1
|
Farokhi-Fard A, Rahmati S, Hashemi Aval NS, Barkhordari F, Bayat E, Komijani S, Aghamirza Moghim Aliabadi H, Davami F. Anti-IL-1RAP scFv-mSA-S19-TAT fusion carrier as a multifunctional platform for versatile delivery of biotinylated payloads to myeloid leukemia cells. Sci Rep 2024; 14:25080. [PMID: 39443595 PMCID: PMC11500005 DOI: 10.1038/s41598-024-76851-7] [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: 08/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive blood cancer with frequently poor clinical outcomes. This heterogeneous malignancy encompasses genetically, molecularly, and even clinically different subgroups. This makes it difficult to develop therapeutic agents that are effective for all subtypes of the disease. Therefore, a selective, universal, and adaptable delivery platform capable of carrying various types of anti-neoplastic agents is an unmet requirement in this area. Two multifunctional fusion proteins were designed for the delivery of biotinylated cargoes to human myeloid leukemia cells by fusing an anti-IL-1RAP single-chain antibody with streptavidin (tetramer or monomer), a cell-penetrating peptide (CPP), and an endosomolytic peptide in a single biomacromolecule. The designed fusions were analyzed primarily in silico, and the biofunctionality of the selected fusion was fully characterized via several binding assays, hemolysis assay, confocal microscopy and cell cytotoxicity assay after production via the Escherichia coli (E. coli) system. The refolded protein exhibited desirable binding activity to leukemic cells, pure antigen and biotinylated BSA. Further analyses revealed efficient cellular uptake, endosomolytic activity, and nuclear penetration without any detectable cytotoxicity toward normal epithelial cells. The described platform seems to have great potential for targeted delivery of different therapeutics to malignant myeloid cells.
Collapse
MESH Headings
- Humans
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/genetics
- Recombinant Fusion Proteins/genetics
- Biotinylation
- Cell-Penetrating Peptides/chemistry
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- Cell Line, Tumor
- Drug Delivery Systems
- Streptavidin/chemistry
- Drug Carriers/chemistry
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/drug therapy
- Leukemia, Myeloid/therapy
Collapse
Affiliation(s)
- Aref Farokhi-Fard
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Saman Rahmati
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | | | | | - Elham Bayat
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Samira Komijani
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
- Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran, Iran.
| |
Collapse
|
2
|
Lee Y, English EL, Schwartzmann CM, Liu Y, Krueger JM. Sleep loss-induced oncogenic pathways are mediated via the neuron-specific interleukin-1 receptor accessory protein (AcPb). Brain Behav Immun 2024; 123:411-421. [PMID: 39343106 DOI: 10.1016/j.bbi.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/04/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024] Open
Abstract
Interleukin-1β (IL1), a pleiotropic cytokine, is involved in sleep regulation, tumor ontogeny, and immune responses. IL1 receptor adaptor proteins, including the IL1 receptor accessory protein (AcP), and its neuron-specific isoform, AcPb, are required for IL1 signaling. The AcPb isoform is resultant from alternate splicing of the AcP transcript. Our previous studies using AcPb null (AcPb-/-) mice characterized its participation in sleep regulation and emergent neuronal/glial network properties. Here, we investigated the impact of acute sleep disruption (SD) on brain cancer-related pathways in wild-type (WT) and AcPb-/- mice, employing RNA sequencing methods. In WT mice, SD increased AcPb mRNA levels, but not AcP mRNA, confirming prior similar work in rats. Transcriptome and pathway enrichment analyses demonstrated significant alterations in cancer, immune, and viral disease-related pathways in WT mice after SD, which were attenuated in AcPb-/- mice including multiple upregulated Src phosphorylation-signaling-dependent genes associated with cancer progression and metastasis. Our RNAseq findings, were analyzed within the context of The Cancer Genome Atlas Program (TCGA) data base; revealing an upregulation of sleep- and cancer-linked genes (e.g., IL-17B, IL-17RA, LCN2) across various tumors, including brain tumors, compared to normal tissues. Sleep-linked factors, identified through TCGA analyses, significantly impact patient prognosis and survival, particularly in low-grade glioma (LGG) and glioblastoma multiforme (GBM) patients. Overall, our findings suggest that SD promotes a pro-tumor environment through AcPb-modulated pathways.
Collapse
Affiliation(s)
- Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA 99202, USA; Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA.
| | - Erika L English
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA
| | - Catherine M Schwartzmann
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Yiyong Liu
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; Genomics Core, Washington State University, Spokane, WA, USA
| | - James M Krueger
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA; Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
3
|
Tao BF, Zhu HQ, Qi LN, Zhong JH, Mai RY, Ma L. Preoperative monocyte-to-lymphocyte ratio as a prognosis predictor after curative hepatectomy for intrahepatic cholangiocarcinoma. BMC Cancer 2024; 24:1179. [PMID: 39333901 PMCID: PMC11437980 DOI: 10.1186/s12885-024-12929-9] [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: 10/29/2023] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Several inflammatory indicators have been reported to have predictive value in many types of malignant cancer. This research was aimed to explore the ability of the monocyte-to-lymphocyte ratio (MLR) to predict prognosis in patients with intrahepatic cholangiocarcinoma (ICC) who subjected to curative hepatectomy. METHODS This retrospective analysis included 196 patients with ICC who underwent curative hepatectomy between May 2018 and April 2023. The predictive abilities of the preoperative MLR in assessing overall survival (OS) and disease-free survival (DFS) in those patients were compared with other inflammation-based scores, including monocyte-to-white ratio, neutrophil-to-lymphocyte ratio, neutrophil-to-white ratio, platelet-to-lymphocyte ratio, platelet-to-white ratio, and systemic immune-inflammation index, as well as tumor markers, like carcinoembryonic antigen (CEA) and carbohydrate antigen 19 - 9 (CA19-9). RESULTS The area under the time-dependent receiver operating characteristic curve indicated that the preoperative MLR had higher predictive efficiency in contrast with other inflammation-based scores and tumor markers in assessing OS and DFS. Stratifying patients according to the optimal cut-off value for the preoperative MLR, the data showed that both OS and DFS in the high MLR group were significantly worse than those in the low MLR group (p < 0.05 for all). Univariable and multivariable Cox analyses revealed that the preoperative MLR was an independent risk factor for OS and DFS in patients with ICC. In addition to predicting OS in patients with high CEA levels and predicting DFS in patients with high CA19-9 levels, patients with different CEA and CA19-9 levels were divided into completely different OS and DFS subgroups based on the risk stratification of the preoperative MLR. CONCLUSIONS Our results demonstrated that the preoperative MLR was a good prognosis indicator to predict DFS and OS following curative hepatectomy in patients with ICC.
Collapse
Affiliation(s)
- Bin-Feng Tao
- Hepatobiliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, He Di Rd. #71, Nanning, 530021, China
| | - Hai-Qiang Zhu
- Hepatobiliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, He Di Rd. #71, Nanning, 530021, China
| | - Lu-Nan Qi
- Hepatobiliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, He Di Rd. #71, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Jian-Hong Zhong
- Hepatobiliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, He Di Rd. #71, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Rong-Yun Mai
- Hepatobiliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, He Di Rd. #71, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China.
| | - Liang Ma
- Hepatobiliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, He Di Rd. #71, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China.
| |
Collapse
|
4
|
Huang L, Wu C, Xu D, Cui Y, Tang J. IL1RAP Exacerbates Sepsis-Induced Pulmonary and Spleen Injury Through Regulating CD4 + T Lymphocyte Differentiation. Immunol Invest 2024; 53:574-585. [PMID: 38329477 DOI: 10.1080/08820139.2024.2312898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
BACKGROUND Complex pathophysiological the specific mechanism of sepsis on CD4+ T-cell responses is less well understood. IL1 receptor accessory protein (IL1RAP) was found to be involved in activating host immune responses. METHOD Cecum ligation and puncture (CLP) was utilized to build a mouse sepsis model. The experiment was randomly divided into four groups: Sham, CLP, CLP + shNC, and CLP + shIL1RAP group. RESULTS qRT-PCR suggested mRNA levels of IL1RAP were decreased when IL1RAP was knocked down with the mRNA levels of IL-1β, NF-κB, and p38 decreased. Histopathology showed severe pathological damage with alveolar integrity lost, red blood cells in the alveoli, massive inflammatory cell infiltration, and the alveolar wall was thickening in the CLP group. The inflammatory cytokine levels of TNF-α, IL-1β, and IFN-γ were elevated in CLP mice by ELISA. The counts of CD4+ T cells were decreased in sepsis mice in peripheral blood, spleen, and BALF by flow cytometry. However, the above was blocked down when using shIL1RAP. Western blot suggested sh IL1RAP inhibited IL-1β, NF-κB, and p38 protein expressions. CONCLUSIONS We defined IL1RAP as a new target gene through NF-κB/MAPK pathways regulating CD4+ T lymphocyte differentiation mediated the progression of sepsis, which is potentially exploitable for immunotherapy.
Collapse
Affiliation(s)
- Liou Huang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Chunrong Wu
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Dan Xu
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yuhui Cui
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Zarezadeh Mehrabadi A, Shahba F, Khorramdelazad H, Aghamohammadi N, Karimi M, Bagherzadeh K, Khoshmirsafa M, Massoumi R, Falak R. Interleukin-1 receptor accessory protein (IL-1RAP): A magic bullet candidate for immunotherapy of human malignancies. Crit Rev Oncol Hematol 2024; 193:104200. [PMID: 37981104 DOI: 10.1016/j.critrevonc.2023.104200] [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: 04/05/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
IL-1, plays a role in some pathological inflammatory conditions. This pro-inflammatory cytokine also has a crucial role in tumorigenesis and immune responses in the tumor microenvironment (TME). IL-1 receptor accessory protein (IL-1RAP), combined with IL-1 receptor-1, provides a functional complex for binding and signaling. In addition to the direct role of IL-1, some studies demonstrated that IL1-RAP has essential roles in the progression, angiogenesis, and metastasis of solid tumors such as gastrointestinal tumors, lung carcinoma, glioma, breast and cervical cancers. This molecule also interacts with FLT-3 and c-Kit tyrosine kinases and is involved in the pathogenesis of hematological malignancies such as acute myeloid lymphoma. Additionally, IL-1RAP interacts with solute carrier family 3 member 2 (SLC3A2) and thereby increasing the resistance to anoikis and metastasis in Ewing sarcoma. This review summarizes the role of IL-1RAP in different types of cancers and discusses its targeting as a novel therapeutic approach for malignancies.
Collapse
Affiliation(s)
- Ali Zarezadeh Mehrabadi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Faezeh Shahba
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Nazanin Aghamohammadi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Milad Karimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kowsar Bagherzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Khoshmirsafa
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden.
| | - Reza Falak
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Deng J, Zhang W, Xu M, Liu X, Ren T, Li S, Sun Q, Xue C, Zhou J. Value of spectral CT parameters in predicting the efficacy of neoadjuvant chemotherapy for gastric cancer. Clin Radiol 2024; 79:51-59. [PMID: 37914603 DOI: 10.1016/j.crad.2023.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/26/2023] [Accepted: 08/30/2023] [Indexed: 11/03/2023]
Abstract
AIM To investigate the value of pre-chemotherapy spectral computed tomography (CT) parameters in predicting neoadjuvant chemotherapy (NAC) response in gastric cancer (GC). MATERIALS AND METHODS Sixty patients with GC who received NAC and underwent spectral CT examination before chemotherapy were enrolled retrospectively and divided into a responsive group and a non-responsive group according to the postoperative pathological tumour regression grade. Clinical characteristics were collected. The iodine concentration (IC), water concentration (WC), and effective atomic number (Eff-Z) of the portal venous phases were measured before chemotherapy, and IC was normalised to that of the aorta to provide the normalised IC (NIC). An independent samples t-test, Mann-Whitney U-test, or chi-square test was used to analyse the differences between the two groups, and the receiver operating curve (ROC) was used to evaluate the predictive performance of different variables. RESULTS The neutrophil-to-lymphocyte ratio (NLR) was lower in the responsive group than in the non-responsive group (p<0.05). IC, NIC, and Eff-Z values were significantly higher in the responsive group than in the non-responsive group (p<0.01). The areas under the ROC curves for the NLR, IC, NIC, and Eff-Z were 0.694, 0.688, 0.799, and 0.690, respectively. The combination of NIC, Eff-Z, and NLR values showed good diagnostic performance in predicting response to NAC in GC, with an area under the ROC curve of 0.857, 76.92% sensitivity, 80% accuracy, and 85.71% specificity. CONCLUSION Spectral CT parameters may serve as non-invasive tools for predicting the response to NAC in patients with GC.
Collapse
Affiliation(s)
- J Deng
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - W Zhang
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - M Xu
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - X Liu
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - T Ren
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - S Li
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - Q Sun
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - C Xue
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - J Zhou
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China.
| |
Collapse
|
7
|
Chatterjee A, Azevedo-Martins JM, Stachler MD. Interleukin-33 as a Potential Therapeutic Target in Gastric Cancer Patients: Current Insights. Onco Targets Ther 2023; 16:675-687. [PMID: 37583706 PMCID: PMC10424681 DOI: 10.2147/ott.s389120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/06/2023] [Indexed: 08/17/2023] Open
Abstract
Gastric cancer is a significant global health problem as it is the fifth most prevalent cancer worldwide and the fourth leading cause of cancer-related mortality. While cytotoxic chemotherapy remains the primary treatment for advanced GC, response rates are limited. Recent progresses, focused on molecular signalling within gastric cancer, have ignited new hope for potential therapeutic targets that may improve survival and/or reduce the toxic effects of traditional therapies. Carcinomas are generally initiated when critical regulatory genes get mutated, but the progression to malignancy is usually supported by the non-neoplastic cells that create a conducive environment for transformation and progression to occur. Interleukin 33 (IL-33) functions as a dual activity cytokine as it is also a nuclear factor. IL-33 is usually present in the nuclei of the cells. Upon tissue damage, it is released into the extracellular space and binds to its receptor, suppression of tumorigenicity 2 (ST2) L, which is expressed on the membranes of the target cells. IL-33 signalling activates the T Helper 2 (Th2) immune response among other responses. Although the studies on the role of IL-33 in gastric cancer are still in the early stages, they have revealed potentially important (though sometimes conflicting) functions or roles in cancer development and progression. The pro-tumorigenic roles include induction and the recruitment of tumor-associated immune cells, promoting metaplasia progression, and inducing stem cell like and EMT properties in gastric cancer cells. Therapeutic interventions to disrupt these functions may provide a unique strategy for gastric cancer prevention and treatment. This review aims to provide a summary of the role of IL-33 in GC, state its multiple functions in relation to GC, and show potential avenues for promising therapeutic investigation.
Collapse
Affiliation(s)
- Annesha Chatterjee
- University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| | | | - Matthew D Stachler
- University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| |
Collapse
|
8
|
Yu F, Xing J, Li L, Xiang M. CircCRIM1 mediates proliferation, migration, and invasion of trophoblast cell through regulating miR-942-5p/IL1RAP axis. Am J Reprod Immunol 2023; 90:e13699. [PMID: 37382169 DOI: 10.1111/aji.13699] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/20/2023] [Accepted: 03/16/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Preeclampsia (PE) is a severe complication that occurs during pregnancy and a main cause of perinatal mortality of mothers as well as infants, which is characterized by abnormal placental trophoblast. Previous study reported that aberrant circular RNA (circRNA) was involved in the pathogenesis and progression of PE. Herein, we aimed to investigate the role of circCRIM1 and explore the mechanism of circCRIM1 in PE. METHODS The quantitative real-time PCR (qRT-PCR) was conducted to determine the relative expression of circCRIM1, miR-942-5p, and IL1RAP in tissues and cells. Cell proliferation viability was assessed by both MTT and EdU assays. Cell cycle distribution was analyzed using flow cytometry. Transwell assay was performed to test the cell migration and invasion. The protein levels of CyclinD1, MMP9, MMP2, and IL1RAP were measured by western blot. The putative binding sites between miR-942-5p and circCRIM1 or IL1RAP 3'UTR were verified by dual-luciferase reporter gene assay. Rescue experiment was performed to confirm that miR-942-5p/IL1RAP axis was functional target of circCRIM1 in trophoblast cells. RESULTS CircCRIM1 was upregulated in placenta tissues of PE and its expression was inversely related to infant weight. Overexpression of circCRIM1 suppressed proliferation, migration, and invasion and reduced the protein levels of CyclinD1, MMP9, MMP2 of trophoblast cells, whereas its knockdown exerted the opposite effect. CircCRIM1 could interact with miR-942-5p, and introduction of miR-942-5p partially abated the inhibitory effect of circCRIM1 on trophoblast cell behaviors. IL1RAP was directly targeted and negatively regulated by miR-942-5p. miR-942-5p played its regulatory role on cell proliferation, migration, and invasion of trophoblast by IL1RAP. Further analysis showed that circCRIM1 modulated IL1RAP expression via sponging miR-942-5p. CONCLUSION The results of the present study demonstrated that circCRIM1 inhibited the proliferation, migration, and invasion of trophoblast cells through sponging miR-942-5p and up-regulating IL1RAP, providing a possible new mechanism of PE.
Collapse
Affiliation(s)
- Fen Yu
- Department of Gynecology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jie Xing
- Department of Obstetrics, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Lingyun Li
- Department of Obstetrics, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Mi Xiang
- Department of Obstetrics, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Baker KJ, Buskiewicz E, Finucane M, Chelliah A, Burke L, Houston A, Brint E. IL-36 expression is increased in NSCLC with IL-36 stimulation of lung cancer cells promoting a pro-tumorigenic phenotype. Cytokine 2023; 165:156170. [PMID: 36931148 DOI: 10.1016/j.cyto.2023.156170] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023]
Abstract
The IL-36 cytokines are a recently described subset of the IL-1 family of cytokines, and have been shown to play a role in the pathogenesis of respiratory diseases such as asthma and COPD. Given the common aetiological links between COPD and lung cancer development, as well as the involvement of other IL-1 family members in lung tumorigenesis, the aim of this work was to investigate the role of IL-36 cytokines in the pathogenesis of lung cancer. In this study we demonstrate that expression of IL-36 cytokines and receptor mRNA and protein are significantly increased in lung cancer tissue compared to adjacent non-tumour tissue. In vitro assays showed that stimulation of two lung cancer cell lines, SKMES-1 human squamous cell and LLC murine lung cancer, with IL-36R agonists resulted in increased cellular migration and proliferation. All IL-36 cytokines induced the expression of pro-inflammatory chemokines in both lung cancer cell lines with synergistic effects identified upon co-stimulation of cells with IL-17, IL-22 and TNFα. Furthermore, we report that IL-36 cytokines induce protein expression of the immune checkpoint inhibitor protein PD-L1 on lung cancer cells. Taken together, this data indicates that targeting IL-36R signalling may be a useful targeted therapy for lung cancer patients with IL-36R+ cancer cells.
Collapse
Affiliation(s)
- Kevin James Baker
- Department of Pathology, UCC, Cork, Ireland; Department of Medicine, UCC, Cork, Ireland
| | | | - Méabh Finucane
- Department of Pathology, UCC, Cork, Ireland; Department of Medicine, UCC, Cork, Ireland
| | | | - Louise Burke
- Department of Pathology, UCC, Cork, Ireland; Dept. Of Pathology CUH, Ireland
| | - Aileen Houston
- Department of Medicine, UCC, Cork, Ireland; APC Microbiome Ireland, UCC, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, UCC, Cork, Ireland; APC Microbiome Ireland, UCC, Cork, Ireland.
| |
Collapse
|
10
|
Pasvenskaite A, Liutkeviciene R, Gedvilaite G, Vilkeviciute A, Liutkevicius V, Uloza V. The survival rate of laryngeal squamous cell carcinoma: impact of IL1RAP rs4624606, IL1RL1 rs1041973, IL-6 rs1800795, BLK rs13277113, and TIMP3 rs9621532 single nucleotide polymorphisms. Discov Oncol 2023; 14:8. [PMID: 36682035 PMCID: PMC9867797 DOI: 10.1007/s12672-023-00619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/16/2023] [Indexed: 01/23/2023] Open
Abstract
PURPOSE Results of laryngeal squamous cell carcinoma (LSCC) treatment and the 5 year survival rate of these patients remain poor. To purify therapeutic targets, investigation of new specific and prognostic blood-based markers for LSCC development is essential. METHODS In the present study, we evaluated five single nucleotide polymorphisms (SNPs): IL1RAP rs4624606, IL1RL1 rs1041973, IL-6 rs1800795, BLK rs13277113, and TIMP3 rs9621532, and determined their associations with the patients' 5 year survival rate. Also, we performed a detailed statistical analysis of different LSCC patients' characteristics impact on their survival rate. RESULTS Three hundred fifty-three LSCC patients and 538 control subjects were included in this study. The multivariable Cox regression analysis revealed a significant association between patients' survival rate and distribution of IL1RAP rs4624606 variants: patients carrying AT genotype at IL1RAP rs4624606 had a lower risk of death (p = 0.044). Also, it was revealed that tumor size (T) (p = 0.000), tumor differentiation grade (G) (p = 0.015), and IL1RAP rs4624606 genotype (p = 0.044) were effective variables in multivariable Cox regression analysis prognosing survival of LSCC patients. The specific-LSCC 5 year survival rate was 77%. CONCLUSIONS In summary, our findings indicate that the genotypic distribution of IL1RAP rs4624606 influences the 5 year survival rate of LSCC patients. The results of the present study facilitate a more complete understanding of LSCC at the biological level, thus providing the base for the identification of new specific and prognostic blood-based markers for LSCC development.
Collapse
Affiliation(s)
- Agne Pasvenskaite
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| | - Rasa Liutkeviciene
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), Kaunas, Lithuania
| | - Greta Gedvilaite
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), Kaunas, Lithuania
| | - Alvita Vilkeviciute
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), Kaunas, Lithuania
| | - Vykintas Liutkevicius
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| | - Virgilijus Uloza
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| |
Collapse
|
11
|
Zhao M, Wu J, Xu J, Li A, Mei Y, Ge X, Yin G, Liu X, Wei L, Xu Q. Association of environmental exposure to chromium with differential DNA methylation: An epigenome-wide study. Front Genet 2023; 13:1043486. [PMID: 36685967 PMCID: PMC9845398 DOI: 10.3389/fgene.2022.1043486] [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: 09/13/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction: Previous studies have reported that chromium (Cr)-induced epigenetic alterations and DNA methylation play a vital role in the pathogenesis of diseases induced by chromium exposure. Epigenomic analyses have been limited and mainly focused on occupational chromium exposure; their findings are not generalizable to populations with environmental Cr exposure. Methods: We identified the differential methylation of genes and regions to elucidate the mechanisms of toxicity related to environmental chromium exposure. DNA methylation was measured in blood samples collected from individuals in Cr-contaminated (n = 10) and unexposed areas (n = 10) by using the Illumina Infinium HumanMethylation850K array. To evaluate the relationship between chromium levels in urine and CpG methylation at 850 thousand sites, we investigated differentially methylated positions (DMPs) and differentially methylated regions (DMRs) by using linear models and DMRcate method, respectively. The model was adjusted for biologically relevant variables and estimated cell-type compositions. Results: At the epigenome-wide level, we identified five CpGs [cg20690919 (p FDR =0.006), cg00704664 (p FDR =0.024), cg10809143 (p FDR =0.043), cg27057652 (p FDR =0.047), cg05390480 (p FDR =0.024)] and one DMR (chr17: 19,648,718-19,648,972), annotated to ALDH3A1 genes (p < 0.05) as being significantly associated with log2 transformed urinary chromium levels. Discussion: Environmental chromium exposure is associated with DNA methylation, and the significant DMPs and DMR being annotated to cause DNA damage and genomic instability were found in this work. Research involving larger samples is required to further explore the epigenetic effect of environmental chromium exposure on health outcomes through DNA methylation.
Collapse
Affiliation(s)
- Meiduo Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jingtao Wu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ang Li
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yayuan Mei
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoyu Ge
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Guohuan Yin
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaolin Liu
- Department of Epidemiology and Biostatistics, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lanping Wei
- Jinzhou Central Hospital, Jinzhou, Liaoning, China
| | - Qun Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,Center of Environmental and Health Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,*Correspondence: Qun Xu,
| |
Collapse
|
12
|
Frenay J, Bellaye PS, Oudot A, Helbling A, Petitot C, Ferrand C, Collin B, Dias AMM. IL-1RAP, a Key Therapeutic Target in Cancer. Int J Mol Sci 2022; 23:ijms232314918. [PMID: 36499246 PMCID: PMC9735758 DOI: 10.3390/ijms232314918] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer is a major cause of death worldwide and especially in high- and upper-middle-income countries. Despite recent progress in cancer therapies, such as chimeric antigen receptor T (CAR-T) cells or antibody-drug conjugate (ADC), new targets expressed by the tumor cells need to be identified in order to selectively drive these innovative therapies to tumors. In this context, IL-1RAP recently showed great potential to become one of these new targets for cancer therapy. IL-1RAP is highly involved in the inflammation process through the interleukins 1, 33, and 36 (IL-1, IL-33, IL-36) signaling pathways. Inflammation is now recognized as a hallmark of carcinogenesis, suggesting that IL-1RAP could play a role in cancer development and progression. Furthermore, IL-1RAP was found overexpressed on tumor cells from several hematological and solid cancers, thus confirming its potential involvement in carcinogenesis. This review will first describe the structure and genetics of IL-1RAP as well as its role in tumor development. Finally, a focus will be made on the therapies based on IL-1RAP targeting, which are now under preclinical or clinical development.
Collapse
Affiliation(s)
- Jame Frenay
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Pierre-Simon Bellaye
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Alexandra Oudot
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Alex Helbling
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Camille Petitot
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Christophe Ferrand
- INSERM UMR1098, EFS BFC, Université de Bourgogne Franche-Comté, 25000 Besançon, France
- CanCell Therapeutics, 25000 Besançon, France
| | - Bertrand Collin
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR CNRS 6302, 21000 Dijon, France
| | - Alexandre M M Dias
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| |
Collapse
|
13
|
Trad R, Warda W, Alcazer V, Neto da Rocha M, Berceanu A, Nicod C, Haderbache R, Roussel X, Desbrosses Y, Daguindau E, Renosi F, Roumier C, Bouquet L, Biichle S, Guiot M, Seffar E, Caillot D, Depil S, Robinet E, Salma Y, Deconinck E, Deschamps M, Ferrand C. Chimeric antigen receptor T-cells targeting IL-1RAP: a promising new cellular immunotherapy to treat acute myeloid leukemia. J Immunother Cancer 2022; 10:jitc-2021-004222. [PMID: 35803613 PMCID: PMC9272123 DOI: 10.1136/jitc-2021-004222] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
Background Acute myeloid leukemia (AML) remains a very difficult disease to cure due to the persistence of leukemic stem cells (LSCs), which are resistant to different lines of chemotherapy and are the basis of refractory/relapsed (R/R) disease in 80% of patients with AML not receiving allogeneic transplantation. Methods In this study, we showed that the interleukin-1 receptor accessory protein (IL-1RAP) protein is overexpressed on the cell surface of LSCs in all subtypes of AML and confirmed it as an interesting and promising target in AML compared with the most common potential AML targets, since it is not expressed by the normal hematopoietic stem cell. After establishing the proof of concept for the efficacy of chimeric antigen receptor (CAR) T-cells targeting IL-1RAP in chronic myeloid leukemia, we hypothesized that third-generation IL-1RAP CAR T-cells could eliminate AML LSCs, where the medical need is not covered. Results We first demonstrated that IL-1RAP CAR T-cells can be produced from AML T-cells at the time of diagnosis and at relapse. In vitro and in vivo, we showed the effectiveness of IL-1RAP CAR T-cells against AML cell lines expressing different levels of IL-1RAP and the cytotoxicity of autologous IL-1RAP CAR T-cells against primary cells from patients with AML at diagnosis or at relapse. In patient-derived relapsed AML xenograft models, we confirmed that IL-1RAP CAR T-cells are able to circulate in peripheral blood and to migrate in the bone marrow and spleen, are cytotoxic against primary AML cells and increased overall survival. Conclusion In conclusion, our preclinical results suggest that IL-1RAP CAR T-based adoptive therapy could be a promising strategy in AML treatment and it warrants the clinical investigation of this CAR T-cell therapy.
Collapse
Affiliation(s)
- Rim Trad
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Walid Warda
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France.,CanCell Therapeutics, Besancon, France
| | | | - Mathieu Neto da Rocha
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France.,CanCell Therapeutics, Besancon, France
| | - Ana Berceanu
- Clinical Hematology, C.H. Univ Jean Minjoz, Besancon, France
| | | | | | - Xavier Roussel
- Clinical Hematology, C.H. Univ Jean Minjoz, Besancon, France
| | | | | | - Florain Renosi
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | | | - Lucie Bouquet
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Sabeha Biichle
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Melanie Guiot
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Evan Seffar
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Denis Caillot
- Clinical Hematology, CHU François Mitterrand, Dijon, France
| | | | | | - Yahya Salma
- Laboratory of Applied Biotechnology (LBA3B), Lebanese University, Tripoli, Lebanon
| | - Eric Deconinck
- Clinical Hematology, C.H. Univ Jean Minjoz, Besancon, France
| | - Marina Deschamps
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France.,CanCell Therapeutics, Besancon, France
| | - Christophe Ferrand
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France .,CanCell Therapeutics, Besancon, France
| |
Collapse
|
14
|
Zhang J, Zhang N, Fu X, Wang W, Liu H, McKay MJ, Dejkriengkraikul P, Nie Y. Bioinformatic analysis of cancer-associated fibroblast related gene signature as a predictive model in clinical outcomes and immune characteristics of gastric cancer. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:698. [PMID: 35845527 PMCID: PMC9279800 DOI: 10.21037/atm-22-2810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
Background Gastric cancer (GC) has a high incidence and high mortality rate among Asian countries, and distinguishing predictive prognosis biomarkers for GC are essential. Cancer-associated fibroblasts (CAFs) play a significant role in the progression, immune evasion, and therapeutic resistance of GC. Therefore, CAF-associated genes might have huge potential as prognostic biomarkers for predicting tumor progression and survival rate in GC pateints. Methods A sum of 1,134 GC patients from the The Cancer Genome Atlas Stomach Adenocarcinoma (TCGA-STAD), GSE62254, and GSE84437 datasets as well as GC cohorts from Xijing hospital were included. Firstly, we performed univariate Cox regression analysis to identify CAF-associated prognostic genes. Subsequently, the Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis was used to develop a CAF gene signature (CAFGS) in the TCGA-STAD training cohort. CAFGS’s predictive performance was examined in both the training and validation cohorts, and the relationship between CAFGS and the tumor microenvironment (TME) was investigated by ssGSEA, CIBERSORT, TIMER, and ESTIMATE. Finally, a nomogram of CAFGS was established. Results Ten CAF-associated genes (ANGPTL4, CPNE8, CST2, HTR1F, IL1RAP, NR1D1, NTAN1, OLFML2B, TMEM259, and VTN) were identified to develop CAFGS. A high CAFGS score represented a worse outcome for GC patients in four cohorts, and a strong correlation was found between CAFGS and the infiltration of immune cells. We showed that CAFs contribute to immune evasion and unfavorable prognoses of GC patients by promoting the formation of an immunosuppressive microenvironment, and a high level of CAF infiltration may attenuate the efficacy of immunotherapy. The nomogram based on CAFGS showed reasonable predictive ability and may deliver great clinical net benefits. Conclusions We established a CAFGS model with 10 CAF-associated genes that had a great predictive value for GC prognosis and survival rate evaluation. This study could provide a novel insight for investigating the role of CAFs in GC.
Collapse
Affiliation(s)
- Jiehao Zhang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Nannan Zhang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xin Fu
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Weizhen Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.,College of Life Sciences, Northwest University, Xi'an, China
| | - Hui Liu
- College of Life Sciences, Northwest University, Xi'an, China
| | - Michael J McKay
- Northern Cancer Service, North West Cancer Centre, Burnie, Tasmania, Australia.,Rural Clinical School, The University of Tasmania, Northwest Regional Hospital, Burnie, Tasmania, Australia
| | - Pornngarm Dejkriengkraikul
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
15
|
Clinical Value of PLR, MLR, and NWR in Neoadjuvant Chemotherapy for Locally Advanced Gastric Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8005975. [PMID: 35664642 PMCID: PMC9162812 DOI: 10.1155/2022/8005975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/25/2022]
Abstract
Objective The clinical value of platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR), and neutrophil-to-white blood cell ratio (NWR) in predicting the prognosis of patients with locally advanced gastric cancer after neoadjuvant chemotherapy (NACT) was studied. Methods A total of 131 patients with locally advanced gastric cancer treated with neoadjuvant chemotherapy in our hospital from May 2015 to June 2018 were selected as the study subjects, and all were treated with neoadjuvant chemotherapy. The relationship between the values of PLR, MLR, and NWR and the efficacy of neoadjuvant chemotherapy and clinical staging was analyzed; all patients were followed up for 3 years. Patients were divided into death group and survival group according to the survival of patients. The predictive value of PLR, MLR, and NWR values for patients' prognosis was analyzed, and the survival rates of patients with different PLR, MLR, and NWR values were compared. Results The effective rate of neoadjuvant chemotherapy in patients with locally advanced gastric cancer was 62.60% (82/131), and the PLR, MLR, and NWR values in the effective group were lower than those in the ineffective group (P < 0.05). The AUC of combined PLR, MLR, and NWR in evaluating the efficacy of neoadjuvant chemotherapy was greater than that of PLR and NWR alone (P < 0.05). The PLR value of patients with stage IIIa, IIIb, and IIIc was greater than that of patients with stage II, the MLR value of patients with stage IIIb and IIIc was greater than that of patients with stage II and IIIa, and the NMR value of patients with stage IIIc was greater than that of patients with stage II, IIIa, and IIIb (P < 0.05). PLR, MLR, and NWR values were positively correlated with clinical stage (P < 0.05). The PLR, MLR, and NWR values in the survival group were lower than those in the death group (P < 0.05). The AUC of combined PLR, MLR, and NWR in predicting the prognosis of patients was greater than that of MLR and NWR alone (P < 0.05). The survival rate of patients with PLR ≥ 162.11 (36.21%) was lower than that of patients with PLR < 162.11 (80.82%), and the survival rate of patients with MLR ≥ 0.31 (42.86%) was lower than that of patients with MLR < 0.31 (74.67%), and the survival rate of patients with NWR ≥ 0.62 (45.00%) was lower than that of patients with NWR < 0.62 (74.65%) (P < 0.05). Conclusions PLR, MLR, and NWR values are correlated with clinical stage, and the combined detection has value in evaluating the clinical efficacy of neoadjuvant chemotherapy and predicting the prognosis of patients with locally advanced gastric cancer.
Collapse
|