1
|
Mechahougui H, Chevallay M, Cauchy F, Chaveau N, Puppa G, Koessler T, Monig S. Complete response of a metastatic microsatellite-stable gastric cancer after neoadjuvant chemoimmunotherapy: should we still operate? A case report and review of the literature. Front Oncol 2024; 14:1440046. [PMID: 39634260 PMCID: PMC11614720 DOI: 10.3389/fonc.2024.1440046] [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: 05/28/2024] [Accepted: 10/16/2024] [Indexed: 12/07/2024] Open
Abstract
Gastric cancer often presents at an advanced stage in Western populations due to a lack of screening programs, leading to poor prognoses. Historically, palliative chemotherapy resulted in a median survival of 9.9 months. However, the introduction of the FLOT regimen and immunotherapy has significantly altered treatment outcomes. Oligometastatic gastric cancer, defined as metastasis limited to a single organ or a few sites, has emerged as a distinct subgroup with improved survival when treated with a combination of systemic and local therapies. We present the case of a 54-year-old male patient diagnosed with microsatellite-stable (MSS) oligometastatic gastric adenocarcinoma, including liver and peritoneal metastases, who achieved a complete pathological response following neoadjuvant chemoimmunotherapy with FOLFOX and nivolumab. Despite unfavorable prognostic factors, such as liver involvement and positive peritoneal cytology, the patient responded well to the treatment, allowing curative surgery. Postoperative histology confirmed complete regression of both the primary tumor and metastases, with no recurrence observed at the 1-year follow-up. This case shows the potential of combined chemoimmunotherapy to convert previously inoperable MSS gastric cancer to surgical candidates. Further research is needed to better define patient selection criteria and assess long-term outcomes for these patients.
Collapse
Affiliation(s)
- Hiba Mechahougui
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Mickael Chevallay
- Department of Visceral Surgery, Geneva University Hospital, Geneva, Switzerland
| | - François Cauchy
- Department of Visceral Surgery, Geneva University Hospital, Geneva, Switzerland
| | - Nicolas Chaveau
- Division of Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Giacomo Puppa
- Division of Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Thibaud Koessler
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Stefan Monig
- Department of Visceral Surgery, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
2
|
Provenzano L, Gwee YX, Conca V, Lonardi S, Bozzarelli S, Tamburini E, Passardi A, Zaniboni A, Tosi F, Aprile G, Nasca V, Boccaccino A, Ambrosini M, Vetere G, Carullo M, Guaglio M, Battaglia L, Zhao JJ, Chia DKA, Yong WP, Tan P, So J, Kim G, Shabbir A, Ong CAJ, Casella F, Cremolini C, Bencivenga M, Sundar R, Pietrantonio F. Unveiling the prognostic significance of malignant ascites in advanced gastrointestinal cancers: a marker of peritoneal carcinomatosis burden. Ther Adv Med Oncol 2024; 16:17588359241289517. [PMID: 39502404 PMCID: PMC11536604 DOI: 10.1177/17588359241289517] [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: 05/25/2024] [Accepted: 09/19/2024] [Indexed: 11/08/2024] Open
Abstract
Background Ascites is common in advanced gastrointestinal cancers with peritoneal metastases (PM) and negatively impacts patient survival. No study to date has specifically evaluated the relationship between ascites, PM and survival outcomes in metastatic colorectal cancer (mCRC) and metastatic gastric cancer (mGC). Objectives This study aims to investigate and elucidate the relationship between malignant ascites, PM and survival outcomes in both mCRC and mGC patients. Design This is a retrospective analysis of prospectively collected clinical trial data of mCRC and mGC patients with PM. Methods We performed two pooled analyses, firstly of two Italian randomized trials enrolling patients with mCRC eligible for systemic therapy (TRIBE2; VALENTINO), and secondly of gastric cancer and peritoneal metastasis (GCPM) patients who underwent bi-directional therapeutic treatment comprising systemic and peritoneal-directed therapies. Results Of 900 mCRC patients, 39 (4.3%) had PM with malignant ascites. Compared to the group without PM, median progression-free and overall survival were significantly inferior in the ascites group (hazard ratio (HR) for progression-free survival (PFS) 1.68, 95% confidence interval (CI): 1.21-2.35, p = 0.007; HR for overall survival (OS) 2.14, 95% CI: 1.57-3.01, p < 0.001), but not in the group of PM without ascites (HR for PFS 1.10, 95% CI: 0.91 - 1.34; HR for OS 1.04, 95% CI: 0.84 - 1.30). Of 170 patients with GCPM, those with ascites had higher median Peritoneal Cancer Index scores (23 vs 9, p < 0.001). Median OS was significantly inferior among those with ascites compared to those without (13.0 vs 21.0 months, HR 1.71, 95% CI: 1.16-2.52, p = 0.007). Conclusion Ascites identifies a subgroup of patients with PM and poor outcomes, for whom tailored research are needed.
Collapse
Affiliation(s)
- Leonardo Provenzano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Yong Xiang Gwee
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Singapore
| | - Veronica Conca
- Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Sara Lonardi
- Medical Oncology 3 Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Silvia Bozzarelli
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’, Meldola, Italy
| | - Alberto Zaniboni
- Unity of Oncology, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Federica Tosi
- Department of Hematology, Oncology and Molecular Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Azienda ULSS8, Vicenza, Italy
| | - Vincenzo Nasca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Boccaccino
- Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Margherita Ambrosini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | - Marcello Guaglio
- Colorectal Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luigi Battaglia
- Colorectal Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Joseph Jonathan Zhao
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Singapore
| | - Daryl Kai Ann Chia
- Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
| | - Wei Peng Yong
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Patrick Tan
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jimmy So
- Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Surgical Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
| | - Guowei Kim
- Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
- Division of Surgical Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
| | - Asim Shabbir
- Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
- Division of Surgical Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore, Singapore
| | - Chin-Ann Johnny Ong
- Department of Sarcoma, Peritoneal and Rare Tumors (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
- Unity of Oncology, University Hospital of Pisa, Pisa, Italy
| | - Maria Bencivenga
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
- Department of Surgery, Dentistry, Pediatrics and Gynaecology, Upper GI Surgery Unit, University of Verona, Verona, Italy
| | - Raghav Sundar
- Department of Haematology–Oncology, National University Cancer Institute, 1E Kent Ridge Road, Singapore 119228, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.Venezian, 1, Milan 20133, Italy
| |
Collapse
|
3
|
Du WY, Masuda H, Nagaoka K, Yasuda T, Kuge K, Seto Y, Kakimi K, Nomura S. Janus kinase inhibitor overcomes resistance to immune checkpoint inhibitor treatment in peritoneal dissemination of gastric cancer in C57BL/6 J mice. Gastric Cancer 2024; 27:971-985. [PMID: 38805119 PMCID: PMC11335826 DOI: 10.1007/s10120-024-01514-5] [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: 02/21/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Cancer immunotherapy aims to unleash the immune system's potential against cancer cells, providing sustained relief for tumors responsive to immune checkpoint inhibitors (ICIs). While promising in gastric cancer (GC) trials, the efficacy of ICIs diminishes in the context of peritoneal dissemination. Our objective is to identify strategies to enhance the impact of ICI treatment specifically for cases involving peritoneal dissemination in GC. METHODS The therapeutic efficacy of anti-PD1, CTLA4 treatment alone, or in combination was assessed using the YTN16 peritoneal dissemination tumor model. Peritoneum and peritoneal exudate cells were collected for subsequent analysis. Immunohistochemical staining, flow cytometry, and bulk RNA-sequence analyses were conducted to evaluate the tumor microenvironment (TME). A Janus kinase inhibitor (JAKi) was introduced based on the pathway analysis results. RESULTS Anti-PD1 and anti-CTLA4 combination treatment (dual ICI treatment) demonstrated therapeutic efficacy in certain mice, primarily mediated by CD8 + T cells. However, in mice resistant to dual ICI treatment, even with CD8 + T cell infiltration, most of the T cells exhibited an exhaustion phenotype. Notably, resistant tumors displayed abnormal activation of the Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) pathway compared to the untreated group, with observed infiltration of macrophages, neutrophils, and Tregs in the TME. The concurrent administration of JAKi rescued CD8 + T cells function and reshaped the immunosuppressive TME, resulting in enhanced efficacy of the dual ICI treatment. CONCLUSION Dual ICI treatment exerts its anti-tumor effects by increasing tumor-specific CD8 + T cell infiltration, and the addition of JAKi further improves ICI resistance by reshaping the immunosuppressive TME.
Collapse
Affiliation(s)
- Wan-Ying Du
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Hiroki Masuda
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
- Department of Gastrointestinal Surgery, Nippon Medical School, Tokyo, Japan
| | - Koji Nagaoka
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Tomohiko Yasuda
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
- Department of Gastrointestinal Surgery, Nippon Medical School, Tokyo, Japan
| | - Komei Kuge
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
- Department of Gastrointestinal Surgery, Nippon Medical School, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
- Department of Clinical Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-Ku, Tokyo, 142-8501, Japan.
| |
Collapse
|
4
|
Chen S, Wang Z. Integration of mult-omics and nucleotide metabolism reprogramming signature analysis reveals gastric cancer immunological and prognostic features. Cancer Cell Int 2024; 24:212. [PMID: 38880869 PMCID: PMC11180389 DOI: 10.1186/s12935-024-03396-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Gastric cancer is a frequent and lethal solid tumor that has a poor prognosis and treatment result. Reprogramming of nucleotide metabolism is a characteristic of cancer development and progression. METHODS We used a variety of machine learning techniques to create a novel nucleotide metabolism-related index (NMRI) using gastric cancer sample data obtained from the TCGA and GEO databases. This index is based on genes associated to nucleotide metabolism. Gastric cancer patients were categorized into high and low NMRI groups based on NMRI results. The clinical features, tumor immune microenvironment, response to chemotherapy, and response to immunotherapy were then thoroughly examined. In vitro experiments were then used to confirm the biological role of SERPINE1 in gastric cancer. RESULTS The four nucleotide metabolism-related genes that make up NMRI (GAMT, ORC1, CNGB3, and SERPINE1) were verified in an external dataset and are a valid predictor of prognosis for patients with gastric cancer. The high NMRI group was more responsive to immunotherapy and had greater levels of immune cell infiltration than the low NMRI group. The proliferation and migration of stomach cancer was shown to be decreased by SERPINE1 knockdown in vitro. CONCLUSIONS This study's NMRI can reliably predict a patient's prognosis for stomach cancer and pinpoint the patient group that will benefit from immunotherapy, offering important new information on the clinical treatment of stomach cancer.
Collapse
Affiliation(s)
- Shaofei Chen
- Gastrointestinal Surgery, Wuhan Union Hospital, Wuhan, 430022, Hubei, China
| | - Zhiyong Wang
- Gastrointestinal Surgery, Wuhan Union Hospital, Wuhan, 430022, Hubei, China.
| |
Collapse
|
5
|
Jeong SA, Kim S, Lee IS, Yoo MW, Kim BS. Does total omentectomy prevent peritoneal seeding for advanced gastric cancer with serosal invasion? Surg Endosc 2024; 38:97-104. [PMID: 37917161 DOI: 10.1007/s00464-023-10514-y] [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] [Accepted: 10/08/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Radical gastrectomy is composed of gastrectomy, lymph node dissection, and omentectomy. Total omentectomy (TO) is expected to reduce the incidence of peritoneal recurrence. We aimed to investigate the necessity of TO for advanced gastric cancer (AGC) with serosal invasion. METHODS We retrospectively reviewed 310 patients who underwent radical gastrectomy with TO and 93 patients who underwent partial omentectomy (PO) for gastric cancer with serosal invasion between August, 2005 and December, 2017. Finally, 91 patients in the PO group and 91 in the TO group were enrolled based on a 1:1 propensity-score matching analysis. We evaluated surgical and oncological outcomes, including 5-year overall and recurrence-free survival rates. RESULTS There was no statistically significant difference between the two groups in postoperative complications. Recurrence sites showed similar patterns in both groups, including peritoneal recurrence (PO vs. TO, 18.7% vs. 28.6%; p = 0.188). Five-year overall survival was better in the PO group (p = 0.018), while 5-year recurrence-free survival was similar in both groups (p = 0.066). CONCLUSION TO might not be an essential part of preventing peritoneal recurrence for AGC with serosal invasion. PO could be considered a radical gastrectomy for T4a gastric cancer.
Collapse
Affiliation(s)
- Seong-A Jeong
- Department of Surgery, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, 25440, Korea
| | - Sehee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - In-Seob Lee
- Department of Gastrointestinal Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Moon-Won Yoo
- Department of Gastrointestinal Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Beom Su Kim
- Department of Gastrointestinal Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
6
|
Peng X, Cai Z, Chen D, Ye F, Hong L. Prognostic significance and immune characteristics of APOE in gastric cancer. Aging (Albany NY) 2023; 15:13840-13853. [PMID: 38054821 PMCID: PMC10756126 DOI: 10.18632/aging.205265] [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: 06/26/2023] [Accepted: 10/23/2023] [Indexed: 12/07/2023]
Abstract
Gastric cancer (GC) is a prevalent malignancy affecting the digestive system, and it is the second leading cause of cancer-related mortality worldwide. Immunotherapy presents a potential lifeline for patients with advanced gastric cancer, emphasizing the need to find new molecular targets that improve the response to immunotherapy. In our research, we conducted a comprehensive bioinformatic analysis to investigate the expression profiles of apolipoprotein E (APOE) transcription. Subsequently, we examined the correlation between APOE transcription and the prognosis of GC patients. Additionally, we evaluated the connection between APOE transcription and immune cells abundance. To validate our findings, we conducted immunohistochemistry experiment to ascertain the level of APOE protein in GC patients and assessed its prognostic role in a cohort of 97 GC individuals. Our results revealed that APOE is increased in GC tissues, and APOE displays diagnostic potential in distinguishing GC from normal tissues. Notably, upregulated APOE expression in GC patients is associated with unfavorable overall survival. Differential APOE expression was further observed across different immune subtypes of GC, indicating its involvement in immune cell activation and infiltration. Moreover, we detected increased APOE protein expression in GC tissues, which exhibited a strong correlation with poor survival outcomes. In light of these findings, APOE has become a crucial prognostic molecular with immunomodulatory function in GC. These results underscore the significance of APOE across various cancer types, including GC, and provide valuable insights into its role from both a bioinformatics and clinical perspective.
Collapse
Affiliation(s)
- Xiulan Peng
- Department of Oncology, The Second Affiliated Hospital of Jianghan University, Wuhan, Hubei 430050, China
| | - Zhen Cai
- Department of Operation Room, The Second Affiliated Hospital of Jianghan University, Wuhan, Hubei 430050, China
| | - Duansi Chen
- Department of Oncology, Suizhou Zengdu Hospital, Suizhou, Hubei 441300, China
| | - Fei Ye
- Department of Radiology, The Second Affiliated Hospital of Jianghan University, Wuhan, Hubei 430050, China
| | - Lifeng Hong
- Department of Cardiology, The Second Affiliated Hospital of Jianghan University, Wuhan, Hubei 430050, China
| |
Collapse
|
7
|
Xiang Y, Wan F, Ren Y, Yang D, Xiang K, Zhu B, Ruan X, Li S, Zhang L, Liu X, Si Y, Liu Y. Polyphyllin VII induces autophagy-dependent ferroptosis in human gastric cancer through targeting T-lymphokine-activated killer cell-originated protein kinase. Phytother Res 2023; 37:5803-5820. [PMID: 37632389 DOI: 10.1002/ptr.7986] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/20/2023] [Accepted: 08/06/2023] [Indexed: 08/28/2023]
Abstract
T-lymphokine-activated killer cell-originated protein kinase (TOPK) is a serine-threonine kinase that is overexpressed in gastric cancer (GC) and promotes tumor progression. Polyphyllin VII (PPVII), a pennogenin isolated from the rhizomes of Paris polyphylla, shows anticancer effects. Here, we explored the antitumor activity and mechanism of PPVII in GC. Ferroptosis was detected by transmission electron microscope, malondialdehyde, and iron determination assays. Autophagy and its upstream signaling pathway were detected by Western blot, and gene alterations. The binding of PPVII and TOPK was examined through microscale thermophoresis and drug affinity responsive target stability assays. An in vivo mouse model was performed to evaluate the therapeutic of PPVII. PPVII inhibits GC by inducing autophagy-mediated ferroptosis. PPVII promotes the degradation of ferritin heavy chain 1, which is responsible for autophagy-mediated ferroptosis. PPVII activates the Unc-51-like autophagy-activating kinase 1 (ULK1) upstream of autophagy. PPVII inhibits the activity of TOPK, thereby weakening the inhibition of downstream ULK1. PPVII stabilizes the dimer of the inactive form of TOPK by direct binding. PPVII inhibits tumor growth without causing obvious toxicity in vivo. Collectively, this study suggests that PPVII is a potential agent for the treatment of GC by targeting TOPK to activate autophagy-mediated ferroptosis.
Collapse
Affiliation(s)
- Yuchen Xiang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Fang Wan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yuliang Ren
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Dan Yang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ke Xiang
- Gucheng People's Hospital, Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Bingxin Zhu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuzhi Ruan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shuzhen Li
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
| | - Liang Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
8
|
Pan J, Gao Y. Prognostic significance and immune characteristics of GPR27 in gastric cancer. Aging (Albany NY) 2023; 15:9144-9166. [PMID: 37702614 PMCID: PMC10522374 DOI: 10.18632/aging.205023] [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: 05/19/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023]
Abstract
Gastric cancer (GC) is one of the most typical cancerous neoplasms occurring in the digestive system. For advanced GC, immunotherapy is the final option for them to prolong survival time. Hence, we aimed to identify new molecular targets to enhance the immunotherapy response in GC individuals. Then we applied bioinformatic analysis to explore the expression profiles of G-protein-coupled receptor 27 (GPR27) transcription and GPR27 methylation. The associations between survival of GC patients and GPR27 transcription and methylation were then analyzed. We also studied the link between GPR27 expression and levels of immune cell infiltration. Finally, we gained insights into the prognostic role of GPR27 protein in 97 cases of GC individuals. According to datasets gained from TCGA, GPR27 mRNA is expressed lower in GC tissues. Down-regulation of GPR27 transcription was related with better survival in GC individuals, and GPR27 cg03024619 had the most significant prognostic value (HR=0.553, P<0.0001). In addition, the expression level of GPR27 has a clear interaction with immune cells' infiltration and their markers. Single-cell analysis displayed that GPR27 is mainly expressed in macrophages. Finally, down-regulation of GPR27 protein was observed in GC tissues and correlated with better survival outcomes. GPR27 can serve as an important prognostic biomarker and exert an immunomodulatory role in GC. Our findings highlight the significance of GPR27 in a variety of cancers, including GC, and provide clues for a better understanding of GPR27 from bioinformatics and clinically validated perspective.
Collapse
Affiliation(s)
- Jun Pan
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yuanjun Gao
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| |
Collapse
|
9
|
Yang A, Zhang Z, Chaurasiya S, Park AK, Jung A, Lu J, Kim SI, Priceman S, Fong Y, Woo Y. Development of the oncolytic virus, CF33, and its derivatives for peritoneal-directed treatment of gastric cancer peritoneal metastases. J Immunother Cancer 2023; 11:e006280. [PMID: 37019471 PMCID: PMC10083877 DOI: 10.1136/jitc-2022-006280] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) that metastasizes to the peritoneum is fatal. CF33 and its genetically modified derivatives show cancer selectivity and oncolytic potency against various solid tumors. CF33-hNIS and CF33-hNIS-antiPDL1 have entered phase I trials for intratumoral and intravenous treatments of unresectable solid tumors (NCT05346484) and triple-negative breast cancer (NCT05081492). Here, we investigated the antitumor activity of CF33-oncolytic viruses (OVs) against GC and CF33-hNIS-antiPDL1 in the intraperitoneal (IP) treatment of GC peritoneal metastases (GCPM). METHODS We infected six human GC cell lines AGS, MKN-45, MKN-74, KATO III, SNU-1, and SNU-16 with CF33, CF33-GFP, or CF33-hNIS-antiPDL1 at various multiplicities of infection (0.01, 0.1, 1.0, and 10.0), and performed viral proliferation and cytotoxicity assays. We used immunofluorescence imaging and flow cytometric analysis to verify virus-encoded gene expression. We evaluated the antitumor activity of CF33-hNIS-antiPDL1 following IP treatment (3×105 pfu × 3 doses) in an SNU-16 human tumor xenograft model using non-invasive bioluminescence imaging. RESULTS CF33-OVs showed dose-dependent infection, replication, and killing of both diffuse and intestinal subtypes of human GC cell lines. Immunofluorescence imaging showed virus-encoded GFP, hNIS, and anti-PD-L1 antibody scFv expression in CF33-OV-infected GC cells. We confirmed GC cell surface PD-L1 blockade by virus-encoded anti-PD-L1 scFv using flow cytometry. In the xenograft model, CF33-hNIS-antiPDL1 (IP; 3×105 pfu × 3 doses) treatment significantly reduced peritoneal tumors (p<0.0001), decreased amount of ascites (62.5% PBS vs 25% CF33-hNIS-antiPDL1) and prolonged animal survival. At day 91, seven out of eight mice were alive in the virus-treated group versus one out of eight in the control group (p<0.01). CONCLUSIONS Our results show that CF33-OVs can deliver functional proteins and demonstrate effective antitumor activity in GCPM models when delivered intraperitoneally. These preclinical results will inform the design of future peritoneal-directed therapy in GCPM patients.
Collapse
Affiliation(s)
- Annie Yang
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Zhifang Zhang
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Shyambabu Chaurasiya
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Anthony K Park
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Audrey Jung
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Saul Priceman
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
10
|
Graversen M, Detlefsen S, Ainsworth AP, Fristrup CW, Knudsen AO, Pfeiffer P, Tarpgaard LS, Mortensen MB. Treatment of Peritoneal Metastasis with Pressurized Intraperitoneal Aerosol Chemotherapy: Results from the Prospective PIPAC-OPC2 Study. Ann Surg Oncol 2023; 30:2634-2644. [PMID: 36602663 DOI: 10.1245/s10434-022-13010-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Pressurized Intraperitoneal Aerosol chemotherapy (PIPAC) is a local treatment for peritoneal metastasis (PM). Prospective data are scarce and evaluation of treatment response remains difficult. This study evaluated the use of the Peritoneal Regression Grading score (PRGS) and its prognostic value. PATIENTS AND METHODS This was a prospective, controlled phase II trial in patients with PM from gastrointestinal, gynaecological, hepatopancreatobiliary, primary peritoneal, or unknown primary cancer. Patients in performance status 0-1, with a non-obstructed gastrointestinal tract, and a maximum of one extraperitoneal metastasis were eligible. Colorectal or appendiceal PM had PIPAC with oxaliplatin, other primaries had PIPAC with cisplatin and doxorubicin. Biopsies were taken at each PIPAC and evaluated using the PRGS. Quality-of-life questionnaires were reported at baseline and after three PIPACs. RESULTS One hundred ten patients were treated with 336 PIPACs (median 3, range 1-12). One hundred patients had prior palliative chemotherapy and 45 patients received bidirectional treatment. Complete or major histological response to treatment (PRGS 1-2) was observed in 38 patients (61%) who had three PIPACs, which was the only independent prognostic factor in a multivariate analysis. The median overall survival (mOS) from PIPAC 1 was 10 months, while patients with PM from gastric, colorectal, and pancreatic cancer had a mOS of 7.4, 16.7, and 8.2 months, respectively. Global health scores were significantly reduced, but patients were less fatigued, nauseated, constipated, and had better appetite after three PIPACs. CONCLUSIONS PIPAC with oxaliplatin or cisplatin and doxorubicin was able to induce a major or complete histological response during three PIPACs, which may provide significant prognostic information, both at baseline and after treatment.
Collapse
Affiliation(s)
- Martin Graversen
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark. .,Department of Surgery, Odense University Hospital, Odense, Denmark. .,OPEN-Open Patient data Explorative Network, Odense University Hospital, Odense, Region of Southern Denmark, Denmark. .,OPAC-Odense Pancreas Center, Odense University Hospital, Odense, Denmark. .,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| | - S Detlefsen
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,OPAC-Odense Pancreas Center, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| | - A P Ainsworth
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark.,OPAC-Odense Pancreas Center, Odense University Hospital, Odense, Denmark
| | - C W Fristrup
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark.,OPAC-Odense Pancreas Center, Odense University Hospital, Odense, Denmark
| | - A O Knudsen
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark
| | - P Pfeiffer
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,OPAC-Odense Pancreas Center, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark
| | - L S Tarpgaard
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark
| | - M B Mortensen
- Odense PIPAC Center, Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark.,OPAC-Odense Pancreas Center, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
11
|
Yamamoto M, Kurino T, Matsuda R, Jones HS, Nakamura Y, Kanamori T, Tsuji AB, Sugyo A, Tsuda R, Matsumoto Y, Sakurai Y, Suzuki H, Sano M, Osada K, Uehara T, Ishii Y, Akita H, Arano Y, Hisaka A, Hatakeyama H. Delivery of aPD-L1 antibody to i.p. tumors via direct penetration by i.p. route: Beyond EPR effect. J Control Release 2022; 352:328-337. [PMID: 36280153 DOI: 10.1016/j.jconrel.2022.10.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/28/2022] [Accepted: 10/18/2022] [Indexed: 11/08/2022]
Abstract
Chemotherapy for peritoneal dissemination is poorly effective owing to limited drug transfer from the blood to the intraperitoneal (i.p.) compartment after intravenous (i.v.) administration. i.p. chemotherapy has been investigated to improve drug delivery to tumors; however, the efficacy continues to be debated. As anticancer drugs have low molecular weight and are rapidly excreted through the peritoneal blood vessels, maintaining the i.p. concentration as high as expected is a challenge. In this study, we examined whether i.p. administration is an efficient route of administration of high-molecular-weight immune checkpoint inhibitors (ICIs) for the treatment of peritoneal dissemination using a model of peritoneal disseminated carcinoma. After i.p. administration, the amount of anti-PD-L1 antibody transferred into i.p. tumors increased by approximately eight folds compared to that after i.v. administration. Intratumoral distribution analysis revealed that anti-PD-L1 antibodies were delivered directly from the i.p. space to the surface of tumor tissue, and that they deeply penetrated the tumor tissues after i.p. administration; in contrast, after i.v. administration, anti-PD-L1 antibodies were only distributed around blood vessels in tumor tissues via the enhanced permeability and retention (EPR) effect. Owing to the enhanced delivery, the therapeutic efficacy of anti-PD-L1 antibody in the peritoneal dissemination models was also improved after i.p. administration compared to that after i.v. administration. This is the first study to clearly demonstrate an EPR-independent delivery of ICIs to i.p. tumors by which ICIs were delivered in a massive amount to the tumor tissue via direct penetration after i.p. administration.
Collapse
Affiliation(s)
- Mayu Yamamoto
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Taiki Kurino
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Reiko Matsuda
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Haleigh Sakura Jones
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Yoshito Nakamura
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Taisei Kanamori
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Atushi B Tsuji
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Aya Sugyo
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Ryota Tsuda
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Yui Matsumoto
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Yu Sakurai
- Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Hiroyuki Suzuki
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Makoto Sano
- Division of Medical Research Planning and Development, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kensuke Osada
- Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Tomoya Uehara
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Yukimoto Ishii
- Division of Medical Research Planning and Development, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; Laboratory of DDS design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Yasushi Arano
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Hiroto Hatakeyama
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan.
| |
Collapse
|
12
|
Gastrectomy with or without Complete Omentectomy for Advanced Gastric Cancer: A Meta-Analysis. Medicina (B Aires) 2022; 58:medicina58091241. [PMID: 36143918 PMCID: PMC9503724 DOI: 10.3390/medicina58091241] [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] [Received: 07/27/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 02/03/2023] Open
Abstract
Background and Objectives: Surgery remains the only possible curative treatment for advanced gastric cancer (AGC). Peritoneal metastases are estimated to occur in approximately 55–60% AGC patients. Greater omentum is the most common metastatic area in AGC. At present, omentectomy alone or bursectomy are usually carried out during gastric cancer surgery. We performed a meta-analysis in order to evaluate long-term and short-term outcomes among AGC patients, who have undergone radical gastrectomy with or without complete omentectomy (CO). Materials and Methods: We performed a systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Meta-analysis was performed by use of RevMan (Computer program) Version 5.4. Results: The eight included studies covered an approximately 20 years long study period (2000–2018). Almost all included studies were retrospective ones and originated from Asian countries. Meta-analysis indicated gastrectomy without CO as significantly associated with longer 3-year (RR: 0.94, 95% CI: 0.90–0.98, p = 0.005) and 5-year overall survivals (OS) (RR: 0.93, 95% CI: 0.88–0.98, p = 0.007). Moreover, we found longer operative time (MD: 24.00, 95% CI: −0.45–48.45, p = 0.05) and higher estimated blood loss (MD: 194.76, 95% CI: 96.40–293.13, p = 0.0001) in CO group. Conclusions: Non-complete omentectomy (NCO) group had a statistically greater rate in 3-year and 5-year OSs than the CO group, while the CO group had significantly longer operative time and higher estimated blood loss than the NCO group. Further randomized, possibly multi-center trials may turn out of paramount importance in confirming our results.
Collapse
|
13
|
Sahin A, Ozturk SC. Intraperitoneal administration of PLGA nanoparticles could deliver the cargo to tumor-associated macrophages with less spreading peritoneal macrophages in the treatment of peritoneal carcinomatosis. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
14
|
Mari V, Angerilli V, Munari G, Scarpa M, Bao QR, Pucciarelli S, Fassan M, Spolverato G. Molecular Determinants of Peritoneal Dissemination in Gastric Adenocarcinoma. Dig Dis 2022; 41:49-65. [PMID: 35940137 DOI: 10.1159/000526333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/25/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Peritoneal dissemination represents a poor prognostic indicator in gastric cancer. Despite a comprehensive molecular characterization of this disease, no peritoneal dissemination-specific signature has been identified, limiting the tailoring of the surgical and oncological treatments. In this review, we outline the available literature focusing on the role of the different molecular pathways involved in the acquisition of peritoneal metastatic dissemination. SUMMARY According to our results, several molecular determinants are associated with peritoneal carcinomatosis and are involved in several cellular and molecular carcinogenetic processes. However, a comprehensive understanding of the complex molecular landscape of gastric carcinosis is still lacking. KEY MESSAGES More efforts should be made toward the integration of molecular and histologic data to perform a risk prediction assessment of peritoneal dissemination based on molecular profiling and histological evaluation.
Collapse
Affiliation(s)
- Valentina Mari
- Department of Surgical, General Surgery 3, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, Padua, Italy
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Giada Munari
- Veneto Institute of Oncology (I.O.V. IRCSS), Padua, Italy
| | - Marco Scarpa
- Department of Surgical, General Surgery 3, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, Padua, Italy
| | - Quoc Riccardo Bao
- Department of Surgical, General Surgery 3, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, Padua, Italy
| | - Salvatore Pucciarelli
- Department of Surgical, General Surgery 3, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, Padua, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
- Veneto Institute of Oncology (I.O.V. IRCSS), Padua, Italy
| | - Gaya Spolverato
- Department of Surgical, General Surgery 3, Oncological and Gastroenterological Sciences (DISCOG), University of Padua, Padua, Italy
| |
Collapse
|
15
|
Luo Z, Luo Y, Xiao K. A-Kinase Interacting Protein 1 Promotes Cell Invasion and Stemness via Activating HIF-1α and β-Catenin Signaling Pathways in Gastric Cancer Under Hypoxia Condition. Front Oncol 2022; 11:798557. [PMID: 35355804 PMCID: PMC8959465 DOI: 10.3389/fonc.2021.798557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/23/2021] [Indexed: 01/16/2023] Open
Abstract
Background A-Kinase interacting protein 1 (AKIP1) relates to gastric cancer growth, metastasis, and prognosis, while its regulation on gastric cancer invasion and stemness under hypoxia microenvironment is not reported. Therefore, this study aimed to explore this topic to uncover AKIP1’s role in gastric cancer under hypoxia. Methods Gastric cancer cell lines AGS and MKN45 were cultured under hypoxia condition, then transfected with AKIP1 or negative control (NC) overexpression plasmid or AKIP1 or NC knockdown plasmid. Furthermore, rescue experiments were conducted by transfecting HIF-1α or β-catenin overexpression plasmid, combined with AKIP1 or NC knockdown plasmid. Afterward, cell invasion, CD133+ cell proportion, sphere number/1,000 cells, and HIF-1α and β-catenin pathways were measured. Results The invasive cell count, CD133+ cell proportion, and sphere number/1,000 cells were enhanced in both AGS cells and MKN45 cells under hypoxia, and AKIP1 expression was also elevated. AKIP1 knockdown inhibited cell invasion, CD133+ cell proportion, sphere number/1,000 cells, HIF-1α, vascular endothelial growth factor (VEGF), β-catenin, and calcium-binding protein (CBP) expressions in AGS cells and MKN45 cells under hypoxia, while AKIP1 overexpression presented with the opposite effect. Then, in rescue experiments, HIF-1α overexpression and β-catenin overexpression both promoted cell invasion, CD133+ cell proportion, and sphere number/1,000 cells, which also attenuated the effect of AKIP1 knockdown on these functions in AGS cells and MKN45 cells. Conclusion AKIP1 promotes cell invasion and stemness via activating HIF-1α and β-catenin signaling pathways in gastric cancer under hypoxia condition.
Collapse
Affiliation(s)
- Zhenqin Luo
- Department of Comprehensive Chemotherapy, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuhang Luo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, The University of South China, Hengyang, China
| | - Ke Xiao
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|