1
|
Yu J, Cai W, Zhou T, Men B, Chen S, Tu D, Guo W, Wang J, Zhao F, Wang Y. CEACAM1 increased the lymphangiogenesis through miR-423-5p and NF- kB in Non-Small Cell Lung Cancer. Biochem Biophys Rep 2024; 40:101833. [PMID: 39398537 PMCID: PMC11470192 DOI: 10.1016/j.bbrep.2024.101833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Background Lung cancer causes significant mortality, with invasion and metastasis being the main features that cause most cancer deaths. Lymph node metastasis is the primary metastatic route in non-small cell carcinoma (NSCLC) and influences the staging and prognosis of NSCLC. Cumulative studies have reported that Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is involved in the progression of various cancers. However, few studies have discussed the function of CEACAM1 in lymphangiogenesis in NSCLC. Here, we examined how CEACAM1 influences lymphangiogenesis in NSCLC. Methods A total of 30 primary squamous cell carcinoma (LUSC) patients diagnosed with LN metastasis were prospectively selected. LUSC tumor tissues, para-cancerous tissues, and positive lymph node tissues were harvested. The expression and subcellular location of CEACAM1, CD31, and LVYE1 in clinical samples were detected by immunohistochemistry. Next, the CEACAM1 and hsa-miR-423-5p expressions were detected by qPCR. The protein expression of lymphangiogenesis-associated proteins and critical cytokines of the NF-κB pathway in HDLECs was detected by Western blot. A tube formation assay was performed to detect the lymphangiogenesis in different groups. The interaction between CEACAM1 and hsa-miR-423-5p was verified using a dual luciferase assay. Results CEACAM1 was found to be a potential gene associated with lung cancer prognosis. It was positively correlated with angiogenesis and lymphangiogenesis. Then, we detected the function of CEACAM1 in lymphangiogenesis and found that CEACAM1 promoted lymphangiogenesis. hsa-miR-423-5p overexpression inhibited lymphangiogenesis via targeting CEACAM1. Finally, we observed that CEACAM1 can activate the NF-κB pathway and, therefore, promote lymphangiogenesis. Conclusion We found that CEACAM1 enhanced lymphangiogenesis in NSCLC via NF-kB activation and was repressed by miR-423-5p. This suggests the value of CEACAM1 as a new therapeutic marker in NSCLC.
Collapse
Affiliation(s)
- Jie Yu
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Wenke Cai
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Tao Zhou
- Department of Respiration, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Bo Men
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Shunqiong Chen
- Department of Respiration, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Dong Tu
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Wei Guo
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Jicui Wang
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Feipeng Zhao
- Department of Plastic and Maxillofacial Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
2
|
Matsunaga T, Saito H, Osaki T, Fukuda K, Fukumoto Y, Takahashi S, Taniguchi K, Iwamoto A, Kuroda H, Katano K, Shimizu S, Shishido Y, Miyatani K, Sakamoto T, Fujiwara Y. Using the geriatric nutritional risk index to predict outcomes in older patients with remnant gastric cancer after gastrectomy: a retrospective multicenter study in Japan. Surg Today 2024; 54:1360-1368. [PMID: 38652300 DOI: 10.1007/s00595-024-02850-w] [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: 02/08/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE This study investigated the prognostic value of the geriatric nutritional risk index (GNRI) in patients undergoing curative gastrectomy for remnant gastric cancer (RGC). METHODS This multicenter retrospective study included 105 patients with RGC of ≥ 65 years of age who underwent curative gastrectomy at 10 institutions in Japan between January 2000 and December 2016. Postoperative complications, overall survival (OS), and disease-specific survival (DSS) were analyzed. RESULTS Receiver operating curve analyses indicated that the optimal cutoff value of the GNRI for OS was 95.4. Patients were categorized into high and low GNRI groups based on the optimal GNRI cutoff value. The GNRI was significantly correlated with body mass index (p < 0.001), amount of bleeding (p = 0.021), Clavien-Dindo grade 5 postoperative complications (p = 0.040), death caused by primary disease (p = 0.010), and death caused by other diseases (p = 0.002). The OS and DSS were significantly worse in the low GNRI group. A low GNRI and T3 or deeper tumor invasion were independent prognostic factors for OS and DSS. CONCLUSIONS The GNRI is a promising predictor of both short- and long-term outcomes in older patients with RGC.
Collapse
Affiliation(s)
- Tomoyuki Matsunaga
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Yonago, 683-8504, Japan.
| | - Hiroaki Saito
- Department of Surgery, Japanese Red Cross Tottori Hospital, Tottori, 680-8517, Japan
| | - Tomohiro Osaki
- Department of Surgery, Tottori Prefectural Central Hospital, Tottori, 680-0901, Japan
| | - Kenji Fukuda
- Department of Surgery, Sanin Rosai Hospital, Yonago, 683-8605, Japan
| | - Yoji Fukumoto
- Division of Gastroenterology, Matsue City Hospital, Matsue, 690-8509, Japan
| | - Sadamu Takahashi
- National Hospital Organization, Hamada Medical Center, Hamada, 697-8511, Japan
| | - Kenjiro Taniguchi
- Department of Surgery, Yonago Medical Center of National Hospital Organization, Yonago, 683-0006, Japan
| | - Akemi Iwamoto
- Division of Digestive Surgery, Tottori Prefectural Kousei Hospital, Kurayoshi, 682-0804, Japan
| | - Hirohiko Kuroda
- Department of Surgery, Japanese Red Cross Masuda Hospital, Masuda, 698-8501, Japan
| | - Kuniyuki Katano
- Department of Surgery, Nanbu Town National Health Insurance Saihaku Hospital, Nanbu, 683-0323, Japan
| | - Shota Shimizu
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Yonago, 683-8504, Japan
| | - Yuji Shishido
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Yonago, 683-8504, Japan
| | - Kozo Miyatani
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Yonago, 683-8504, Japan
| | - Teruhisa Sakamoto
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Yonago, 683-8504, Japan
| | - Yoshiyuki Fujiwara
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, Yonago, 683-8504, Japan
| |
Collapse
|
3
|
Gupta R, Das CK, Nair SS, Pedraza-Bermeo AM, Zahalka AH, Kyprianou N, Bhardwaj N, Tewari AK. From foes to friends: rethinking the role of lymph nodes in prostate cancer. Nat Rev Urol 2024; 21:687-700. [PMID: 39095580 DOI: 10.1038/s41585-024-00912-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 08/04/2024]
Abstract
Clinically localized prostate cancer is often treated with radical prostatectomy combined with pelvic lymph node dissection. Data suggest that lymph node dissection does improve disease staging, but its therapeutic value has often been debated, with few studies showing that lymph node removal directly improves oncological outcomes; however, lymph nodes are an important first site of antigen recognition and immune system activation and the success of many currently used immunological therapies hinges on this dogma. Evidence, particularly in the preclinical setting, has demonstrated that the success of immune checkpoint inhibitors is dampened by the removal of tumour-draining lymph nodes. Thus, whether lymph nodes are truly 'foes' or whether they are actually 'friends' in oncological care is an important idea to discuss.
Collapse
Affiliation(s)
- Raghav Gupta
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chandan K Das
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sujit S Nair
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ali H Zahalka
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashutosh K Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Abate M, Drebin H, Shimada S, Fei T, McKinley S, Poruk K, Ferguson B, Neuwirth M, Tang LH, Vardhana S, Strong VE. Feasibility and Efficacy of Sentinel Lymph Node Mapping in Gastric Cancer. Ann Surg Oncol 2024; 31:6959-6969. [PMID: 39097552 DOI: 10.1245/s10434-024-15642-w] [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: 09/15/2023] [Accepted: 06/07/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Lymph node metastasis is a critical prognostic factor for patients with gastric carcinoma (GC). Sentinel lymph node (SLN) mapping has the potential to identify the initial site of draining lymph node metastasis and reduce the extent of surgical lymphadenectomy. This study aimed to evaluate the diagnostic accuracy of SLN mapping in GC. METHODS The study enrolled 129 GC patients undergoing total or partial gastrectomy with D2 lymphadenectomy and indocyanine green fluorescence-guided SLN mapping. The primary outcomes were the negative predictive value (NPV) and sensitivity of SLN mapping. The secondary outcomes were clinicopathologic factors associated with SLN mapping accuracy and successful SLN mapping. RESULTS The SLN detection rate in this study was 86.8 %. The study had an overall NPV of 83.1 % and an overall sensitivity of 65.8 %. The NPV was found to be significantly higher in the patients with no lymphovascular invasion (LVI) than in those with LVI (96.0 % vs 59.3 %; p < 0.001) and in the patients whose pathologic T (pT) stage lower than 3 than in those whose T stage was 3 or higher (92.0 % vs 66.7 %; p = 0.009). The sensitivity of SLN mapping was 50 % in the patients with no LVI and 33 % in the patients with a pT stage lower than 3. CONCLUSION The study results showed that for patients with early-stage GC with no LVI, negative SLN findings may represent a potential additive predictor indicating the absence of regional LN metastasis. However, given the low sensitivity rates noted, further research is needed to identify specific patient populations that may benefit from SLN mapping in GC.
Collapse
Affiliation(s)
- Miseker Abate
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, USA
| | - Harrison Drebin
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shoji Shimada
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sophia McKinley
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katherine Poruk
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ben Ferguson
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Madalyn Neuwirth
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura H Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Santosha Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vivian E Strong
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
5
|
Jung YJ, Kim SJ, Seo HS, Lee HH, Song KY, Kim SG. Low Absolute Lymphocyte Count Correlates with Lymph Node Metastases and Worse Survival of Patients with Gastric Cancer. Ann Surg Oncol 2024; 31:6951-6958. [PMID: 39090494 DOI: 10.1245/s10434-024-15874-w] [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: 01/11/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Several studies have found that the absolute lymphocyte (ALC) or neutrophil count predicts the survival of patients with solid tumors, and that the neutrophil-to-lymphocyte ratio and the prognostic nutritional index are useful markers of gastric cancer prognosis. However, it remains unclear whether the ALC is prognostic of lymph node (LN) metastasis in patients with gastric cancer. In this study, we aimed to explore the impact of ALC on prognosis and distinctive clinical characteristics in patients with gastric cancer. PATIENTS AND METHODS The medical records of patients with gastric adenocarcinomas who underwent radical gastrectomy with curative intent at Seoul St. Mary's Hospital and Yeouido St. Mary's Hospital between January 2010 and December 2017 were reviewed. Of these, 4149 patients for whom preoperative white blood cell, neutrophil, and lymphocyte counts were available were enrolled. RESULTS In all 4149 patients, ALC gradually decreased as the pN stage increased. Those with an ALC of less than 1360 cells/μL were defined as a low-ALC group, and advanced cT and cN stages were the strongest risk factors for LN metastasis in both univariate and multivariate analyses; undifferentiated tumor histology and a low ALC were also significant risk factors. Patients of all stages in the ALC-low group exhibited poorer prognoses. The ALC-low group also exhibited a higher recurrence rate in a greater proportion of LNs. CONCLUSIONS In patients with gastric cancer, as the preoperative ALC decreases, the incidence of LN metastasis increases. A low ALC is associated with a high recurrence rate, particularly in LNs.
Collapse
Affiliation(s)
- Yoon Ju Jung
- Division of Gastrointestinal Surgery, Department of Surgery, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - So Jung Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Seok Seo
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Han Hong Lee
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyo Young Song
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Geun Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
6
|
Capuano A, Vescovo M, Canesi S, Pivetta E, Doliana R, Nadin MG, Yamamoto M, Tsukamoto T, Nomura S, Pilozzi E, Palumbo A, Canzonieri V, Cannizzaro R, Scanziani E, Baldassarre G, Mongiat M, Spessotto P. The extracellular matrix protein EMILIN-1 impacts on the microenvironment by hampering gastric cancer development and progression. Gastric Cancer 2024; 27:1016-1030. [PMID: 38941035 PMCID: PMC11335817 DOI: 10.1007/s10120-024-01528-z] [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: 03/06/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND The contribution of the tumor microenvironment and extracellular matrix to the aggressive biology of Gastric Cancer (GC) has been recently characterized; however, the role of EMILIN-1 in this context is unknown. EMILIN-1 is an essential structural element for the maintenance of lymphatic vessel (LV) integrity and displays anti-proliferative properties as demonstrated in skin and colon cancer. Given the key role of LVs in GC progression, the aim of this study was to investigate the role of EMILIN-1 in GC mouse models. METHODS We used the syngeneic YTN16 cells which were injected subcutaneously and intraperitoneally in genetically modified EMILIN-1 mice. In alternative, carcinogenesis was induced using N-Methyl-N-nitrosourea (MNU). Mouse-derived samples and human biopsies were analyzed by IHC and IF to the possible correlation between EMILIN-1 expression and LV pattern. RESULTS Transgenic mice developed tumors earlier compared to WT animals. 20 days post-injection tumors developed in EMILIN-1 mutant mice were larger and displayed a significant increase of lymphangiogenesis. Treatment of transgenic mice with MNU associated with an increased number of tumors, exacerbated aggressive lesions and higher levels of LV abnormalities. A significant correlation between the levels of EMILIN-1 and podoplanin was detected also in human samples, confirming the results obtained with the pre-clinical models. CONCLUSIONS This study demonstrates for the first time that loss of EMILIN-1 in GC leads to lymphatic dysfunction and proliferative advantages that sustain tumorigenesis, and assess the use of our animal model as a valuable tool to verify the fate of GC upon loss of EMILIN-1.
Collapse
Affiliation(s)
- Alessandra Capuano
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Maddalena Vescovo
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Simone Canesi
- Dipartimento di Medicina Veterinaria e Scienze Animali (DIVAS), Università Degli Studi di Milano, Milan, Italy
| | - Eliana Pivetta
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
- Clinical Pathology Unit, Ospedale Santa Maria Degli Angeli, Pordenone, Italy
| | - Roberto Doliana
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Maria Grazia Nadin
- Oncological Gastroenterology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Aviano, Italy
| | - Masami Yamamoto
- Laboratory of Physiological Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Department of Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Japan
| | - Sachiyo Nomura
- Department of Clinical Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Azienda Ospedaliero-Universitaria Sant'Andrea, Rome, Italy
| | - Antonio Palumbo
- Pathology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Aviano, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Renato Cannizzaro
- Oncological Gastroenterology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Eugenio Scanziani
- Dipartimento di Medicina Veterinaria e Scienze Animali (DIVAS), Università Degli Studi di Milano, Milan, Italy
| | - Gustavo Baldassarre
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Maurizio Mongiat
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Paola Spessotto
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy.
| |
Collapse
|
7
|
Jain V, Sakhuja P, Agarwal AK, Sirdeshmukh R, Siraj F, Gautam P. Lymph Node Metastasis in Gastrointestinal Carcinomas: A View from a Proteomics Perspective. Curr Oncol 2024; 31:4455-4475. [PMID: 39195316 DOI: 10.3390/curroncol31080333] [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: 04/16/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 08/29/2024] Open
Abstract
Lymph node metastasis (LNM) is one of the major prognostic factors in human gastrointestinal carcinomas (GICs). The lymph node-positive patients have poorer survival than node-negative patients. LNM is directly associated with the recurrence and poor survival of patients with GICs. The early detection of LNM in patients and designing effective therapies to suppress LNM may significantly impact the survival of these patients. The rapid progress made in proteomic technologies could be successfully applied to identify molecular targets for cancers at high-throughput levels. LC-MS/MS analysis enables the identification of proteins involved in LN metastasis, which can be utilized for diagnostic and therapeutic applications. This review summarizes the studies on LN metastasis in GICs using proteomic approaches to date.
Collapse
Affiliation(s)
- Vaishali Jain
- Indian Council of Medical Research, National Institute of Pathology, New Delhi 110029, India
- Faculty of Health Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Puja Sakhuja
- Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India
| | - Anil Kumar Agarwal
- Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India
| | - Ravi Sirdeshmukh
- Faculty of Health Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
- Institute of Bioinformatics, International Tech Park, Bangalore 560066, India
| | - Fouzia Siraj
- Indian Council of Medical Research, National Institute of Pathology, New Delhi 110029, India
| | - Poonam Gautam
- Indian Council of Medical Research, National Institute of Pathology, New Delhi 110029, India
| |
Collapse
|
8
|
Fabbi M, Milani MS, Giacopuzzi S, De Werra C, Roviello F, Santangelo C, Galli F, Benevento A, Rausei S. Adherence to Guidelines for Diagnosis, Staging, and Treatment for Gastric Cancer in Italy According to the View of Surgeons and Patients. J Clin Med 2024; 13:4240. [PMID: 39064280 PMCID: PMC11277783 DOI: 10.3390/jcm13144240] [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: 06/25/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Background: Despite the strong declining trends in incidence and mortality over the last decades, gastric cancer (GC) is still burdened with high mortality, even in high-income countries. To improve GC prognosis, several guidelines have been increasingly published with indications about the most appropriate GC management. The Italian Society of Digestive System Pathology (SIPAD) and Gastric Cancer Italian Research Group (GIRCG) designed a survey for both surgeons and patients with the purpose of evaluating the degree of application and adherence to guidelines in GC management in Italy. Materials and Methods: Between January and May 2022, a questionnaire has been administered to a sample of Italian surgeons and, in a simplified version, to members of the Patient Association "Vivere Senza Stomaco" (patients surgically treated for GC between 2008 and 2021) to investigate the diagnosis, staging, and treatment issues. Results: The survey has been completed by 125 surgeons and 125 patients. Abdominal CT with gastric hydro-distension before treatment was not widespread in both groups (47% and 42%, respectively). The rate of surgeons stating that they do not usually perform minimally invasive gastrectomy was 15%, but the rate of patients who underwent a minimally invasive approach was 22% (between 2011 and 2022). The percentage of surgeons declaring to perform extended lymphadenectomy (>D2) was 97%, although a limited lymph node dissection rate was observed in about 35% of patients. Conclusions: This survey shows several important discrepancies from surgical attitudes declared by surgeons and real data derived from the reports available to the patients, suggesting heterogeneous management in clinical practice and, thus, a not rigorous adherence to the guidelines.
Collapse
Affiliation(s)
- Manrica Fabbi
- Department of General Surgery, Cittiglio-Angera Hospital, ASST Settelaghi, 21033 Varese, Italy; (M.S.M.); (S.R.)
| | - Marika Sharmayne Milani
- Department of General Surgery, Cittiglio-Angera Hospital, ASST Settelaghi, 21033 Varese, Italy; (M.S.M.); (S.R.)
| | - Simone Giacopuzzi
- General and Upper GI Surgery Division, Department of Surgery, University of Verona, 37134 Verona, Italy;
| | - Carlo De Werra
- Department of Advanced Biomedical Sciences, Federico II University Hospital, 80131 Naples, Italy;
| | - Franco Roviello
- Department of Medical Surgical Sciences and Neurosciences, Section of General Surgery and Surgical Oncology, Istituto Toscano Tumori (ITT), University Hospital of Siena, University of Siena, 53100 Siena, Italy;
| | | | - Federica Galli
- Department of General Surgery, Gallarate Hospital, ASST Valle Olona, 21013 Gallarate, Italy; (F.G.); (A.B.)
| | - Angelo Benevento
- Department of General Surgery, Gallarate Hospital, ASST Valle Olona, 21013 Gallarate, Italy; (F.G.); (A.B.)
| | - Stefano Rausei
- Department of General Surgery, Cittiglio-Angera Hospital, ASST Settelaghi, 21033 Varese, Italy; (M.S.M.); (S.R.)
| |
Collapse
|
9
|
Chen S, Ding P, Zhao Q. Comparison of the predictive performance of three lymph node staging systems for late-onset gastric cancer patients after surgery. Front Surg 2024; 11:1376702. [PMID: 38919979 PMCID: PMC11196640 DOI: 10.3389/fsurg.2024.1376702] [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: 01/26/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Introduction Lymph node (LN) status is a vital prognostic factor for patients. However, there has been limited focus on predicting the prognosis of patients with late-onset gastric cancer (LOGC). This study aimed to investigate the predictive potential of the log odds of positive lymph nodes (LODDS), lymph node ratio (LNR), and pN stage in assessing the prognosis of patients diagnosed with LOGC. Methods The LOGC data were obtained from the Surveillance, Epidemiology, and End Results database. This study evaluated and compared the predictive performance of three LN staging systems. Univariate and multivariate Cox regression analyses were carried out to identify prognostic factors for overall survival (OS). Three machine learning methods, namely, LASSO, XGBoost, and RF analyses, were subsequently used to identify the optimal LN staging system. A nomogram was built to predict the prognosis of patients with LOGC. The efficacy of the model was demonstrated through receiver operating characteristic (ROC) curve analysis and decision curve analysis. Results A total of 4,743 patients with >16 removed lymph nodes were ultimately included in this investigation. Three LN staging systems demonstrated significant performance in predicting survival outcomes (P < 0.001). The LNR exhibited the most important prognostic ability, as evidenced by the use of three machine learning methods. Utilizing independent factors derived from multivariate Cox regression analysis, a nomogram for OS was constructed. Discussion The calibration, C-index, and AUC revealed their excellent predictive performance. The LNR demonstrated a more powerful performance than other LN staging methods in LOGC patients after surgery. Our novel nomogram exhibited superior clinical feasibility and may assist in patient clinical decision-making.
Collapse
Affiliation(s)
- Sheng Chen
- Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Ping’an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| |
Collapse
|
10
|
Xie Y, Sang Q, Da Q, Niu G, Deng S, Feng H, Chen Y, Li YY, Liu B, Yang Y, Dai W. Improving diagnosis and outcome prediction of gastric cancer via multimodal learning using whole slide pathological images and gene expression. Artif Intell Med 2024; 152:102871. [PMID: 38685169 DOI: 10.1016/j.artmed.2024.102871] [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: 04/25/2023] [Revised: 03/08/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024]
Abstract
For the diagnosis and outcome prediction of gastric cancer (GC), machine learning methods based on whole slide pathological images (WSIs) have shown promising performance and reduced the cost of manual analysis. Nevertheless, accurate prediction of GC outcome may rely on multiple modalities with complementary information, particularly gene expression data. Thus, there is a need to develop multimodal learning methods to enhance prediction performance. In this paper, we collect a dataset from Ruijin Hospital and propose a multimodal learning method for GC diagnosis and outcome prediction, called GaCaMML, which is featured by a cross-modal attention mechanism and Per-Slide training scheme. Additionally, we perform feature attribution analysis via integrated gradient (IG) to identify important input features. The proposed method improves prediction accuracy over the single-modal learning method on three tasks, i.e., survival prediction (by 4.9% on C-index), pathological stage classification (by 11.6% on accuracy), and lymph node classification (by 12.0% on accuracy). Especially, the Per-Slide strategy addresses the issue of a high WSI-to-patient ratio and leads to much better results compared with the Per-Person training scheme. For the interpretable analysis, we find that although WSIs dominate the prediction for most samples, there is still a substantial portion of samples whose prediction highly relies on gene expression information. This study demonstrates the great potential of multimodal learning in GC-related prediction tasks and investigates the contribution of WSIs and gene expression, respectively, which not only shows how the model makes a decision but also provides insights into the association between macroscopic pathological phenotypes and microscopic molecular features.
Collapse
Affiliation(s)
- Yuzhang Xie
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qingqing Sang
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasm, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Da
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Guoshuai Niu
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shijie Deng
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Haoran Feng
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasm, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yunqin Chen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China; Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203, China
| | - Yuan-Yuan Li
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China; Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203, China
| | - Bingya Liu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasm, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yang Yang
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Wentao Dai
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasm, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China; Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203, China.
| |
Collapse
|
11
|
Zhou JW, Zhang YB, Huang ZY, Yuan YP, Jin J. Identification of differentially expressed mRNAs as novel predictive biomarkers for gastric cancer diagnosis and prognosis. World J Gastrointest Oncol 2024; 16:1947-1964. [PMID: 38764850 PMCID: PMC11099425 DOI: 10.4251/wjgo.v16.i5.1947] [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: 12/13/2023] [Revised: 01/04/2024] [Accepted: 03/14/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) has a high mortality rate worldwide. Despite significant progress in GC diagnosis and treatment, the prognosis for affected patients still remains unfavorable. AIM To identify important candidate genes related to the development of GC and identify potential pathogenic mechanisms through comprehensive bioinformatics analysis. METHODS The Gene Expression Omnibus database was used to obtain the GSE183136 dataset, which includes a total of 135 GC samples. The limma package in R software was employed to identify differentially expressed genes (DEGs). Thereafter, enrichment analyses of Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were performed for the gene modules using the clusterProfile package in R software. The protein-protein interaction (PPI) networks of target genes were constructed using STRING and visualized by Cytoscape software. The common hub genes that emerged in the cohort of DEGs that was retrieved from the GEPIA database were then screened using a Venn Diagram. The expression levels of these overlapping genes in stomach adenocarcinoma samples and non-tumor samples and their association with prognosis in GC patients were also obtained from the GEPIA database and Kaplan-Meier curves. Moreover, real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting were performed to determine the mRNA and protein levels of glutamic-pyruvic transaminase (GPT) in GC and normal immortalized cell lines. In addition, cell viability, cell cycle distribution, migration and invasion were evaluated by cell counting kit-8, flow cytometry and transwell assays. Furthermore, we also conducted a retrospective analysis on 70 GC patients diagnosed and surgically treated in Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University between January 2017 to December 2020. The tumor and adjacent normal samples were collected from the patients to determine the potential association between the expression level of GPT and the clinical as well as pathological features of GC patients. RESULTS We selected 19214 genes from the GSE183136 dataset, among which there were 250 downregulated genes and 401 upregulated genes in the tumor samples of stage III-IV in comparison to those in tumor samples of stage I-II with a P-value < 0.05. In addition, GO and KEGG results revealed that the various upregulated DEGs were mainly enriched in plasma membrane and neuroactive ligand-receptor interaction, whereas the downregulated DEGs were primarily enriched in cytosol and pancreatic secretion, vascular smooth muscle contraction and biosynthesis of the different cofactors. Furthermore, PPI networks were constructed based on the various upregulated and downregulated genes, and there were a total 15 upregulated and 10 downregulated hub genes. After a comprehensive analysis, several hub genes, including runt-related transcription factor 2 (RUNX2), salmonella pathogenicity island 1 (SPI1), lysyl oxidase (LOX), fibrillin 1 (FBN1) and GPT, displayed prognostic values. Interestingly, it was observed that GPT was downregulated in GC cells and its upregulation could suppress the malignant phenotypes of GC cells. Furthermore, the expression level of GPT was found to be associated with age, lymph node metastasis, pathological staging and distant metastasis (P < 0.05). CONCLUSION RUNX2, SPI1, LOX, FBN1 and GPT were identified key hub genes in GC by bioinformatics analysis. GPT was significantly associated with the prognosis of GC, and its upregulation can effectively inhibit the proliferative, migrative and invasive capabilities of GC cells.
Collapse
Affiliation(s)
- Jian-Wei Zhou
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| | - Yi-Bing Zhang
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| | - Zhi-Yang Huang
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| | - Yu-Ping Yuan
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| | - Jie Jin
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| |
Collapse
|
12
|
Didehvar DS, Lanza MR, Atherton MJ, Lenz JA. Malignant transformation and subsequent leptomeningeal carcinomatosis of a gastric polyp in a dog. J Vet Intern Med 2024; 38:1744-1750. [PMID: 38587203 PMCID: PMC11099795 DOI: 10.1111/jvim.17072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/27/2024] [Indexed: 04/09/2024] Open
Abstract
Progressive carcinogenesis of a gastric polyp with transformation to gastric adenocarcinoma and subsequent development of leptomeningeal carcinomatosis is described in an adult male Scottish terrier. Presenting clinical signs consisted of vomiting with intermittent hematemesis. Surgical biopsies over the course of 14 months documented the progression from gastric polyp to minimally invasive gastric carcinoma to invasive gastric adenocarcinoma, a pathogenesis not previously documented in veterinary oncology. The patient ultimately developed neurologic pathology and was euthanized, and necropsy evaluation identified widespread carcinomatosis with accompanying leptomeningeal metastasis. As in humans, gastric polyps in dogs rarely have malignant potential.
Collapse
Affiliation(s)
- Dillon S. Didehvar
- Department of Clinical Science & Advanced MedicineSchool of Veterinary Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Matthew R. Lanza
- Department of PathobiologySchool of Veterinary Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Present address:
Department of Comparative Medicine, College of MedicinePennsylvania State UniversityHersheyPennsylvaniaUSA
| | - Matthew J. Atherton
- Department of Clinical Science & Advanced MedicineSchool of Veterinary Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biomedical SciencesSchool of Veterinary Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jennifer A. Lenz
- Department of Clinical Science & Advanced MedicineSchool of Veterinary Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
13
|
Pang S, Wang W, Zhou J, Jiang X, Lin R. Inverse association between age and risk of lymph node metastasis in patients with early gastric cancer: a surveillance, epidemiology, and end results analysis. J Cancer 2024; 15:2829-2836. [PMID: 38577611 PMCID: PMC10988299 DOI: 10.7150/jca.94542] [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: 01/22/2024] [Accepted: 03/13/2024] [Indexed: 04/06/2024] Open
Abstract
Background: Recent studies have shown that young patients with gastric cancer are at a more advanced stage and have poor survival, but the cause is still unclear. The prognosis of gastric cancer is closely related to LNM, but the relationship between age and LNM in early gastric cancer (EGC) is currently unclear. Therefore, we aimed to study the relationship between age and the risk of LNM in EGC. Materials and Methods: We screened out patients with EGC who underwent surgery from the SEER research database from 1975 to 2016, and retrospectively analyzed the proportion of LNM in different age groups. We grouped age into 18-39, 40-49, 50-59, 60-69, 70-79, and ≥ 80 years old, and used univariate analysis and multivariate logistic regression to analyze the correlation between age and LNM. Results: We included 9231 patients with EGC, with LNM rates of 20.3%, 23.3%, 21.0%, 19.8%, 18.1%, and 13.2% in the age groups of 18-39 years old (2.3%), 40-49 (6.1%), 50-59 years old (15.7%), 60-69 years old (24.8%), 70-79 years old (27.2%) and ≥80 years old (23.9%), respectively. We found that when older than 39 years old, the risk of LNM and postoperative survival time of EGC patients decrease (p<0.001). Multivariate analysis results showed that age, tumor size, the number of retrieved lymph nodes (rN), tumor grade, and tumor location were related to LNM. Conclusions: This study found that age in patients with EGC was inversely related to the risk of LNM, and positively correlated with postoperative survival. For older patients with EGC, endoscopic treatment is more appropriate. For young patients with EGC, LNM should be considered when choosing endoscopic treatment.
Collapse
Affiliation(s)
| | | | | | | | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
14
|
Wu M, Yang X, Liu Y, Han F, Li X, Wang J, Guo D, Tang X, Lin L, Liu C. Development and validation of a deep learning model for predicting postoperative survival of patients with gastric cancer. BMC Public Health 2024; 24:723. [PMID: 38448849 PMCID: PMC10916254 DOI: 10.1186/s12889-024-18221-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Deep learning (DL), a specialized form of machine learning (ML), is valuable for forecasting survival in various diseases. Its clinical applicability in real-world patients with gastric cancer (GC) has yet to be extensively validated. METHODS A combined cohort of 11,414 GC patients from the Surveillance, Epidemiology and End Results (SEER) database and 2,846 patients from a Chinese dataset were utilized. The internal validation of different algorithms, including DL model, traditional ML models, and American Joint Committee on Cancer (AJCC) stage model, was conducted by training and testing sets on the SEER database, followed by external validation on the Chinese dataset. The performance of the algorithms was assessed using the area under the receiver operating characteristic curve, decision curve, and calibration curve. RESULTS DL model demonstrated superior performance in terms of the area under the curve (AUC) at 1, 3, and, 5 years post-surgery across both datasets, surpassing other ML models and AJCC stage model, with AUCs of 0.77, 0.80, and 0.82 in the SEER dataset and 0.77, 0.76, and 0.75 in the Chinese dataset, respectively. Furthermore, decision curve analysis revealed that the DL model yielded greater net gains at 3 years than other ML models and AJCC stage model, and calibration plots at 3 years indicated a favorable level of consistency between the ML and actual observations during external validation. CONCLUSIONS DL-based model was established to accurately predict the survival rate of postoperative patients with GC.
Collapse
Affiliation(s)
- Mengjie Wu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xiaofan Yang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yuxi Liu
- Department of Medical Records, Office for DRGs (Diagnosis Related Groups), Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127 Dongming Rd, PO Box 0061, Zhengzhou, Henan Province, 450008, China
| | - Feng Han
- Department of Medical Records, Office for DRGs (Diagnosis Related Groups), Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127 Dongming Rd, PO Box 0061, Zhengzhou, Henan Province, 450008, China
| | - Xi Li
- Department of Medical Records, Office for DRGs (Diagnosis Related Groups), Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127 Dongming Rd, PO Box 0061, Zhengzhou, Henan Province, 450008, China
| | - Jufeng Wang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Dandan Guo
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiance Tang
- Department of Medical Records, Office for DRGs (Diagnosis Related Groups), Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127 Dongming Rd, PO Box 0061, Zhengzhou, Henan Province, 450008, China
| | - Lu Lin
- Translational Medicine Research Center, People's Hospital of Henan University of Chinese Medicine, Zhengzhou People's Hospital, Zhengzhou, Henan, 450003, China
| | - Changpeng Liu
- Department of Medical Records, Office for DRGs (Diagnosis Related Groups), Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127 Dongming Rd, PO Box 0061, Zhengzhou, Henan Province, 450008, China.
| |
Collapse
|
15
|
Sadeghi R, Taheri R, Jangjoo A, Pakdel A, Arjmand MH, Motiei MR, Memar B, Aliakbarian M. Evaluation of the diagnostic value of Sentinel Lymph Node in patients with gastric adenocarcinoma. ASIA OCEANIA JOURNAL OF NUCLEAR MEDICINE & BIOLOGY 2024; 12:21-26. [PMID: 38164234 PMCID: PMC10757058 DOI: 10.22038/aojnmb.2023.70461.1491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 01/03/2024]
Abstract
Objectives Sentinel lymph node biopsy (SLNB) has been proven as a safe and efficient procedure in some cancers like breast cancer and melanoma with a reduction of complications and side effects of unnecessary lymphadenectomy in many patients. However, the diagnostic value of SLNB in gastric cancer is a point of debate. This study evaluated the diagnostic value of SLNB using radiotracer and isosulphan blue dye injection in patients with Gastric Adenocarcinomas (GA). Methods This descriptive study was performed at Imam-Reza HOSPITAL on 39 patients diagnosed with GA with no lymphatic metastasis using two methods: the combination of radionuclide with isosulphan together (R&I) method compared with the isosulphan alone method. Lymphatic dissection was performed in all patients. The pathological results were compared between the sentinel lymph nodes (SLN) and other lymph nodes and their accordance rate was calculated. Results In the T1 group, the sentinel lymph node biopsy detection rate was 100% for the combination of the R&I method and 60% for the isosulphan method and the false negative rate was zero. These values respectively were 88.8% and 88.8% in the T2 group with a false negative rate of 75%. In the T3 group, the values were 100% for the combination of the R&I method and 93.7% for the isosulphan method with a false negative rate of 40%. In the combination of the R&I method, the sensitivity, specificity, and positive and negative predictive values were 57.9, 100, 100, and 69.2 percent respectively. Conclusion Based on the false negative rate (47.4%), SLNB by injection of isosulphan blue dye alone is not a diagnostic enough value for predicting lymph node metastasis in GA. Although, SLNB by combination of the R&I had better accuracy compared to the isosulphan alone, more studies with larger samples are needed to prove this result.
Collapse
Affiliation(s)
- Ramin Sadeghi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Taheri
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Jangjoo
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Akbar Pakdel
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad-Hassan Arjmand
- Transplant Research Center, Clinical research institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Motiei
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahram Memar
- Department of Pathology, Emam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Aliakbarian
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Transplant Research Center, Clinical research institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Lu C, Xie L, Qiu S, Jiang T, Wang L, Chen Z, Xia Y, Lv J, Li Y, Li B, Gu C, Xu Z. Small Extracellular Vesicles Derived from Helicobacter Pylori-Infected Gastric Cancer Cells Induce Lymphangiogenesis and Lymphatic Remodeling via Transfer of miR-1246. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2308688. [PMID: 37946695 DOI: 10.1002/smll.202308688] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Indexed: 11/12/2023]
Abstract
Lymph node metastasis (LNM) is a significant barrier to the prognosis of patients with gastric cancer (GC). Helicobacter pylori (H. pylori)-positive GC patients experience a higher rate of LNM than H. pylori-negative GC patients. However, the underlying mechanism remains unclear. Based on the findings of this study, H. pylori-positive GC patients have greater lymphangiogenesis and lymph node immunosuppression than H. pylori-negative GC patients. In addition, miR-1246 is overexpressed in the plasma small extracellular vesicles (sEVs) of H. pylori-positive GC patients, indicating a poor prognosis. Functionally, sEVs derived from GC cells infected with H. pylori deliver miR-1246 to lymphatic endothelial cells (LECs) and promote lymphangiogenesis and lymphatic remodeling. Mechanistically, miR-1246 suppresses GSK3β expression and promotes β-Catenin and downstream MMP7 expression in LECs. miR-1246 also stabilizes programmed death ligand-1 (PD-L1) by suppressing GSK3β and induces the apoptosis of CD8+ T cells. Overall, miR-1246 in plasma sEVs may be a novel biomarker and therapeutic target in GC-LNM.
Collapse
Affiliation(s)
- Chen Lu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Li Xie
- Department of General Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Shengkui Qiu
- Department of General Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
| | - Tianlu Jiang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Luyao Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Zetian Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yiwen Xia
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Jialun Lv
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Ying Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Chao Gu
- Department of General Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215000, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| |
Collapse
|
17
|
Mahenthiran AK, Logan CD, Janczewski LM, Valukas C, Warwar S, Silver CM, Feinglass J, Merkow RP, Bentrem DJ, Odell DD. Evaluation of Nationwide Trends in Nodal Sampling Guideline Adherence for Gastric Cancer: 2005-2017. J Surg Res 2023; 291:514-526. [PMID: 37540969 PMCID: PMC10529819 DOI: 10.1016/j.jss.2023.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/15/2023] [Accepted: 07/04/2023] [Indexed: 08/06/2023]
Abstract
INTRODUCTION Surgical resection is the primary curative treatment for localized gastric cancer. A multitude of research supports surgical nodal sampling guidelines. Though there are known disparities in adherence to nodal sampling, it is unclear how hospital program-level disparities have changed over time. The purpose of this study is to evaluate trends in program-level disparities in adherence to gastric cancer nodal sampling guidelines. METHODS Patients who underwent resection of gastric cancer from 2005 to 2017 were identified in the National Cancer Database. Patients treated at academic programs were compared to those treated at nonacademic programs, and rates and trends of adherence to nodal sampling guidelines (defined as ≥15 lymph nodes) were determined. Adjusted multivariable analysis was used to determine likelihood of nodal sampling adherence while controlling for sociodemographic, clinical, hospital, and travel distance characteristics. RESULTS A total of 55,421 patients were included with 27,201 (49.1%) of patients meeting adherence criteria for lymph node sampling. Academic programs treated 44.4% of the total cohort. Overall, lymph node sampling criteria were met in 59.2% of patients treated at high-volume academic programs and 37.0% of patients treated at low-volume nonacademic programs (incidence rate ratios 0.67, 95% confidence interval 0.63-0.72 versus high-volume academic programs). Adherence rates improved from 2005 to 2017 for both low-volume nonacademic programs (27.8% in 2005 to 50.1% in 2017) and high-volume academic programs (46.0% in 2005 to 69.8% in 2017, P < 0.001). CONCLUSIONS Though adherence rates have improved from 2005 to 2017, high-volume academic programs were more likely to adhere to lymph node sampling guidelines for gastric cancer.
Collapse
Affiliation(s)
- Ashorne K Mahenthiran
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Charles D Logan
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Surgery, Canning Thoracic Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Lauren M Janczewski
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Catherine Valukas
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Samantha Warwar
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Casey M Silver
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joe Feinglass
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ryan P Merkow
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David J Bentrem
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David D Odell
- Department of Surgery, Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Surgery, Canning Thoracic Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
18
|
Wen JY, Li X, Chen JN, Chen J, Zhang JY, Du Y, Zhu WH, Chen YJ, Yang RH, Shao CK. CD45 - erythroid progenitor cells promote lymph node metastasis in gastric cancer by inducing a hybrid epithelial/mesenchymal state in lymphatic endothelial cells. Gastric Cancer 2023; 26:918-933. [PMID: 37676622 DOI: 10.1007/s10120-023-01425-x] [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: 02/15/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND AND AIMS Specific mechanisms of lymph node (LN) metastasis in early-stage gastric cancer (GC) have not been elucidated. The role of anemia, a vital clinical feature of GC, in LN metastasis is also unclear. Since the number of erythroid progenitor cells (EPCs) is increased in chronic anemia, we investigated its association with LN metastasis in GC. METHODS Flow cytometry and immunofluorescence analyses were performed to sort and study EPCs from the circulation and tumors of patients with stage I-III GC. The effect of these EPCs on the activation of T and B cells and on the functions of lymphatic endothelial cells (LECs) was determined, and their ability to promote LN metastasis was evaluated using a footpad-popliteal LN metastasis model based on two human adenocarcinoma GC cell lines in nude mice. The prognostic value of EPCs was also analyzed. RESULTS The proportion of CD45- EPCs was higher in the mononuclear cells in the circulation, tumors, and LNs of GC patients with LN metastasis (N+) than in those of GC patients without LN metastasis (N0). In N+ patients, CD45- EPCs were more abundant in metastatic LNs than in non-metastatic LNs. Lymphatic vessel endothelial hyaluronan receptor 1 immunoreactivity in tumors revealed that CD45- EPCs were positively associated with nodal stages and lymph vessel density. Furthermore, CD45- EPCs increased LEC proliferation and migration through their S100A8/A9 heterodimer-induced hybrid epithelial/mesenchymal (E/M) state; however, they did not influence the invasion and tubulogenesis of LECs or T and B cell proliferation. CD45- EPCs promoted LN metastasis in vivo; the S100A8/A9 heterodimer mimicked this phenomenon. Finally, CD45- EPCs predicted the overall and disease-free survival of stage I-III GC patients after radical resection. CONCLUSIONS The CD45- EPCs accumulated in GC tissues and metastatic LNs and promoted LN metastasis via the S100A8/9-induced hybrid E/M state of LECs, which was the specific mechanism of LN metastasis in the early stages of GC.
Collapse
Affiliation(s)
- Jing-Yun Wen
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Xing Li
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Jian-Ning Chen
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Jie Chen
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Jing-Yue Zhang
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Yu Du
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Wei-Hang Zhu
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Yong-Jian Chen
- Department of Medical Oncology and Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Ri-Hong Yang
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Chun-Kui Shao
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
19
|
Zhu KG, Yang J, Zhu Y, Zhu Q, Pan W, Deng S, He Y, Zuo D, Wang P, Han Y, Zhang HY. The microprotein encoded by exosomal lncAKR1C2 promotes gastric cancer lymph node metastasis by regulating fatty acid metabolism. Cell Death Dis 2023; 14:708. [PMID: 37903800 PMCID: PMC10616111 DOI: 10.1038/s41419-023-06220-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/01/2023]
Abstract
Lymph node metastasis (LNM) is the prominent route of gastric cancer dissemination, and usually leads to tumor progression and a dismal prognosis of gastric cancer. Although exosomal lncRNAs have been reported to be involved in tumor development, whether secreted lncRNAs can encode peptides in recipient cells remains unknown. Here, we identified an exosomal lncRNA (lncAKR1C2) that was clinically correlated with lymph node metastasis in gastric cancer in a VEGFC-independent manner. Exo-lncAKR1C2 secreted from gastric cancer cells was demonstrated to enhance tube formation and migration of lymphatic endothelial cells, and facilitate lymphangiogenesis and lymphatic metastasis in vivo. By comparing the metabolic characteristics of LN metastases and primary focuses, we found that LN metastases of gastric cancer displayed higher lipid metabolic activity. Moreover, exo-lncAKR1C2 encodes a microprotein (pep-AKR1C2) in lymphatic endothelial cells and promotes CPT1A expression by regulating YAP phosphorylation, leading to enhanced fatty acid oxidation (FAO) and ATP production. These findings highlight a novel mechanism of LNM and suggest that the microprotein encoded by exosomal lncAKR1C2 serves as a therapeutic target for advanced gastric cancer.
Collapse
Affiliation(s)
- Ke-Gan Zhu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Jiayu Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Yuehong Zhu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Qihang Zhu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Wen Pan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Siyu Deng
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yi He
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Duo Zuo
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Peiyun Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Yueting Han
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Hai-Yang Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China.
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
20
|
Baldari L, Boni L, Cassinotti E. Lymph node mapping with ICG near-infrared fluorescence imaging: technique and results. MINIM INVASIV THER 2023; 32:213-221. [PMID: 37261486 DOI: 10.1080/13645706.2023.2217916] [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: 01/16/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023]
Abstract
PURPOSE Lymphadenectomy represents a fundamental step during gastrointestinal cancer resection, as the removal of an adequate number of lymph nodes is crucial to define the stage of the disease and prognosis. Lymphadenectomy during gastric and colorectal resection and adrenalectomy for cancer are technically demanding and can be associated with risk of bleeding. To date, lymphadenectomy is often performed without any visual aid. Indocyanine green fluorescence for lymph node mapping can provide better intraoperative visualization. The purpose of this review is to report the current evidence on this topic. MATERIALS AND METHODS A systematic research of the electronic databases Medline, Embase and Google Scholar was conducted from the inception to December 2022. RESULTS This review summarizes the current evidence of techniques and results of fluorescence guided lymphatic mapping during gastrointestinal and adrenal surgery. CONCLUSION According to this review, ICG guided lymphadenectomy for gastrointestinal tumours and adrenocortical carcinoma is feasible and safe. In gastrointestinal tumours it allows higher number of harvested lymph nodes.
Collapse
Affiliation(s)
- Ludovica Baldari
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luigi Boni
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Cassinotti
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
21
|
Abate M, Walch H, Arora K, Vanderbilt CM, Fei T, Drebin H, Shimada S, Maio A, Kemel Y, Stadler ZK, Schmeltz J, Sihag S, Ku GY, Gu P, Tang L, Vardhana S, Berger MF, Brennan MF, Schultz ND, Strong VE. Unique Genomic Alterations and Microbial Profiles Identified in Patients With Gastric Cancer of African, European, and Asian Ancestry: A Novel Path for Precision Oncology. Ann Surg 2023; 278:506-518. [PMID: 37436885 PMCID: PMC10527605 DOI: 10.1097/sla.0000000000005970] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
OBJECTIVE Here, we characterize differences in the genetic and microbial profiles of GC in patients of African (AFR), European, and Asian ancestry. BACKGROUND Gastric cancer (GC) is a heterogeneous disease with clinicopathologic variations due to a complex interplay of environmental and biological factors, which may affect disparities in oncologic outcomes.. METHODS We identified 1042 patients with GC with next-generation sequencing data from an institutional Integrated Mutation Profiling of Actionable Cancer Targets assay and the Cancer Genomic Atlas group. Genetic ancestry was inferred from markers captured by the Integrated Mutation Profiling of Actionable Cancer Targets and the Cancer Genomic Atlas whole exome sequencing panels. Tumor microbial profiles were inferred from sequencing data using a validated microbiome bioinformatics pipeline. Genomic alterations and microbial profiles were compared among patients with GC of different ancestries. RESULTS We assessed 8023 genomic alterations. The most frequently altered genes were TP53 , ARID1A , KRAS , ERBB2 , and CDH1 . Patients of AFR ancestry had a significantly higher rate of CCNE1 alterations and a lower rate of KRAS alterations ( P < 0.05), and patients of East Asian ancestry had a significantly lower rate of PI3K pathway alterations ( P < 0.05) compared with other ancestries. Microbial diversity and enrichment did not differ significantly across ancestry groups ( P > 0.05). CONCLUSIONS Distinct patterns of genomic alterations and variations in microbial profiles were identified in patients with GC of AFR, European, and Asian ancestry. Our findings of variation in the prevalence of clinically actionable tumor alterations among ancestry groups suggest that precision medicine can mitigate oncologic disparities.
Collapse
Affiliation(s)
- Miseker Abate
- Department of Surgery, Memorial Sloan Kettering Cancer Center (MSK), New York, NY
- Human Oncology and Pathogenesis Program, MSK
- Department of Surgery, Weill Cornell Medicine
| | - Henry Walch
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, MSK
| | - Kanika Arora
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, MSK
| | | | - Teng Fei
- Department of Epidemiology and Biostatistics, MSK
| | - Harrison Drebin
- Department of Surgery, Memorial Sloan Kettering Cancer Center (MSK), New York, NY
- Human Oncology and Pathogenesis Program, MSK
| | - Shoji Shimada
- Department of Surgery, Memorial Sloan Kettering Cancer Center (MSK), New York, NY
- Human Oncology and Pathogenesis Program, MSK
| | - Anna Maio
- Niehaus Center of Inherited Cancer Genomics, MSK
| | - Yelena Kemel
- Niehaus Center of Inherited Cancer Genomics, MSK
| | - Zsofia K. Stadler
- Niehaus Center of Inherited Cancer Genomics, MSK
- Department of Medicine, MSK
- Department of Medicine, Weill Cornell Medicine
| | | | - Smita Sihag
- Department of Surgery, Memorial Sloan Kettering Cancer Center (MSK), New York, NY
- Department of Surgery, Weill Cornell Medicine
| | - Geoffrey Y. Ku
- Department of Medicine, MSK
- Department of Medicine, Weill Cornell Medicine
| | | | - Laura Tang
- Department of Pathology and Laboratory Medicine, MSK
- Department of Pathology and Laboratory Medicine, WCM
| | - Santosha Vardhana
- Human Oncology and Pathogenesis Program, MSK
- Department of Medicine, MSK
- Department of Medicine, Weill Cornell Medicine
| | - Michael F. Berger
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, MSK
- Department of Pathology and Laboratory Medicine, MSK
- Department of Pathology and Laboratory Medicine, WCM
| | - Murray F. Brennan
- Department of Surgery, Memorial Sloan Kettering Cancer Center (MSK), New York, NY
- Department of Surgery, Weill Cornell Medicine
| | | | - Vivian E. Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center (MSK), New York, NY
- Department of Surgery, Weill Cornell Medicine
| |
Collapse
|
22
|
Xiao J, Wang G, Zhu C, Liu K, Wang Y, Shen K, Fan H, Ma X, Xu Z, Yang L. A thirty-three gene-based signature predicts lymph node metastasis and prognosis in patients with gastric cancer. Heliyon 2023; 9:e17017. [PMID: 37484383 PMCID: PMC10361117 DOI: 10.1016/j.heliyon.2023.e17017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 07/25/2023] Open
Abstract
Recently, several studies have indicated the great potential of gene expression signature of the primary tumor in predicting lymph node metastasis; however, few current gene biomarkers can predict lymph node status and prognosis in gastric cancer (GC). Thus, we used the RNA-seq data from The Cancer Genome Atlas (TCGA) to identify differentially expressed genes between pathological lymph node-negative (pN0) and positive (pN+) patients and to establish a gene signature that could predict lymph node metastasis. Meanwhile, the robustness of identified gene signatures was validated in an independent dataset Asian Cancer Research Group (n = 300). In this study, our thirty-three gene-based signature was highly correlated with lymph node metastasis and could successfully discriminate pN + patients in the training set (Area under the receiver operating characteristic curve = 0.951). Moreover, Disease-free survival (P = 0.0029) and overall survival (P = 0.026) were significantly worse in high-risk compared with low-risk patients overall and when confined to pN0 patients only (P < 0.0001). Of note, this gene signature also proved useful in predicting lymph node status and survival in the validation cohort. The present study suggests a thirty-three gene-based signature that could effectively predict lymph node metastasis and prognosis in GC.
Collapse
Affiliation(s)
- Jian Xiao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Gang Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chuming Zhu
- Department of General Surgery, Liyang People's Hospital, Liyang Branch Hospital of Jiangsu Province Hospital, Liyang, Jiangsu Province, China
| | - Kanghui Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yuanhang Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kuan Shen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hao Fan
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiang Ma
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of General Surgery, Liyang People's Hospital, Liyang Branch Hospital of Jiangsu Province Hospital, Liyang, Jiangsu Province, China
| |
Collapse
|
23
|
Marano L, Carbone L, Poto GE, Restaino V, Piccioni SA, Verre L, Roviello F, Marrelli D. Extended Lymphadenectomy for Gastric Cancer in the Neoadjuvant Era: Current Status, Clinical Implications and Contentious Issues. Curr Oncol 2023; 30:875-896. [PMID: 36661716 PMCID: PMC9858164 DOI: 10.3390/curroncol30010067] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Despite its decreasing incidence, gastric cancer remains an important global healthcare problem due to its overall high prevalence and high mortality rate. Since the MAGIC and FNLCC/FFCD trials, the neoadjuvant chemotherapy has been recommended throughout Europe in gastric cancer. Potential benefits of preoperative treatments include a higher rate of R0 resection achieved by downstaging the primary tumor, a likely effect on micrometastases and isolated tumor cells in the lymph nodes, and, as a result, improved cancer-related survival. Nevertheless, distortion of anatomical planes of dissection, interstitial fibrosis, and sclerotic tissue changes may increase surgical difficulty. The collection of at least twenty-five lymph nodes after neoadjuvant therapy would seem to ensure removal of undetectable node metastasis and reduce the likelihood of locoregional recurrence. It is not what you take but what you leave behind that defines survival. Therefore, para-aortic lymph node dissection is safe and effective after neoadjuvant chemotherapy, in both therapeutic and prophylactic settings. In this review, the efficacy of adequate lymph node dissection, also in a neoadjuvant setting, has been investigated in the key studies conducted to date on the topic.
Collapse
Affiliation(s)
| | - Ludovico Carbone
- Unit of Surgical Oncology, Department of Medicine Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | | | | | | | | | | | | |
Collapse
|
24
|
The Tumor Microenvironment in Tumorigenesis and Therapy Resistance Revisited. Cancers (Basel) 2023; 15:cancers15020376. [PMID: 36672326 PMCID: PMC9856874 DOI: 10.3390/cancers15020376] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Tumorigenesis is a complex and dynamic process involving cell-cell and cell-extracellular matrix (ECM) interactions that allow tumor cell growth, drug resistance and metastasis. This review provides an updated summary of the role played by the tumor microenvironment (TME) components and hypoxia in tumorigenesis, and highlight various ways through which tumor cells reprogram normal cells into phenotypes that are pro-tumorigenic, including cancer associated- fibroblasts, -macrophages and -endothelial cells. Tumor cells secrete numerous factors leading to the transformation of a previously anti-tumorigenic environment into a pro-tumorigenic environment. Once formed, solid tumors continue to interact with various stromal cells, including local and infiltrating fibroblasts, macrophages, mesenchymal stem cells, endothelial cells, pericytes, and secreted factors and the ECM within the tumor microenvironment (TME). The TME is key to tumorigenesis, drug response and treatment outcome. Importantly, stromal cells and secreted factors can initially be anti-tumorigenic, but over time promote tumorigenesis and induce therapy resistance. To counter hypoxia, increased angiogenesis leads to the formation of new vascular networks in order to actively promote and sustain tumor growth via the supply of oxygen and nutrients, whilst removing metabolic waste. Angiogenic vascular network formation aid in tumor cell metastatic dissemination. Successful tumor treatment and novel drug development require the identification and therapeutic targeting of pro-tumorigenic components of the TME including cancer-associated- fibroblasts (CAFs) and -macrophages (CAMs), hypoxia, blocking ECM-receptor interactions, in addition to the targeting of tumor cells. The reprogramming of stromal cells and the immune response to be anti-tumorigenic is key to therapeutic success. Lastly, this review highlights potential TME- and hypoxia-centered therapies under investigation.
Collapse
|
25
|
Omeroglu S, Gulmez S, Yazici P, Demir U, Guven O, Capkinoglu E, Uzun O, Senger AS, Polat E, Duman M. Clinical significance of the largest histopathological metastatic lymph node size for postoperative course of patients undergoing surgery for gastric cancer. Front Surg 2023; 10:1105189. [PMID: 36874461 PMCID: PMC9982115 DOI: 10.3389/fsurg.2023.1105189] [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: 11/23/2022] [Accepted: 01/31/2023] [Indexed: 02/19/2023] Open
Abstract
Aim The aim of this study was to investigate the effect of the largest metastatic lymph node (MLN) size on postoperative outcomes of patients with stage II-III gastric cancer (GC). Methods A total of 163 patients with stage II/III GC who underwent curative surgery were included in this single-center retrospective study. The lymph nodes were counted, each lymph node was analyzed for metastatic involvement by histopathological examination, and the diameter of the largest metastatic lymph node was recorded. The severity of postoperative complications was assessed by Clavien-Dindo classification system. Two groups of 163 patients were defined according to ROC analysis with cut-off value of histopathologically maximum MLN diameter. A comparative analysis of demographic and clinicopathological characteristics of the patients and their postoperative outcomes were performed. Results The median hospital stay was significantly longer in patients with major complications compared to patients without major complications [18 days (IQR: 13-24) vs. 8 days (IQR: 7-11); (p < 0.001)]. The median MLN size was significantly larger in deceased patients compared to survived [1.3 cm (IQR: 0.8-1.6) vs. 0.9 cm (IQR: 0.6-1.2), respectively; (p < 0.001)]. The cut-off value of MLN size predicting mortality was found as 1.05 cm. MLN size ≥1.05 cm had nearly 3.5 times more negative impact on survival. Conclusions The largest metastatic lymph node size had a significant association with survival outcomes. Particularly, MLN size over 1.05 cm was associated with worse survival outcomes. However, the largest MLN was not shown to have any effect on major complications. Further prospective and large-scale studies are required to draw more precise conclusions.
Collapse
Affiliation(s)
- Sinan Omeroglu
- Department of General Surgery, University of Health Sciences Sisli Hamidiye Etfal Research and Training Hospital, Istanbul, Türkiye
| | - Selcuk Gulmez
- Department of Gastrointestinal Surgery, University of Health Sciences Kosuyolu High Specialization Education and Research Hospital, Istanbul, Türkiye
| | - Pinar Yazici
- Department of General Surgery, University of Health Sciences Sisli Hamidiye Etfal Research and Training Hospital, Istanbul, Türkiye
| | - Uygar Demir
- Department of General Surgery, University of Health Sciences Sisli Hamidiye Etfal Research and Training Hospital, Istanbul, Türkiye
| | - Onur Guven
- Department of General Surgery, University of Health Sciences Sisli Hamidiye Etfal Research and Training Hospital, Istanbul, Türkiye
| | - Emir Capkinoglu
- Department of General Surgery, University of Health Sciences Sisli Hamidiye Etfal Research and Training Hospital, Istanbul, Türkiye
| | - Orhan Uzun
- Department of Gastrointestinal Surgery, University of Health Sciences Kosuyolu High Specialization Education and Research Hospital, Istanbul, Türkiye
| | - Aziz Serkan Senger
- Department of Gastrointestinal Surgery, University of Health Sciences Kosuyolu High Specialization Education and Research Hospital, Istanbul, Türkiye
| | - Erdal Polat
- Department of Gastrointestinal Surgery, University of Health Sciences Kosuyolu High Specialization Education and Research Hospital, Istanbul, Türkiye
| | - Mustafa Duman
- Department of Gastrointestinal Surgery, University of Health Sciences Kosuyolu High Specialization Education and Research Hospital, Istanbul, Türkiye
| |
Collapse
|
26
|
Qiao W, Sha S, Song J, Chen Y, Lian G, Wang J, Zhou X, Peng L, Li L, Tian F, Jing C. Association between multiple coagulation-related factors and lymph node metastasis in patients with gastric cancer: A retrospective cohort study. Front Oncol 2023; 13:1099857. [PMID: 36910598 PMCID: PMC9996287 DOI: 10.3389/fonc.2023.1099857] [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: 11/16/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023] Open
Abstract
Background Patients with tumors generally present with accompanying activation of the coagulation system, which may be related to tumor stage. To our knowledge, few studies have examined the activation of the coagulation system in reference to lymph node metastasis within gastric cancer. This study aimed to investigate the correlation between multiple coagulation-related factors and lymph node metastasis in patients with gastric cancer after excluding the influence of tumor T stage. Materials and methods We retrospectively evaluated the relationship between lymph node metastasis and coagulation-related factors in 516 patients with T4a stage gastric cancer. We further analyzed influencing factors for lymph node metastasis and verified the predictive value of maximum amplitude (MA, a parameter of thromboelastography which is widely used to assess the strength of platelet-fibrinogen interaction in forming clots) in reference to lymph node metastasis. Results Platelet counts (P=0.011), fibrinogen levels (P=0.002) and MA values (P=0.006) were statistically significantly higher in patients with T4a stage gastric cancer presenting with lymph node metastasis than in those without lymph node metastasis. Moreover, tumor N stage was statistically significantly and positively correlated with platelet count (P<0.001), fibrinogen level (P=0.003), MA value (P<0.001), and D-dimer level (P=0.010). The MA value was an independent factor for lymph node metastasis (β=0.098, 95% CI: 1.020-1.193, P=0.014) and tumor N stage (β=0.059, 95% CI: 0.015-0.104, P=0.009), and could be used to predict the presence of lymph node metastasis in patients with gastric cancer (sensitivity 0.477, specificity 0.783, P=0.006). The independent influencing factors for MA value mainly included platelet levels, fibrinogen levels, D-dimer and hemoglobin levels; we found no statistically significant correlations with tumor diameter, tumor area, and other evaluated factors. Conclusion We conclude that MA value is an independent influencing factor for lymph node metastasis and tumor N stage in patients with T4a stage gastric cancer. The MA value has important value in predicting the presence or absence of lymph node metastasis in patients with gastric cancer. Clinical trial registration http://www.chictr.org.cn, identifier ChiCTR2200064936.
Collapse
Affiliation(s)
- Wenhao Qiao
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Shengxu Sha
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Jiyuan Song
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yuezhi Chen
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.,Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Guodong Lian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.,Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Junke Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xinxiu Zhou
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lipan Peng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.,Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.,Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Feng Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.,Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.,Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
27
|
Lu X, Fu Y, Gu L, Zhang Y, Liao AY, Wang T, Jia B, Zhou D, Liao L. Integrated proteome and phosphoproteome analysis of gastric adenocarcinoma reveals molecular signatures capable of stratifying patient outcome. Mol Oncol 2022; 17:261-283. [PMID: 36520032 PMCID: PMC9892830 DOI: 10.1002/1878-0261.13361] [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: 05/26/2022] [Revised: 11/04/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Metastasis is one of the main causes of low survival rate of gastric cancer patients. Exploring key proteins in the progression of gastric adenocarcinoma (GAC) may provide new candidates for prognostic biomarker development and therapeutic intervention. We applied quantitative mass spectrometry to compare the proteome and phosphoproteome of primary tumor tissues between GAC patients with and without lymph node metastasis (LNM). We then performed an integrated analysis of the proteomic and transcriptomic data to reveal the molecular features. We quantified a total of 5536 proteins, and we found 218 upregulated and 49 downregulated proteins in tumor samples from patients with LNM compared to those without LNM. Clustering analysis identified a number of hub proteins that have been previously shown to play important roles in gastric cancer progression. We also found that two extracellular proteins, TNXB and SPON1, are overexpressed in patients with LNM, which correlates with poor survival of GAC patients. Overexpression of TNXB and SPON1 was validated by western blotting and immunohistochemistry. Furthermore, treating gastric cancer cells with anti-TNXB antibody significantly reduced cell migration. Finally, quantitative phosphoproteomic analysis combined with activity-based kinase capture revealed a number of activated kinases in primary tumor tissues from patients with LNM, among which GSK3 might be a new target that warrants further study. Our study provides a snapshot of the proteome and phosphoproteome of GAC tumor tissues that have metastatic potential, and identifies potential biomarkers for GAC progression.
Collapse
Affiliation(s)
- Xue Lu
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Yunyun Fu
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Lei Gu
- Department of General Surgery, Shanghai Tenth People's Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yunpeng Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesEast China Normal UniversityShanghaiChina
| | | | | | - Bin Jia
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityChina
| | - Donglei Zhou
- Department of Gastric SurgeryFudan University Shanghai Cancer CenterChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesEast China Normal UniversityShanghaiChina
| |
Collapse
|
28
|
Xiao J, Shen K, Liu K, Wang Y, Fan H, Cheng Q, Zhou X, Hu L, Wang G, Xu Z, Yang L. Obesity promotes lipid accumulation in lymph node metastasis of gastric cancer: a retrospective case‒control study. Lipids Health Dis 2022; 21:123. [PMID: 36397145 PMCID: PMC9673345 DOI: 10.1186/s12944-022-01734-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022] Open
Abstract
Background The connection between obesity, lipid accumulation, and lymph node metastasis (LNM) in gastric cancer (GC) is unclear. Methods The association of body mass index (BMI) and serum lipid levels with LNM was measured by calculating the odds ratio (OR) and 95% confidence interval (CI) in 1,058 eligible GC patients with a mean age of 61.4 years. Meanwhile, differentially expressed genes (DEGs) were identified between lymph node metastasis-positive (N +) and -negative (N0) groups using public RNA-seq data. Neutral lipids in human GC samples were detected by Oil red O staining. The expression of cluster of differentiation 36 (CD36), fatty acid synthase (FASN), and lipoprotein lipase (LPL) was detected by immunohistochemistry (IHC) and quantitative real-time PCR. Results Compared with normal-weight patients, overweight (OR = 2.02, 95% CI = 1.26–3.23) and obese (OR = 1.83, 95% CI = 1.15–2.91) patients showed increased ORs for LNM. However, no significant results were obtained for serum lipids in the multivariable-adjusted model (P > 0.05). Subgroup analysis suggested that increased low-density lipoprotein cholesterol was a risk factor in females (OR = 1.27, 95% CI = 1.02–1.59). Functional enrichment analysis of DEGs revealed a connection between lipid metabolism and LNM. Meanwhile, lipid staining showed a mass of lipids in obese N + tumor samples, and IHC analysis indicated an increase in LPL and CD36 expression in N + cases, implying a crucial role for exogenous lipid supply in LNM. Conclusions High BMI significantly increases the risk of LNM in GC and promotes lipid accumulation in GC cells in LNM. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-022-01734-7.
Collapse
|
29
|
Zhu Q, Li L, Jiao X, Xiong J, Zhai S, Zhu G, Cheng P, Qu J. Rare metastasis of gastric cancer to the axillary lymph node: A case report. Front Oncol 2022; 12:995738. [PMID: 36387206 PMCID: PMC9641636 DOI: 10.3389/fonc.2022.995738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/26/2022] [Indexed: 10/31/2023] Open
Abstract
Lymph node metastasis of gastric cancer is more common, metastatic lymph nodes are often around the stomach, and metastasis is carried out in a certain order, but gastric cancer metastasis to axillary lymph nodes is very rare. Due to the small number of patients with this kind of metastasis, its clinical features and treatment are not very clear. We initially thought that the enlarged axillary lymph nodes were inflammatory lesions. Axillary lymph node biopsy was later diagnosed as gastric cancer metastases to axillary lymph nodes. The patient refused further treatment and died 11 months after the second operation because of multiple systemic metastases. We believe that metastasis of gastric cancer to axillary lymph nodes is rare and the prognosis is poor. In clinical work, the possibility of metastatic lymph nodes should be considered in patients with a history of gastric cancer with enlarged axillary lymph nodes.
Collapse
Affiliation(s)
- Qingshun Zhu
- Department of Clinical Medical College, Weifang Medical University, Weifang, China
| | - Lei Li
- Department of Clinical Medical College, Weifang Medical University, Weifang, China
| | - Xuguang Jiao
- Department of General Surgery, The First Affiliated Hospital of Weifang Medical University (Weifang people’s Hospital), Weifang, China
| | - Jinqiu Xiong
- Department of General Surgery, The First Affiliated Hospital of Weifang Medical University (Weifang people’s Hospital), Weifang, China
| | - Shengyong Zhai
- Department of General Surgery, The First Affiliated Hospital of Weifang Medical University (Weifang people’s Hospital), Weifang, China
| | - Guangxu Zhu
- Department of General Surgery, The First Affiliated Hospital of Weifang Medical University (Weifang people’s Hospital), Weifang, China
| | - PeiPei Cheng
- Department of General Surgery, The First Affiliated Hospital of Weifang Medical University (Weifang people’s Hospital), Weifang, China
| | - Jianjun Qu
- Department of General Surgery, The First Affiliated Hospital of Weifang Medical University (Weifang people’s Hospital), Weifang, China
| |
Collapse
|
30
|
Kuroda K, Sakurai K, Kunimoto T, Kubo N, Hasegawa T, Inoue T. Ingenuity and tips for laparoscopic local resection for local recurrence of early gastric cancer after endoscopic submucosal dissection: A case report. Int J Surg Case Rep 2022; 99:107650. [PMID: 36115119 PMCID: PMC9568766 DOI: 10.1016/j.ijscr.2022.107650] [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: 08/17/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION AND IMPORTANCE The presence of severe comorbidities is associated with poor surgical outcomes in patients who undergo curative resection for gastric cancer. Herein, we present a report of laparoscopic local resection (LLR) for the local recurrence of gastric cancer in a patient with severe comorbidities. CASE PRESENTATION A 73-year-old man underwent endoscopic submucosal dissection (ESD) for an early gastric cancer located at the greater curvature of the gastric antrum. One year after the ESD, follow-up gastroscopy revealed a local recurrence. CLINICAL DISCUSSION LLR was performed owing to severe comorbidities such as chronic heart failure, severe chronic obstructive pulmonary disease, and diabetes mellitus. We confirmed the location of the tumor with an endoscope, and full-thickness stitches were placed outside the tumor to prevent displacement between the submucosal and serosal muscular layers. Full-thickness resection of the stomach wall was performed along the stitch points using an ultrasonic shear. The edge of the resection line was closed using a laparoscopic stapling system. The patient had an uneventful postoperative course and was discharged 8 days after the operation. The patient had no disease recurrence and late complications such as digestive symptom and weight loss at the 9-month follow-up after the operation. CONCLUSION LLR might be a suitable treatment option for high-risk patients with gastric cancer. During full-thickness resection of the stomach wall, our method enables the maintenance of a negative margin without the need for ESD.
Collapse
Affiliation(s)
- Kenji Kuroda
- Department of Gastroenterological Surgery, Osaka City General Hospital, Osaka, Japan.
| | - Katsunobu Sakurai
- Department of Gastroenterological Surgery, Osaka City General Hospital, Osaka, Japan
| | | | - Naoshi Kubo
- Department of Gastroenterological Surgery, Osaka City General Hospital, Osaka, Japan
| | - Tsuyoshi Hasegawa
- Department of Gastroenterological Surgery, Osaka City General Hospital, Osaka, Japan
| | - Toru Inoue
- Department of Gastroenterological Surgery, Osaka City General Hospital, Osaka, Japan
| |
Collapse
|
31
|
Fang C, Zhang X, Li C, Liu F, Liu H. Troponin C-1 Activated by E2F1 Accelerates Gastric Cancer Progression via Regulating TGF-β/Smad Signaling. Dig Dis Sci 2022; 67:4444-4457. [PMID: 34797443 DOI: 10.1007/s10620-021-07287-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Troponin C-1 (TNNC1) has been previously characterized as an oncogenic gene. AIMS This study aimed to reveal the roles of TNNC1 in gastric cancer and the potential underlying mechanisms. METHODS TNNC1 siRNAs and TNNC1 overexpression plasmid were used to alter its expression in AGS, MKN45, and HGC-27 cells. CCK-8 assay, colony formation, EdU assay, flow cytometry, transwell assay, and scratch test were conducted to measure the phenotype changes. In vivo effects of TNNC1 silence were confirmed by using a xenograft mouse model. Bioinformatics analysis was conducted to screen out the transcription factor and downstream signaling of TNNC1. RESULTS TNNC1 was highly expressed in gastric cancer tissues and cell lines, and its expression was associated with poor prognosis. TNNC1 silence suppressed the proliferation, migration, and invasion of AGS and MKN45 cells. However, TNNC1 silence induced apoptosis by mediating the cleavage of caspase-3 and caspase-9. Overexpression of TNNC1 in HGC-27 cells led to the contrary effects. The anti-tumor effects of TNNC1 silence were also confirmed in a xenograft animal model. E2F1 was validated as an upstream transcription factor of TNNC1. Effects of TNNC1 silence on AGS cell migration and invasion were attenuated by E2F1 overexpression. Besides, TGF-β/Smad was a downstream signaling pathway of TNNC1. The anti-tumor impacts of TNNC1 silence were weaken by SB431542 (a specific inhibitor of TGF-β signaling) while accelerated by TGF-β. CONCLUSION TNNC1 activated by E2F1 functioned as an oncogenic gene through regulating TGF-β/Smad signaling. TNNC1 was suggested as a potential molecular drug target of gastric cancer.
Collapse
Affiliation(s)
- Can Fang
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, 717 Jinbu Street, Muping District, Yantai, 264100, Shandong, People's Republic of China
| | - Xinxin Zhang
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, 717 Jinbu Street, Muping District, Yantai, 264100, Shandong, People's Republic of China
| | - Chengyan Li
- Department of Digestive Endoscopy Room, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong, People's Republic of China
| | - Fang Liu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, 717 Jinbu Street, Muping District, Yantai, 264100, Shandong, People's Republic of China
| | - Hui Liu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, 717 Jinbu Street, Muping District, Yantai, 264100, Shandong, People's Republic of China.
| |
Collapse
|
32
|
Detection Rate and Prognosis of Lymph Nodes in Gastric Cancer Using Nano Carbon Combined with In Vitro Anatomical Sorting. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:4540176. [PMID: 36052030 PMCID: PMC9427233 DOI: 10.1155/2022/4540176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/07/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022]
Abstract
In this study, we are going to investigate the effect of nano carbon combined with ex vitro anatomical sorting on the detection rate of lymph nodes (LNs) in gastric cancer (GC) along with the analysis of the correlation between LNs detection rate and patients’ prognosis. The clinical data of patients undergoing radical gastrectomy in Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University from January 2018 to January 2019 were examined retrospectively. According to whether they adopt nano carbon tracing and specimen sorting method, patients were divided into nano carbon and control groups. The respective rate of detection and correlation of total and positive LNs, respectively, clinical treatment, tumor marker level, and long-term prognosis were matched between these groups. At the same time, the effects of the nano carbon tracer on the detection of total and positive LNs were evaluated. In nano carbon group, more LN specimens could be detected, and the number of positive LNs increased significantly. In addition, in patients with different infiltration stages and LN substations, more LNs could be detected in the nano carbon group for examination, and the detection rate of LNs with diameter less than 5 mm was also more. Furthermore, LNs (preferably positive in number) were correlated positively with the attained LNs number. Otherwise, the use of nano carbon suspension could better label LNs in each substation, especially N1 station, and improve micro-LN detection rate. At the same time, the positive metastasis rate in black-stained LNs was higher (31.67% vs. 13.51%). In relation to the clinical prognosis, CEA’s level, i.e., CA199 and CA125, in the nano carbon group is controlled more effectively. Their condition was not easy to progress and relapse, and their mortality was further reduced. As a result, nano carbon, coupled with ex vitro anatomical sorting, may considerably enhance the detection rate of total and positive LNs, thereby improving the accuracy of clinical staging in GC patients, which has a good influence on their long-term prognosis.
Collapse
|
33
|
Xue XQ, Yu WJ, Shi X, Shao XL, Wang YT. 18F-FDG PET/CT-based radiomics nomogram for the preoperative prediction of lymph node metastasis in gastric cancer. Front Oncol 2022; 12:911168. [PMID: 36003788 PMCID: PMC9393365 DOI: 10.3389/fonc.2022.911168] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/13/2022] [Indexed: 11/27/2022] Open
Abstract
Objective Lymph node metastasis (LNM) is not only one of the important factors affecting the prognosis of gastric cancer but also an important basis for treatment decisions. The purpose of this study was to investigate the value of the radiomics nomogram based on preoperative 18F-deoxyglucose (FDG) PET/CT primary lesions and clinical risk factors for predicting LNM in gastric cancer (GC). Methods We retrospectively analyzed radiomics features of preoperative 18F-FDG PET/CT images in 224 gastric cancer patients from two centers. The prediction model was developed in the training cohort (n = 134) and validated in the internal (n = 59) and external validation cohorts (n = 31). The least absolute shrinkage and selection operator (LASSO) regression was used to select features and build radiomics signatures. The radiomics feature score (Rad-score) was calculated and established a radiomics signature. Multivariate logistic regression analysis was used to screen independent risk factors for LNM. The minimum Akaike’s information criterion (AIC) was used to select the optimal model parameters to construct a radiomics nomogram. The performance of the nomogram was assessed with calibration, discrimination, and clinical usefulness. Results There was no significant difference between the internal verification and external verification of the clinical data of patients (all p > 0.05). The areas under the curve (AUCs) (95% CI) for predicting LNM based on the 18F-FDG PET/CT radiomics signature in the training cohort, internal validation cohort, and external validation cohort were 0.792 (95% CI: 0.712–0.870), 0.803 (95% CI: 0.681–0.924), and 0.762 (95% CI: 0.579–0.945), respectively. Multivariate logistic regression showed that carbohydrate antigen (CA) 19-9 [OR (95% CI): 10.180 (1.267–81.831)], PET/CT diagnosis of LNM [OR (95% CI): 6.370 (2.256–17.984)], PET/CT Rad-score [OR (95% CI): 16.536 (5.506–49.660)] were independent influencing factors of LNM (all p < 0.05), and a radiomics nomogram was established based on those factors. The AUCs (95% CI) for predicting LNM were 0.861 (95% CI: 0.799–0.924), 0.889 (95% CI: 0.800–0.976), and 0.897 (95% CI: 0.683–0.948) in the training cohort, the internal validation cohort, and the external validation cohort, respectively. Decision curve analysis (DCA) indicated that the 18F-FDG PET/CT-based radiomics nomogram has good clinical utility. Conclusions Radiomics nomogram based on the primary tumor of 18F-FDG PET/CT could facilitate the preoperative individualized prediction of LNM, which is helpful for risk stratification in GC patients.
Collapse
Affiliation(s)
- Xiu-qing Xue
- Department of Nuclear Medicine, The First People’s Hospital of Yancheng, Yancheng, China
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Wen-Ji Yu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
| | - Xun Shi
- Department of Nuclear Medicine, The First People’s Hospital of Yancheng, Yancheng, China
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Xiao-Liang Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
| | - Yue-Tao Wang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- *Correspondence: Yue-Tao Wang,
| |
Collapse
|
34
|
Zhao J, Li K, Wang Z, Ke Q, Li J, Zhang Y, Zhou X, Zou Y, Song C. Efficacy and safety of indocyanine green tracer-guided lymph node dissection in minimally invasive radical gastrectomy for gastric cancer: A systematic review and meta-analysis. Front Oncol 2022; 12:884011. [PMID: 35992827 PMCID: PMC9388933 DOI: 10.3389/fonc.2022.884011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Background The implementation of indocyanine green (ICG) tracer-guided lymph node dissection is still in the preliminary stages of laparoscopic surgery, and its safety and efficacy for gastric cancer remain unclear. Methods A systematic review was conducted in PubMed, Embase, Web of Science, the Cochrane Library, and Scopus to identify relevant subjects from inception to June 2022. The core indicators were the total number of harvested lymph nodes and the safety of the laparoscopic gastrectomy with ICG. A meta-analysis was performed to estimate the pooled weighted mean difference (WMD) and 95% confidence interval (CI). Results Thirteen studies and 2,027 participants were included (642 for the ICG-group and 1,385 for the non-ICG group). The mean number of lymph nodes dissected in the ICG group was significantly greater than that in the non-ICG group (WMD = 6.24, 95% CI: 4.26 to 8.22, P <0.001). However, there was no significant difference in the mean number of positive lymph nodes dissected between the ICG and the non-ICG groups (WMD = 0.18, 95% CI: −0.70 to 1.07, P = 0.879). Additionally, ICG gastrectomy did not increase the risk in terms of the operative time, estimated blood loss, and postoperative complications. Conclusion ICG tracer with favorable safety increases the number of harvested lymph nodes but not the number of positive lymph nodes in laparoscopic gastrectomy. More high-quality, large-sample-size randomized controlled trials are still needed to enhance this evidence.
Collapse
Affiliation(s)
- Jixiang Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ke Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Surgical Oncology, Capital Medical University, Beijing, China
| | - Zikang Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterolog, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingqing Ke
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiapu Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yizhen Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaojiang Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunzhi Zou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Surgical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- *Correspondence: Yunzhi Zou, ; Conghua Song,
| | - Conghua Song
- Department of Gastroenterology, The Affiliated Hospital (Group) of Putian University, Putian, China
- *Correspondence: Yunzhi Zou, ; Conghua Song,
| |
Collapse
|
35
|
Tham E, Sestito M, Markovich B, Garland-Kledzik M. Current and future imaging modalities in gastric cancer. J Surg Oncol 2022; 125:1123-1134. [PMID: 35481912 DOI: 10.1002/jso.26875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/24/2022]
Abstract
Gastric adenocarcinoma treatment can include endoscopic mucosal resection, surgery, chemotherapy, radiation, and palliative measures depending on staging. Both invasive and noninvasive staging techniques have been used to dictate the best treatment pathway. Here, we review the current imaging modalities used in gastric cancer as well as novel techniques to accurately stage and screen these patients.
Collapse
Affiliation(s)
- Elwin Tham
- Department of Surgical Oncology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Michael Sestito
- Department of Surgical Oncology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Brian Markovich
- Department of Diagnostic Radiology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Mary Garland-Kledzik
- Department of Surgical Oncology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| |
Collapse
|
36
|
Miftode S, Bruns H. Misclassification of nodal stage in gastric cancer: 16 lymph nodes is not enough. SURGICAL AND EXPERIMENTAL PATHOLOGY 2022. [DOI: 10.1186/s42047-022-00109-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
In gastric cancer, nodal stage plays an important role. Insufficient lymph node harvesting or incomplete examination may lead to misclassification and affect postoperative strategy and group survival. This study’s objective was to determine the minimum number of examined lymph nodes needed in gastric cancer and compare this to the minimum lymph node count according to the current Union for International Cancer Control (UICC) classification using real world data.
Methods
Based on anatomical data, expected mean lymph node counts and their 95% confidence intervals for complete D2 lymphadenectomy were calculated. Using stochastic analysis, a threshold for correct classification in 95% of cases was determined. Survival data of nodal negative gastric cancer patients was extracted from the Surveillance, Epidemiology, and End Results (SEER)-Database for 2010–2017. Patients reaching at least the calculated theoretical threshold were compared to the minimum threshold according to the current UICC classification.
Results
The expected lymph node count was 30 (95% CI: 28–32; range 17–52), corresponding to a 27 lymph nodes. In nodal negative patients with exactly 16 and at least 27 examined lymph nodes, relative 5 year survival was 79 and 89% in T1/T2 and 39 and 64% T3/T4 gastric cancer, respectively. Theoretically, when only 16 lymph nodes are analyzed, nodal negative staging may be incorrect in up to 47% of cases.
Conclusions
A minimum threshold of 16 examined lymph nodes cannot be justified. Retrospective analysis confirmed systematic misclassification of patients with insufficient lymphadenectomy in nodal negative gastric cancer patients. Correct lymphadenectomy and thorough examination of the surgical specimen is mandatory.
Collapse
|
37
|
Li T, Cao H, Wu S, Zhong P, Ding J, Wang J, Wang F, He Z, Huang GL. Phosphorylated ATF1 at Thr184 promotes metastasis and regulates MMP2 expression in gastric cancer. J Transl Med 2022; 20:169. [PMID: 35397606 PMCID: PMC8994398 DOI: 10.1186/s12967-022-03361-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 03/26/2022] [Indexed: 12/24/2022] Open
Abstract
Background Studies have revealed an important role of activating transcription factor 1 (ATF1) and phosphorylated ATF1 at Ser63 in tumors. Our previous study identified Thr184 as a novel phosphorylation site of ATF1. However, the role of phosphorylated ATF1 at Thr184 (p-ATF1-T184) in tumor is unclear. This study figured out the role of p-ATF1-T184 in the metastasis of gastric cancer (GC) and in the regulation of Matrix metallopeptidase 2 (MMP2). Methods Immunohistochemical analysis (IHC) was performed to analyze the level of p-ATF1-T184 and its relationship with clinicopathological characteristics. Wound scratch test, Transwell assay were used to observe the role of p-ATF1-T184 in the invasion and metastasis of GC. The regulation of MMP2 by p-ATF1-T184 was investigated by a series of experiments including quantitative RT-PCR, western blot, gelatin zymography assay, Chromatin immunoprecipitation (ChIP), luciferase reporter assay and cycloheximide experiment. The Cancer Genome Atlas (TCGA) data were used to analyze the expression and prognostic role of ATF1 and MMP2 in GC. Mass spectrometry (MS) following co-immunoprecipitation (co-IP) assay was performed to identify potential upstream kinases that would phosphorylate ATF1 at Thr184. Results High expression level of p-ATF1-T184 was found and significantly associated with lymph node metastasis and poor survival in a GC cohort of 126 patients. P-ATF1-T184 promoted migration and invasion of gastric cancer cells. Phosphorylation of ATF1-T184 could regulate the mRNA, protein expression and extracellular activity of MMP2. P-ATF1-T184 further increased the DNA binding ability, transcription activity, and stabilized the protein expression of ATF1. Moreover, TCGA data and IHC results suggested that the mRNA level of ATF1 and MMP2, and protein level of p-ATF1-T184 and MMP2 could be prognosis markers of GC. Two protein kinase related genes, LRBA and S100A8, were identified to be correlated with the expression ATF1 in GC. Conclusion Our results indicated that p-ATF1-T184 promoted metastasis of GC by regulating MMP2. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03361-3.
Collapse
|
38
|
Song S, Liu B, Zeng X, Wu Y, Chen H, Wu H, Gu J, Gao X, Ruan Y, Wang H. Reticulon 2 promotes gastric cancer metastasis via activating endoplasmic reticulum Ca2+ efflux-mediated ERK signalling. Cell Death Dis 2022; 13:349. [PMID: 35428758 PMCID: PMC9012842 DOI: 10.1038/s41419-022-04757-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 03/01/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022]
Abstract
Gastric cancer ranks fourth for mortality globally among various malignant tumours, and invasion and metastasis are the major reason leading to its poor prognosis. Recently, accumulating studies revealed the role of reticulon proteins in cell growth and transmigration. However, the expression and biological function of reticulon proteins in human gastric cancer remain largely unclear. Herein, we explored the potential role of reticulon 2 (RTN2) in the progression of gastric cancer. Tissue microarray was used to determine the expression levels of RTN2 in 267 gastric cancer patients by immunohistochemistry. Gastric cancer cell lines were utilised to examine the influences of RTN2 on cellular migration and invasion abilities, epithelial-to-mesenchymal transition (EMT) and signalling pathway. In vivo studies were also performed to detect the effect of RTN2 on tumour metastasis. We found that RTN2 expression was notably upregulated in tumour tissues compared to pericarcinomatous tissues. High RTN2 expression was positively correlated with patients’ age, vessel invasion, tumour invasion depth, lymph node metastasis and TNM stage. Besides, high RTN2 staining intensity was associated with adverse survival which was further identified as an independent prognostic factor for gastric cancer patients by multivariate analysis. And the predictive accuracy was also improved when incorporated RTN2 into the TNM-staging system. RTN2 could promote the proliferation, migration and invasion of gastric cancer cells in vitro and lung metastasis in vivo. Mechanistically, RTN2 interacted with IP3R, and activated ERK signalling pathway via facilitating Ca2+ release from the endoplasmic reticulum, and subsequently drove EMT in gastric cancer cells. These results proposed RTN2 as a novel promotor and potential molecular target for gastric cancer therapies.
Collapse
|
39
|
Sivacoumarane S, Dutta S, Dubashi B, Adithan S, Toi PC, Nelamangala Ramakrishnaiah VP. Role of Neoadjuvant Chemotherapy in Locally Advanced Carcinoma Stomach: An Analysis of the Short-Term Outcomes. Cureus 2022; 14:e23936. [PMID: 35535287 PMCID: PMC9079323 DOI: 10.7759/cureus.23936] [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] [Accepted: 04/07/2022] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapy (NACT) in carcinoma stomach was introduced in an effort to eliminate micro-metastasis and to improve resectablity before surgery which improves R0 resection rates. We aimed to study the short term outcomes of neoadjuvant chemotherapy on the Tumor Node Metastasis (TNM) stage and the operative outcomes including R0 resection rate in locally advanced gastric cancer. METHODS We prospectively included patients with locally advanced adenocarcinoma stomach staged by contrast-enhanced computed tomography (CECT) in our study. Patients in Group I were started on neoadjuvant chemotherapy (epirubicin, oxaliplatin, and capecitabine). Surgery was done following response assessment CECT. Patients in Group II underwent upfront surgery. We assessed R0 resection rate, number of harvested and metastatic lymph nodes, lymph node ratio, duration of surgery, blood loss, hospital stay and complications between two groups. Response to NACT was assessed in Group I. RESULTS Out of 47 patients who received NACT, two patients had complete response (4.2%), 13 had partial response (27.7%), 10 had stable disease (21.3%) and 22 patients had progressive disease (46.8%). We found no significant difference in the rate of R0 resection between the two groups (88.2% in NACT group vs 85.1% in surgery group, P=0.55). CONCLUSIONS The rate of R0 resection does not significantly improve with neoadjuvant chemotherapy. In view of high progression rates, patient selection is required when NACT is planned in carcinoma stomach which are surgically resectable at presentation. We await survival analysis to further validate the role of NACT.
Collapse
Affiliation(s)
| | - Souradeep Dutta
- Surgery, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Biswajit Dubashi
- Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Subathra Adithan
- Radiodiagnosis, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Pampa C Toi
- Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | | |
Collapse
|
40
|
Wang M, Yu W, Cao X, Gu H, Huang J, Wu C, Wang L, Sha X, Shen B, Wang T, Yao Y, Zhu W, Huang F. Exosomal CD44 Transmits Lymph Node Metastatic Capacity Between Gastric Cancer Cells via YAP-CPT1A-Mediated FAO Reprogramming. Front Oncol 2022; 12:860175. [PMID: 35359362 PMCID: PMC8960311 DOI: 10.3389/fonc.2022.860175] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
Background Lymph node metastasis (LNM) commonly occurs in gastric cancer (GC) and is tightly associated with poor prognosis. Exosome-mediated lymphangiogenesis has been considered an important driver of LNM. Whether exosomes directly transmit the LNM phenotype between GC cells and its mechanisms remain elusive. Methods A highly lymphatic metastatic GC cell line (HGC-27-L) was established by serial passage of parental HGC-27 cells in BALB/c nude mice. The capacities of migration, invasion and LNM; fatty acid oxidation (FAO) levels; and the role of exosome-transferred LNM phenotype were compared among HGC-27-L, HGC-27 and primary GC cell line AGS. Exosomes derived from GC cells and sera were separately isolated using ultracentrifugation and ExoQuick exosome precipitation solution, and were characterized by transmission electron microscopy, Nanosight and western blotting. Transwell assay and LNM models were conducted to evaluate the capacities of migration, invasion and LNM of GC cells in vitro and in vivo. β-oxidation rate and CPT1 activity were measured to assess FAO. CPT1A inhibitor etomoxir was used to determine the role of FAO. Label-free LC-MS/MS proteome analysis screened the differential protein profiling between HGC-27-exosomes and AGS-exosomes. Small interference RNAs and YAP inhibitor verteporfin were used to elucidate the role and mechanism of exosomal CD44. TCGA data analysis, immunochemistry staining and ELISA were performed to analyze the expression correlation and clinical significance of CD44/YAP/CPT1A. Results FAO was increased in lymphatic metastatic GC cells and indispensable for sustaining LNM capacity. Lymphatic metastatic GC cell-exosomes conferred LNM capacity on primary GC cells in an FAO-dependent way. Mechanistically, CD44 was identified to be enriched in HGC-27-exosomes and was a critical cargo protein regulating exosome-mediated transmission, possibly by modulating the RhoA/YAP/Prox1/CPT1A signaling axis. Abnormal expression of CD44/YAP/CPT1A in GC tissues was correlated with each other and associated with LNM status, stages, invasion and poor survival. Serum exosomal CD44 concentration was positively correlated with tumor burden in lymph nodes. Conclusions We uncovered a novel mechanism: exosomal CD44 transmits LNM capacity between GC cells via YAP-CPT1A-mediated FAO reprogramming from the perspective of exosomes-transferred LNM phenotype. This provides potential therapeutic targets and a non-invasive biomarker for GC patients with LNM.
Collapse
Affiliation(s)
- Mei Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wanjun Yu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaoli Cao
- Department of Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Hongbing Gu
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Jiaying Huang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Chen Wu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Lin Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xin Sha
- Department of Surgery, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Bo Shen
- Department of Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Ting Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yongliang Yao
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China
| | - Wei Zhu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Feng Huang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China
- Department of Clinical Laboratory, Maternal and Child Health Care Hospital of Kunshan, Suzhou, China
| |
Collapse
|
41
|
Zhang C, Yang J, Chen Y, Jiang F, Liao H, Liu X, Wang Y, Kong G, Zhang X, Li J, Gao J, Shen L. miRNAs derived from plasma small extracellular vesicles predict organo-tropic metastasis of gastric cancer. Gastric Cancer 2022; 25:360-374. [PMID: 35031872 DOI: 10.1007/s10120-021-01267-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Peritoneum, liver and lymph node are the most common metastatic sites of gastric cancer (GC). Biomarkers for GC's organo-tropic metastasis remained largely unknown, which was investigated in this study from the perspective of small extracellular vesicle (sEV)-derived miRNAs. METHODS Plasma from treatment-naïve GC patients including no metastasis (M0), peritoneal metastasis (PM), hepatic metastasis (HM) and distant lymph node metastasis (dLNM)) were divided into one discovery (N = 40), one training (N = 40) and one validating cohort (N = 86), then assessed by sEV-miRNA-sequencing and sEV-miRNA-qPCR. Functional explorations were also performed for verification. RESULTS The expression profiles of sEV-miRNAs varied greatly across different metastatic patterns. Based on logistic regression models, we constructed signatures for M0 (hsa-miR-186-5p/hsa-miR-200c-3p/hsa-miR-429/hsa-miR-5187-5p/hsa-miR-548ae-5p), PM (hsa-miR-200c-3p/hsa-miR-429), HM (hsa-miR-200c-3p/hsa-miR-429) and dLNM (hsa-miR-324-5p/hsa-miR-374a-5p/hsa-miR-429/hsa-miR-548ae-5p). These signatures vigorously characterized organo-tropic metastasis (all displaying AUC > 0.8, consistency ≥ 75%), and effectively conjectured the risk of future metastasis within 5 years (accuracy 45.5% for occurrence, 70% for organotropism, P = 0.002 for prognostic diversity). Additionally, we explored these seven biomarker miRNAs' impact on GC's in vitro motility and discussed their potential involvement in cancer-related biological processes and pathways. CONCLUSIONS Our work highlighted that plasma sEV-miRNAs powerfully characterized and predicted the organo-tropic metastasis of GC and provided new insight into the applications of sEV-based liquid biopsy in clinical practice.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jing Yang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Fangli Jiang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Haiyan Liao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, 518116, China
| | - Xiang Liu
- Department of R&D, Echo Biotech Co., Ltd, Beijing, People's Republic of China
| | - Yuan Wang
- Department of R&D, Echo Biotech Co., Ltd, Beijing, People's Republic of China
| | - Guanyi Kong
- Department of R&D, Echo Biotech Co., Ltd, Beijing, People's Republic of China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jing Gao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, 518116, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
| |
Collapse
|
42
|
Zhuo N, Liu C, Zhang Q, Li J, Zhang X, Gong J, Lu M, Peng Z, Zhou J, Wang X, Jiao X, Wang Y, Wang Y, Gao M, Shen L, Lu Z. Characteristics and Prognosis of Acquired Resistance to Immune Checkpoint Inhibitors in Gastrointestinal Cancer. JAMA Netw Open 2022; 5:e224637. [PMID: 35348710 PMCID: PMC8965636 DOI: 10.1001/jamanetworkopen.2022.4637] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
IMPORTANCE With the expanding use of immune checkpoint inhibitors (ICIs) in gastrointestinal (GI) cancer, the occurrence of acquired resistance (AR) has gradually emerged. However, the progression patterns and survival of patients with AR to ICIs are still unknown. OBJECTIVE To explore the characteristics and prognosis of AR after ICI therapy in patients with advanced GI cancer. DESIGN, SETTING, AND PARTICIPANTS This cohort study screened patients with advanced GI cancer treated with ICIs between January 14, 2016, and December 31, 2020, at Peking University Cancer Hospital. Initial response was defined as complete response, partial response, or stable disease longer than 6 months as assessed by Response Evaluation Criteria in Solid Tumors version 1.1. Progression was also based on Response Evaluation Criteria in Solid Tumors version 1.1. Progression or death after the initial response was defined as AR. Oligoprogression of AR was defined as 2 or more disease sites progression. The current status of AR in GI cancer and the patterns of AR and its prognosis were evaluated. The site of AR and subsequent management were also assessed. Data were analyzed from June to August 2021. EXPOSURES Patients in the cohort were treated with mono-ICI or combination therapy. MAIN OUTCOMES AND MEASURES Kaplan-Meier analyses and log-rank tests were conducted for overall survival analyses. Univariate and multivariate Cox analyses were conducted to determine the prognostic implications of each variable. RESULTS Of the 1124 patients who received ICIs, 373 (33.2%) patients (282 men [75.6%]; median [IQR] age, 62 [54-68] years) achieved an initial response, and 173 (46.4%) patients (137 men [79.2%]; median [IQR] age, 61 [54-67] years) developed AR. Almost all patients (167 patients [96.5%]) developed AR within 24.0 months. Progression patterns of AR were most commonly oligoprogression (122 patients [70.5%]) rather than polymetastatic progression (38 patients [22.0%]) and were associated with a good prognosis (38.5 vs 14.0 months; hazard ratio, 0.37; 95% CI, 0.18-0.74; P < .001). Lymph nodes (101 patients [58.4%]) appeared to be the most common site of AR. Management after AR was mainly systemic therapy (96 patients [55.5%]). CONCLUSIONS AND RELEVANCE Oligoprogression was the most common pattern of AR progression, and lymph nodes were the most susceptible site for AR. Further study will be needed to determine the most favorable management for AR.
Collapse
Affiliation(s)
- Na Zhuo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Chang Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Qi Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Xicheng Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Xi Jiao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Yujiao Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Yanni Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Mengting Gao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| |
Collapse
|
43
|
Li T, Liu T, Zhao Z, Xu X, Zhan S, Zhou S, Jiang N, Zhu W, Sun R, Wei F, Feng B, Guo H, Yang R. The Lymph Node Microenvironment May Invigorate Cancer Cells With Enhanced Metastatic Capacities. Front Oncol 2022; 12:816506. [PMID: 35295999 PMCID: PMC8918682 DOI: 10.3389/fonc.2022.816506] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/02/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer metastasis, a typical malignant biological behavior involving the distant migration of tumor cells from the primary site to other organs, contributed majorly to cancer-related deaths of patients. Although constant efforts have been paid by researchers to elucidate the mechanisms of cancer metastasis, we are still far away from the definite answer. Recently, emerging evidence demonstrated that cancer metastasis is a continuous coevolutionary process mediated by the interactions between tumor cells and the host organ microenvironment, and epigenetic reprogramming of metastatic cancer cells may confer them with stronger metastatic capacities. The lymph node served as the first metastatic niche for many types of cancer, and the appearance of lymph node metastasis predicted poor prognosis. Importantly, multiple immune cells and stromal cells station and linger in the lymph nodes, which constitutes the complexity of the lymph node microenvironment. The active cross talk between cancer cells and immune cells could happen unceasingly within the metastatic environment of lymph nodes. Of note, diverse immune cells have been found to participate in the formation of malignant properties of tumor, including stemness and immune escape. Based on these available evidence and data, we hypothesize that the metastatic microenvironment of lymph nodes could drive cancer cells to metastasize to further organs through epigenetic mechanisms.
Collapse
Affiliation(s)
- Tianhang Li
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tianyao Liu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zihan Zhao
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xinyan Xu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shoubin Zhan
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shengkai Zhou
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ning Jiang
- Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Wenjie Zhu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Rui Sun
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fayun Wei
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Baofu Feng
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Rong Yang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
44
|
Pang HY, Liang XW, Chen XL, Zhou Q, Zhao LY, Liu K, Zhang WH, Yang K, Chen XZ, Hu JK. Assessment of indocyanine green fluorescence lymphography on lymphadenectomy during minimally invasive gastric cancer surgery: a systematic review and meta-analysis. Surg Endosc 2022; 36:1726-1738. [PMID: 35079880 DOI: 10.1007/s00464-021-08830-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND In recent years, indocyanine green fluorescence lymphography has been introduced for lymphatic mapping in gastric cancer surgery. The aim of this study was to investigate the efficacy of ICGFL in lymph node dissection during minimally invasive surgery for gastric cancer. METHODS A systematic review of electronic databases including PubMed, Embase, Web of Science, the Cochrane Library, and China National Knowledge Infrastructure was performed from the inception to January 2021 for all studies comparing ICGFL with non-ICGFL in GC patients undergoing minimal access gastrectomy. The primary outcome was the total number of harvested lymph nodes. The secondary endpoints were the number of metastatic LNs, operative time, estimated blood loss, and postoperative complications. The registration number of this protocol is PROSPERO CRD42020203443. RESULTS A total of 13 studies including 1882 participants were included. In this meta-analysis, the use of ICGFL was associated with a higher number of harvested LNs (40.33 vs. 33.40; MD = 6.93; 95%CI: 4.28 to 9.58; P < 0.0001; I2 = 86%). No significant difference was found between the ICGFL and control groups in terms of metastatic LNs (2.63 vs. 2.42; MD = 0.21; 95%CI: -0.46 to 0.87; P = 0.54; I2 = 0%). In addition, the use of ICGFL could be safely performed without increasing the operative time (P = 0.49), estimated blood loss (P = 0.26) and postoperative complications (P = 0.54). CONCLUSION The use of ICGFL may be a useful tool facilitating complete lymph node dissection during minimally invasive GC resection. However, more high-quality RCTs with large sample size are needed to validate this issue.
Collapse
Affiliation(s)
- Hua-Yang Pang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Xian-Wen Liang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Xiao-Long Chen
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Quan Zhou
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Lin-Yong Zhao
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Kai Liu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Wei-Han Zhang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Kun Yang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Xin-Zu Chen
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China
| | - Jian-Kun Hu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, No. 37 Guo Xue Xiang Street, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
45
|
Kinami S, Saito H, Takamura H. Significance of Lymph Node Metastasis in the Treatment of Gastric Cancer and Current Challenges in Determining the Extent of Metastasis. Front Oncol 2022; 11:806162. [PMID: 35071010 PMCID: PMC8777129 DOI: 10.3389/fonc.2021.806162] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/13/2021] [Indexed: 12/16/2022] Open
Abstract
The stomach exhibits abundant lymphatic flow, and metastasis to lymph nodes is common. In the case of gastric cancer, there is a regularity to the spread of lymph node metastasis, and it does not easily metastasize outside the regional nodes. Furthermore, when its extent is limited, nodal metastasis of gastric cancer can be cured by appropriate lymph node dissection. Therefore, identifying and determining the extent of lymph node metastasis is important for ensuring accurate diagnosis and appropriate surgical treatment in patients with gastric cancer. However, precise detection of lymph node metastasis remains difficult. Most nodal metastases in gastric cancer are microscopic metastases, which often occur in small-sized lymph nodes, and are thus difficult to diagnose both preoperatively and intraoperatively. Preoperative nodal diagnoses are mainly made using computed tomography, although the specificity of this method is low because it is mainly based on the size of the lymph node. Furthermore, peripheral nodal metastases cannot be palpated intraoperatively, nodal harvesting of resected specimens remains difficult, and the number of lymph nodes detected vary greatly depending on the skill of the technician. Based on these findings, gastrectomy with prophylactic lymph node dissection is considered the standard surgical procedure for gastric cancer. In contrast, several groups have examined the value of sentinel node biopsy for accurately evaluating nodal metastasis in patients with early gastric cancer, reporting high sensitivity and accuracy. Sentinel node biopsy is also important for individualizing and optimizing the extent of uniform prophylactic lymph node dissection and determining whether patients are indicated for function-preserving curative gastrectomy, which is superior in preventing post-gastrectomy symptoms and maintaining dietary habits. Notably, advancements in surgical treatment for early gastric cancer are expected to result in individualized surgical strategies with sentinel node biopsy. Chemotherapy for advanced gastric cancer has also progressed, and conversion gastrectomy can now be performed after downstaging, even in cases previously regarded as inoperable. In this review, we discuss the importance of determining lymph node metastasis in the treatment of gastric cancer, the associated difficulties, and the need to investigate strategies that can improve the diagnosis of lymph node metastasis.
Collapse
Affiliation(s)
- Shinichi Kinami
- Department of Surgical Oncology, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Kahoku-gun, Japan
- Department of General and Gastroenterologic Surgery, Kanazawa Medical University Himi Municipal Hospital, Himi City, Japan
| | - Hitoshi Saito
- Department of General and Gastroenterologic Surgery, Kanazawa Medical University Himi Municipal Hospital, Himi City, Japan
| | - Hiroyuki Takamura
- Department of Surgical Oncology, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Kahoku-gun, Japan
| |
Collapse
|
46
|
Elucidated tumorigenic role of MAML1 and TWIST1 in gastric cancer is associated with Helicobacter pylori infection. Microb Pathog 2021; 162:105304. [PMID: 34818576 DOI: 10.1016/j.micpath.2021.105304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) has a fundamental role in tumor initiation, progression, and metastasis. Helicobacter pylori (HP) induces EMT and thus causes gastric cancer (GC) by deregulating multiple signaling pathways involved in EMT. TWIST1 and MAML1 have been confirmed to be critical inducers of EMT via diverse signaling pathways such as Notch signaling. This study aimed to investigate for the first time possible associations between TWIST1/MAML1 mRNA expression levels, HP infection, and clinicopathological characteristics in GC patients. METHOD TWIST1 and MAML1 mRNA expression levels were evaluated in tumoral and adjacent normal tissues in 73 GC patients using the quantitative reverse transcription PCR (RT-qPCR) method. PCR technique was also applied to examine the infection with HP in GC samples. RESULTS Upregulation of TWIST1 and MAML1 expression was observed in 35 (48%) and 34 (46.6%) of 73 tumor samples, respectively. Co-overexpression of these genes was found in 26 of 73 (35.6%) tumor samples; meanwhile, there was a significant positive correlation between MAML1 and TWIST1 mRNA expression levels (P < 0.001). MAML1 overexpression exhibited meaningful associations with advanced tumor stages (P = 0.006) and nodal metastases (P ˂ 0.001). 34 of 73 (46.6%) tumors tested positive for HP, and meanwhile, MAML1 expression was positively related with T (P = 0.05) and grade (P = 0.0001) in these HP-positive samples. Increased TWIST1 expression was correlated with patient sex (P = 0.035) and advanced tumor grade (P = 0.017) in HP-infected tumors. Furthermore, TWIST1 and MAML1 expression levels were inversely linked with histologic grade in HP-negative tumor samples (P = 0.021 and P = 0.048, respectively). CONCLUSION We propose TWIST1 and MAML1 as potential biomarkers of advanced-stage GC that determine the characteristics and aggressiveness of the disease. Based on accumulating evidence and our findings, they can be introduced as promising therapeutic targets to modify functional abnormalities in cells that promote GC progression. Moreover, HP may enhance GC growth and metastasis by disrupting TWIS1/MAML1 expression patterns and related pathways.
Collapse
|
47
|
Diabetes mellitus promoted lymph node metastasis in gastric cancer: a 15-year single-institution experience. Chin Med J (Engl) 2021; 135:950-961. [PMID: 34711717 PMCID: PMC9276325 DOI: 10.1097/cm9.0000000000001795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: Previous studies have revealed that diabetes mellitus (DM) promotes disease progress of gastric cancer (GC). This study aimed to further investigating whether DM advanced lymph nodes (LNs) metastasis in GC. Methods: The clinicopathologic data of GC patients with >15 examined LN (ELN) between October 2004 and December 2019 from a prospectively maintained database were included. The observational outcomes included the number (N3b status) and anatomical distribution (N3 stations) of metastatic LN (MLN). Results: A total of 2142 eligible patients were included in the study between October 2004 and December 2019. N3 stations metastasis (26.8% in DM vs. 19.3% in non-DM, P = 0.026) and N3b status (18.8% in DM vs. 12.8% in non-DM, P = 0.039) were more advanced in the DM group, and multivariate logistic regression analyses confirmed that DM was an independent factor of developing N3 stations metastasis (odds ratio [OR] = 1.771, P = 0.011) and N3b status (OR = 1.752, P = 0.028). Also, multivariate analyses determined DM was independently associated with more MLN (β = 1.424, P = 0.047). The preponderance of N3 stations metastasis (DM vs. non-DM, T1–2: 2.2% vs. 4.9%, T3: 29.0% vs. 20.3%, T4a: 38.9% vs. 25.8%, T4b: 50.0% vs. 36.6%; ELN16–29: 8.6% vs. 10.4%, ELN30–44: 27.9% vs. 20.5%, ELN ≥ 45: 37.7% vs. 25.3%), N3b status (DM vs. non-DM, T1–2: 0% vs. 1.7%, T3: 16.1% vs. 5.1%, T4a: 27.8% vs. 19.1%, T4b: 44.0% vs. 28.0%; ELN16–29: 8.6% vs. 7.9%, ELN30–44: 18.0% vs. 11.8%, ELN ≥ 45: 26.4% vs. 17.3%), and the number of MLN (DM vs. non-DM, T1–2: 0.4 vs. 1.1, T3: 8.6 vs. 5.2, T4a: 9.7 vs. 8.6, T4b: 17.0 vs. 12.8; ELN16–29: 3.6 vs. 4.6, ELN30–44: 5.8 vs. 5.5, ELN ≥ 45: 12.0 vs. 7.7) of DM group increased with the advancement of primary tumor depth stage and raising of ELN. Conclusions: DM was an independent risk factor for promoting LN metastasis. The preponderance of LN involvement in the DM group was aggravated with the advancement of tumor depth.
Collapse
|
48
|
Regulation of follistatin-like 3 expression by miR-486-5p modulates gastric cancer cell proliferation, migration and tumor progression. Aging (Albany NY) 2021; 13:20302-20318. [PMID: 34425560 PMCID: PMC8436905 DOI: 10.18632/aging.203412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 08/02/2021] [Indexed: 12/23/2022]
Abstract
Cancer development and progression can be regulated by the levels of endogenous factors. Gastric cancer is an aggressive disease state with poor patient prognosis, needing the development of new diagnostics and therapeutic strategies. We investigated the close association between follistatin-like 3 (FSTL3) and different cancers, and focused on its role in gastric cancer cell function. Using cancer bioinformatics, we found that FSTL3 expression is elevated in a large majority of the 33 cancers we analyzed in publicly available cancer databases. Elevated levels of FSTL3 is associated with poor patient prognosis in gastric cancer. In a comparison of normal gastric epithelial cells and gastric cancer cell lines, FSTL3 expression was consistently elevated in gastric cancer cells. Overexpression of FSTL3 promoted gastric cancer cell viability, proliferation and migration. Conversely, FSTL3 knockdown inhibits these cellular processes. Using bioinformatics, we found that the FSTL3 mRNA has a potential binding site in the 3'-UTR for a small microRNA, miR-486-5p. Further bioinformatics revealed significant negative correlation between FSTL3 and miR-486-5p levels. Using luciferase reporter constructs, we provide evidence that the 3'UTR from the FSTL3 mRNA can confer downregulation in the presence of miR-486-5p. These studies lead us to conclude that FSTL3 has oncogenic properties and increased expression of this gene product promotes gastric cancer development and progression.
Collapse
|
49
|
Then EO, Grantham T, Deda X, Ramachandran R, Gaduputi V. Metastatic Gastric Cancer to the Colon. World J Oncol 2021; 12:127-131. [PMID: 34349858 PMCID: PMC8297054 DOI: 10.14740/wjon1375] [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: 03/28/2021] [Accepted: 04/29/2021] [Indexed: 12/09/2022] Open
Abstract
Gastric cancer is one of the most common and deadly cancers worldwide, especially amongst older males. Current data suggest gastric cancer is the fifth most common neoplasm and the third most deadly cancer, with an estimated 783,000 deaths in 2018. Risk factors associated with the development of gastric cancer include obesity, gastroesophageal reflux disease, Helicobacter pylori infection, and low socioeconomic status. Diagnosis of gastric cancer can be accomplished by endoscopy, which allows the clinician to obtain a biopsy specimen. Endoscopic ultrasound is also an important modality that is helpful in assessing tumor invasion. The most common sites of metastatic gastric cancer in descending order are the liver, peritoneum, lung and bone. Rarely will gastric cancer metastasize to the colon. Here we present a rare case of colonic metastasis of a primary gastric adenocarcinoma.
Collapse
Affiliation(s)
- Eric Omar Then
- Division of Gastroenterology and Hepatology, The Brooklyn Hospital Center, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA.,Division of Gastroenterology and Hepatology, SBH Health System, Bronx, NY, USA
| | - Tyler Grantham
- Department of Internal Medicine, St. George's University School of Medicine, Grenada, West Indies
| | - Xheni Deda
- Division of Gastroenterology and Hepatology, SBH Health System, Bronx, NY, USA
| | - Rajarajeshwari Ramachandran
- Division of Gastroenterology and Hepatology, The Brooklyn Hospital Center, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
| | - Vinaya Gaduputi
- Division of Gastroenterology and Hepatology, SBH Health System, Bronx, NY, USA
| |
Collapse
|
50
|
Chang CW, Chen CY. Prognostic factors of advanced gastric cancer. J Chin Med Assoc 2021; 84:557-558. [PMID: 33871396 DOI: 10.1097/jcma.0000000000000526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Chen-Wang Chang
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan, ROC
- MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan, ROC
- MacKay Medical College, New Taipei City, Taiwan, ROC
| | - Chih-Yen Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine and Institute of Emergency and Critical Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan, ROC
| |
Collapse
|