariate and multivariate analyses. The performance of the nomogram was evaluated using Harrell's C-index, the area under the receiver operating characteristic curve (AUC), decision curve analysis (DCA), and calibration plots. RESULTS This study analyzed 22 potential variables of OPM in 111 gastric cancer patients who underwent laparoscopic exploration or peritoneal lavage cytology examination. Logistic regression analysis results showed that Lauren classification, CLDN18.2 score and CA125 were independent risk factors for OPM in patients with gastric cancer. We developed a simple and easy-to-use prediction nomogram of occult peritoneal metastasis in advanced gastric cancer. This nomogram had an excellent diagnostic performance. The AUC of the bootstrap model in the training set was 0.771 and in the validation set was 0.711. This model showed a good fitting and calibration and positive net benefits in decision curve analysis. CONCLUSION We have developed a prediction nomogram of OPM for gastric cancer. This novel nomogram has the potential to enhance diagnostic accuracy for occult peritoneal metastasis in gastric cancer patients.
Collapse
Affiliation(s)
- Hengfei Gao
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Kangkang Ji
- Department of Gastrointestinal, Fuyang People's Hospital, Fuyang, China
| | - Linsen Bao
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Hao Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Chen Lin
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Min Feng
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Medical School of Nanjing University, Nanjing, China.
| | - Liang Tao
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Medical School of Nanjing University, Nanjing, China.
| | - Meng Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
19
|
van Hootegem SJM, de Pasqual CA, Eyck BM, Mostert B, Bradshaw A, Phillips AW, Lagarde SM, Wijnhoven BPL. Clinical impact of routine response assessment after preoperative chemotherapy in patients with gastric cancer. BJS Open 2023; 7:zrad093. [PMID: 37738366 PMCID: PMC10516457 DOI: 10.1093/bjsopen/zrad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 09/24/2023] Open
Affiliation(s)
| | - Carlo A de Pasqual
- Department of Surgery, Erasmus MC University, Rotterdam, The Netherlands
- General and Upper GI Surgery Division, University Hospital of Verona, Verona, Italy
| | - Ben M Eyck
- Department of Surgery, Erasmus MC University, Rotterdam, The Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Centre, Rotterdam, The Netherlands
| | - Alexander Bradshaw
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle-upon-Tyne, UK
| | - Alexander W Phillips
- Northern Oesophagogastric Unit, Royal Victoria Infirmary, Newcastle-Upon-Tyne, UK
- School of Medical Education, Newcastle University, Newcastle-upon-Tyne, UK
| | - Sjoerd M Lagarde
- Department of Surgery, Erasmus MC University, Rotterdam, The Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus MC University, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Liu Z, Tian H, Zhu Z. Application of Circulating Tumor Cells and Interleukin-6 in Preoperative Prediction of Peritoneal Metastasis of Advanced Gastric Cancer. J Inflamm Res 2023; 16:3033-3047. [PMID: 37497064 PMCID: PMC10366674 DOI: 10.2147/jir.s414786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/16/2023] [Indexed: 07/28/2023] Open
Abstract
Background The purpose of this study was to explore the clinical significance of circulating tumor cells (CTCs) and cytokines in peripheral blood in preoperative prediction of peritoneal metastasis (PM) in advanced gastric cancer (AGC). Methods The clinicopathological characteristics of 282 patients with AGC were retrospectively analyzed. The patients were divided into training and validation groups according to the time of receiving treatment. We used univariate analysis and multivariate logistic regression analysis to screen out the independent risk factors of PM in AGC. Then, we incorporated independent risk factors into the nomogram, and evaluated the discriminative ability. Results The levels of CTCs and interleukin-6 (IL-6) of AGC patients with PM were higher than those without PM (P<0.05). Moreover, the levels of CTCs and IL-6 in the occult peritoneal metastasis (OPM) group and the CT-positive PM group were higher than those in the negative PM (P<0.05). Multivariate logistic regression analysis showed that IL-6 > 12.22 pg/mL, CTCs > 4/5mL, CA724 > 6 IU/mL, CA125 > 35 U/mL and tumor size > 5 cm were independent risk factors for PM of AGC. The area under the ROC curve of the nomogram were 0.898 and 0.926 in the training and validation sets, respectively. The clinical decision curve showed that the nomogram had good clinical utility. Conclusion CTCs and IL-6 in peripheral blood are promising biomarkers for predicting the risk of PM in AGC. The nomogram constructed from five risk factors can effectively assess the risk of PM in AGC patients individually.
Collapse
Affiliation(s)
- Zitao Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Huakai Tian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Zhengming Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
21
|
Schena CA, Laterza V, De Sio D, Quero G, Fiorillo C, Gunawardena G, Strippoli A, Tondolo V, de'Angelis N, Alfieri S, Rosa F. The Role of Staging Laparoscopy for Gastric Cancer Patients: Current Evidence and Future Perspectives. Cancers (Basel) 2023; 15:3425. [PMID: 37444535 DOI: 10.3390/cancers15133425] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/18/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
A significant proportion of patients diagnosed with gastric cancer is discovered with peritoneal metastases at laparotomy. Despite the continuous improvement in the performance of radiological imaging, the preoperative recognition of such an advanced disease is still challenging during the diagnostic work-up, since the sensitivity of CT scans to peritoneal carcinomatosis is not always adequate. Staging laparoscopy offers the chance to significantly increase the rate of promptly diagnosed peritoneal metastases, thus reducing the number of unnecessary laparotomies and modifying the initial treatment strategy of gastric cancer. The aim of this review was to provide a comprehensive summary of the current literature regarding the role of staging laparoscopy in the management of gastric cancer. Indications, techniques, accuracy, advantages, and limitations of staging laparoscopy and peritoneal cytology were discussed. Furthermore, a focus on current evidence regarding the application of artificial intelligence and image-guided surgery in staging laparoscopy was included in order to provide a picture of the future perspectives of this technique and its integration with modern tools in the preoperative management of gastric cancer.
Collapse
Affiliation(s)
- Carlo Alberto Schena
- Digestive Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Unit of Colorectal and Digestive Surgery, DIGEST Department, Beaujon University Hospital, AP-HP, University of Paris Cité, Clichy, 92110 Paris, France
| | - Vito Laterza
- Digestive Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Davide De Sio
- Digestive Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giuseppe Quero
- Digestive Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Digestive Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Claudio Fiorillo
- Digestive Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gayani Gunawardena
- Department of Digestive Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonia Strippoli
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Vincenzo Tondolo
- Digestive Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Nicola de'Angelis
- Unit of Colorectal and Digestive Surgery, DIGEST Department, Beaujon University Hospital, AP-HP, University of Paris Cité, Clichy, 92110 Paris, France
| | - Sergio Alfieri
- Digestive Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Digestive Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Fausto Rosa
- Department of Digestive Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Emergency and Trauma Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
22
|
Chen X, Wu Z, He Y, Hao Z, Wang Q, Zhou K, Zhou W, Wang P, Shan F, Li Z, Ji J, Fan Y, Li Z, Yue S. Accurate and Rapid Detection of Peritoneal Metastasis from Gastric Cancer by AI-Assisted Stimulated Raman Molecular Cytology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2300961. [PMID: 37114845 PMCID: PMC10375130 DOI: 10.1002/advs.202300961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/04/2023] [Indexed: 06/19/2023]
Abstract
Peritoneal metastasis (PM) is the mostcommon form of distant metastasis and one of the leading causes of death in gastriccancer (GC). For locally advanced GC, clinical guidelines recommend peritoneal lavage cytology for intraoperative PM detection. Unfortunately, current peritoneal lavage cytology is limited by low sensitivity (<60%). Here the authors established the stimulated Raman molecular cytology (SRMC), a chemical microscopy-based intelligent cytology. The authors firstly imaged 53 951 exfoliated cells in ascites obtained from 80 GC patients (27 PM positive, 53 PM negative). Then, the authors revealed 12 single cell features of morphology and composition that are significantly different between PM positive and negative specimens, including cellular area, lipid protein ratio, etc. Importantly, the authors developed a single cell phenotyping algorithm to further transform the above raw features to feature matrix. Such matrix is crucial to identify the significant marker cell cluster, the divergence of which is finally used to differentiate the PM positive and negative. Compared with histopathology, the gold standard of PM detection, their SRMC method could reach 81.5% sensitivity, 84.9% specificity, and the AUC of 0.85, within 20 minutes for each patient. Together, their SRMC method shows great potential for accurate and rapid detection of PM from GC.
Collapse
Affiliation(s)
- Xun Chen
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 100191, Beijing, China
- School of Engineering Medicine, Beihang University, 100191, Beijing, China
| | - Zhouqiao Wu
- Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 100142, Beijing, China
| | - Yexuan He
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 100191, Beijing, China
| | - Zhe Hao
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 100191, Beijing, China
| | - Qi Wang
- Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 100142, Beijing, China
| | - Keji Zhou
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 100191, Beijing, China
| | - Wanhui Zhou
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 100191, Beijing, China
| | - Pu Wang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 100191, Beijing, China
| | - Fei Shan
- Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 100142, Beijing, China
| | - Zhongwu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, 100142, Beijing, China
| | - Jiafu Ji
- Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 100142, Beijing, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 100191, Beijing, China
- School of Engineering Medicine, Beihang University, 100191, Beijing, China
| | - Ziyu Li
- Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 100142, Beijing, China
| | - Shuhua Yue
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 100191, Beijing, China
| |
Collapse
|
23
|
Li J, Cong L, Sun X, Li X, Chen Y, Cai J, He M, Zhang X, Tang L. CT characteristics for predicting prognosis of gastric cancer with synchronous peritoneal metastasis. Front Oncol 2023; 12:1061806. [PMID: 36713539 PMCID: PMC9874217 DOI: 10.3389/fonc.2022.1061806] [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: 10/05/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction To explore the CT characteristics for the prediction of long term survival in gastric cancer patients with synchronous peritoneal metastasis (PM). Materials and methods Sixty-six patients diagnosed as gastric cancer with synchronous peritoneum metastasis were enrolled in this retrospective study. Ten anatomic peritoneal regions were evaluated to check for the signs of PM on CT. One positive area equaled one score. The CT characteristic-based PM score (CT-PMS) was the sum of the total points assigned to all 10 regions, with a range of 0-10. The triple tract dilatation (TTD) sign caused by peritoneal metastasis, the presence of extensive lymph node metastasis (ELM), and the grade of ascites were recorded. The overall survival (OS) was used as the prognostic indicator. The performance of the CT characteristics was assessed by the Kaplan-Meier analysis and Cox proportional hazards model, while its reproducibility was evaluated by Kappa statistic and weighted Kappa statistic. Results Patients with a CT-PMS of 3-10 had significantly poorer OS (P = .02). Patients with either the presence of TTD sign, or ELM had a trend toward unfavorable OS (both P = .07), and when CT-PMS of 3-10 was detected simultaneously, the survival was further reduced (P = .00 for TTD sign; P = .01 for ELM). The grade of ascites failed to show a significant correlation with OS. The interobserver reproducibility for assessing the CT-PMS, the presence of TTD sign, the presence of ELM, and the grade of ascites had a substantial to almost perfect agreement. Conclusion The prognosis of gastric cancer patients with PM has a correlation with the extent of metastasis dissemination on baseline CT. A CT-PMS of 3-10 is associated with a worse prognosis than that of 0-2. The presence of TTD sign and ELM may help further select patients with extraordinarily poor prognoses.
Collapse
Affiliation(s)
- Jiazheng Li
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lin Cong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xuefeng Sun
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,Department of Radiology, The Affiliated Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Xiaoting Li
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jieyuan Cai
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Meng He
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,*Correspondence: Xiaotian Zhang, ; Lei Tang,
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China,*Correspondence: Xiaotian Zhang, ; Lei Tang,
| |
Collapse
|
24
|
SAKAMOTO E, RAMOS MFKP, PEREIRA MA, DIAS AR, RIBEIRO U, ZILBERSTEIN B, NAHAS SC. STAGING LAPAROSCOPY IS STILL A VALUABLE TOOL FOR OPTIMAL GASTRIC CANCER MANAGEMENT. ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2023; 35:e1700. [PMID: 36629683 PMCID: PMC9830676 DOI: 10.1590/0102-672020220002e1700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/30/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Complete surgical resection is the main determining factor in the survival of advanced gastric cancer patients, but is not indicated in metastatic disease. The peritoneum is a common site of metastasis and preoperative imaging techniques still fail to detect it. AIM The aim of this study was to evaluate the role of staging laparoscopy in the staging of advanced gastric cancer patients in a Western tertiary cancer center. METHODS A total of 130 patients with gastric adenocarcinoma who underwent staging laparoscopy from 2009 to 2020 were evaluated from a prospective database. Clinicopathological characteristics were analyzed to identify factors associated with the presence of peritoneal metastasis and were also evaluated the accuracy and strength of agreement between computed tomography and staging laparoscopy in detecting peritoneal metastasis and the change in treatment strategy after the procedure. RESULTS The peritoneal metastasis was identified in 66 (50.76%) patients. The sensitivity, specificity, and accuracy of computed tomography in detecting peritoneal metastasis were 51.5, 87.5, and 69.2%, respectively. According to the Kappa coefficient, the concordance between staging laparoscopy and computed tomography was 38.8%. In multivariate analysis, ascites (p=0.001) and suspected peritoneal metastasis on computed tomography (p=0.007) were statistically correlated with peritoneal metastasis. In 40 (30.8%) patients, staging and treatment plans changed after staging laparoscopy (32 patients avoided unnecessary laparotomy, and 8 patients, who were previously considered stage IVb by computed tomography, were referred to surgical treatment). CONCLUSION The staging laparoscopy demonstrated an important role in the diagnosis of peritoneal metastasis, even with current advances in imaging techniques.
Collapse
Affiliation(s)
- Erica SAKAMOTO
- Universidade de São Paulo, University Hospital, Department of Gastroenterology of Faculty of Medicine – São Paulo (SP), Brazil
| | | | - Marina Alessandra PEREIRA
- Universidade de São Paulo, University Hospital, Department of Gastroenterology of Faculty of Medicine – São Paulo (SP), Brazil
| | - André Roncon DIAS
- Universidade de São Paulo, University Hospital, Department of Gastroenterology of Faculty of Medicine – São Paulo (SP), Brazil
| | - Ulysses RIBEIRO
- Universidade de São Paulo, University Hospital, Department of Gastroenterology of Faculty of Medicine – São Paulo (SP), Brazil
| | - Bruno ZILBERSTEIN
- Universidade de São Paulo, University Hospital, Department of Gastroenterology of Faculty of Medicine – São Paulo (SP), Brazil
| | - Sergio Carlos NAHAS
- Universidade de São Paulo, University Hospital, Department of Gastroenterology of Faculty of Medicine – São Paulo (SP), Brazil
| |
Collapse
|
25
|
Kim TH, Kim IH, Kang SJ, Choi M, Kim BH, Eom BW, Kim BJ, Min BH, Choi CI, Shin CM, Tae CH, Gong CS, Kim DJ, Cho AEH, Gong EJ, Song GJ, Im HS, Ahn HS, Lim H, Kim HD, Kim JJ, Yu JI, Lee JW, Park JY, Kim JH, Song KD, Jung M, Jung MR, Son SY, Park SH, Kim SJ, Lee SH, Kim TY, Bae WK, Koom WS, Jee Y, Kim YM, Kwak Y, Park YS, Han HS, Nam SY, Kong SH. Korean Practice Guidelines for Gastric Cancer 2022: An Evidence-based, Multidisciplinary Approach. J Gastric Cancer 2023; 23:3-106. [PMID: 36750993 PMCID: PMC9911619 DOI: 10.5230/jgc.2023.23.e11] [Citation(s) in RCA: 155] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/22/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Gastric cancer is one of the most common cancers in Korea and the world. Since 2004, this is the 4th gastric cancer guideline published in Korea which is the revised version of previous evidence-based approach in 2018. Current guideline is a collaborative work of the interdisciplinary working group including experts in the field of gastric surgery, gastroenterology, endoscopy, medical oncology, abdominal radiology, pathology, nuclear medicine, radiation oncology and guideline development methodology. Total of 33 key questions were updated or proposed after a collaborative review by the working group and 40 statements were developed according to the systematic review using the MEDLINE, Embase, Cochrane Library and KoreaMed database. The level of evidence and the grading of recommendations were categorized according to the Grading of Recommendations, Assessment, Development and Evaluation proposition. Evidence level, benefit, harm, and clinical applicability was considered as the significant factors for recommendation. The working group reviewed recommendations and discussed for consensus. In the earlier part, general consideration discusses screening, diagnosis and staging of endoscopy, pathology, radiology, and nuclear medicine. Flowchart is depicted with statements which is supported by meta-analysis and references. Since clinical trial and systematic review was not suitable for postoperative oncologic and nutritional follow-up, working group agreed to conduct a nationwide survey investigating the clinical practice of all tertiary or general hospitals in Korea. The purpose of this survey was to provide baseline information on follow up. Herein we present a multidisciplinary-evidence based gastric cancer guideline.
Collapse
Affiliation(s)
- Tae-Han Kim
- Department of Surgery, Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Joo Kang
- Department of Internal Medicine, Seoul National University Hospital Healthcare System Gangnam Center Seoul, Seoul, Korea
| | - Miyoung Choi
- National Evidence-based Healthcare Collaborating Agency (NECA), Seoul, Korea
| | - Baek-Hui Kim
- Department of Pathology, Korea University Guro Hospital, Seoul, Korea
| | - Bang Wool Eom
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Bum Jun Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang, Korea
| | - Byung-Hoon Min
- Department of Medicine, Samsung Medical Center, Seoul, Korea
| | - Chang In Choi
- Department of Surgery, Pusan National University Hospital, Pusan, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seungnam, Korea
| | - Chung Hyun Tae
- Department of Internal Medicine, Ewha Woman's University College of Medicine, Seoul, Korea
| | - Chung Sik Gong
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center and University of Ulsan College of Medicine, Seoul, Korea
| | - Dong Jin Kim
- Department of Surgery, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | - Eun Jeong Gong
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Geum Jong Song
- Department of Surgery, Soonchunhyang University, Cheonan, Korea
| | - Hyeon-Su Im
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Hye Seong Ahn
- Department of Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Hyun Lim
- Department of Gastroenterology, Hallym University Sacred Heart Hospital, University of Hallym College of Medicine, Anyang, Korea
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae-Joon Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jeong Il Yu
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul, Korea
| | - Jeong Won Lee
- Department of Nuclear Medicine, Catholic Kwandong University, College of Medicine, Incheon, Korea
| | - Ji Yeon Park
- Department of Surgery, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jwa Hoon Kim
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Kyoung Doo Song
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| | - Minkyu Jung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University Health System, Seoul, Korea
| | - Mi Ran Jung
- Department of Surgery, Chonnam National University Medical School, Gwangju, Korea
| | - Sang-Yong Son
- Department of Surgery, Ajou University School of Medicine, Suwon, Korea
| | - Shin-Hoo Park
- Department of Surgery, Korea University Anam Hospital, Seoul, Korea
| | - Soo Jin Kim
- Department of Radiology, National Cancer Center, Goyang, Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Woo Kyun Bae
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yeseob Jee
- Department of Surgery, Dankook University Hospital, Cheonan, Korea
| | - Yoo Min Kim
- Department of Surgery, Severance Hospital, Seoul, Korea
| | - Yoonjin Kwak
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Young Suk Park
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hye Sook Han
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea.
| | - Su Youn Nam
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea.
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University Hospital and Seoul National University College of Medicine Cancer Research Institute, Seoul, Korea.
| |
Collapse
|
26
|
Kubo N, Cho H, Lee D, Yang H, Kim Y, Khalayleh H, Yoon HM, Ryu KW, Hanna GB, Coit DG, Hakamada K, Kim YW. Risk prediction model of peritoneal seeding in advanced gastric cancer: A decision tool for diagnostic laparoscopy. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2022; 49:853-861. [PMID: 36586786 DOI: 10.1016/j.ejso.2022.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/07/2022] [Accepted: 12/23/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Selective diagnostic laparoscopy in gastric cancer patients at high risk of peritoneal metastasis is essential for optimal treatment planning. In this study available clinicopathologic factors predictive of peritoneal seeding in advanced gastric cancer (AGC) were identified, and this information was translated into a clinically useful tool. METHODS Totally 2833 patients underwent surgery for AGC between 2003 and 2013. The study identified clinicopathologic factors associated with the risk of peritoneal seeding for constructing nomograms using a multivariate logistic regression model with backward elimination. A nomogram was constructed to generate a numerical value indicating risk. Accuracy was validated using bootstrapping and cross-validation. RESULTS The proportion of seeding positive was 12.7% in females and 9.6% in males. Of 2833 patients who underwent surgery for AGC, 300 (10.6%) were intraoperatively identified with peritoneal seeding. Multivariate analysis revealed the following factors associated with peritoneal seeding: high American Society of Anesthesiologists score, fibrinogen, Borrmann type 3 or 4 tumors, the involvement of the middle, anterior, and greater curvature, cT3 or cT4cN1 or cN2 or cN3, cM1, and the presence of ascites or peritoneal thickening or plaque or a nodule on the peritoneal wall on computed tomography. The bootstrap analysis revealed a robust concordance between mean and final parameter estimates. The area under the ROC curve for the final model was 0.856 (95% CI, 0.835-0.877), which implies good performance. CONCLUSIONS This nomogram provides effective risk estimates of peritoneal seeding from gastric cancer and can facilitate individualized decision-making regarding the selective use of diagnostic laparoscopy.
Collapse
Affiliation(s)
- Norihito Kubo
- Center for Gastric Cancer, National Cancer Center, Korea; Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Japan
| | - Hyunsoon Cho
- Department of Cancer Control and Population Science, Graduate School of Cancer Science and Policy, National Cancer Center, Korea
| | - Dahhay Lee
- Department of Cancer Control and Population Science, Graduate School of Cancer Science and Policy, National Cancer Center, Korea
| | - Hannah Yang
- Center for Gastric Cancer, National Cancer Center, Korea; Division of Biology and Biological Engineering, California Institute of Technology Pasadena, California, 91125, USA
| | - Youngsook Kim
- Center for Gastric Cancer, National Cancer Center, Korea
| | - Harbi Khalayleh
- Center for Gastric Cancer, National Cancer Center, Korea; Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Surgery, Kaplan Medical Center, Israel
| | - Hong Man Yoon
- Center for Gastric Cancer, National Cancer Center, Korea
| | - Keun Won Ryu
- Center for Gastric Cancer, National Cancer Center, Korea
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College of London, United Kingdom
| | - Daniel G Coit
- Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, USA
| | - Kenichi Hakamada
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Japan
| | - Young-Woo Kim
- Center for Gastric Cancer, National Cancer Center, Korea; Department of Cancer Control and Population Science, Graduate School of Cancer Science and Policy, National Cancer Center, Korea.
| |
Collapse
|
27
|
Cao R, Tang L, Fang M, Zhong L, Wang S, Gong L, Li J, Dong D, Tian J. Artificial intelligence in gastric cancer: applications and challenges. Gastroenterol Rep (Oxf) 2022; 10:goac064. [PMID: 36457374 PMCID: PMC9707405 DOI: 10.1093/gastro/goac064] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 08/10/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors with high mortality. Accurate diagnosis and treatment decisions for GC rely heavily on human experts' careful judgments on medical images. However, the improvement of the accuracy is hindered by imaging conditions, limited experience, objective criteria, and inter-observer discrepancies. Recently, the developments of machine learning, especially deep-learning algorithms, have been facilitating computers to extract more information from data automatically. Researchers are exploring the far-reaching applications of artificial intelligence (AI) in various clinical practices, including GC. Herein, we aim to provide a broad framework to summarize current research on AI in GC. In the screening of GC, AI can identify precancerous diseases and assist in early cancer detection with endoscopic examination and pathological confirmation. In the diagnosis of GC, AI can support tumor-node-metastasis (TNM) staging and subtype classification. For treatment decisions, AI can help with surgical margin determination and prognosis prediction. Meanwhile, current approaches are challenged by data scarcity and poor interpretability. To tackle these problems, more regulated data, unified processing procedures, and advanced algorithms are urgently needed to build more accurate and robust AI models for GC.
Collapse
Affiliation(s)
| | | | - Mengjie Fang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China
| | - Lianzhen Zhong
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
| | - Siwen Wang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
| | - Lixin Gong
- College of Medicine and Biological Information Engineering School, Northeastern University, Shenyang, Liaoning, P. R. China
| | - Jiazheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Di Dong
- Corresponding authors. Di Dong, CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing 100190, P. R. China. Tel: +86-13811833760; ; Jie Tian, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China. Tel: +86-10-82618465;
| | - Jie Tian
- Corresponding authors. Di Dong, CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing 100190, P. R. China. Tel: +86-13811833760; ; Jie Tian, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China. Tel: +86-10-82618465;
| |
Collapse
|
28
|
Guan G, Li Z, Wang Q, Ying X, Shan F, Li Z. Risk factors associated with peritoneal carcinomatosis of gastric cancer in staging laparoscopy: A systematic review and meta-analysis. Front Oncol 2022; 12:955181. [PMID: 36387230 PMCID: PMC9650136 DOI: 10.3389/fonc.2022.955181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022] Open
Abstract
Background The optimal indications of staging laparoscopy in gastric cancer to detect peritoneal carcinomatosis are still controversial. We performed this systematic review and meta-analysis to quantify the relevance of the preoperative factors with peritoneal carcinomatosis to explore the indications of staging laparoscopy. Materials and methods Systematic searches were conducted using Medline, Embase, and the Cochrane Library in December 2021. On the basis of calculating the odds ratio (OR) of each factor, we quantified the association between the risk factors and peritoneal carcinomatosis such as clinical T/N stage, Borrmann type, and tumor markers, using meta-analysis with a random-effects model. Results A total of 21 case-control studies and one cohort study were obtained. T stage, N stage, and differentiation degree were most widely studied, with OR values of 2.96 (95% CI: 1.87–4.69), 1.22 (95% CI: 0.86–1.73), and 1.91 (95% CI: 1.42–2.56), respectively. Among all the factors, elevated CA125 (OR = 19.45, 95% CI: 4.71–80.30), Borrmann type IV (OR = 7.68, 95% CI: 3.62–16.27), and large tumor diameter (OR = 5.12, 95% CI: 2.55–10.31) had the highest OR. In particular, CA125 had the best predictability for peritoneal carcinomatosis but was only mentioned by three articles. Conclusions There was a cognitive gap between the awareness and importance of risk factors for peritoneal carcinomatosis. In addition to T4 stage, patients with factors with high OR, such as Borrmann type IV, large tumor diameter, and elevated CA125, should undergo staging laparoscopy.
Collapse
|
29
|
Zhang Q, Yuan Y, Li S, Li Z, Jing G, Lu J, Shao C, Hao Q, Lu Y, Shen F. A CT-Based Radiomics Model for Evaluating Peritoneal Cancer Index in Peritoneal Metastasis Cases: A Preliminary Study. Acad Radiol 2022:S1076-6332(22)00492-5. [DOI: 10.1016/j.acra.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/20/2022] [Accepted: 09/02/2022] [Indexed: 01/17/2023]
|
30
|
Li Z, Guan G, Liu Z, Li J, Ying X, Shan F, Li Z. Predicting peritoneal carcinomatosis of gastric cancer: A simple model to exempt low-risk patients from unnecessary staging laparoscopy. Front Surg 2022; 9:916001. [PMID: 35937608 PMCID: PMC9349356 DOI: 10.3389/fsurg.2022.916001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/28/2022] [Indexed: 11/21/2022] Open
Abstract
Background Peritoneal carcinomatosis (PC) of gastric cancer indicates a poor outcome and is mainly diagnosed by staging laparoscopy (SL). This study was designed to develop a risk stratification model based on the number of risk factors to exempt low-risk patients from unnecessary SL. Methods This was a retrospective cohort study based on a single institution between January 2015 and December 2019. SL is indicated for patients of advanced locoregional stage, and clinicopathologic characteristics of 535 consecutive patients were included. PC-associated variables were identified by logistic regression analysis. A risk stratification model based on the number of risk factors was constructed, and we defined its predictive value with a receiver operating characteristic (ROC) curve and negative predictive value. Results In total, 15.9% of included patients were found to have PC during SL. Borrmann type IV, elevated CA125, and tumour diameter ≥5 cm were independent risk factors of PC. These three factors combined with cT4 were selected as predictive factors, and the number of predictive variables was significantly related to the possibility of PC (2.0%, 12.8%, 20.0%, 54.2%, and 100%, respectively). When the cutoff value is more than one predictive factor, the negative predictive value is 98.0%, with an area under the curve of 0.780. This model could exempt 29.8% of unnecessary SL compared to the indication of the current NCCN guideline. Conclusions We constructed a simple model to predict the probability of PC using the number of predictive factors. It is recommended that patients without any of these factors should be exempt from SL.
Collapse
|
31
|
Guo Z, Guo H, Tian Y, Zhang Z, Zhao Q. Nomograms for Predicting Disease-Free Survival in Patients With Siewert Type II/III Adenocarcinoma of the Esophagogastric Junction Receiving Neoadjuvant Therapy and Radical Surgery. Front Oncol 2022; 12:908229. [PMID: 35756688 PMCID: PMC9213656 DOI: 10.3389/fonc.2022.908229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/19/2022] [Indexed: 11/15/2022] Open
Abstract
Objective This study aimed to develop prognostic prediction models for patients with Siewert type II/III adenocarcinoma of the esophagogastric junction (AEG) who received neoadjuvant therapy (neoadjuvant chemoradiotherapy or neoadjuvant chemotherapy) and radical surgery. A baseline nomogram and a post-operative nomogram were constructed before neoadjuvant therapy and after surgery. The predictive performance of the constructed nomograms was internally validated and compared to the TNM staging system. Materials and Methods A total of 245 patients diagnosed with Siewert type II/III AEG and treated with neoadjuvant therapy followed by radical surgery at The Fourth Hospital of Hebei Medical University between January 2011 and December 2017 were enrolled. The variables before neoadjuvant therapy were defined as baseline factors, while the variables of baseline factors along with the variables of treatment and postoperative pathology were defined as post-operative factors. To construct the corresponding nomograms, independent predictors of baseline and post-operative factors were identified. The C-index and a time-dependent receiver operating characteristic curve were used to evaluate the model’s discrimination ability. The calibration ability of the model was determined by comparing the probability of predicted free-recurrence to the actual free-recurrence. Decision curve analysis (DCA) was used to determine the clinical usefulness of the nomogram. Results Among the baseline factors, age, cT stage, cN stage, Borrmann type, and staging laparoscopy were independent prognostic predictors. In contrast, among the post-operative factors, age, cN stage, staging laparoscopy, ypT stage, clinical response, number of positive lymph nodes, number of negative lymph nodes, laurén classification, and lymphatic, or perineural invasion (VELPI) were independent prognostic predictors. The two nomograms were constructed using the independent predictors of prognosis. The C-indexes for the baseline and post-operative nomograms were 0.690 (95% CI, 0.644-0.736) and 0.817 (95% CI, 0.782-0.853), respectively. The AUCs of the baseline nomogram at 3 and 5 years were both greater than cTNM (73.1 vs 58.8, 76.1 vs 55.7). Similarly, the AUCs of the post-operative nomogram were both greater than ypTNM (85.2 vs 69.1, 88.2 vs 71.3) at 3 and 5 years. The calibration curves indicated that both models had a high degree of calibration ability. By comparing the DCA at 3 and 5 years, we determined that the two nomograms constructed had better clinical utility than the TNM staging system. Conclusions The constructed nomograms have a more accurate predictive ability than the eighth edition TNM staging system, which can be useful for treatment selection and follow-up monitoring of patients.
Collapse
Affiliation(s)
- Zhenjiang Guo
- Third Surgery Department, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Gastrointestinal Surgery, Hengshui People's Hospital, Hengshui, China
| | - Honghai Guo
- Third Surgery Department, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuan Tian
- Third Surgery Department, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ze Zhang
- Third Surgery Department, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qun Zhao
- Third Surgery Department, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
32
|
Bao D, Yang Z, Chen S, Li K, Hu Y. Construction of a Nomogram Model for Predicting Peritoneal Dissemination in Gastric Cancer Based on Clinicopathologic Features and Preoperative Serum Tumor Markers. Front Oncol 2022; 12:844786. [PMID: 35719995 PMCID: PMC9198602 DOI: 10.3389/fonc.2022.844786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 05/04/2022] [Indexed: 12/24/2022] Open
Abstract
Background Peritoneal dissemination (PD) is the most common mode of metastasis for advanced gastric cancer (GC) with poor prognosis. It is of great significance to accurately predict preoperative PD and develop optimal treatment strategies for GC patients. Our study assessed the diagnostic potential of serum tumor markers and clinicopathologic features, to improve the accuracy of predicting the presence of PD in GC patients. Methods In our study, 1264 patients with GC at Fudan University Shanghai Cancer Center and Wenzhou people’s hospital from 2018 to 2020 were retrospectively analyzed, including 316 cases of PD and 948 cases without PD. All patients underwent enhanced CT scan or magnetic resonance imaging (MRI) before surgery and treatment. Clinicopathological features, including tumor diameter and tumor stage (depth of tumor invasion, nearby lymph node metastasis and distant metastasis), were obtained by imaging examination. The independent risk factors for PD were screened through univariate and multivariate logistic regression analyses, and the results were expressed with 95% confidence intervals (CIs). A model of PD diagnosis and prediction was established by using Cox proportional hazards regression model of training set. Furthermore, the accuracy of the prediction model was verified by ROC curve and calibration plots. Results Univariate analysis showed that PD in GC was significantly related to tumor diameter (odds ratio (OR)=12.06, p<0.0006), depth of invasion (OR=14.55, p<0.0001), lymph node metastases (OR=5.89, p<0.0001), carcinoembryonic antigen (CEA) (OR=2.50, p<0.0001), CA125 (OR=11.46, p<0.0001), CA72-4 (OR=4.09, p<0.0001), CA19-9 (OR=2.74, p<0.0001), CA50 (OR=5.20, p<0.0001) and CA242 (OR=3.83, p<0.0001). Multivariate analysis revealed that clinical invasion depth and serum marker of CA125 and CA72-4 were independent risk factors for PD. The prediction model was established based on the risk factors using the R program. The area under the curve (AUC) of the receiver operating characteristics (ROC) was 0.931 (95% CI: 0.900–0.960), with the accuracy, sensitivity and specificity values of 90.5%, 86.2% and 82.2%, respectively. Conclusion The nomogram model constructed using CA125, CA72-4 and depth of invasion increases the accuracy and sensitivity in predicting the incidence of PD in GC patients and can be used as an important tool for preoperative diagnosis.
Collapse
Affiliation(s)
- Dandan Bao
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
| | - Zhangwei Yang
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
| | - Senrui Chen
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
| | - Keqin Li
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
| | - Yiren Hu
- Department of General Surgery, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China.,Department of General Surgery, Medical College of Soochow University, Soochow, China
| |
Collapse
|
33
|
AKÇAKAYA A. Staging Laparoscopy in Stomach Cancer. BEZMIALEM SCIENCE 2022. [DOI: 10.14235/bas.galenos.2021.04696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
34
|
Masci GM, Ciccarelli F, Mattei FI, Grasso D, Accarpio F, Catalano C, Laghi A, Sammartino P, Iafrate F. Role of CT texture analysis for predicting peritoneal metastases in patients with gastric cancer. Radiol Med 2022; 127:251-258. [PMID: 35066804 DOI: 10.1007/s11547-021-01443-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/21/2021] [Indexed: 12/29/2022]
Abstract
PURPOSE Aim of the study was to perform CT texture analysis in patients with gastric cancer (GC) to investigate potential role of radiomics for predicting the occurrence of peritoneal metastases (PM). MATERIALS AND METHODS In this single-centre retrospective study, patients with gastric adenocarcinoma and surgically confirmed presence or absence of PM were, respectively, enrolled in group PM and group non-PM. Patients with T1-staging, previous treatment or presence of imaging artifacts were excluded from the study. Pre-operative CT examinations were evaluated. Acquisition protocol consisted of gastric distension with water, pre-contrast and arterial phases on upper abdomen and portal phase on thorax and whole abdomen. Texture analysis was performed on portal phase images: the region of interest was manually drawn along the margins of the primitive lesion on each slice and the volume of interest of the whole tumour was obtained. A total of 38 texture parameters were extracted and analysed. ROC curves were performed on significant texture features (p < 0.05). Multiple logistic regression was conducted on features with the best AUC to identify differentiating variables for both groups. RESULTS A total of 90 patients were evaluated (group PM, n = 45; group non-PM, n = 45). T2/T3 tumours were prevalent in group non-PM, T4 was significantly associated with group PM. Significant differences between the two groups were observed for 22/38 texture parameters. Volume and GLRLM_LRHGE showed the greatest AUC in ROC curve analysis (0.737 and 0.734, respectively) and were found to be independent differentiating variables of group PM in the multiple regression analysis (OR 8.44, [95% CI, 1.52-46.8] and OR 18.99 [95% CI, 84-195.31], respectively). CONCLUSIONS Our preliminary results suggest the potential value of CT texture analysis for predicting the risk of PM from GC, which may be helpful to stratify patients and address them to the most appropriate treatment.
Collapse
Affiliation(s)
- Giorgio Maria Masci
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Fabio Ciccarelli
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Fabrizio Ivo Mattei
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Damiano Grasso
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Fabio Accarpio
- Department of Surgery "Pietro Valdoni", Policlinico Umberto I, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Carlo Catalano
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Andrea Laghi
- Department of Medical-Surgical and Translational Medicine-Radiology Unit, "Sapienza" University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Paolo Sammartino
- Department of Surgery "Pietro Valdoni", Policlinico Umberto I, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Franco Iafrate
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
35
|
Liu D, Zhang W, Hu F, Yu P, Zhang X, Yin H, Yang L, Fang X, Song B, Wu B, Hu J, Huang Z. A Bounding Box-Based Radiomics Model for Detecting Occult Peritoneal Metastasis in Advanced Gastric Cancer: A Multicenter Study. Front Oncol 2021; 11:777760. [PMID: 34926287 PMCID: PMC8678129 DOI: 10.3389/fonc.2021.777760] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/09/2021] [Indexed: 02/05/2023] Open
Abstract
Purpose To develop a bounding box (BBOX)-based radiomics model for the preoperative diagnosis of occult peritoneal metastasis (OPM) in advanced gastric cancer (AGC) patients. Materials and Methods 599 AGC patients from 3 centers were retrospectively enrolled and were divided into training, validation, and testing cohorts. The minimum circumscribed rectangle of the ROIs for the largest tumor area (R_BBOX), the nonoverlapping area between the tumor and R_BBOX (peritumoral area; PERI) and the smallest rectangle that could completely contain the tumor determined by a radiologist (M_BBOX) were used as inputs to extract radiomic features. Multivariate logistic regression was used to construct a radiomics model to estimate the preoperative probability of OPM in AGC patients. Results The M_BBOX model was not significantly different from R_BBOX in the validation cohort [AUC: M_BBOX model 0.871 (95% CI, 0.814–0.940) vs. R_BBOX model 0.873 (95% CI, 0.820–0.940); p = 0.937]. M_BBOX was selected as the final radiomics model because of its extremely low annotation cost and superior OPM discrimination performance (sensitivity of 85.7% and specificity of 82.8%) over the clinical model, and this radiomics model showed comparable diagnostic efficacy in the testing cohort. Conclusions The BBOX-based radiomics could serve as a simpler reliable and powerful tool for the preoperative diagnosis of OPM in AGC patients. And M_BBOX-based radiomics is simpler and less time consuming.
Collapse
Affiliation(s)
- Dan Liu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Weihan Zhang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fubi Hu
- Department of Radiology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Pengxin Yu
- Institute of Advanced Research, Infervision, Beijing, China
| | - Xiao Zhang
- Department of Radiology, People's Hospital of Leshan, Leshan, China
| | - Hongkun Yin
- Institute of Advanced Research, Infervision, Beijing, China
| | - Lanqing Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Fang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Bing Wu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiankun Hu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zixing Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Kwak MH, Yang SM, Yun SK, Kim S, Choi MG, Park JM. Identification and validation of LGR5-binding peptide for molecular imaging of gastric cancer. Biochem Biophys Res Commun 2021; 580:93-99. [PMID: 34628260 DOI: 10.1016/j.bbrc.2021.09.073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/28/2021] [Indexed: 12/27/2022]
Abstract
Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) is a stem cell marker in gastric cancer. In this study, we aimed to produce the LGR5-targeting peptide probe for the use of molecular imaging for gastric cancer. We used phage display libraries to produce a LGR5-specific peptide probe. This peptide was validated for targeting gastric cancer with in vitro and in vivo studies. This peptide was tagged with fluorescein isothiocyanate (FITC) and cyanine 5.5 (Cy5.5). We used two normal and three gastric cancer cell lines. Immunocytochemistry (ICC) and fluorescence-activated cell sorting (FACS) analysis were used to validate the target specificity of the peptide. After three rounds of bio-panning, we found a novel 7-mer peptides, IPQILSI (IPQ∗). FITC-conjugated IPQ∗ showed 2 to 10 times higher fluorescence in gastric cancer cells vs. control cells in ICC. This discrimination was consistently observed using Cy5.5-conjugated IPQ∗ in ICC. FACS analysis showed right shift of peak point in gastric cancers compared to the control cells. In the peritoneal metastasis animal model, we could find Cy5.5-conjugated IPQ∗ accumulated specifically to gastric tumors. In conclusion, IPQ∗ peptide showed a specific probe for gastric cancer diagnosis. This probe can be applied to theragnosis for gastric cancer diagnosis including peritoneal metastasis.
Collapse
Affiliation(s)
- Moon Hwa Kwak
- Catholic Photomedicine Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea; Department of Medical Life Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung Mok Yang
- Catholic Photomedicine Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seul Ki Yun
- Catholic Photomedicine Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea; Department of Medical Life Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sol Kim
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Myung-Gyu Choi
- Catholic Photomedicine Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea; Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jae Myung Park
- Catholic Photomedicine Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea; Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
37
|
Shimura T, Toden S, Kandimalla R, Toiyama Y, Okugawa Y, Kanda M, Baba H, Kodera Y, Kusunoki M, Goel A. Genomewide Expression Profiling Identifies a Novel miRNA-based Signature for the Detection of Peritoneal Metastasis in Patients With Gastric Cancer. Ann Surg 2021; 274:e425-e434. [PMID: 31663973 PMCID: PMC7577555 DOI: 10.1097/sla.0000000000003647] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE This study aimed to conduct a genomewide transcriptomic profiling to develop a microRNA (miRNA)-based signature for the identification of peritoneal metastasis (PM) in patients with gastric cancer (GC). SUMMARY BACKGROUND DATA Even though PM in patients with GC has long been recognized to associate with poor prognosis, currently there is lack of availability of molecular biomarkers for its robust diagnosis. METHODS We performed a systematic biomarker discovery by analyzing miRNA expression profiles in primary tumors from GC patients with and without PM, and subsequently validated the expression of candidate miRNA biomarkers in 3 independent clinical cohorts of 354 patients with advanced GC. RESULTS Five miRNAs (miR-30a-5p, -134-5p, -337-3p, -659-3p, and -3917) were identified during the initial discovery phase; three of which (miR-30a-5p, -659-3p, and -3917) were significantly overexpressed in the primary tumors from PM-positive patients in the testing cohort (P = 0.002, 0.04, and 0.007, respectively), and distinguished patients with versus without peritoneal metastasis with the value of area under the curve (AUC) of 0.82. Furthermore, high expression of these miRNAs also associated with poor prognosis (hazard ratio = 2.18, P = 0.04). The efficacy of the combination miRNA signature was subsequently validated in an independent validation cohort (AUC = 0.74). Finally, our miRNA signature when combined together with the macroscopic Borrmann's type score offered a much superior diagnostic in all 3 cohorts (AUC = 0.87, 0.76, and 0.79, respectively). CONCLUSIONS We have established an miRNA-based signature that have a potential to identify peritoneal metastasis in GC patients.
Collapse
Affiliation(s)
- Tadanobu Shimura
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Department of Molecular Diagnostics, Therapeutics and Translational Medicine, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Shusuke Toden
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Department of Molecular Diagnostics, Therapeutics and Translational Medicine, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Raju Kandimalla
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Department of Molecular Diagnostics, Therapeutics and Translational Medicine, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Yoshinaga Okugawa
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masato Kusunoki
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Ajay Goel
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Department of Molecular Diagnostics, Therapeutics and Translational Medicine, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
38
|
Shimada H, Fukagawa T, Haga Y, Okazumi S, Oba K. Clinical TNM staging for esophageal, gastric, and colorectal cancers in the era of neoadjuvant therapy: A systematic review of the literature. Ann Gastroenterol Surg 2021; 5:404-418. [PMID: 34337289 PMCID: PMC8316742 DOI: 10.1002/ags3.12444] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/06/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
AIM Clinical staging is vital for selecting appropriate candidates and designing neoadjuvant treatment strategies for advanced tumors. The aim of this review was to evaluate diagnostic abilities of clinical TNM staging for gastrointestinal, gastrointestinal cancers. METHODS We conducted a systematic review of recent publications to evaluate the accuracy of diagnostic modalities on gastrointestinal cancers. A systematic literature search was performed in PubMed/MEDLINE using the keywords "TNM staging," "T4 staging," "distant metastases," "esophageal cancer," "gastric cancer," and "colorectal cancer," and the search terms used in Cochrane Reviews between January 2005 to July 2020. Articles focusing on preoperative diagnosis of: (a) depth of invasion; (b) lymph node metastases; and (c) distant metastases were selected. RESULTS After a full-text search, a final set of 55 studies (17 esophageal cancer studies, 26 gastric cancer studies, and 12 colorectal cancer studies) were used to evaluate the accuracy of clinical TNM staging. Positron emission tomography-computed tomography (PET-CT) and/or magnetic resonance imaging (MRI) were the best modalities to assess distant metastases. Fat and fiber mode of CT may be useful for T4 staging of esophageal cancer, CT was a partially reliable modality for lymph node staging in gastric cancer, and CT combined with MRI was the most reliable modality for liver metastases from colorectal cancer. CONCLUSION The most reliable diagnostic modality differed among gastrointestinal cancers depending on the type of cancer. Therefore, we propose diagnostic algorithms for clinical staging for each type of cancer.
Collapse
Affiliation(s)
- Hideaki Shimada
- Department of Gastroenterological SurgeryToho University Graduate School of MedicineTokyoJapan
| | - Takeo Fukagawa
- Department of SurgeryTeikyo University School of MedicineTokyoJapan
| | - Yoshio Haga
- Department of SurgeryJapan Community Healthcare Organization Amakusa Central General HospitalAmakusaJapan
| | - Shin‐ichi Okazumi
- Department of Gastroenterological SurgeryToho University Graduate School of MedicineTokyoJapan
- Department of SurgeryToho University Sakura Medical CenterSakuraJapan
| | - Koji Oba
- Department of BiostatisticsSchool of Public HealthGraduate School of MedicineThe University of TokyoTokyoJapan
- Interfaculty Initiative in Information StudiesGraduate School of Interdisciplinary Information StudiesThe University of TokyoTokyoJapan
| |
Collapse
|
39
|
Chen Y, Xi W, Yao W, Wang L, Xu Z, Wels M, Yuan F, Yan C, Zhang H. Dual-Energy Computed Tomography-Based Radiomics to Predict Peritoneal Metastasis in Gastric Cancer. Front Oncol 2021; 11:659981. [PMID: 34055627 PMCID: PMC8160383 DOI: 10.3389/fonc.2021.659981] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/26/2021] [Indexed: 01/06/2023] Open
Abstract
Objective To develop and validate a dual-energy computed tomography (DECT) derived radiomics model to predict peritoneal metastasis (PM) in patients with gastric cancer (GC). Methods This retrospective study recruited 239 GC (non-PM = 174, PM = 65) patients with histopathological confirmation for peritoneal status from January 2015 to December 2019. All patients were randomly divided into a training cohort (n = 160) and a testing cohort (n = 79). Standardized iodine-uptake (IU) images and 120-kV-equivalent mixed images (simulating conventional CT images) from portal-venous and delayed phases were used for analysis. Two regions of interest (ROIs) including the peritoneal area and the primary tumor were independently delineated. Subsequently, 1691 and 1226 radiomics features were extracted from the peritoneal area and the primary tumor from IU and mixed images on each phase. Boruta and Spearman correlation analysis were used for feature selection. Three radiomics models were established, including the R_IU model for IU images, the R_MIX model for mixed images and the combined radiomics model (the R_comb model). Random forest was used to tune the optimal radiomics model. The performance of the clinical model and human experts to assess PM was also recorded. Results Fourteen and three radiomics features with low redundancy and high importance were extracted from the IU and mixed images, respectively. The R_IU model showed significantly better performance to predict PM than the R_MIX model in the training cohort (AUC, 0.981 vs. 0.917, p = 0.034). No improvement was observed in the R_comb model (AUC = 0.967). The R_IU model was the optimal radiomics model which showed no overfitting in the testing cohort (AUC = 0.967, p = 0.528). The R_IU model demonstrated significantly higher predictive value on peritoneal status than the clinical model and human experts in the testing cohort (AUC, 0.785, p = 0.005; AUC, 0.732, p <0.001, respectively). Conclusion DECT derived radiomics could serve as a non-invasive and easy-to-use biomarker to preoperatively predict PM for GC, providing opportunity for those patients to tailor appropriate treatment.
Collapse
Affiliation(s)
- Yong Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqi Xi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwu Yao
- Department of Radiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingyun Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihan Xu
- Department of DI CT Collaboration, Siemens Healthineers Ltd, Shanghai, China
| | - Michael Wels
- Department of Diagnostic Imaging Computed Tomography Image Analytics, Siemens Healthcare GmbH, Forchheim, Germany
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Yan
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
40
|
Yüksel C, Erşen O, Basceken SI, Mercan Ü, Yalkın Ö, Culcu S, Bakırarar B, Bayar S, Ünal AE, Demirci S. The role of laparoscopic staging for the management of gastric cancer. POLISH JOURNAL OF SURGERY 2021; 93:1-8. [PMID: 33949319 DOI: 10.5604/01.3001.0014.7360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AİM Staging laparoscopy enables us to perform palliative treatment, neo-adjuvant therapy for curative resection or direct curative resection and making a decision with minimal morbidity by avoiding from unnecessary laparotomies. In the present study, the importance of staging lapafoscopy was retrospectively investigated by using clinical and pathologic data. METHODS Data of 70 out of 350 patients who underwent diagnostic laparoscopy due to gastric cancer at Surgical Oncology department between August 2013 and January 2020 were retrospectively analyzed. RESULTS Peritoneal biopsy was positive for malignity in 41 (58.5%) and negative in 29 (41.5%) of the patients who underwent SL. Peritoneal cytology (PC) results were negative in 32 (45.7%) patients and positive in 38 (54.3%) patients. Peritoneal biopsy and cytology results were concurrently positive in 35 patients and concurrently negative in 26 patients. CONCLUSİONS In conclusion, even the most developed imaging methods cannot provide 100% staging, therefore SL plays an important role in treatment of gastric cancer and laparoscopic staging is essential as a simple, inexpensive, safe and well tolerated method in patients who have the suspicion of peritoneal disease and who cannot be clearly evaluated with pre-operative methods.
Collapse
Affiliation(s)
- Cemil Yüksel
- University of Health Science, Ankara Abdurrahman Yurtaslan Oncology Training and Research Hospital
| | - Ogün Erşen
- Ankara University School of Medicine Department of Surgical Oncology
| | | | - Ümit Mercan
- Ankara University School of Medicine, Department of Surgical Oncology
| | - Ömer Yalkın
- Bursa State Hospital Surgical Oncology Clinic
| | - Serdar Culcu
- University of Health Science, Ankara Abdurrahman Yurtaslan Oncology Training and Research Hospital
| | | | - Sancar Bayar
- Ankara University School of Medicine, Department of Surgical Oncology
| | - Ali Ekrem Ünal
- Ankara University School of Medicine Department of Surgical Oncology
| | - Salim Demirci
- Ankara University School of Medicine, Department of Surgical Oncology
| |
Collapse
|
41
|
Jiang Y, Liang X, Wang W, Chen C, Yuan Q, Zhang X, Li N, Chen H, Yu J, Xie Y, Xu Y, Zhou Z, Li G, Li R. Noninvasive Prediction of Occult Peritoneal Metastasis in Gastric Cancer Using Deep Learning. JAMA Netw Open 2021; 4:e2032269. [PMID: 33399858 PMCID: PMC7786251 DOI: 10.1001/jamanetworkopen.2020.32269] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022] Open
Abstract
IMPORTANCE Occult peritoneal metastasis frequently occurs in patients with advanced gastric cancer and is poorly diagnosed with currently available tools. Because the presence of peritoneal metastasis precludes the possibility of curative surgery, there is an unmet need for a noninvasive approach to reliably identify patients with occult peritoneal metastasis. OBJECTIVE To assess the use of a deep learning model for predicting occult peritoneal metastasis based on preoperative computed tomography images. DESIGN, SETTING, AND PARTICIPANTS In this multicenter, retrospective cohort study, a deep convolutional neural network, the Peritoneal Metastasis Network (PMetNet), was trained to predict occult peritoneal metastasis based on preoperative computed tomography images. Data from a cohort of 1225 patients with gastric cancer who underwent surgery at Sun Yat-sen University Cancer Center (Guangzhou, China) were used for training purposes. To externally validate the model, data were collected from 2 independent cohorts comprising a total of 753 patients with gastric cancer who underwent surgery at Nanfang Hospital (Guangzhou, China) or the Third Affiliated Hospital of Southern Medical University (Guangzhou, China). The status of peritoneal metastasis for all patients was confirmed by pathological examination of pleural specimens obtained during surgery. Detailed clinicopathological data were collected for each patient. Data analysis was performed between September 1, 2019, and January 31, 2020. MAIN OUTCOMES AND MEASURES The area under the receiver operating characteristic curve (AUC) and decision curve were analyzed to evaluate performance in predicting occult peritoneal metastasis. RESULTS A total of 1978 patients (mean [SD] age, 56.0 [12.2] years; 1350 [68.3%] male) were included in the study. The PMetNet model achieved an AUC of 0.946 (95% CI, 0.927-0.965), with a sensitivity of 75.4% and a specificity of 92.9% in external validation cohort 1. In external validation cohort 2, the AUC was 0.920 (95% CI, 0.848-0.992), with a sensitivity of 87.5% and a specificity of 98.2%. The discrimination performance of PMetNet was substantially higher than conventional clinicopathological factors (AUC range, 0.51-0.63). In multivariable logistic regression analysis, PMetNet was an independent predictor of occult peritoneal metastasis. CONCLUSIONS AND RELEVANCE The findings of this cohort study suggest that the PMetNet model can serve as a reliable noninvasive tool for early identification of patients with clinically occult peritoneal metastasis, which will inform individualized preoperative treatment decision-making and may avoid unnecessary surgery and complications. These results warrant further validation in prospective studies.
Collapse
Affiliation(s)
- Yuming Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Xiaokun Liang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaodong Zhang
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Na Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Hao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaoqin Xie
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
42
|
A Nomogram Based on Clinicopathologic Features and Preoperative Hematology Parameters to Predict Occult Peritoneal Metastasis of Gastric Cancer: A Single-Center Retrospective Study. DISEASE MARKERS 2020; 2020:1418978. [PMID: 33376558 PMCID: PMC7746455 DOI: 10.1155/2020/1418978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/22/2020] [Accepted: 11/22/2020] [Indexed: 01/19/2023]
Abstract
Background In patients with gastric cancer (GC), peritoneal metastasis is an indication of the end stage and often indicates a poor outcome. The diagnosis of peritoneal metastasis, especially occult peritoneal metastasis (OPM), remains a challenge for surgeons. This study was designed to explore the relationship between OPM and clinicopathological characteristics and preoperative hematological parameters in patients with GC and to develop a nomogram to predict the probability of OPM before surgery. Methods A total of 672 patients with GC from our center were included, including 583 OPM-negative and 89 OPM-positive patients. These patients were divided into training and validation groups based on when they received treatment. OPM was diagnosed during surgery in patients without any signs of metastasis through imaging examination. Predictive factors were screened by least absolute shrinkage and selection operator logistic regression of all 18 characteristics. The nomogram of OPM was constructed based on these filtered variables. The discriminative and calibration performance of the model were simultaneously evaluated. Results A total of six variables, including tumor size, degree of differentiation, depth of invasion, Glasgow prognosis score, and plasma levels of CA125 and fibrinogen, were selected for integration into the final predictive nomogram. The area under curve (AUC) of the nomogram with six factors was 0.906 (95% confidence interval (CI): 0.872-0.941) and 0.889 (95% CI: 0.795-0.984) in the training and validation groups, respectively. Calibration plots of the nomogram in the two sets revealed a good consistency between predicted and actual probabilities. Decision curve analysis showed that the nomogram had a positive net benefit among all threshold probabilities between 0% and 82%. This nomogram was superior to models incorporating only clinicopathologic or hematologic features. Conclusion Both clinicopathological and preoperative hematological parameters are significantly associated with OPM. The nomogram constructed with six factors could be used to calculate the probability of OPM and identify the high-risk population in GC. This may be helpful for early detection of OPM in patients with GC.
Collapse
|
43
|
Zhang C, Chen Z, Chong X, Chen Y, Wang Z, Yu R, Sun T, Chen X, Shao Y, Zhang X, Gao J, Shen L. Clinical implications of plasma ctDNA features and dynamics in gastric cancer treated with HER2-targeted therapies. Clin Transl Med 2020; 10:e254. [PMID: 33377634 PMCID: PMC7737756 DOI: 10.1002/ctm2.254] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is confronted with limited options for precision medicine. Human epidermal growth factor receptor 2 (HER2) is the principal druggable target of GC, yet proper biomarkers for response/resistance prediction remain unveiled. METHODS From 40 GC patients received HER2-targeted therapy, a total of 327 peripheral blood plasma specimens was collected including baseline and treatment time points. Circulating tumor DNA (ctDNA) was extracted and sequenced with a target panel of 425 genes. Experimental validation of resistant mutations was carried out in NIH-3T3 cell line. RESULTS Genomic features, including ERBB2 copy number variation (CNV), total copy number load, and tumor mutation burdens (TMBs), dynamically changed along with the treatment process and correlated with disease progression. Plasma ctDNA-based diagnosis was more sensitive than conventional computed tomography scanning in 40% of investigated patients, gaining additional time for clinical management. Compared to baseline, new gene alterations were emerged in 12 patients who developed drug resistance during treatment. ERBB2 mutations potentially related to Pyrotinib resistance were identified in plasma ctDNA of one patient and functional analysis of their downstream signaling pathways was carried out in NIH-3T3 cell line. TMB exhibited more power than ERBB2 CNV in predicting treatment responses and prognosis for HER2-targeted therapy in GC patients. Interestingly, survival analysis indicated that patients harboring both HER2 (ERBB2) positivity and high TMB might gain more therapeutic benefits from immune checkpoint inhibitors instead of HER2-targeted regimens that required further studies and validations CONCLUSIONS: Our work showed that the dynamic surveillance of plasma ctDNA genomic features provided instructive information for the precision medication of GC patients.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Gastrointestinal OncologyKey laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Peking University Cancer Hospital & InstituteBeijingChina
| | - Zuhua Chen
- Department of OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoyi Chong
- Department of Gastrointestinal OncologyKey laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Peking University Cancer Hospital & InstituteBeijingChina
| | - Yang Chen
- Department of Gastrointestinal OncologyKey laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Peking University Cancer Hospital & InstituteBeijingChina
| | - Zhenghang Wang
- Department of Gastrointestinal OncologyKey laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Peking University Cancer Hospital & InstituteBeijingChina
| | - Ruoying Yu
- Translational Medicine Research InstituteGeneseeq Technology IncTorontoOntarioCanada
| | | | - Xiaoxi Chen
- Translational Medicine Research InstituteGeneseeq Technology IncTorontoOntarioCanada
| | - Yang Shao
- Nanjing Geneseeq Technology IncNanjingChina
| | - Xiaotian Zhang
- Department of Gastrointestinal OncologyKey laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Peking University Cancer Hospital & InstituteBeijingChina
| | - Jing Gao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
| | - Lin Shen
- Department of Gastrointestinal OncologyKey laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Peking University Cancer Hospital & InstituteBeijingChina
| |
Collapse
|
44
|
Xu SJ, Lin GS, Ling HJ, Guo RJ, Chen J, Liao YM, Lin T, Zhou YJ. Nomogram to Predict Preoperative Occult Peritoneal Metastasis of Gastrointestinal Stromal Tumors (GIST) Based on Imaging and Inflammatory Indexes. Cancer Manag Res 2020; 12:11713-11721. [PMID: 33239911 PMCID: PMC7681585 DOI: 10.2147/cmar.s275422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/06/2020] [Indexed: 12/27/2022] Open
Abstract
Background Preoperative imaging examination is the primary method for diagnosing metastatic gastrointestinal stromal tumor (GIST), but it is associated with a high rate of missed diagnosis. Therefore, it is important to establish an accurate model for predicting occult peritoneal metastasis (PM) of GIST. Patients and Methods GIST patients seen between April 2002 and December 2018 were selected from an institutional database. Using multivariate logistic regression analyses, we created a nomogram to predict occult PM of GIST and validated it with an independent cohort from the same center. The concordance index (C-index), decision curve analysis (DCA) and a clinical impact curve (CIC) were used to evaluate its predictive ability. Results A total of 522 eligible GIST patients were enrolled in this study and divided into training (n=350) and validation cohorts (n=172). Factors associated with occult PM were included in the model: tumor size (odds ratio [OR] 1.194 95% confidence interval [CI], 1.034-1.378; p=0.016), primary location (OR 7.365 95% CI, 2.192-24.746; p=0.001), tumor capsule (OR 4.282 95% CI, 1.209-15.166; p=0.024), Alb (OR 0.813 95% CI, 0.693-0.954; p=0.011) and FIB (OR 2.322 95% CI, 1.410-3.823; p=0.001). The C-index was 0.951 (95% CI, 0.917-0.985) in the training cohort and 0.946 (95% CI, 0.900-0.992) in the validation cohort. In the training cohort, the prediction model had a sensitivity of 82.8%, a specificity of 93.8%, a positive predictive value of 54.7%, and a negative predictive value of 98.4%; the validation cohort values were 94.7%, 85.0%, 43.9% and 99.2%, respectively. DCA and CIC results showed that the nomogram had clinical value in predicting occult PM in GIST patients. Conclusion Imaging and inflammatory indexes are significantly associated with microscopic metastases of GIST. A nomogram including these factors would have an excellent ability to predict occult PM.
Collapse
Affiliation(s)
- Shao-Jun Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Guo-Sheng Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Hong-Jian Ling
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Ren-Jie Guo
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Jie Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Yi-Ming Liao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Tao Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Yong-Jian Zhou
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| |
Collapse
|
45
|
Huang Z, Liu D, Chen X, He D, Yu P, Liu B, Wu B, Hu J, Song B. Deep Convolutional Neural Network Based on Computed Tomography Images for the Preoperative Diagnosis of Occult Peritoneal Metastasis in Advanced Gastric Cancer. Front Oncol 2020; 10:601869. [PMID: 33224893 PMCID: PMC7667265 DOI: 10.3389/fonc.2020.601869] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/12/2020] [Indexed: 02/05/2023] Open
Abstract
We aimed to develop a deep convolutional neural network (DCNN) model based on computed tomography (CT) images for the preoperative diagnosis of occult peritoneal metastasis (OPM) in advanced gastric cancer (AGC). A total of 544 patients with AGC were retrospectively enrolled. Seventy-nine patients were confirmed with OPM during surgery or laparoscopy. CT images collected during the initial visit were randomly split into a training cohort and a testing cohort for DCNN model development and performance evaluation, respectively. A conventional clinical model using multivariable logistic regression was also developed to estimate the pretest probability of OPM in patients with gastric cancer. The DCNN model showed an AUC of 0.900 (95% CI: 0.851-0.953), outperforming the conventional clinical model (AUC = 0.670, 95% CI: 0.615-0.739; p < 0.001). The proposed DCNN model demonstrated the diagnostic detection of occult PM, with a sensitivity of 81.0% and specificity of 87.5% using the cutoff value according to the Youden index. Our study shows that the proposed deep learning algorithm, developed with CT images, may be used as an effective tool to preoperatively diagnose OPM in AGC.
Collapse
Affiliation(s)
- Zixing Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Liu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xinzu Chen
- State Key Laboratory of Biotherapy, Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Du He
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengxin Yu
- Institute of Advanced Research, Infervision, Beijing, China
| | - Baiyun Liu
- Institute of Advanced Research, Infervision, Beijing, China
| | - Bing Wu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiankun Hu
- State Key Laboratory of Biotherapy, Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Liu F, Huang C, Xu Z, Su X, Zhao G, Ye J, Du X, Huang H, Hu J, Li G, Yu P, Li Y, Suo J, Zhao N, Zhang W, Li H, He H, Sun Y. Morbidity and Mortality of Laparoscopic vs Open Total Gastrectomy for Clinical Stage I Gastric Cancer: The CLASS02 Multicenter Randomized Clinical Trial. JAMA Oncol 2020; 6:1590-1597. [PMID: 32815991 PMCID: PMC7441466 DOI: 10.1001/jamaoncol.2020.3152] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/28/2020] [Indexed: 02/05/2023]
Abstract
IMPORTANCE The safety of laparoscopic total gastrectomy (LTG) for the treatment of gastric cancer remains uncertain given the lack of high-level clinical evidence. OBJECTIVE To compare the safety of LTG for clinical stage I gastric cancer with that of conventional open total gastrectomy (OTG). DESIGN, SETTING, AND PARTICIPANTS The Chinese Laparoscopic Gastrointestinal Surgery Study (CLASS) Group CLASS02 study was a prospective, multicenter, open-label, noninferiority, randomized clinical trial that compared the safety of LTG vs OTG with lymphadenectomy for patients with clinical stage I gastric cancer. From January 2017 to September 2018, a total of 227 patients were enrolled. Final follow-up was in October 2018. INTERVENTIONS Eligible patients were randomized to LTG (n = 113) or OTG (n = 114) by an interactive web response system. MAIN OUTCOMES AND MEASURES The primary outcome was the morbidity and mortality within 30 days following surgeries between LTG and OTG with a noninferiority margin of 10%. The secondary outcomes were recovery courses and postoperative hospital stays. RESULTS A total of 214 patients were analyzed for morbidity and mortality (105 patients in the LTG group and 109 patients in the OTG group). The mean (SD) age was 59.8 (9.4) years in the LTG group and 59.4 (9.2) years in the OTG group, and most were male (LTG group, 75 of 105 [71.4%]; OTG group, 80 of 109 [73.4%]). The overall morbidity and mortality rates were not significantly different between the groups (rate difference, -1.1%; 95% CI, -11.8% to 9.6%). Intraoperative complications occurred in 3 patients (2.9%) in the LTG group and 4 patients (3.7%) in the OTG group (rate difference, -0.8%; 95% CI, -6.5% to 4.9%). In addition, there was no significant difference in the overall postoperative complication rate of 18.1% in the LTG group and 17.4% in the OTG group (rate difference, 0.7%; 95% CI, -9.6% to 11.0%). One patient in the LTG group died from intra-abdominal bleeding secondary to splenic artery hemorrhage. However, there was no significant difference in mortality between the LTG group and the OTG group (rate difference, 1.0%; 95% CI, -2.5% to 5.2%), and the distribution of complication severity was similar between the 2 groups. CONCLUSIONS AND RELEVANCE The results of the CLASS02 trial showed that the safety of LTG with lymphadenectomy by experienced surgeons for clinical stage I gastric cancer was comparable to that of OTG. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03007550.
Collapse
Affiliation(s)
- Fenglin Liu
- Zhongshan Hospital, Department of General Surgery, Fudan University, Shanghai, China
| | - Changming Huang
- Union Hospital, Department of General Surgery, Fujian Medical University, Fuzhou, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xiangqian Su
- Beijing Cancer Hospital, Department of General Surgery, Peking University, Beijing, China
| | - Gang Zhao
- Renji Hospital, Department of General Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Jianxin Ye
- Department of General Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaohui Du
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Hua Huang
- Shanghai Cancer Center, Department of General Surgery, Fudan University, Shanghai, China
| | - Jiankun Hu
- West China Hospital, Department of General Surgery, Sichuan University, Chengdu, China
| | - Guoxin Li
- Nanfang Hospital, Department of General Surgery, Southern Medical University, Guangzhou, China
| | - Peiwu Yu
- Department of General Surgery, The First Hospital Affiliated to AMU, Chongqing, China
| | - Yong Li
- Department of General Surgery, Guangdong General Hospital, Guangzhou, China
| | - Jian Suo
- Department of General Surgery, The First Hospital of Jilin University, Changchun, China
| | - Naiqing Zhao
- Department of Biostatistics, Fudan University School of Public Health, Shanghai, China
| | - Wei Zhang
- Department of Biostatistics, Fudan University School of Public Health, Shanghai, China
| | - Haojie Li
- Zhongshan Hospital, Department of General Surgery, Fudan University, Shanghai, China
| | - Hongyong He
- Zhongshan Hospital, Department of General Surgery, Fudan University, Shanghai, China
| | - Yihong Sun
- Zhongshan Hospital, Department of General Surgery, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Abstract
Neoadjuvant or perioperative therapy with radical surgery is a meaningful approach to improve the prognosis of gastric cancer. The FLOT regimen (5-fluorouracil, leucovorin, oxaliplatin, docetaxel) has been established as a perioperative concept in Germany and is also increasingly being used internationally. The prognostic significance of neoadjuvant chemoradiotherapy has to be awaited based on the results of currently active studies. The microsatellite instability (MSI) status has increasingly gained in importance with respect to decision-making processes in the interdisciplinary tumor board as patients with MSI tumors probably do not benefit from neoadjuvant therapy. Patients with only initial stages of locally advanced MSI tumors (cT2, N0) and those with comorbidities could be spared from neoadjuvant therapy. This article critically deals with the current state of the art concepts as well as with ongoing studies with respect to neoadjuvant and perioperative treatment of gastric cancer. For this purpose, the essential already published and the active studies are presented.
Collapse
|
48
|
Huang W, Zhou K, Jiang Y, Chen C, Yuan Q, Han Z, Xie J, Yu S, Sun Z, Hu Y, Yu J, Liu H, Xiao R, Xu Y, Zhou Z, Li G. Radiomics Nomogram for Prediction of Peritoneal Metastasis in Patients With Gastric Cancer. Front Oncol 2020; 10:1416. [PMID: 32974149 PMCID: PMC7468436 DOI: 10.3389/fonc.2020.01416] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
Objective: The aim of this study is to evaluate whether radiomics imaging signatures based on computed tomography (CT) could predict peritoneal metastasis (PM) in gastric cancer (GC) and to develop a nomogram for preoperative prediction of PM status. Methods: We collected CT images of pathological T4 gastric cancer in 955 consecutive patients of two cancer centers to analyze the radiomics features retrospectively and then developed and validated the prediction model built from 292 quantitative image features in the training cohort and two validation cohorts. Lasso regression model was applied for selecting feature and constructing radiomics signature. Predicting model was developed by multivariable logistic regression analysis. Radiomics nomogram was developed by the incorporation of radiomics signature and clinical T and N stage. Calibration, discrimination, and clinical usefulness were used to evaluate the performance of the nomogram. Results: In training and validation cohorts, PM status was associated with the radiomics signature significantly. It was found that the radiomics signature was an independent predictor for peritoneal metastasis in multivariable logistic analysis. For training and internal and external validation cohorts, the area under the receiver operating characteristic curves (AUCs) of radiomics signature for predicting PM were 0.751 (95%CI, 0.703-0.799), 0.802 (95%CI, 0.691-0.912), and 0.745 (95%CI, 0.683-0.806), respectively. Furthermore, for training and internal and external validation cohorts, the AUCs of radiomics nomogram for predicting PM were 0.792 (95%CI, 0.748-0.836), 0.870 (95%CI, 0.795-0.946), and 0.815 (95%CI, 0.763-0.867), respectively. Conclusions: CT-based radiomics signature could predict peritoneal metastasis, and the radiomics nomogram can make a meaningful contribution for predicting PM status in GC patient preoperatively.
Collapse
Affiliation(s)
- Weicai Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kangneng Zhou
- School of Computer and Communication Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Han
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingjing Xie
- Center for Drug and Clinical Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shitong Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zepang Sun
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruoxiu Xiao
- School of Computer and Communication Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Nakamura N, Kinami S, Tomita Y, Miyata T, Fujita H, Takamura H, Ueda N, Kosaka T. The neutrophil/lymphocyte ratio as a predictor of successful conversion surgery for stage IV gastric cancer: a retrospective study. BMC Cancer 2020; 20:363. [PMID: 32349701 PMCID: PMC7191721 DOI: 10.1186/s12885-020-06884-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 04/21/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Conversion surgery (CS) following a response to chemotherapy occasionally leads to prolonged survival in patients with stage IV gastric cancer (GC). This study aimed to evaluate the predictive value of the neutrophil/lymphocyte ratio (NLR) for the success of CS in patients with stage IV GC. METHODS We retrospectively analyzed data of 50 patients with stage IV GC who received systemic chemotherapy between January 2009 and December 2017 at the Kanazawa Medical University Hospital. The successful CS group included the patients who underwent R0 or R1 resection with CS, and the failed CS group included the patients who did not undergo CS after chemotherapy or those who, despite undergoing CS, had to additionally undergo R2 resection. Clinicopathological characteristics were examined in both groups. Univariate and multivariate analyses were performed to identify pretherapeutic parameters that were independently associated with the achievement of successful CS. RESULTS The number of patients in the successful and failed CS groups were 12 and 38, respectively. On univariate analysis, gender (P = 0.01), NLR (P = 0.003), albumin levels (P = 0.004), and absence of peritoneal metastasis (P = 0.004) were found to be significantly correlated with a successful CS. On multivariate analysis, NLR < 4 and absence of peritoneal metastasis were independently correlated with a successful CS (P = 0.02 and P = 0.002, respectively). In patients without peritoneal metastasis, successful CS rates in patients with NLR < 4 were significantly higher than those in patients with NLR ≥ 4 (61.1% vs. 10.0%, P = 0.005). CONCLUSIONS The NLR was a significant independent predictor of the achievement of successful CS in stage IV GC patients, especially among the patients without peritoneal metastasis. Patients with a low NLR could have higher possibility of achieving successful CS.
Collapse
Affiliation(s)
- Naohiko Nakamura
- Department of Surgical Oncology, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan.
| | - Shinichi Kinami
- Department of Surgical Oncology, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Yasuto Tomita
- Department of Surgical Oncology, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Takashi Miyata
- Department of Surgical Oncology, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Hideto Fujita
- Department of Surgical Oncology, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Hiroyuki Takamura
- Department of Surgical Oncology, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Nobuhiko Ueda
- Department of Surgical Oncology, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Takeo Kosaka
- Department of Surgical Oncology, Kanazawa Medical University Hospital, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| |
Collapse
|
50
|
Dong D, Tang L, Li ZY, Fang MJ, Gao JB, Shan XH, Ying XJ, Sun YS, Fu J, Wang XX, Li LM, Li ZH, Zhang DF, Zhang Y, Li ZM, Shan F, Bu ZD, Tian J, Ji JF. Development and validation of an individualized nomogram to identify occult peritoneal metastasis in patients with advanced gastric cancer. Ann Oncol 2020; 30:431-438. [PMID: 30689702 PMCID: PMC6442651 DOI: 10.1093/annonc/mdz001] [Citation(s) in RCA: 302] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Occult peritoneal metastasis (PM) in advanced gastric cancer (AGC) patients is highly possible to be missed on computed tomography (CT) images. Patients with occult PMs are subject to late detection or even improper surgical treatment. We therefore aimed to develop a radiomic nomogram to preoperatively identify occult PMs in AGC patients. Patients and methods A total of 554 AGC patients from 4 centers were divided into 1 training, 1 internal validation, and 2 external validation cohorts. All patients’ PM status was firstly diagnosed as negative by CT, but later confirmed by laparoscopy (PM-positive n = 122, PM-negative n = 432). Radiomic signatures reflecting phenotypes of the primary tumor (RS1) and peritoneum region (RS2) were built as predictors of PM from 266 quantitative image features. Individualized nomograms of PM status incorporating RS1, RS2, or clinical factors were developed and evaluated regarding prediction ability. Results RS1, RS2, and Lauren type were significant predictors of occult PM (all P < 0.05). A nomogram of these three factors demonstrated better diagnostic accuracy than the model with RS1, RS2, or clinical factors alone (all net reclassification improvement P < 0.05). The area under curve yielded was 0.958 [95% confidence interval (CI) 0.923–0.993], 0.941 (95% CI 0.904–0.977), 0.928 (95% CI 0.886–0.971), and 0.920 (95% CI 0.862–0.978) for the training, internal, and two external validation cohorts, respectively. Stratification analysis showed that this nomogram had potential generalization ability. Conclusion CT phenotypes of both primary tumor and nearby peritoneum are significantly associated with occult PM status. A nomogram of these CT phenotypes and Lauren type has an excellent prediction ability of occult PM, and may have significant clinical implications on early detection of occult PM for AGC.
Collapse
Affiliation(s)
- D Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Radiology Department, , Peking University Cancer Hospital & Institute, Beijing; University of Chinese Academy of Sciences, Beijing
| | - L Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Radiology Department, , Peking University Cancer Hospital & Institute, Beijing
| | - Z-Y Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - M-J Fang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing; University of Chinese Academy of Sciences, Beijing
| | - J-B Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - X-H Shan
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang
| | - X-J Ying
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - Y-S Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Radiology Department, , Peking University Cancer Hospital & Institute, Beijing
| | - J Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Radiology Department, , Peking University Cancer Hospital & Institute, Beijing
| | - X-X Wang
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang
| | - L-M Li
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Z-H Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming
| | - D-F Zhang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming
| | - Y Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - Z-M Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - F Shan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - Z-D Bu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - J Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing; University of Chinese Academy of Sciences, Beijing; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China.
| | - J-F Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing.
| |
Collapse
|