1
|
Díaz del Arco C, Fernández Aceñero MJ, Ortega Medina L. Liquid biopsy for gastric cancer: Techniques, applications, and future directions. World J Gastroenterol 2024; 30:1680-1705. [PMID: 38617733 PMCID: PMC11008373 DOI: 10.3748/wjg.v30.i12.1680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/01/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
After the study of circulating tumor cells in blood through liquid biopsy (LB), this technique has evolved to encompass the analysis of multiple materials originating from the tumor, such as nucleic acids, extracellular vesicles, tumor-educated platelets, and other metabolites. Additionally, research has extended to include the examination of samples other than blood or plasma, such as saliva, gastric juice, urine, or stool. LB techniques are diverse, intricate, and variable. They must be highly sensitive, and pre-analytical, patient, and tumor-related factors significantly influence the detection threshold, diagnostic method selection, and potential results. Consequently, the implementation of LB in clinical practice still faces several challenges. The potential applications of LB range from early cancer detection to guiding targeted therapy or immunotherapy in both early and advanced cancer cases, monitoring treatment response, early identification of relapses, or assessing patient risk. On the other hand, gastric cancer (GC) is a disease often diagnosed at advanced stages. Despite recent advances in molecular understanding, the currently available treatment options have not substantially improved the prognosis for many of these patients. The application of LB in GC could be highly valuable as a non-invasive method for early diagnosis and for enhancing the management and outcomes of these patients. In this comprehensive review, from a pathologist's perspective, we provide an overview of the main options available in LB, delve into the fundamental principles of the most studied techniques, explore the potential utility of LB application in the context of GC, and address the obstacles that need to be overcome in the future to make this innovative technique a game-changer in cancer diagnosis and treatment within clinical practice.
Collapse
Affiliation(s)
- Cristina Díaz del Arco
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - M Jesús Fernández Aceñero
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Luis Ortega Medina
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| |
Collapse
|
2
|
Ge Z, Dai S, Yu H, Zhao J, Yang W, Tan W, Sun J, Gan Q, Liu L, Wang Z. Nanomechanical Analysis of Living Small Extracellular Vesicles to Identify Gastric Cancer Cell Malignancy Based on a Biomimetic Peritoneum. ACS NANO 2024; 18:6130-6146. [PMID: 38349890 PMCID: PMC10906078 DOI: 10.1021/acsnano.3c02285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 02/03/2024] [Accepted: 02/08/2024] [Indexed: 02/15/2024]
Abstract
Gastric cancer is one of the most prevalent digestive malignancies. The lack of effective in vitro peritoneal models has hindered the exploration of the potential mechanisms behind gastric cancer's peritoneal metastasis. An accumulating body of research indicates that small extracellular vesicles (sEVs) play an indispensable role in peritoneal metastasis of gastric cancer cells. In this study, a biomimetic peritoneum was constructed. The biomimetic model is similar to real peritoneum in internal microstructure, composition, and primary function, and it enables the recurrence of peritoneal metastasis process in vitro. Based on this model, the association between the mechanical properties of sEVs and the invasiveness of gastric cancer was identified. By performing nanomechanical analysis on sEVs, we found that the Young's modulus of sEVs can be utilized to differentiate between malignant clinical samples (ascites) and nonmalignant clinical samples (peritoneal lavage). Furthermore, patients' ascites-derived sEVs were verified to stimulate the mesothelial-to-mesenchymal transition, thereby promoting peritoneal metastasis. In summary, nanomechanical analysis of living sEVs could be utilized for the noninvasive diagnosis of malignant degree and peritoneal metastasis of gastric cancer. This finding is expected to contribute future treatments.
Collapse
Affiliation(s)
- Zhixing Ge
- State
Key Laboratory of Robotics, Shenyang Institute
of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes
for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Songchen Dai
- Department
of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110016, China
- Key
Laboratory of Precision Diagnosis and Treatment of Gastrointestinal
Tumors, Ministry of Education, Shenyang 110016, China
| | - Haibo Yu
- State
Key Laboratory of Robotics, Shenyang Institute
of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes
for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| | - Junhua Zhao
- Department
of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110016, China
- Key
Laboratory of Precision Diagnosis and Treatment of Gastrointestinal
Tumors, Ministry of Education, Shenyang 110016, China
| | - Wenguang Yang
- School of
Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China
| | - Wenjun Tan
- State
Key Laboratory of Robotics, Shenyang Institute
of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes
for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingxu Sun
- Department
of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110016, China
- Key
Laboratory of Precision Diagnosis and Treatment of Gastrointestinal
Tumors, Ministry of Education, Shenyang 110016, China
| | - Quan Gan
- State
Key Laboratory of Robotics, Shenyang Institute
of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes
for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Lianqing Liu
- State
Key Laboratory of Robotics, Shenyang Institute
of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes
for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| | - Zhenning Wang
- Department
of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110016, China
- Key
Laboratory of Precision Diagnosis and Treatment of Gastrointestinal
Tumors, Ministry of Education, Shenyang 110016, China
| |
Collapse
|
3
|
Chia DKA, Demuytere J, Ernst S, Salavati H, Ceelen W. Effects of Hyperthermia and Hyperthermic Intraperitoneal Chemoperfusion on the Peritoneal and Tumor Immune Contexture. Cancers (Basel) 2023; 15:4314. [PMID: 37686590 PMCID: PMC10486595 DOI: 10.3390/cancers15174314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Hyperthermia combined with intraperitoneal (IP) drug delivery is increasingly used in the treatment of peritoneal metastases (PM). Hyperthermia enhances tumor perfusion and increases drug penetration after IP delivery. The peritoneum is increasingly recognized as an immune-privileged organ with its own distinct immune microenvironment. Here, we review the immune landscape of the healthy peritoneal cavity and immune contexture of peritoneal metastases. Next, we review the potential benefits and unwanted tumor-promoting effects of hyperthermia and the associated heat shock response on the tumor immune microenvironment. We highlight the potential modulating effect of hyperthermia on the biomechanical properties of tumor tissue and the consequences for immune cell infiltration. Data from translational and clinical studies are reviewed. We conclude that (mild) hyperthermia and HIPEC have the potential to enhance antitumor immunity, but detailed further studies are required to distinguish beneficial from tumor-promoting effects.
Collapse
Affiliation(s)
- Daryl K. A. Chia
- Department of Surgery, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Jesse Demuytere
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Sam Ernst
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Hooman Salavati
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Wim Ceelen
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Department of GI Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
4
|
Kimura Y, Ohzawa H, Miyato H, Kaneko Y, Kuchimaru T, Takahashi R, Yamaguchi H, Kurashina K, Saito S, Hosoya Y, Lefor AK, Sata N, Kitayama J. Intraperitoneal transfer of microRNA-29b-containing small extracellular vesicles can suppress peritoneal metastases of gastric cancer. Cancer Sci 2023; 114:2939-2950. [PMID: 36939028 PMCID: PMC10323101 DOI: 10.1111/cas.15793] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/21/2023] Open
Abstract
Small extracellular vesicles (sEV) contain various microRNAs (miRNAs) and play crucial roles in the tumor metastatic process. Although miR-29b levels in peritoneal exosomes were markedly reduced in patients with peritoneal metastases (PM), their role has not been fully clarified. In this study, we asked whether the replacement of miR-29b can affect the development of PM in a murine model. UE6E7T-12, human bone marrow-derived mesenchymal stem cells (BMSCs), were transfected with miR-29b-integrating recombinant lentiviral vector and sEV were isolated from culture supernatants using ultracentrifugation. The sEV contained markedly increased amounts of miR-29b compared with negative controls. Treatment with transforming growth factor-β1 decreased the expression of E-cadherin and calretinin with increased expression of vimentin and fibronectin on human omental tissue-derived mesothelial cells (HPMCs). However, the effects were totally abrogated by adding miR-29b-rich sEV. The sEV inhibited proliferation and migration of HPMCs by 15% (p < 0.005, n = 6) and 70% (p < 0.005, n = 6), respectively, and inhibited adhesion of NUGC-4 and MKN45 to HPMCs by 90% (p < 0.0001, n = 5) and 77% (p < 0.0001, n = 5), respectively. MicroRNA-29b-rich murine sEV were similarly obtained using mouse BMSCs and examined for in vivo effects with a syngeneic murine model using YTN16P, a highly metastatic clone of gastric cancer cell. Intraperitoneal (IP) transfer of the sEV every 3 days markedly reduced the number of PM from YTN16P in the mesentery (p < 0.05, n = 6) and the omentum (p < 0.05, n = 6). Bone marrow mesenchymal stem cell-derived sEV are a useful carrier for IP administration of miR-29b, which can suppress the development of PM of gastric cancer.
Collapse
Affiliation(s)
- Yuki Kimura
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Hideyuki Ohzawa
- Department of Clinical OncologyJichi Medical University HospitalShimotsukeJapan
| | - Hideyo Miyato
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Yuki Kaneko
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | | | - Rei Takahashi
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Hironori Yamaguchi
- Department of Clinical OncologyJichi Medical University HospitalShimotsukeJapan
| | - Kentaro Kurashina
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Shin Saito
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Yoshinori Hosoya
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | | | - Naohiro Sata
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
| | - Joji Kitayama
- Department of SurgeryJichi Medical University HospitalShimotsukeJapan
- Center for Clinical ResearchJichi Medical University HospitalShimotsukeJapan
| |
Collapse
|
5
|
Chen Y, Cai G, Jiang J, He C, Chen Y, Ding Y, Lu J, Zhao W, Yang Y, Zhang Y, Wu G, Wang H, Zhou Z, Teng L. Proteomic profiling of gastric cancer with peritoneal metastasis identifies a protein signature associated with immune microenvironment and patient outcome. Gastric Cancer 2023; 26:504-516. [PMID: 36930369 PMCID: PMC10284991 DOI: 10.1007/s10120-023-01379-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 02/24/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Peritoneal metastasis (PM) frequently occurs in patients with gastric cancer (GC) and is a major cause of mortality. Risk stratification for PM can optimize decision making in GC treatment. METHODS A total of 25 GC patients (13 with synchronous, 6 with metachronous PM and 6 PM-free) were included in this study. Quantitative proteomics by high-depth tandem mass tags labeling and whole-exome sequencing were conducted in primary GC and PM samples. Proteomic signature and prognostic model were established by machine learning algorithms in PM and PM-free GC, then validated in two external cohorts. Tumor-infiltrating immune cells in GC were analyzed by CIBERSORT. RESULTS Heterogeneity between paired primary and PM samples was observed at both genomic and proteomic levels. Compared to primary GC, proteome of PM samples was enriched in RNA binding and extracellular exosomes. 641 differently expressed proteins (DEPs) between primary GC of PM group and PM-free group were screened, which were enriched in extracellular exosome and cell adhesion pathways. Subsequently, a ten-protein signature was derived based on DEPs by machine learning. This signature was significantly associated with patient prognosis in internal cohort and two external proteomic datasets of diffuse and mixed type GC. Tumor-infiltrating immune cell analysis showed that the signature was associated with immune microenvironment of GC. CONCLUSIONS We characterized proteomic features that were informative for PM progression of GC. A protein signature associated with immune microenvironment and patient outcome was derived, and it could guide risk stratification and individualized treatment.
Collapse
Affiliation(s)
- Yanyan Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79# Qingchun Road, Hangzhou, 310003, China
| | - Guoxin Cai
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Junjie Jiang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao He
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79# Qingchun Road, Hangzhou, 310003, China
| | - Yiran Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79# Qingchun Road, Hangzhou, 310003, China
| | - Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Lu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79# Qingchun Road, Hangzhou, 310003, China
| | - Wenyi Zhao
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yan Yang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79# Qingchun Road, Hangzhou, 310003, China
| | - Yiqin Zhang
- Department of Informatics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guanghao Wu
- School of Clinical Medicine, Hangzhou Normal University Medical College, Hangzhou, China
| | - Haiyong Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79# Qingchun Road, Hangzhou, 310003, China
| | - Zhan Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79# Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
6
|
Ouyang L, Li J, Chen X, Huang H, Tian Y, Li X, Pang D, Wei X, Xie J, Wang L, Liu D, Tu P, Li J, Hu Z. Chinese dragon's blood ethyl acetate extract suppresses gastric cancer progression through induction of apoptosis and autophagy mediated by activation of MAPK and downregulation of the mTOR-Beclin1 signalling cascade. Phytother Res 2023; 37:689-701. [PMID: 36245270 DOI: 10.1002/ptr.7652] [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: 06/20/2022] [Revised: 07/24/2022] [Accepted: 08/02/2022] [Indexed: 11/11/2022]
Abstract
Gastric cancer (GC) is a malignancy with high morbidity and mortality. Chinese dragon's blood is a traditional Chinese medicine derived from the red resin of Dracaena cochinchinensis (Lour.) S. C. Chen. However, the antigastric cancer effect of Chinese dragon's blood has not yet been reported. Herein, we demonstrated that Chinese dragon's blood ethyl acetate extract (CDBEE) suppressed the proliferative and metastatic potential of human gastric cancer MGC-803 and HGC-27 cells. CDBEE suppressed epithelial-mesenchymal transition in MGC-803 and HGC-27 cells. Moreover, CDBEE induced apoptotic and autophagic cell death in MGC-803 and HGC-27 cells. The cytotoxicity of CDBEE in human gastric epithelial GES-1 cells was dramatically weaker than that in human gastric cancer cells. Mechanistically, the activation of the mitogen-activated protein kinase (MAPK) signalling pathway was involved in the growth inhibition of MGC-803 and HGC-27 cells by CDBEE. Additionally, CDBEE-induced autophagic cell death was mediated by downregulation of the mammalian target of rapamycin (mTOR)-Beclin1 signalling cascade and upregulation of the ATG3/ATG7-LC3 signalling cascade. Importantly, CDBEE exhibited potent anti-GC efficacy in vivo without obvious toxicity or side effects. Therefore, CDBEE may be a promising candidate drug for the treatment of gastric cancer, especially for GC patients with aberrant MAPK signalling or mTOR signalling.
Collapse
Affiliation(s)
- Lishan Ouyang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaqi Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaonan Chen
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Huiming Huang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Tian
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xingxing Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Daoran Pang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,Innovation Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuejiao Wei
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jinxin Xie
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Longyan Wang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dongxiao Liu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
NAMBARA SHO, MASUDA TAKAAKI, HIROSE KOSUKE, HU QINGJIANG, TOBO TARO, OZATO YUKI, KURASHIGE JUNJI, HIRAKI YOSHIKI, HISAMATSU YUICHI, IGUCHI TOMOHIRO, SUGIMACHI KEISHI, OKI EIJI, YOSHIZUMI TOMOHARU, MIMORI KOSHI. Rab27b, a Regulator of Exosome Secretion, Is Associated With Peritoneal Metastases in Gastric Cancer. Cancer Genomics Proteomics 2023; 20:30-39. [PMID: 36581340 PMCID: PMC9806673 DOI: 10.21873/cgp.20362] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/02/2022] [Accepted: 10/05/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND/AIM Peritoneal metastasis (PM) of gastric cancer (GC) leads to poor clinical outcomes. Tumor-derived exosomes promote metastasis via communication between tumor cells and host cells. In this study, we investigated the effect of Rab27, which is required for exosome secretion, on the PM of GC. MATERIALS AND METHODS We established a stable knockdown of two Rab27 homologs, Rab27a and Rab27b, in human GC cells (58As9) with a high potential of PM. We examined the level of exosome secretion from Rab27-knockdown 58As9 cells by Western blotting and the ability of Rab27b knockdown to suppress PM in 58As9 cells using a mouse xenograft model. In vitro proliferation and invasion assays were performed in the Rab27b-knockdown cells. Next, Rab27b expression was evaluated in human GC tissues by immunohistochemistry. Finally, we assessed the clinicopathological and prognostic significance of Rab27b expression by RT-qPCR in both our and other TCGA datasets of GC. RESULTS Rab27a and Rab27b knockdown in 58As9 cells decreased the secretion of exosomes, characterized by the endocytic marker CD63. Rab27b knockdown decreased PM in vivo without affecting the in vitro proliferation or invasion ability of 58As9 cells. In human GC tissues, Rab27b was overexpressed in tumor cells. The overall and recurrence-free survival rates were significantly lower in GC patients with high compared to low Rab27b mRNA expression in our and other TCGA datasets. CONCLUSION Rab27b expression potentially serves as a poor prognostic biomarker, possibly affecting PM via exosome secretion from GC cells.
Collapse
Affiliation(s)
- SHO NAMBARA
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan,Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - TAKAAKI MASUDA
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - KOSUKE HIROSE
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - QINGJIANG HU
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan,Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - TARO TOBO
- Department of Clinical Laboratory Medicine, Kyushu University Beppu Hospital, Beppu, Japan
| | - YUKI OZATO
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - JUNJI KURASHIGE
- Department of Surgery, National Hospital Organization, Kumamoto Medical Center, Kumamoto, Japan
| | - YOSHIKI HIRAKI
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - YUICHI HISAMATSU
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - TOMOHIRO IGUCHI
- Department of Surgery, Saiseikai Fukuoka General Hospital, Fukuoka, Japan
| | - KEISHI SUGIMACHI
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan,Department of Gastroenterological Surgery, National Kyushu Cancer Center, Fukuoka, Japan
| | - EIJI OKI
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - TOMOHARU YOSHIZUMI
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - KOSHI MIMORI
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| |
Collapse
|
8
|
He H, Wu J, Liang M, Xiao Y, Wei X, Cao Y, Chen Z, Lin T, Ye M. Cuprous oxide-based nanocrystals with combined chemo/chemodynamic therapy to increase tumor drug sensitivity by reducing mitochondria-derived adenosine-triphosphate. Drug Deliv 2022; 29:3134-3141. [PMID: 36154354 PMCID: PMC9518603 DOI: 10.1080/10717544.2022.2121450] [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] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) tumor is a serious disease with high mortality rates and morbidity rates worldwide. Chemotherapy is a key treatment for GI, however, systematic side effects and inevitable drug resistance complicate the situation. In the process of therapy, P-glycoprotein (P-gp) could remove chemotherapy drugs from cells, thus causing multi-drug resistance. Chemodynamic therapy (CDT) utilizing Fenton chemistry has been used for cancer therapy, along with various combination therapies. The reactive oxygen species produced by CDT could inhibit P-gp’s efflux pump function, which reduce chemoagents excretion and reverse drug resistance. In the present study, we developed novel nanocrystals (Cu2O@Pt NCs) to overcome drug resistance by reducing mitochondria-derived ATP through chemo/CDT in GI cancer. Furthermore, in vivo results in tumor-bearing mice demonstrated that treatment with Cu2O@Pt NCs with CDT and chemotherapy could achieve the most effective antitumor therapeutic effect with the least amounts of adverse effects. As a result, Cu2O@Pt NCs could provide a promising strategy for chemo/CDT-synergistic therapy.
Collapse
Affiliation(s)
- Haoran He
- The Second People's Hospital of Nanhai District, Guangdong Provincial People's Hosptial's Nanhai Hospital, Foshan, Guangdong Province, China
| | - Jiaming Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province, China
| | - Min Liang
- Department of Oncology, Innovation Centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yao Xiao
- Department of Oncology, Innovation Centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xuejian Wei
- The Second People's Hospital of Nanhai District, Guangdong Provincial People's Hosptial's Nanhai Hospital, Foshan, Guangdong Province, China
| | - Yuqin Cao
- The Second People's Hospital of Nanhai District, Guangdong Provincial People's Hosptial's Nanhai Hospital, Foshan, Guangdong Province, China
| | - Zhiheng Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang Province, China
| | - Tian Lin
- Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Miaosheng Ye
- The Second People's Hospital of Nanhai District, Guangdong Provincial People's Hosptial's Nanhai Hospital, Foshan, Guangdong Province, China
| |
Collapse
|
9
|
Molecular Docking and Intracellular Translocation of Extracellular Vesicles for Efficient Drug Delivery. Int J Mol Sci 2022; 23:ijms232112971. [PMID: 36361760 PMCID: PMC9659046 DOI: 10.3390/ijms232112971] [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: 08/10/2022] [Revised: 10/07/2022] [Accepted: 10/21/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes, mediate intercellular communication by delivering their contents, such as nucleic acids, proteins, and lipids, to distant target cells. EVs play a role in the progression of several diseases. In particular, programmed death-ligand 1 (PD-L1) levels in exosomes are associated with cancer progression. Furthermore, exosomes are being used for new drug-delivery systems by modifying their membrane peptides to promote their intracellular transduction via micropinocytosis. In this review, we aim to show that an efficient drug-delivery system and a useful therapeutic strategy can be established by controlling the molecular docking and intracellular translocation of exosomes. We summarise the mechanisms of molecular docking of exosomes, the biological effects of exosomes transmitted into target cells, and the current state of exosomes as drug delivery systems.
Collapse
|
10
|
Yu X, He S, Shen J, Huang Q, Yang P, Huang L, Pu D, Wang L, Li L, Liu J, Liu Z, Zhu L. Tumor vessel normalization and immunotherapy in gastric cancer. Ther Adv Med Oncol 2022; 14:17588359221110176. [PMID: 35872968 PMCID: PMC9297465 DOI: 10.1177/17588359221110176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is a common malignant tumor, and patients with GC have a low survival rate due to limited effective treatment methods. Angiogenesis and immune evasion are two key processes in GC progression, and they act synergistically to promote tumor progression. Tumor vascular normalization has been shown to improve the efficacy of cancer immunotherapy, which in turn may be improved through enhanced immune stimulation. Therefore, it may be interesting to identify synergies between immunomodulatory agents and anti-angiogenic therapies in GC. This strategy aims to normalize the tumor microenvironment through the action of the anti-vascular endothelial growth factor while stimulating the immune response through immunotherapy and prolonging the survival of GC patients.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Shan He
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jian Shen
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Qiushi Huang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Peng Yang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Lin Huang
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dan Pu
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Wang
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Lu Li
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Jinghua Liu
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Linyi, Shandong 276000, People's Republic of China
| | - Zelong Liu
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, No. 37, Guo Xue Xiang, Wuhou District, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|