1
|
Zheng L, Li Y, Güngör C, Ge H. Gut microbiota influences colorectal cancer through immune cell interactions: a Mendelian randomization study. Discov Oncol 2025; 16:747. [PMID: 40358736 PMCID: PMC12075717 DOI: 10.1007/s12672-025-02486-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the most prevalent malignant tumor of the digestive system globally, posing a significant threat to human health and quality of life. Recent studies have established associations between gut microbiota and immune cells with CRC; however, the mechanisms by which gut microbiota influence the development and progression of CRC through immune mediators remain poorly understood. METHODS We conducted a two-sample, bidirectional Mendelian randomization analysis. We utilized 731 immune cell types and 473 gut microbial species along with colorectal cancer statistics from published summary statistics from genome-wide association studies (GWAS).The analysis employed several methodologies, including inverse variance-weighted (IVW) analysis, MR-Egger regression, the weighted median method, and both weighted and simple model approaches.Sensitivity analyses were performed to confirm the reliability of the Mendelian randomization results, and reverse Mendelian randomization was used to assess the overall impact of CRC on gut microbiota and immune cells. RESULTS Our findings suggest a causal relationship involving nine immunophenotypes and five specific gut microbial taxa with CRC. Notably, the gut microbes Alloprevotella and Holdemania, along with immune cell types CD3 on CD28- CD8br and CD4 + T cells, demonstrated significant causal associations with CRC. Mediation analysis revealed that the association between Alloprevotella and CRC was mediated by CD4 + T cells, with a mediation effect of 6.48%. Additionally, Holdemania was found to mediate its association with CRC through CD3 on CD28- CD8br, exhibiting a mediation effect of 9.29%. Reverse Mendelian randomization did not indicate any causal effect of CRC on specific immune cells or gut microbiota. Two-sided sensitivity analyses revealed no evidence of heterogeneity or horizontal pleiotropy in our findings. CONCLUSIONS This comprehensive Mendelian randomization study enhances our understanding of the mechanisms by which gut microbiota affects CRC through immune cell interactions. Further investigations are warranted to unravel the underlying mechanisms linking gut microbiota, immune cells, and colorectal cancer.
Collapse
Affiliation(s)
- Linyi Zheng
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yuqiang Li
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Cenap Güngör
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Heming Ge
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, 410013, China.
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
2
|
Zhang P, Wang L, Liu H, Lin S, Guo D. Unveiling the crucial role of glycosylation modification in lung adenocarcinoma metastasis through artificial neural network-based spatial multi-omics single-cell analysis and Mendelian randomization. BMC Cancer 2025; 25:249. [PMID: 39948531 PMCID: PMC11823056 DOI: 10.1186/s12885-025-13650-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Investigations into the intricacies of glycosylation modifications, a prevalent post-translational alteration observed in neoplasms, especially remain elusive in the context of lung adenocarcinoma. Through the integration of multiple omics approaches, the investigation aimed to delineate the significance of glycosylation in lung adenocarcinoma, with an objective to pinpoint viable biological targets. METHODS Initial steps involved the identification of genes differentially expressed in relation to glycosylation at the aggregate transcriptome level within lung adenocarcinoma tissues. This was followed by analyses of localization and function employing both single-cell and spatial transcriptomics to provide a more nuanced understanding. In pursuit of elucidating functional disparities in glycosylation patterns, a predictive framework employing artificial neural networks was constructed. To ascertain causal relationships between specific genes and lung adenocarcinoma, Mendelian randomization was applied, culminating in the experimental validation of these genes' roles. RESULTS Analysis at the single-cell level uncovered marked glycosylation modification expressions in metastatic tissues of lung adenocarcinoma. Moreover, tissues of lung adenocarcinoma with elevated expression of genes associated with glycosylation displayed enhanced differentiation and activation across signaling pathways including TGF-β, oxidative stress, and WNT. Through spatial transcriptomics, zones of intense glycosylation modification were pinpointed within tumor nests and proximate to tumor-associated blood vessels. An artificial neural network-derived prognostic model demonstrated outstanding predictive capability, with AUC scores achieving 0.84, 0.83, and 0.89 for 1, 3, and 5-year forecasts, respectively. The group identified as high-risk was characterized by pronounced immunosuppression and diminished responsiveness to immunotherapy. Mendelian randomization analysis pinpointed GLANT2 (OR = 1.3654, p < 0.05) and GYS1 (OR = 1.2668, p < 0.05) as genes contributing to the pathogenesis of lung adenocarcinoma. Cell assays have reaffirmed that the inhibition of GYS1 significantly reduces proliferation and invasion in lung adenocarcinoma cell lines, while also decreasing glycogen storage and the formation of glycosylation end products, indicating suppression of glycosylation processes. These findings identify GYS1 as a prospective glycosylation-linked biological target for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Penngcheng Zhang
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zheiiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Lexin Wang
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Western Institute of Digital-Intelligent Medicine, Chongqing, China
| | - Hanwen Liu
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shengyou Lin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zheiiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China.
| | - Dechao Guo
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Permain J, Hock B, Eglinton T, Purcell R. Functional links between the microbiome and the molecular pathways of colorectal carcinogenesis. Cancer Metastasis Rev 2024; 43:1463-1474. [PMID: 39340753 PMCID: PMC11554747 DOI: 10.1007/s10555-024-10215-5] [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: 04/02/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Colorectal cancer (CRC) is a common cancer, with a concerning rise in early-onset CRC cases, signalling a shift in disease epidemiology. Whilst our understanding of the molecular underpinnings of CRC has expanded, the complexities underlying its initiation remain elusive, with emerging evidence implicating the microbiome in CRC pathogenesis. This review synthesizes current knowledge on the intricate interplay between the microbiome, tumour microenvironment (TME), and molecular pathways driving CRC carcinogenesis. Recent studies have reported how the microbiome may modulate the TME and tumour immune responses, consequently influencing cancer progression, and whilst specific bacteria have been linked with CRC, the underlying mechanisms remains poorly understood. By elucidating the functional links between microbial landscapes and carcinogenesis pathways, this review offers insights into how bacteria orchestrate diverse pathways of CRC development, shedding light on potential therapeutic targets and personalized intervention strategies.
Collapse
Affiliation(s)
- Jessica Permain
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand
| | - Barry Hock
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Timothy Eglinton
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand
| | - Rachel Purcell
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
4
|
Sun Q, Yang Z, Qiu M, Wang S, Zhao X, Pang W, Liu R, Wang Y, Wang H, Hao J, Gao M. Inflammatory factor TNFα-induced circDMD mediates R-loop formation to promote tumorigenesis. Int J Biol Macromol 2024; 280:135689. [PMID: 39288863 DOI: 10.1016/j.ijbiomac.2024.135689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Chronic inflammation has been associated with the development of cancer in various anatomical sites. However, the crosstalk between inflammatory factors and circular RNAs (circRNAs) in tumorigenesis is unclear. Here, we revealed that circDMD was upregulated in Tumor necrosis factor alpha-like (TNFα)-induced HeLa cells. circDMD promoted the expression and nuclear translocation of Nuclear factor kappa B subunit (NF-κB) to activate downstream factors. circDMD absorbed miR-4711-5p to increase Lysine demethylase 5 A (KDM5A) expression, which reduced Suppressor of cytokine signaling 1 (SOCS1) to decrease the ubiquitination of Rela proto-oncogene (P65). In addition, circDMD promoted Fms related receptor tyrosine kinase 4 (VEGFR3) expression through the formation of an R-loop in its promoter. circDMD promoted tumor proliferation, metastasis and autophagy by activating the NF-κB pathways in vitro and in tumors derived from HeLa cells in vivo. Taken together, our results indicated that the expression of circDMD is induced by TNFα and contributes to tumorigenesis in cervical cancer (CC), which might help elucidate the regulatory effects of circRNAs on tumorigenesis.
Collapse
Affiliation(s)
- Qi Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China; Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China
| | - Zhen Yang
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Clinical Laboratory, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China.
| | - Minghan Qiu
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin 300321, China
| | - Shoujun Wang
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Thyroid and Breast Surgery, Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center of Nankai University, Tianjin 300321, China
| | - Xingli Zhao
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Hematology, Oncology Center, Tianjin Union Medical Center of Nankai University, Tianjin 300321, China
| | - Wenwen Pang
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Clinical Laboratory, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China
| | - Ruxue Liu
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin 300321, China
| | - Yayun Wang
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin 300321, China
| | - Huaqing Wang
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Oncology, Tianjin Union Medical Center of Nankai University, Tianjin 300321, China
| | - Jie Hao
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Thyroid and Breast Surgery, Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center of Nankai University, Tianjin 300321, China.
| | - Ming Gao
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, Tianjin 300121, China; Department of Thyroid and Breast Surgery, Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center of Nankai University, Tianjin 300321, China.
| |
Collapse
|
5
|
Alshamsan B, Elshenawy MA, Aseafan M, Fahmy N, Badran A, Elhassan T, Alsayed A, Suleman K, Al-Tweigeri T. Prognostic significance of the neutrophil to lymphocyte ratio in locally advanced breast cancer. Oncol Lett 2024; 28:429. [PMID: 39049989 PMCID: PMC11268088 DOI: 10.3892/ol.2024.14562] [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: 01/12/2024] [Accepted: 05/28/2024] [Indexed: 07/27/2024] Open
Abstract
The present study aimed to clarify the prognostic role of the pre-treatment neutrophil-to-lymphocyte ratio (NLR) for the response to neoadjuvant chemotherapy (NAC) in locally advanced breast cancer (LABC). Due to conflicting results in currently available data, the specific focus of the present study was on evaluating the associations between the pre-treatment NLR and the rate of achieving a pathological complete response (pCR) and survival outcomes. For the present study, data from a cohort of 465 consecutive patients with LABC who underwent NAC at King Feisal Specialist Hospital and Research Center (Riyadh, Saudi Arabia) between 2005 and 2014 were obtained from a prospective BC database and analyzed. Patients were stratified into two groups based on an optimal NLR cut-off determined using the receiver operating characteristic curve. Logistic regression analyses were conducted to assess variables associated with pCR, and Cox regression analyses were used to assess variables associated with survival outcomes. The low pre-treatment NLR group (≤2.2) was found to exhibit a higher likelihood of achieving a pCR (odds ratio, 2.59; 95% CI, 1.52-4.38; P<0.001), along with higher 5-year disease-free survival (DFS) [75.8 vs. 64.9%; hazard ratio (HR), 0.69; 95% CI, 0.50-0.94; P=0.02] and 5-year overall survival (OS; 90.3 vs. 81.9; HR, 0.62; 95% CI, 0.39-0.98; P=0.04) rates compared with those in the high NLR group (>2.2). Sub-group analysis revealed that the observed significance in survival outcomes was driven by the triple-negative BC (TNBC) subgroup. Patients with residual TNBC disease and a high pre-treatment NLR were observed to have lower 5-year DFS (44.4 vs. 75.0%; P=0.02) and 5-year OS (55.9 vs. 84.5%; P=0.055) rates compared with those with residual TNBC disease and a low NLR. To conclude, data from the present study suggest that the pre-treatment NLR can serve as a viable independent prognostic factor for pCR following NAC in patients with LABC and for survival outcomes, particularly for patients with TNBC.
Collapse
Affiliation(s)
- Bader Alshamsan
- Department of Medicine, College of Medicine, Qassim University, Buraydah, Qassim 52571, Kingdom of Saudi Arabia
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| | - Mahmoud A. Elshenawy
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
- Department of Clinical Oncology, Faculty of Medicine, Menoufia University, Shebeen El-Kom 32511, Egypt
| | - Mohamed Aseafan
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
- Department of Internal Medicine, Section of Oncology, Security Forces Hospital, Riyadh 11481, Kingdom of Saudi Arabia
| | - Nermin Fahmy
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
- Department of Clinical Oncology and Nuclear Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ahmed Badran
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
- Department of Clinical Oncology, Ain Shams University Hospitals, Ain Shams 11517, Egypt
| | - Tusneem Elhassan
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| | - Adher Alsayed
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| | - Kausar Suleman
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| | - Taher Al-Tweigeri
- Section of Medical Oncology, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Kingdom of Saudi Arabia
| |
Collapse
|
6
|
Zabihi MR, Farhadi B, Akhoondian M. Complement protein expression changes in various conditions of breast cancer: in-silico analyses-experimental research. Ann Med Surg (Lond) 2024; 86:5152-5161. [PMID: 39239051 PMCID: PMC11374204 DOI: 10.1097/ms9.0000000000002216] [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: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 09/07/2024] Open
Abstract
Introduction Breast cancer is the most prevalent cancer diagnosed in females worldwide. The known biomarkers are insufficient to understand the actual prognosis of breast cancer, and identifying new biomarkers is desirable and valuable data to improve the patient's survival. Many inflammatory biomarkers, such as the complement system, can be regarded as prognostic values and as potent inflammatory mediators; complement proteins have a critical role in tumorigenesis. In the current study, the authors aim to investigate complement protein expression changes, particularly complement 3 (C3), complement 7 (C7), complement factor B (CFB), and complement factor D (CFD), in various conditions of breast cancer using in-silico tools. Methods The intent data were extracted using webtools, including; Kaplan-Meier plotter, BcGenExMiner, UALCAN, cbioportal, GeneMania, and Enrichr. To select valid data, a P greater than 0.05 was considered. Results The current study clarified that 21 complement genes correlated to survival conditions. Also, down or upregulation of extracted genes and breast cancer statuses were identified. Additionally, expression level difference of complement genes in various breast cancer four stages was detected. Ultimately, co-expression genes with complement genes were extracted and networked. Conclusion Changes in the expression of complement proteins can strongly correlate to breast cancer's prognosis, status, and survival. Furthermore, considering the vital role of CFD and CFB complement proteins in the alternative pathway in different stages of breast cancer, CFD and CFB can be regarded as reliable prognostic values for diagnosis.
Collapse
Affiliation(s)
- Mohammad Reza Zabihi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences
| | - Bahar Farhadi
- School of Medicine, Islamic Azad University, Mashhad Branch, Mashhad
| | - Mohammad Akhoondian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
7
|
Nobin H, Garvin S, Hagman H, Nodin B, Jirström K, Brunnström H. The prognostic value of programmed death-ligand 1 (PD-L1) expression in resected colorectal cancer without neoadjuvant therapy - differences between antibody clones and cell types. BMC Cancer 2024; 24:1051. [PMID: 39187798 PMCID: PMC11346183 DOI: 10.1186/s12885-024-12812-7] [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: 06/14/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) expression on tumor cells is associated with poor prognosis in several malignancies, while partly contradictory and inconclusive results have been presented for colorectal cancer (CRC). This study aimed to evaluate PD-L1 as a prognostic biomarker in CRC by comparing three different antibody clones. METHODS Patients surgically treated for CRC between January 1st, 2007, and December 31st, 2015, in Kalmar County, Sweden, were retrospectively included. Tissue microarrays from 862 primary tumors without neoadjuvant treatment were assessed for immunohistochemical expression of PD-L1 in tumor cells (TC) and immune cells (IC) using clones 73-10, SP263, and 22C3. Cox regression proportional hazard models were used to estimate hazard ratios for overall survival (OS) and disease-free interval (DFI) in univariable and multivariable analyses, with 1% and 5% set as cut-offs for positive expression in TC and IC respectively. RESULTS PD-L1 expression in TC was found in 89 (10%) cases for clone 73-10, 76 (9%) for clone SP263, and 38 (4%) for clone 22C3, while the numbers for IC were 317 (37%) cases for clone 73-10, 264 (31%) for clone SP263, and 89 (10%) for clone 22C3. PD-L1 expression in IC was associated with prolonged OS and DFI in univariable analysis for all three clones. The link to prolonged DFI remained in multivariable analysis for 73-10 and SP263, but only for 73-10 regarding OS. PD-L1 expression in TC was not prognostic of OS in any analysis, while it was associated with prolonged DFI for SP263, and a trend was seen for 73-10. The link to prolonged DFI remained for SP263 and was strengthened for 73-10 in multivariable analysis. CONCLUSIONS The prognostic value of PD-L1 expression in both IC and TC differs between antibody clones, with 73-10 and SP263 being more reliable for prognostic information than 22C3 in resected CRC.
Collapse
Affiliation(s)
- Hampus Nobin
- Department of Pathology, Region Kalmar, Kalmar County Hospital, Kalmar, Sweden.
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, Lund, Sweden.
| | - Stina Garvin
- Department of Clinical Pathology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Helga Hagman
- Department of Clinical Sciences Lund, Division of Oncology and Therapeutic Pathology, Lund University, Lund, Sweden
| | - Björn Nodin
- Department of Clinical Sciences Lund, Division of Oncology and Therapeutic Pathology, Lund University, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Therapeutic Pathology, Lund University, Lund, Sweden
- Department of Genetics, Pathology, and Molecular Diagnostics, Regional University Laboratories, Skåne University Hospital, Lund, Sweden
| | - Hans Brunnström
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, Lund, Sweden
- Department of Genetics, Pathology, and Molecular Diagnostics, Regional University Laboratories, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
8
|
Fang X, Huang X, Lu J, Su D. Causal role of immune cells in thyroid cancer: a bidirectional Mendelian randomization study. Front Immunol 2024; 15:1425873. [PMID: 38953025 PMCID: PMC11215042 DOI: 10.3389/fimmu.2024.1425873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024] Open
Abstract
Background The immune system plays an important role in the development and treatment of thyroid cancer(THCA).However, the correlation between immune cells and THCA has not been systematically studied. Methods This study used a two-sample Mendelian randomization (MR) study to determine the causal relationship between immune cell characteristics and THCA. Based on a large sample of publicly available genetic data, we explored the causal relationship between 731 immune cell characteristics and THCA risk. The 731 immunophenotypes were divided into 7 groups, including B cell panel(n=190),cDC panel(n=64),Maturation stages of T cell panel(n=79),Monocyte panel(n=43),Myeloid cell panel(n=64),TBNK panel(n=124),and Treg panel(n=167). The sensitivity of the results was analyzed, and heterogeneity and horizontal pleiotropy were excluded. Results After FDR correction, the effect of immunophenotype on THCA was not statistically significant. It is worth mentioning, however, that there are some unadjusted low P-values phenotypes. The odds ratio (OR) of CD62L on monocyte on THCA risk was estimated to be 0.953 (95% CI=0.930~0.976, P=1.005×10-4),and which was estimated to be 0.975(95% CI=0.961-0.989, P=7.984×10-4) for Resting Treg%CD4 on THCA risk. Furthermore, THCA was associated with a reduced risk of 5 immunophenotype:CD25 on CD39+ CD4 on Treg (OR=0.871, 95% CI=0.812~0.935, P=1.274×10-4), activated Treg AC (OR=0.884, 95% CI=0.820~0.953, P=0.001), activated & resting Treg % CD4 Treg (OR=0.872, 95%CI=0.811~0.937,P=2.109×10-4),CD28- CD25++ CD8br AC(OR=0.867,95% CI=0.809~0.930,P=6.09×10-5),CD28-CD127-CD25++CD8brAC(OR=0.875,95%CI=0.814~0.942,P=3.619×10-4).THCA was associated with an increased risk of Secreting Treg % CD4 Treg (OR=1.143, 95% CI=1.064~1.229, P=2.779×10-4) and CD19 on IgD+ CD24+ (OR=1.118, 95% CI=1.041~1.120, P=0.002). Conclusions These findings suggest the causal associations between immune cells and THCA by genetic means. Our results may have the potential to provide guidance for future clinical research.
Collapse
Affiliation(s)
- Xianliu Fang
- Department of Medical Imaging Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xiaoxiao Huang
- Rehabilitation Medicine Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianhua Lu
- Department of Medical Imaging Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Danke Su
- Department of Medical Imaging Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| |
Collapse
|
9
|
Wang P, Chen J, Zhong R, Xia Y, Wu Z, Zhang C, Yao H. Recent advances of ultrasound-responsive nanosystems in tumor immunotherapy. Eur J Pharm Biopharm 2024; 198:114246. [PMID: 38479562 DOI: 10.1016/j.ejpb.2024.114246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 04/19/2024]
Abstract
Immunotherapy has revolutionized cancer treatment by boosting the immune system and preventing disease escape mechanisms. Despite its potential, challenges like limited response rates and adverse immune effects impede its widespread clinical adoption. Ultrasound (US), known for its safety and effectiveness in tumor diagnosis and therapy, has been shown to significantly enhance immunotherapy when used with nanosystems. High-intensity focused ultrasound (HIFU) can obliterate tumor cells and elicit immune reactions through the creation of immunogenic debris. Low-intensity focused ultrasound (LIFU) bolsters tumor immunosuppression and mitigates metastasis risk by concentrating dendritic cells. Ultrasonic cavitation (UC) produces microbubbles that can transport immune enhancers directly, thus strengthening the immune response and therapeutic impact. Sonodynamic therapy (SDT) merges nanotechnology with immunotherapy, using specialized sonosensitizers to kill cancer cells and stimulate immune responses, increasing treatment success. This review discusses the integration of ultrasound-responsive nanosystems in tumor immunotherapy, exploring future opportunities and current hurdles.
Collapse
Affiliation(s)
- Penghui Wang
- Department of Ultrasound Medicine, Rui'an people's Hospital (The Third Affiliated Hospital of Wenzhou Medical University), Rui'an 325200, China
| | - Ji Chen
- Department of Ultrasound Medicine, Rui'an people's Hospital (The Third Affiliated Hospital of Wenzhou Medical University), Rui'an 325200, China
| | - Runming Zhong
- Department of Ultrasound Medicine, Rui'an people's Hospital (The Third Affiliated Hospital of Wenzhou Medical University), Rui'an 325200, China
| | - Yuanyuan Xia
- Center For Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou 215004, China
| | - Zhina Wu
- Department of Ultrasound Medicine, Rui'an people's Hospital (The Third Affiliated Hospital of Wenzhou Medical University), Rui'an 325200, China
| | - Chunye Zhang
- Center For Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou 215004, China
| | - Hai Yao
- Center For Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou 215004, China.
| |
Collapse
|
10
|
Hu J, Ascierto P, Cesano A, Herrmann V, Marincola FM. Shifting the paradigm: engaging multicellular networks for cancer therapy. J Transl Med 2024; 22:270. [PMID: 38475820 PMCID: PMC10936124 DOI: 10.1186/s12967-024-05043-8] [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: 10/20/2023] [Accepted: 11/01/2023] [Indexed: 03/14/2024] Open
Abstract
Most anti-cancer modalities are designed to directly kill cancer cells deploying mechanisms of action (MOAs) centered on the presence of a precise target on cancer cells. The efficacy of these approaches is limited because the rapidly evolving genetics of neoplasia swiftly circumvents the MOA generating therapy-resistant cancer cell clones. Other modalities engage endogenous anti-cancer mechanisms by activating the multi-cellular network (MCN) surrounding neoplastic cells in the tumor microenvironment (TME). These modalities hold a better chance of success because they activate numerous types of immune effector cells that deploy distinct cytotoxic MOAs. This in turn decreases the chance of developing treatment-resistance. Engagement of the MCN can be attained through activation of immune effector cells that in turn kill cancer cells or when direct cancer killing is complemented by the production of proinflammatory factors that secondarily recruit and activate immune effector cells. For instance, adoptive cell therapy (ACT) supplements cancer cell killing with the release of homeostatic and pro-inflammatory cytokines by the immune cells and damage associated molecular patterns (DAMPs) by dying cancer cells. The latter phenomenon, referred to as immunogenic cell death (ICD), results in an exponential escalation of anti-cancer MOAs at the tumor site. Other approaches can also induce exponential cancer killing by engaging the MCN of the TME through the release of DAMPs and additional pro-inflammatory factors by dying cancer cells. In this commentary, we will review the basic principles that support emerging paradigms likely to significantly improve the efficacy of anti-cancer therapy.
Collapse
Affiliation(s)
- Joyce Hu
- Sonata Therapeutics, Watertown, MA, 02472, USA.
| | - Paolo Ascierto
- Cancer Immunotherapy and Innovative Therapy, National Tumor Institute, Fondazione G. Pascale, 80131, Naples, Italy
| | | | | | | |
Collapse
|
11
|
Boemi I, Piccini S, Colombo FS, Smiroldo V, Zerbi A, Capretti G, Alloisio M, Trivellin G, Lavezzi E, Mazziotti G, Vitali E, Lania AG. Alteration of the immunophenotype and cytokine profiles in patients affected by neuroendocrine neoplasms. Endocrine 2024; 83:810-823. [PMID: 37845576 DOI: 10.1007/s12020-023-03563-9] [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: 08/01/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
PURPOSE Neuroendocrine neoplasms (NENs) are tumors that arise from cells of the endocrine system and are most common in the gastrointestinal tract, the pancreas, and the lungs. Their incidence is rapidly increasing and the therapeutic options available are limited. METHODS Since the immune system can interfere with tumor growth and response to therapy, using flow cytometry we investigated the immunophenotype in samples of peripheral blood leukocytes from patients with pancreatic (Pan-NENs) and pulmonary NENs (Lung-NENs). Moreover, we performed a multiplex analysis of 13 key cytokines and growth factors essential for the immune response in the plasma of NEN patients and controls. RESULTS Patients presented with a higher percentage of granulocytes, a lower percentage of lymphocytes, and an increase in the granulocytes to lymphocytes ratio compared to healthy donors. These alterations were more marked in patients with metastasis. Somatostatin analogs (SSAs) restored the immunophenotype of patients to that seen in healthy donors. Finally, Pan-NEN patients showed a higher plasma concentration of IP-10, MCP-1, and IL-8 compared to healthy donors, suggesting a potential role for these cytokines as diagnostic biomarkers. CONCLUSION This study highlighted differences in the immunophenotype of patients with Pan- and Lung-NENs compared to healthy individuals; these alterations were partially restored by therapy.
Collapse
Affiliation(s)
- Ilena Boemi
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - Sara Piccini
- Endocrinology, Diabetology, and Andrology Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - Federico S Colombo
- Flow Cytometry Core, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - Valeria Smiroldo
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - Alessandro Zerbi
- Department of Biomedical Sciences, Humanitas University, 20072, Pieve Emanuele, Italy
- Pancreas Surgery Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - Giovanni Capretti
- Department of Biomedical Sciences, Humanitas University, 20072, Pieve Emanuele, Italy
- Pancreas Surgery Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - Marco Alloisio
- Department of Biomedical Sciences, Humanitas University, 20072, Pieve Emanuele, Italy
- Thoracic Surgery Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - Giampaolo Trivellin
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, 20072, Pieve Emanuele, Italy
| | - Elisabetta Lavezzi
- Endocrinology, Diabetology, and Andrology Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - Gherardo Mazziotti
- Endocrinology, Diabetology, and Andrology Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, 20072, Pieve Emanuele, Italy
| | - Eleonora Vitali
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy.
| | - Andrea G Lania
- Endocrinology, Diabetology, and Andrology Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, 20072, Pieve Emanuele, Italy
| |
Collapse
|
12
|
Chen R, Zhu L, Zhang Y, Cui D, Chen R, Guo H, Peng L, Xiao C. Predicting the unpredictable: a robust nomogram for predicting recurrence in patients with ampullary carcinoma. BMC Cancer 2024; 24:212. [PMID: 38360582 PMCID: PMC10870520 DOI: 10.1186/s12885-024-11960-0] [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: 10/12/2023] [Accepted: 02/05/2024] [Indexed: 02/17/2024] Open
Abstract
OBJECTIVE To screen the risk factors affecting the recurrence risk of patients with ampullary carcinoma (AC)after radical resection, and then to construct a model for risk prediction based on Lasso-Cox regression and visualize it. METHODS Clinical data were collected from 162 patients that received pancreaticoduodenectomy treatment in Hebei Provincial Cancer Hospital from January 2011 to January 2022. Lasso regression was used in the training group to screen the risk factors for recurrence. The Lasso-Cox regression and Random Survival Forest (RSF) models were compared using Delong test to determine the optimum model based on the risk factors. Finally, the selected model was validated using clinical data from the validation group. RESULTS The patients were split into two groups, with a 7:3 ratio for training and validation. The variables screened by Lasso regression, such as CA19-9/GGT, AJCC 8th edition TNM staging, Lymph node invasion, Differentiation, Tumor size, CA19-9, Gender, GPR, PLR, Drinking history, and Complications, were used in modeling with the Lasso-Cox regression model (C-index = 0.845) and RSF model (C-index = 0.719) in the training group. According to the Delong test we chose the Lasso-Cox regression model (P = 0.019) and validated its performance with time-dependent receiver operating characteristics curves(tdROC), calibration curves, and decision curve analysis (DCA). The areas under the tdROC curves for 1, 3, and 5 years were 0.855, 0.888, and 0.924 in the training group and 0.841, 0.871, and 0.901 in the validation group, respectively. The calibration curves performed well, as well as the DCA showed higher net returns and a broader range of threshold probabilities using the predictive model. A nomogram visualization is used to display the results of the selected model. CONCLUSION The study established a nomogram based on the Lasso-Cox regression model for predicting recurrence in AC patients. Compared to a nomogram built via other methods, this one is more robust and accurate.
Collapse
Affiliation(s)
- Ruiqiu Chen
- Medical School of Chinese PLA, Beijing, China
- Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People s Liberation Army (PLA) General Hospital, Beijing, China
- The First School of Clinical Medicine, Lanzhou University, No. 1, Donggangxi Rd, Chengguan District, 730000, Lanzhou, Gansu, China
| | - Lin Zhu
- Medical School of Chinese PLA, Beijing, China
- Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People s Liberation Army (PLA) General Hospital, Beijing, China
- The First School of Clinical Medicine, Lanzhou University, No. 1, Donggangxi Rd, Chengguan District, 730000, Lanzhou, Gansu, China
| | - Yibin Zhang
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
| | - Dongyu Cui
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Hao Guo
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li Peng
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Chaohui Xiao
- Faculty of Hepato-Biliary-Pancreatic Surgery, the First Medical Centre, Chinese People s Liberation Army (PLA) General Hospital, Beijing, China.
- Key Laboratory of Digital Hepatobiliary Surgery PLA, Beijing, China.
| |
Collapse
|
13
|
Silva RJG, Grippa WR, Pessanha RM, Enriquez-Martinez OG, Neto LCBS, Lopes-Júnior LC. Neutrophil/Lymphocyte Ratio and Platelet/Lymphocyte Ratio and Their Relationship with Nutritional Status and Quality of Life of Hospitalized Women with Breast Cancer. Nutr Cancer 2024; 76:296-304. [PMID: 38287698 DOI: 10.1080/01635581.2024.2304689] [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: 08/02/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/31/2024]
Abstract
Blood cell biomarkers, such as the neutrophil-lymphocyte ratio (NLR) and the platelet-lymphocyte ratio (PLR), have been recently used as prognostic markers in tumors. In this study, we investigated the association between NLR and PLR with sociodemographic, clinical, anthropometric, and quality of life factors of hospitalized women with non-metastatic breast cancer. A cross-sectional observational study was conducted at a reference center for oncological treatment in Southeast Brazil. Female participants aged over 18 years, with a histopathological diagnosis of stage I, II or III breast cancer, in any phase of antineoplastic treatment, were included. Our study revealed a high risk for participants, with high mean values of NLR and PLR, indicating low antitumor activity and worse prognosis. The binary logistic regression model showed that there was a significant association of the NLR marker and marital status (OR = 3.1; 95%CI = 1.06-8.57; p = 0.03) and, in relation to PLR, a trend was shown for a higher chance in women of black ethnicity to have increased PLR compared to white women (OR = 4.13; 95%CI = 0.96-17.70; p = 0.05). However, the inflammatory markers (NLR and PLR) did not show any significant association with nutritional factors. NLR and PLR are inflammatory biomarkers that can be easily obtained and measured in clinical practice.
Collapse
Affiliation(s)
- Roberto Júnio Gomes Silva
- Graduate Program in Nutrition and Health at the Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil
| | - Wesley Rocha Grippa
- Graduate Program in Public Health at the Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil
| | - Raphael Manhães Pessanha
- Graduate Program in Public Health at the Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil
| | | | | | - Luís Carlos Lopes-Júnior
- Graduate Program in Nutrition and Health at the Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil
- Graduate Program in Public Health at the Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil
| |
Collapse
|
14
|
Zhang R, Chen X, Miao C, Chen Y, Li Y, Shen J, Yuan M, Chen M, Cheng J, Liu S, Sun Q, Wu J. Tumor-associated macrophage-derived exosomal miR-513b-5p is a target of jianpi yangzheng decoction for inhibiting gastric cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117013. [PMID: 37572927 DOI: 10.1016/j.jep.2023.117013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/16/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jianpi Yangzheng decoction (JPYZ) possesses a potential anti-tumor activity in gastric cancer. However, potential effect of JPYZ on regulating tumor-associated macrophage (TAM)-derived exosomes to affect gastric cancer is still unclear. AIM OF STUDY We aimed to clarify the role of tumor-associated macrophage derived exosomes (TAM-exos) in invasive and metastasis of gastric cancer and the mechanism of JPYZ regulate TAM-exos against gastric cancer. MATERIALS AND METHODS Flow cytometry was performed to demonstrate whether JPYZ involved in TAM polarization. After JPYZ treatment, TAM conditioned medium (TAM-CM)/TAM-exos were co-cultured with gastric cancer cells and were detected by wound healing and transwell assay. Transcriptome sequencing and bioinformatics analysis predicted the exosomal miRNA after JPYZ intervention in TAM. miRNA mimic and inhibitor were used to verify the effect of miRNA in exosomes on gastric cancer cells. Q-PCR and luciferase reporter assay were employed to clarify the targeting relationship between miRNA and target gene. Western blot assay detected the expression levels of epithelial-mesenchymal transition (EMT) markers and related signaling pathways proteins. RESULTS We firstly demonstrated that TAM-CM intervened by JPYZ significantly inhibited the invasion and migration of gastric cancer. Furthermore, exosomes in TAM supernatants play a key role in migration of gastric cancer. Meanwhile, transcriptome sequencing and q-PCR revealed that miR-513b-5p expression was significantly reduced in TAM-exos intervened by JPYZ. And miR-513b-5p in TAM aggravated TAM-exos mediated invasion and migration of gastric cancer cells, the inhibitor of miR-513b-5p reversed TAM-exos mediated promotion. Bioinformatics analysis and luciferase reporter assay confirmed that PTEN was a direct target of miR-513b-5p in gastric cancer. MiR-513b-5p inhibited PTEN to activate AKT/mTOR signaling pathway thus promoting gastric cancer invasion and metastasis in vivo and in vitro. Importantly, JPYZ inhibited TAM derived exosomal miR-513b-5p, and alleviated AKT/mTOR activation by PTEN depended manner in gastric cancer. CONCLUSION TAM-exos containing miR-513b-5p lead to gastric cancer invasion and migration. Our findings clarify a novel TAM-exos mechanism of JPYZ for inhibiting gastric cancer progression.
Collapse
Affiliation(s)
- Ruijuan Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Xu Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Chunrun Miao
- Department of Gastroenterology, Dongtai Hospital of Traditional Chinese Medicine, Dongtai, Jiangsu, 224299, China
| | - Yuxuan Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Yaqi Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Junyu Shen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Mengyun Yuan
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Menglin Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Jian Cheng
- BD Bioscience, Becton, Dickinson and Company, Shanghai, 201200, China
| | - Shenlin Liu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Qingmin Sun
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Jian Wu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
15
|
Chakraborty P, Kumar R, Karn S, Raviya DD, Mondal P. Application of Oncolytic Poxviruses: An Emerging Paradigm in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:369-381. [PMID: 38801591 DOI: 10.1007/978-3-031-57165-7_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Despite the significant advancement of new tools and technology in the field of medical biology and molecular biology, the challenges in the treatment of most cancer types remain constant with the problem of developing resistance toward drugs and no substantial enhancement in the overall survival rate of cancer patients. Immunotherapy has shown the most promising results in different clinical and preclinical trials in the treatment of various cancer due to its higher efficacy and minimum collateral damage in many cancer patients as compared to conventional chemotherapy and radiotherapy. An oncolytic virus is a new class of immunotherapy that can selectively replicate in tumor cells and destroy them by the process of cell lysis while exerting minimum or no effect on a normal cell. Besides this, it can also activate the host's innate immune system, which generates an anti-tumor immune response to eliminate the tumor cells. Several wild types and genetically modified viruses have been investigated to show oncolytic behavior. Vaccinia virus has been studied extensively and tested for its promising oncolytic nature on various model systems and clinical trials. Recently, several engineered vaccinia viruses have been developed that express the desired genes encoded for selective penetration in tumor cells and enhanced activation of the immune system for generating anti-tumor immunity. However, further investigation is required to prove their potential and enhance their therapeutic efficacy.
Collapse
Affiliation(s)
- Prasenjit Chakraborty
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India.
| | - Randhir Kumar
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Sanjay Karn
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Dharmiben D Raviya
- Department of Biosciences, School of Science, Indrashil University, Rajpur-Kadi, Mehsana, Gujarat, 382740, India
| | - Priya Mondal
- Laboratory of Cell Biology, National Cancer Institute, National Institute of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
16
|
Guo Z, Wang G, Yun Z, Li Y, Huang B, Jin Q, Chen Y, Xu L, Lv W. Global research trends in tumor stem cell-derived exosomes and tumor microenvironment: visualization biology analysis. J Cancer Res Clin Oncol 2023; 149:17581-17595. [PMID: 37914951 PMCID: PMC10657319 DOI: 10.1007/s00432-023-05450-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023]
Abstract
BANKGROUND The tumor microenvironment (TME) is an internal environment composed of various cells and an extracellular matrix. Cancer stem cell-derived exosomes (CSC-Exos), as essential messengers involved in various tumor processes, are important carriers for bidirectional communication between the tumor microenvironment and tumor cells and play an important role in the tumor microenvironment. Nevertheless, few bibliometric analyses have been systematically studied in this field. METHODS Therefore, we aimed to visualize the research hotspots and trends in this field through bibliometrics to comprehend the future evolution of fundamental and clinical research, as well as to offer insightful information and fresh viewpoints. The Scopus database was used to search the research literature related to exosomes and tumor microenvironments after the establishment of this repository. CiteSpace (version 5.8.R3) and VOSviewer (version 1.6.16) were used for visualization and analysis. RESULTS In this study, a total of 2077 articles and reviews were included, with the number of articles on exosomes and tumor microenvironments significantly increasing yearly. Recent trends showed that the potential value of exosomes as "tumor diagnostics" and "the application prospect of exosomes as therapeutic agents and drug delivery carriers" will receive more attention in the future. CONCLUSIONS We revealed the current status and hotspots of tumor stem cell-derived exosomes and tumor microenvironments globally through bibliometrics. The prospect of the regulatory role of CSC-Exos in TME, the potential value of diagnosis, and the application of drug delivery vectors will all remain cutting-edge research areas in the field of tumor therapy. Meanwhile, this study provided a functional literature analysis for related researchers.
Collapse
Affiliation(s)
- Ziwei Guo
- Department of Infection, China Academy of Chinese Medical Sciences, Guang' anmen Hospital, Beijing, China
| | - Gang Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhangjun Yun
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanbo Li
- Department of Infection, China Academy of Chinese Medical Sciences, Guang' anmen Hospital, Beijing, China
| | - Bohao Huang
- Guang' anmen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Jin
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Luchun Xu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wenliang Lv
- Department of Infection, China Academy of Chinese Medical Sciences, Guang' anmen Hospital, Beijing, China.
| |
Collapse
|
17
|
Ourique F, Kviecinski MR, Bagolin do Nascimento A. Editorial: Nutritional modulation of immune function in cancer. Front Nutr 2023; 10:1290026. [PMID: 37841400 PMCID: PMC10576555 DOI: 10.3389/fnut.2023.1290026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/21/2023] [Indexed: 10/17/2023] Open
Affiliation(s)
- Fabiana Ourique
- Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Maicon Roberto Kviecinski
- Department of Biochemistry, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Amanda Bagolin do Nascimento
- Department of Nutrition, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
18
|
Mitchell MI, Loudig O. Communicator Extraordinaire: Extracellular Vesicles in the Tumor Microenvironment Are Essential Local and Long-Distance Mediators of Cancer Metastasis. Biomedicines 2023; 11:2534. [PMID: 37760975 PMCID: PMC10526527 DOI: 10.3390/biomedicines11092534] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Human tumors are increasingly being described as a complex "ecosystem", that includes many different cell types, secreted growth factors, extracellular matrix (ECM) components, and microvessels, that altogether create the tumor microenvironment (TME). Within the TME, epithelial cancer cells control the function of surrounding stromal cells and the non-cellular ECM components in an intricate orchestra of signaling networks specifically designed for cancer cells to exploit surrounding cells for their own benefit. Tumor-derived extracellular vesicles (EVs) released into the tumor microenvironment are essential mediators in the reprogramming of surrounding stromal cells, which include cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), tumor-infiltrating lymphocytes (TILs), and tumor endothelial cells (TECs), which are responsible for the promotion of neo-angiogenesis, immune cell evasion, and invasion which are essential for cancer progression. Perhaps most importantly, tumor-derived EVs play critical roles in the metastatic dissemination of tumor cells through their two-fold role in initiating cancer cell invasion and the establishment of the pre-metastatic niche, both of which are vital for tumor cell migration, homing, and colonization at secondary tumor sites. This review discusses extracellular vesicle trafficking within the tumor microenvironment and pre-metastatic niche formation, focusing on the complex role that EVs play in orchestrating cancer-to-stromal cell communication in order to promote the metastatic dissemination of cancer cells.
Collapse
Affiliation(s)
| | - Olivier Loudig
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA;
| |
Collapse
|
19
|
Qin R, Ren W, Ya G, Wang B, He J, Ren S, Jiang L, Zhao S. Role of chemokines in the crosstalk between tumor and tumor-associated macrophages. Clin Exp Med 2023; 23:1359-1373. [PMID: 36173487 PMCID: PMC10460746 DOI: 10.1007/s10238-022-00888-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/07/2022] [Indexed: 11/03/2022]
Abstract
Tumor microenvironment (TME) consists of a dynamic network of non-tumoral stromal cells, including cancer-associated fibroblasts, endothelial cells, tumor-associated macrophages (TAMs), B and T cells. In the TME, TAMs support tumor initiation, progression, invasion and metastasis by promoting angiogenesis and immunosuppression of the tumor cells. There is close crosstalk between TAMs and tumor cells. Notably, chemokines are a significant messenger mediating the crosstalk between tumor cells and TAMs. TAMs can promote tumor progression via secretion of chemokines. Various chemokines secreted by tumors are involved in the generation and polarization of TAMs, the infiltration of TAMs in tumors, and the development of TAMs' suppressive function. This paper reviews CCL2-CCR2, CCL3/5-CCR5, CCL15-CCR1, CCL18-CCR8, CX3CL1/CCL26-CX3CR1, CXCL8-CXCR1/2, CXCL12-CXCR4/CXCR7 signaling pathways, their role in the recruitment, polarization and exertion of TAMs, and their correlation with tumor development, metastasis and prognosis. Furthermore, we present the current research progress on modulating the effects of TAMs with chemokine antagonists and discuss the prospects and potential challenges of using chemokine antagonists as therapeutic tools for cancer treatment. The TAMs targeting by chemokine receptor antagonists in combination with chemotherapy drugs, immune checkpoint inhibitors or radiotherapy appears to be a promising approach.
Collapse
Affiliation(s)
- Rui Qin
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Weihong Ren
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Guoqi Ya
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Bei Wang
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jiao He
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shaoxin Ren
- The First Clinical Medical Institute, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Lu Jiang
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shuo Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Wu J, Zhou X, Ren J, Zhang Z, Ju H, Diao X, Jiang S, Zhang J. Glycosyltransferase-related prognostic and diagnostic biomarkers of uterine corpus endometrial carcinoma. Comput Biol Med 2023; 163:107164. [PMID: 37329616 DOI: 10.1016/j.compbiomed.2023.107164] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/19/2023]
Abstract
Uterine corpus endometrial carcinoma (UCEC) has a strong ability of invasion and metastasis, high recurrence rate, and poor survival. Glycosyltransferases are one of the most important enzymes that coordinate the glycosylation process, and abnormal modification of proteins by glycosyltransferases is closely related to the occurrence and development of cancer. However, there were fewer reports on glycosyltransferase related biomarkers in UCEC. In this paper, based on the UCEC transcriptome data published on The Cancer Genome Atlas (TCGA), we predicted the relationship between the expression of glycosyltransferase-related genes (GTs) and the diagnosis and prognosis of UCEC using bioinformatics methods. And validation of model genes by clinical samples. We used 4 methods: generalized linear model (GLM), random forest (RF), support vector machine (SVM) and extreme gradient boosting (XGB) to screen biomarkers with diagnostic significance, and the binary logistic regression was used to establish a diagnostic model for the 2-GTs (AUC = 0.979). And the diagnostic model was validated using a GEO external database (AUC = 0.978). Moreover, a prognostic model for the 6-GTs was developed using univariate, Lasso, and multivariate Cox regression analyses, and the model was made more stable by internal validation using the bootstrap. In addition, risk score is closely related to immune microenvironment (TME), immune infiltration, mutation, immunotherapy and chemotherapy. Overall, this study provides novel biomarkers for the diagnosis and prognosis of UCEC, and the models established by these biomarkers can also provide a good reference for individualized and precision medicine in UCEC.
Collapse
Affiliation(s)
- Jiaoqi Wu
- Department of Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xiaozhu Zhou
- Department of Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Jie Ren
- Department of Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Zhen Zhang
- Department of Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Haoyu Ju
- Department of Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xiaoqi Diao
- Department of Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shuyi Jiang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, 36 SanHao Street, Heping District, Shenyang, 110000, China.
| | - Jing Zhang
- Department of Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
21
|
Li C, Guan R, Li W, Wei D, Cao S, Xu C, Chang F, Wang P, Chen L, Lei D. Single-cell RNA sequencing reveals tumor immune microenvironment in human hypopharygeal squamous cell carcinoma and lymphatic metastasis. Front Immunol 2023; 14:1168191. [PMID: 37503341 PMCID: PMC10369788 DOI: 10.3389/fimmu.2023.1168191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
Background Human hypopharygeal squamous cell carcinoma (HSCC) is a common head and neck cancer with a poor prognosis in advanced stages. The occurrence and development of tumor is the result of mutual influence and co-evolution between tumor cells and tumor microenvironment (TME). Tumor immune microenvironment (TIME) refers to the immune microenvironment surrounding tumor cells. Studying TIME in HSCC could provide new targets and therapeutic strategies for HSCC. Methods We performed single-cell RNA sequencing (scRNA-seq) and analysis of hypopharyngeal carcinoma, paracancerous, and lymphoid tissues from five HSCC patients. Subdivide of B cells, T cells, macrophages cells, and monocytes and their distribution in three kinds of tissues as well as marker genes were analyzed. Different genes of IGHG1 plasma cells and SPP1+ macrophages between HSCC tissues, adjacent normal tissues and lymphatic tissues were analyzed. Additionally, we studied proliferating lymphocytes, T cells exhaustion, and T cell receptor (TCR) repertoire in three kinds of tissues. Results Transcriptome profiles of 132,869 single cells were obtained and grouped into seven cell clusters, including epithelial cells, lymphocytes, mononuclear phagocytics system (MPs), fibroblasts, endothelial cells (ECs), plasmacytoid dendritic cells (pDCs), and mast cells. Tumor metastasis occurred in three lymphoid tissues. Four distinct populations were identified from lymphocytes, including B cells, plasma cells, T cells and proliferating lymphocytes. We found IGHA1 and IGHG1 specific plasma cells significantly overexpressed in HSCC tissues compared with normal hypopharygeal tissues and lymphatic tissues. Five distinct populations from MPs were identified, including macrophages, monocytes, mature dendritic cells (DCs), Type 1 conventional dendritic cells (cDC1) and Type 2 conventional dendritic cells (cDC2). SPP1+ macrophages were significantly overexpressed in HSCC tissues and lymphatic tissues compared with normal hypopharygeal tissues, which are thought to be M2-type macrophages. Exhaustion of CD8+ Teff cells occurred in HSCC tissues. At last, we verified that IgA and IgG1 protein expression levels were significantly up-regulated in HSCC tissues compared to adjacent normal tissues. Conclusion Overall, this study revealed TIME in HSCC and lymphatic metastasis, and provided potential therapeutic targets for HSCC.
Collapse
|
22
|
So YK, Kim Z, Cheong TY, Chung MJ, Baek CH, Son YI, Seok J, Jung YS, Ahn MJ, Ahn YC, Oh D, Cho BH, Chung MK. Detection of Cancer Recurrence Using Systemic Inflammatory Markers and Machine Learning after Concurrent Chemoradiotherapy for Head and Neck Cancers. Cancers (Basel) 2023; 15:3540. [PMID: 37509202 PMCID: PMC10377662 DOI: 10.3390/cancers15143540] [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: 06/01/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Pretreatment values of the neutrophil-to-lymphocyte ratio (NLR) and the platelet-to-lymphocyte ratio (PLR) are well-established prognosticators in various cancers, including head and neck cancers. However, there are no studies on whether temporal changes in the NLR and PLR values after treatment are related to the development of recurrence. Therefore, in this study, we aimed to develop a deep neural network (DNN) model to discern cancer recurrence from temporal NLR and PLR values during follow-up after concurrent chemoradiotherapy (CCRT) and to evaluate the model's performance compared with conventional machine learning (ML) models. Along with conventional ML models such as logistic regression (LR), random forest (RF), and gradient boosting (GB), the DNN model to discern recurrences was trained using a dataset of 778 consecutive patients with primary head and neck cancers who received CCRT. There were 16 input features used, including 12 laboratory values related to the NLR and the PLR. Along with the original training dataset (N = 778), data were augmented to split the training dataset (N = 900). The model performance was measured using ROC-AUC and PR-AUC values. External validation was performed using a dataset of 173 patients from an unrelated external institution. The ROC-AUC and PR-AUC values of the DNN model were 0.828 ± 0.032 and 0.663 ± 0.069, respectively, in the original training dataset, which were higher than the ROC-AUC and PR-AUC values of the LR, RF, and GB models in the original training dataset. With the recursive feature elimination (RFE) algorithm, five input features were selected. The ROC-AUC and PR-AUC values of the DNN-RFE model were higher than those of the original DNN model (0.883 ± 0.027 and 0.778 ± 0.042, respectively). The ROC-AUC and PR-AUC values of the DNN-RFE model trained with a split dataset were 0.889 ± 0.032 and 0.771 ± 0.044, respectively. In the external validation, the ROC-AUC values of the DNN-RFE model trained with the original dataset and the same model trained with the split dataset were 0.710 and 0.784, respectively. The DNN model with feature selection using the RFE algorithm showed the best performance among the ML models to discern a recurrence after CCRT in patients with head and neck cancers. Data augmentation by splitting training data was helpful for model performance. The performance of the DNN-RFE model was also validated with an external dataset.
Collapse
Affiliation(s)
- Yoon Kyoung So
- Department of Otorhinolaryngology-Head & Neck Surgery, Inje University College of Medicine, Ilsan Paik Hospital, Goyang-Si 10380, Republic of Korea
| | - Zero Kim
- Medical AI Research Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Data Convergence and Future Medicine, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Taek Yoon Cheong
- Department of Otorhinolaryngology-Head & Neck Surgery, Inje University College of Medicine, Ilsan Paik Hospital, Goyang-Si 10380, Republic of Korea
| | - Myung Jin Chung
- Medical AI Research Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Data Convergence and Future Medicine, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Chung-Hwan Baek
- Department of Otolaryngology-Head & Neck Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Young-Ik Son
- Department of Otolaryngology-Head & Neck Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Jungirl Seok
- Center for Thyroid Cancer, Department of Otolaryngology-Head and Neck Surgery, Research Institute and Hospital, National Cancer Center, Goyang-si 10408, Republic of Korea
| | - Yuh-Seog Jung
- Center for Thyroid Cancer, Department of Otolaryngology-Head and Neck Surgery, Research Institute and Hospital, National Cancer Center, Goyang-si 10408, Republic of Korea
| | - Myung-Ju Ahn
- Divison of Hematology and Medical Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Yong Chan Ahn
- Department of Radiation Oncology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Dongryul Oh
- Department of Radiation Oncology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Baek Hwan Cho
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam-Si 13488, Republic of Korea
- Institute of Biomedical Informatics, School of Medicine, CHA University, Seongnam-Si 13488, Republic of Korea
| | - Man Ki Chung
- Department of Otolaryngology-Head & Neck Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| |
Collapse
|
23
|
Gu X, Shen H, Xiang Z, Li X, Zhang Y, Zhang R, Su F, Wang Z. Exploring the Correlation Between GPR176, a Potential Target Gene of Gastric Cancer, and Immune Cell Infiltration. Pharmgenomics Pers Med 2023; 16:519-535. [PMID: 37284492 PMCID: PMC10241216 DOI: 10.2147/pgpm.s411199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/26/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction GPR176, an orphan G protein-coupled receptor (GPCR), is essential for the progression of gastrointestinal cancers. However, it is still unclear how GPR176 affects tumor immunity and patient prognosis in gastric cancer (GC). Methods The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were searched in this investigation to assess the expression patterns of GPR176 in GC tissues and normal gastric mucosa. The findings were further verified using immunohistochemical tests and quantitative Real-Time Polymerase Chain Reaction (qRT-PCR). The Kaplan-Meier method, univariate logistic regression, and Cox regression were then used to investigate the relationship between GPR176 and clinical traits. Additionally, the potential correlation between GPR176, immune checkpoint genes, and immune cell infiltration levels was investigated. Results As per the research findings, GC tissues had higher levels of GPR176 than normal tissues. Additionally, individuals with high expression of GPR176 had a worse 10-year overall survival (OS), in contrast with those having a low expression of GPR176 (p < 0.001). The OS of GC can be predicted using a validated nomogram model. The expression of GPR176 demonstrated a negative correlation with CD8+ T cells. When compared to the low-expression group of GPR176, Tumor Immune Dysfunction and Exclusion (TIDE) analysis demonstrated that the high-expression group had a considerably higher risk of immune evasion. A remarkable difference (variation) was observed in the levels of GPR176 expression across both groups, ie, low and high-risk groups, as determined by the immune phenomenon scores (IPS) immunotherapy assessment. Conclusion By examining GPR176 from various biological perspectives, it was determined that GPR176 can act as a predictive biomarker for poor patient prognosis in GC. Additionally, it was observed that GPR176 is capable of suppressing the proliferation of CD8+ T cells and facilitating immune evasion.
Collapse
Affiliation(s)
- Xianhua Gu
- Department of Gynecology Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Honghong Shen
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Zheng Xiang
- Department of Surgical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Xinwei Li
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Yue Zhang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Rong Zhang
- Department of Gynecology Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Fang Su
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Zishu Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| |
Collapse
|
24
|
Ziani I, Ibrahimi A, Bellouki O, Elsayegh H, Abouqal R, Nouini Y, Bouziane A. Pre-therapeutic lymphocytopenia: a new prognostic factor for failure of endovesical BCG-immunotherapy in non-muscle invasive bladder cancer. AFRICAN JOURNAL OF UROLOGY 2023. [DOI: 10.1186/s12301-023-00348-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
Abstract
Background
Inflammation plays a key role in the initiation and development of cancers. The prognostic value of inflammation biomarkers is proven in several urological and non-urological cancers. Knowing that the mechanism of action of endovesical BCG-immunotherapy in the treatment of non-muscle-invasive bladder cancer (NMIBC) is based on inflammation; lymphocytes have a key role in this reaction, particularly in the cytotoxic phase and can be predictive biomarkers of the response to BCG-therapy. The main objective of our work is therefore to study the impact of the number of lymphocytes on the response to endovesical BCG-immunotherapy, and more specifically lymphocytopenia (Lp) as a prognostic factor for BCG-failure.
Methods
Our study is a monocentric retrospective cohort carried for prognostic purposes, including 200 patients neodiagnosed with non-muscle-invasive bladder cancer (Ta -T1 stages), who required adjuvant treatment to TURB by BCG-immunotherapy, over a period of 5 years from January 2012 to December 2016. The cutoff value chosen was 1.67 × 109/L using maximized Log-Rank test. Survival analysis was studied using a Kaplan–Meier model. The comparison between the thresholds (L ≤ Vs > 1.67 × 109/L) concerning the recurrence and progression rates was carried out using the Log-Rank test. The association between lymphocytopenia and BCG-therapy failure was assessed in univariate and multivariate analysis by the Cox model. Statistical analysis was performed using Jamovi statistical software.
Results
One hundred and eight patients had a lymphocyte count > 1.67 × 109/L while 92 had a lymphocyte count ≤ 1.67 × 109/L. The median lymphocyte value was 1.64 (1.19; 2.4). The median survival without failure of BCG treatment was significantly better in the high lymphocyte-count group, with median of 22 months in the > 1.67 × 109/L group versus 11 months until failure in the ≤ 1.67 × 109/L group. A lymphocyte count ≤ 1.67 × 109/L was associated with failure of BCG-therapy in univariate (HR = 4.80, P ≤ 0.001) and multivariate (HR = 1.88, P = 0.025) studies. Other factors associated in the univariate study were found: T1 stage (P = 0.001), high-grade urothelial carcinoma (P = 0.001), multifocal tumor (P = 0.001), tumor size > 3 cm (P = 0.001), concomitant carcinoma in situ (Cis) (P = 0.001) and vascular emboli (P = 0.001). Multivariate study showed significant factors that are, in addition to lymphocytopenia, the presence of T1 stage (P = 0.011) and vascular emboli (P = 0.013).
Conclusion
Our study has shown an association between lymphocytes count and NMIBC progression. Patients with lymphocytopenia carry an increased risk of endovesical BCG-immunotherapy failure. These results should be further validated.
Collapse
|
25
|
Ou J, Zhu M, Ju X, Xu D, Lu G, Li K, Jiang W, Wan C, Zhao Y, Han Y, Tian Y, Niu Z. One-Dimensional Rod-like Tobacco Mosaic Virus Promotes Macrophage Polarization for a Tumor-Suppressive Microenvironment. NANO LETTERS 2023; 23:2056-2064. [PMID: 36695738 DOI: 10.1021/acs.nanolett.2c03809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The phenotype of tumor-associated macrophages plays an important role in their function of regulating the tumor immune microenvironment. The M1-phenotype macrophages display tumor-killing and immune activating functions. Here we show that the tobacco mosaic virus (TMV), a rod-like plant virus, can polarize macrophages to an M1 phenotype and shape a tumor-suppressive microenvironment. RAW 264.7 cells and bone marrow derived-macrophages (BMDMs) can recognize TMV via Toll-like receptor-4, and then the MAPK and NF-κB signaling pathways are activated, leading to the production of pro-inflammatory factors. Furthermore, the in vivo assessments on a subcutaneous co-injection tumor model show that the TMV-polarized BMDMs shape a tumor-suppressive microenvironment, resulting in remarkable delay of 4T1 tumor growth. Another in vivo assessment on an established tumor model indicates the high tumor-metastasis-inhibiting capacity of TMV-polarized BMDMs. This work suggests a role for this plant virus in macrophage-mediated therapeutic approaches and provides a strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Jinzhao Ou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Meng Zhu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Xiaoyan Ju
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Dandan Xu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Guojun Lu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Kejia Li
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Wei Jiang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Chenxiao Wan
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Yuexia Zhao
- Biochemical Engineering College, Beijing Union University, No. 97, North Fourth Ring East Road, Beijing 100023, P.R. China
| | - Yongping Han
- Biochemical Engineering College, Beijing Union University, No. 97, North Fourth Ring East Road, Beijing 100023, P.R. China
| | - Ye Tian
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
| | - Zhongwei Niu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, 29 Zhongguancun East Road, Beijing 100190, P.R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
26
|
Garcia P, Hartman D, Choudry H, Pai RK. CD8 + T-cell Density Is an Independent Predictor of Survival and Response to Adjuvant Chemotherapy in Stage III Colon Cancer. Appl Immunohistochem Mol Morphol 2023; 31:69-76. [PMID: 36508180 PMCID: PMC11199076 DOI: 10.1097/pai.0000000000001094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/12/2022] [Indexed: 12/14/2022]
Abstract
We assessed CD8 + T-cell density in 351 resected stage II to III colon cancers from 2011 to 2015 and correlated the findings with disease-free survival and survival effect of adjuvant chemotherapy. Most tumors (70%) had high/intermediate CD8 + T-cell density, and this was significantly associated with mismatch repair deficiency compared with tumors with low CD8 + T-cell density (28% vs. 13%, P =0.003). Fewer tumors with high/intermediate CD8 + T-cell density had adverse histologic features compared with tumors with low CD8 + T-cell density including high tumor budding (16% vs. 27%) and venous (22% vs. 35%), lymphatic (54% vs. 65%), and perineural (23% vs. 33%) invasion (all with P <0.05). In the stage III cohort, high/intermediate CD8 + T-cell density was an independent predictor of disease-free survival on multivariate analysis (hazard ratio: 0.39, 0.21 to 0.71 95% CI, P =0.002). For stage III patients with high/intermediate CD8 + T-cell density, adjuvant chemotherapy was significantly associated with improved disease-free survival (hazard ratio: 0.28, 0.11 to 0.74 95% CI, P =0.01) whereas stage III patients with low CD8 + T-cell density did not have improved survival with adjuvant chemotherapy. In conclusion, in stage III colon cancer, CD8 + T-cell density is an independent prognostic biomarker for disease-free survival and may help to identify patients who benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Paulo Garcia
- Departments of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Douglas Hartman
- Departments of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Haroon Choudry
- Departments of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Reetesh K. Pai
- Departments of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
27
|
Wen F, Meng F, Li X, Li Q, Liu J, Zhang R, Zhao Y, Zhang Y, Wang X, Ju S, Cui Y, Lu Z. Characterization of prognostic value and immunological roles of RAB22A in hepatocellular carcinoma. Front Immunol 2023; 14:1086342. [PMID: 36936971 PMCID: PMC10021109 DOI: 10.3389/fimmu.2023.1086342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Background The protein-coding gene RAB22A, a member of the RAS oncogene family, is amplified or overexpressed in certain cancers. However, its action mechanism in hepatocellular carcinoma (HCC) remains unclear. Here, we aimed to examine the connection between RAB22A and survival prognosis in HCC and explore the biological significance of RAB22A. Methods A database-based pan-cancer expression analysis of RAB22A was performed. Kaplan-Meier analysis and Cox regression were performed to evaluate the association between RAB22A expression and survival prognosis in HCC. Using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA), various potential biological functions and regulatory pathways of RAB22A in HCC were discovered. Tumor immune infiltration was studied using the single sample gene set enrichment analysis (ssGSEA) method. N6-methyladenosine modifications and the regulatory network of competitive endogenous RNA (ceRNA) were verified in the TCGA cohort. Results RAB22A was upregulated in HCC samples and cell lines. A high RAB22A expression in HCC was strongly correlated with sex, race, age, weight, TNM stage, pathological stage, tumor status, histologic grade, TP53 mutation status, and alpha fetal protein (AFP) levels. Overexpression of RAB22A indicated a poor prognosis was related to overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI). GO and KEGG analyses revealed that the differentially expressed genes related to RAB22A might be involved in the proteasomal protein catabolic process, ncRNA processing, ribosome ribosomal subunit, protein serine/threonine kinase activity, protein serine kinase activity, Endocytosis, and non-alcoholic fatty liver disease. GSEA analyses revealed that the differentially expressed genes related to RAB22A might be involved in the T cell receptor, a co-translational protein, that binds to the membrane, axon guidance, ribosome, phagocytosis, and Eukaryotic translation initiation. RAB22A was correlated with N6-methyladenosine expression in HCC and established RAB22A-related ceRNA regulatory networks. Finally,RAB22A expression was positively connected the levels of infiltrating with T helper cells, Tcm cells, and Th2 cells,In contrast, we observed negatively correlations with cytotoxic cells, DCs, and pDCs cells.Moreover,RAB22A expression showed a strong correlation with various immunomarkergroups in HCC. Conclusions RAB22A is a potential therapeutic target for improving HCC prognosis and is closely related to immune cell infiltration.
Collapse
Affiliation(s)
- Fukai Wen
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanshuai Meng
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuewen Li
- The Department of Inpatient Central Operating Room, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingyu Li
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaming Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunzheng Zhao
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Wang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuai Ju
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yifeng Cui
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Yifeng Cui, ; Zhaoyang Lu,
| | - Zhaoyang Lu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Yifeng Cui, ; Zhaoyang Lu,
| |
Collapse
|
28
|
Sadr S, Ghiassi S, Lotfalizadeh N, Simab PA, Hajjafari A, Borji H. Antitumor Mechanisms of Molecules Secreted by Trypanosoma cruzi in Colon and Breast Cancer: A Review. Anticancer Agents Med Chem 2023; 23:1710-1721. [PMID: 37254546 DOI: 10.2174/1871520623666230529141544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/14/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Molecules secreted by Trypanosoma cruzi (T. cruzi) have beneficial effects on the immune system and can fight against cancer by inhibiting the growth of tumor cells, preventing angiogenesis, and promoting immune activation. OBJECTIVE This study aimed to investigate the effects of molecules secreted by Trypanosoma cruzi on the growth of colon and breast cancer cells, to understand the underlying mechanisms of action. RESULTS Calreticulin from T. cruzi, a 45 kDa protein, participates in essential changes in the tumor microenvironment by triggering an adaptive immune response, exerting an antiangiogenic effect, and inhibiting cell growth. On the other hand, a 21 kDa protein (P21) secreted at all stages of the parasite's life cycle can inhibit cell invasion and migration. Mucins, such as Tn, sialyl-Tn, and TF, are present both in tumor cells and on the surface of T. cruzi and are characterized as common antigenic determinants, inducing a cross-immune response. In addition, molecules secreted by the parasite are used recombinantly in immunotherapy against cancer for their ability to generate a reliable and long-lasting immune response. CONCLUSION By elucidating the antitumor mechanisms of the molecules secreted by T. cruzi, this study provides valuable insights for developing novel therapeutic strategies to combat colon and breast cancer.
Collapse
Affiliation(s)
- Soheil Sadr
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Shakila Ghiassi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Narges Lotfalizadeh
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Pouria Ahmadi Simab
- Department of Pathobiology, Faculty of Veterinary Medicine, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Ashkan Hajjafari
- Department of Pathobiology, Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
29
|
Li Q, Wu J, Zhu M, Tang Y, Jin L, Chen Y, Jin M, Peng Z. A novel risk signature based on autophagy-related genes to evaluate tumor immune microenvironment and predict prognosis in hepatocellular carcinoma. Comput Biol Med 2022. [DOI: 10.1016/j.compbiomed.2022.106437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Zhang Z, Zhou Y, Zhao S, Ding L, Chen B, Chen Y. Nanomedicine-Enabled/Augmented Cell Pyroptosis for Efficient Tumor Nanotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203583. [PMID: 36266982 PMCID: PMC9762308 DOI: 10.1002/advs.202203583] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/23/2022] [Indexed: 05/19/2023]
Abstract
The terrible morbidity and mortality of malignant tumors urgently require innovative therapeutics, especially for apoptosis-resistant tumors. Pyroptosis, a pro-inflammatory form of programmed cell death (PCD), is featured with pore formation in plasma membrane, cell swelling with giant bubbles, and leakage of cytoplasmic pro-inflammatory cytokines, which can remodel the tumor immune microenvironment by stimulating a "cold" tumor microenvironment to be an immunogenic "hot" tumor microenvironment, and consequently augment the therapeutic efficiency of malignant tumors. Benefiting from current advances in nanotechnology, nanomedicine is extensively applied to potentiate, enable, and augment pyroptosis for enhancing cancer-therapeutic efficacy and specificity. This review provides a concentrated summary and discussion of the most recent progress achieved in this emerging field, highlighting the nanomedicine-enabled/augmented specific pyroptosis strategy for favoring the construction of next-generation nanomedicines to efficiently induce PCD. It is highly expected that the further clinical translation of nanomedicine can be accelerated by inducing pyroptotic cell death based on bioactive nanomedicines.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of UltrasoundAffiliated Hospital of Jiangsu UniversityZhenjiang212000P. R. China
| | - Yajun Zhou
- Department of UltrasoundThe Fourth Affiliated HospitalNanjing Medical UniversityNanjing210029P. R. China
| | - Shuangshuang Zhao
- Department of UltrasoundAffiliated Hospital of Jiangsu UniversityZhenjiang212000P. R. China
| | - Li Ding
- Tongji University School of MedicineShanghai Tenth People's HospitalTongji University Cancer CenterShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Baoding Chen
- Department of UltrasoundAffiliated Hospital of Jiangsu UniversityZhenjiang212000P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| |
Collapse
|
31
|
Minabian S, Soleimani S. S, Torabi M, Mohammadi M, Ranjbar H. Evaluation of P53 protein expression in gingival tissues of patients with chronic periodontitis by immunohistochemistry methods. Clin Exp Dent Res 2022; 8:1348-1353. [PMID: 36263737 PMCID: PMC9760160 DOI: 10.1002/cre2.668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Periodontitis is one of the most important periodontal diseases that can be affected by many factors. Although the mechanism of periodontitis development is not yet fully understood, previous studies suggest that apoptosis may be one of the pathological factors that can affect the process of the disease by destroying old and damaged cells. Low expression of P53 protein is one of the reasons for delaying cell death that allows damaged cells to survive longer and gives more time for the chance of mutations and pathogenesis. Because of the important role of P53 in gingival cells of patients with chronic periodontitis, the objective of our study is to evaluate the P53 protein expression in gingival tissues of patients with chronic periodontitis by immunohistochemistry methods. MATERIALS AND METHODS In this cross-sectional study, 35 patients with severe to moderate chronic periodontitis (loss of attachment ≥3 mm, probing depth ≥5 mm) with no treatment and 25 people who were healthy for periodontal problems were examined. Gingival biopsies from marginal and attached gingiva were obtained, prepared, and mounted on slides. Then, the expression of P53 on each slide was evaluated by optic microscopy after using P53 antibodies and staining with hematoxylin-eosin (immunohistochemistry method). Data were analyzed using independent t-test, Mann-Whitney U-test, and Spearman correlation test using SPSS Statistics version 18.0. RESULTS The mean ages of participants in the case and control groups were 37.58 and 32.09, respectively. Our results showed that the expression of P53 was not significant in periodontitis compared to the control group (p > .05). Also, gender could not affect the expression of P53 in both groups (p > .05), and there was no significant relationship between age and P53 gene incidence. CONCLUSION Chronic periodontitis has no significant effect on P53 expression, so changes in apoptosis due to P53 expression in periodontitis are not significant.
Collapse
Affiliation(s)
- Samaneh Minabian
- Oral and Dental Diseases Research CenterKerman University of Medical SciencesKermanIran
| | - Shima Soleimani S.
- Oral and Dental Diseases Research CenterKerman University of Medical SciencesKermanIran
| | - Molook Torabi
- Oral and Dental Diseases Research CenterKerman University of Medical SciencesKermanIran,Oral and Maxillofacial Pathology Department, School of DentistryKerman University of Medical SciencesKermanIran
| | - Mohammad Mohammadi
- Oral and Dental Diseases Research CenterKerman University of Medical SciencesKermanIran,Periodontics Department, School of DentistryKerman University of Medical SciencesKermanIran
| | - Hadi Ranjbar
- Mental Health Research Center, Psychological Health Research InstituteIran University of Medical SciencesTehranIran
| |
Collapse
|
32
|
Wang Z, Tian X, Wang C, Qi X, Gracia‐Sancho J, Dong L. Transforming one organ into another to overcome challenges in tissue engineering. PORTAL HYPERTENSION & CIRRHOSIS 2022; 1:116-124. [DOI: 10.1002/poh2.26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/11/2022] [Indexed: 01/03/2025]
Abstract
AbstractTissue engineering (TE) is promising for the regeneration of failed organs. However, immune rejection, shortage of seed cells, and unintegrated blood vessels restrict the development and clinical application of TE. The last factor is the most challenging and intractable. Harnessing the mature blood vessel network in existing dispensable organs could be a powerful approach to effectively overcome the obstacles. After being remodeled to harbor an immunosuppressive and proregenerative niche, these potential target organs can be transformed into other organs with specific physiological functions, compensating the latter's failed native functions. Organ transformation, such as a hepatized spleen, represents an effective and encouraging TE strategy. In this review, we discuss the current development and obstacles of TE and its feasibility and superiority in organ transformation.
Collapse
Affiliation(s)
- Zhenzhen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
| | - Xuejiao Tian
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences University of Macau Taipa Macau SAR China
| | - Xiaolong Qi
- CHESS Center, Institute of Portal Hypertension The First Hospital of Lanzhou University Lanzhou Gansu China
| | - Jordi Gracia‐Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital University of Bern Bern Switzerland
- Liver Vascular Biology Research Group IDIBAPS Research Institute, CIBEREHD Barcelona Spain
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
- Chemistry and Biomedicine Innovative Center Nanjing University Nanjing Jiangsu China
| |
Collapse
|
33
|
Chan Wah Hak CML, Rullan A, Patin EC, Pedersen M, Melcher AA, Harrington KJ. Enhancing anti-tumour innate immunity by targeting the DNA damage response and pattern recognition receptors in combination with radiotherapy. Front Oncol 2022; 12:971959. [PMID: 36106115 PMCID: PMC9465159 DOI: 10.3389/fonc.2022.971959] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Radiotherapy is one of the most effective and frequently used treatments for a wide range of cancers. In addition to its direct anti-cancer cytotoxic effects, ionising radiation can augment the anti-tumour immune response by triggering pro-inflammatory signals, DNA damage-induced immunogenic cell death and innate immune activation. Anti-tumour innate immunity can result from recruitment and stimulation of dendritic cells (DCs) which leads to tumour-specific adaptive T-cell priming and immunostimulatory cell infiltration. Conversely, radiotherapy can also induce immunosuppressive and anti-inflammatory mediators that can confer radioresistance. Targeting the DNA damage response (DDR) concomitantly with radiotherapy is an attractive strategy for overcoming radioresistance, both by enhancing the radiosensitivity of tumour relative to normal tissues, and tipping the scales in favour of an immunostimulatory tumour microenvironment. This two-pronged approach exploits genomic instability to circumvent immune evasion, targeting both hallmarks of cancer. In this review, we describe targetable DDR proteins (PARP (poly[ADP-ribose] polymerase); ATM/ATR (ataxia-telangiectasia mutated and Rad3-related), DNA-PKcs (DNA-dependent protein kinase, catalytic subunit) and Wee1 (Wee1-like protein kinase) and their potential intersections with druggable immunomodulatory signalling pathways, including nucleic acid-sensing mechanisms (Toll-like receptors (TLR); cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and retinoic acid-inducible gene-I (RIG-I)-like receptors), and how these might be exploited to enhance radiation therapy. We summarise current preclinical advances, recent and ongoing clinical trials and the challenges of therapeutic combinations with existing treatments such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Antonio Rullan
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Emmanuel C. Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Alan A. Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Kevin J. Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
34
|
Bell PD, Pai RK. Immune Response in Colorectal Carcinoma: A Review of Its Significance as a Predictive and Prognostic Biomarker. Histopathology 2022; 81:696-714. [PMID: 35758208 DOI: 10.1111/his.14713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Colorectal carcinoma is a leading cause of cancer-related death worldwide. There is significant prognostic heterogeneity in stage II and III tumours, necessitating the development of new biomarkers to better identify patients at risk of disease progression. Recently, the tumour immune environment, particularly the type and quantity of T lymphocytes, has been shown to be a useful biomarker in predicting prognosis for patients with colorectal carcinoma. In this review, the significance of the immune response in colorectal carcinoma, including its influence on prognosis and response to therapy, will be detailed.
Collapse
Affiliation(s)
- Phoenix D Bell
- Department of Pathology, University of Pittsburgh Medical Centre, Pittsburgh, PA, 15213, USA
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh Medical Centre, Pittsburgh, PA, 15213, USA
| |
Collapse
|
35
|
Yu H, Wang C, Ke S, Bai M, Xu Y, Lu S, Feng Z, Qian B, Xu Y, Zhou M, Li Z, Yin B, Li X, Hua Y, Zhou Y, Pan S, Fu Y, Ma Y. Identification of CFHR4 as a Potential Prognosis Biomarker Associated With lmmune Infiltrates in Hepatocellular Carcinoma. Front Immunol 2022; 13:892750. [PMID: 35812416 PMCID: PMC9257081 DOI: 10.3389/fimmu.2022.892750] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/16/2022] [Indexed: 01/10/2023] Open
Abstract
Background Complement factor H-related 4 (CFHR4) is a protein-coding gene that plays an essential role in multiple diseases. However, the prognostic value of CFHR4 in hepatocellular carcinoma (HCC) is unknown. Methods Using multiple databases, we investigated CFHR4 expression levels in HCC and multiple cancers. The relationship between CFHR4 expression levels and clinicopathological variables was further analyzed. Various potential biological functions and regulatory pathways of CFHR4 in HCC were identified by performing a Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and Gene Set Enrichment Analysis (GSEA). Single-sample gene set enrichment analysis (ssGSEA) was performed to confirm the correlation between CFHR4 expression and immune cell infiltration. The correlations between CFHR4 expression levels in HCC and N6-methyladenosine (m6A) modifications and the competing endogenous RNA (ceRNA) regulatory networks were confirmed in TCGA cohort. Results CFHR4 expression levels were significantly decreased in HCC tissues. Low CFHR4 expression in HCC tissues was significantly correlated with the patients’ sex, race, age, TNM stage, pathological stage, tumor status, residual tumor, histologic grade and alpha fetal protein (AFP) level. GO and KEGG analyses revealed that differentially expressed genes related to CFHR4 may be involved in the synaptic membrane, transmembrane transporter complex, gated channel activity, chemical carcinogenesis, retinol metabolism, calcium signaling pathway, PPAR signaling pathway, insulin and gastric acid secretion. GSEA revealed that the FCGR-activated reaction, PLK1 pathway, ATR pathway, MCM pathway, cascade reactions of PI3K and FGFR1, reactant-mediated MAPK activation and FOXM1 pathway were significantly enriched in HCC with low CFHR4 expression. Moreover, CFHR4 expression was inversely correlated the levels of infiltrating Th2 cells, NK CD56bright cells and Tfh cells. In contrast, we observed positive correlations with the levels of infiltrating DCs, neutrophils, Th17 cells and mast cells. CFHR4 expression showed a strong correlation with various immunomarker groups in HCC. In addition, high CFHR4 expression significantly prolonged the overall survival (OS), disease-specific survival (DSS) and progression-free interval (PFI). We observed a substantial correlation between the expression of CFHR4 and multiple N6-methyladenosine genes in HCC and constructed potential CFHR4-related ceRNA regulatory networks. Conclusions CFHR4 might be a potential therapeutic target for improving the HCC prognosis and is closely related to immune cell infiltration.
Collapse
Affiliation(s)
- Hongjun Yu
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chaoqun Wang
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanjia Ke
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Miaoyu Bai
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanan Xu
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shounan Lu
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhigang Feng
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- The First Department of General Surgery, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, China
| | - Baolin Qian
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Xu
- Department of Pediatrics, Hainan Hospital of PLA General Hospital, Sanya, China
| | - Menghua Zhou
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zihao Li
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Yin
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinglong Li
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongliang Hua
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongzhi Zhou
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shangha Pan
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yao Fu
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Yao Fu, ; Yong Ma,
| | - Yong Ma
- Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Yao Fu, ; Yong Ma,
| |
Collapse
|
36
|
Teng C, Zhu Y, Li Y, Dai L, Pan Z, Wanggou S, Li X. Recurrence- and Malignant Progression-Associated Biomarkers in Low-Grade Gliomas and Their Roles in Immunotherapy. Front Immunol 2022; 13:899710. [PMID: 35677036 PMCID: PMC9168984 DOI: 10.3389/fimmu.2022.899710] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Despite a generally better prognosis than high-grade glioma (HGG), recurrence and malignant progression are the main causes for the poor prognosis and difficulties in the treatment of low-grade glioma (LGG). It is of great importance to learn about the risk factors and underlying mechanisms of LGG recurrence and progression. In this study, the transcriptome characteristics of four groups, namely, normal brain tissue and recurrent LGG (rLGG), normal brain tissue and secondary glioblastoma (sGBM), primary LGG (pLGG) and rLGG, and pLGG and sGBM, were compared using Chinese Glioma Genome Atlas (CGGA) and Genotype-Tissue Expression Project (GTEx) databases. In this study, 296 downregulated and 396 upregulated differentially expressed genes (DEGs) with high consensus were screened out. Univariate Cox regression analysis of data from The Cancer Genome Atlas (TCGA) yielded 86 prognostically relevant DEGs; a prognostic prediction model based on five key genes (HOXA1, KIF18A, FAM133A, HGF, and MN1) was established using the least absolute shrinkage and selection operator (LASSO) regression dimensionality reduction and multivariate Cox regression analysis. LGG was divided into high- and low-risk groups using this prediction model. Gene Set Enrichment Analysis (GSEA) revealed that signaling pathway differences in the high- and low-risk groups were mainly seen in tumor immune regulation and DNA damage-related cell cycle checkpoints. Furthermore, the infiltration of immune cells in the high- and low-risk groups was analyzed, which indicated a stronger infiltration of immune cells in the high-risk group than that in the low-risk group, suggesting that an immune microenvironment more conducive to tumor growth emerged due to the interaction between tumor and immune cells. The tumor mutational burden and tumor methylation burden in the high- and low-risk groups were also analyzed, which indicated higher gene mutation burden and lower DNA methylation level in the high-risk group, suggesting that with the accumulation of genomic mutations and epigenetic changes, tumor cells continued to evolve and led to the progression of LGG to HGG. Finally, the value of potential therapeutic targets for the five key genes was analyzed, and findings demonstrated that KIF18A was the gene most likely to be a potential therapeutic target. In conclusion, the prediction model based on these five key genes can better identify the high- and low-risk groups of LGG and lay a solid foundation for evaluating the risk of LGG recurrence and malignant progression.
Collapse
Affiliation(s)
- Chubei Teng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China.,Department of Neurosurgery, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yongwei Zhu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Yueshuo Li
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Luohuan Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Zhouyang Pan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
37
|
Li Q, Shi Z, Zhang F, Zeng W, Zhu D, Mei L. Symphony of nanomaterials and immunotherapy based on the cancer-immunity cycle. Acta Pharm Sin B 2022; 12:107-134. [PMID: 35127375 PMCID: PMC8799879 DOI: 10.1016/j.apsb.2021.05.031] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/21/2021] [Accepted: 04/25/2021] [Indexed: 02/07/2023] Open
Abstract
The immune system is involved in the initiation and progression of cancer. Research on cancer and immunity has contributed to the development of several clinically successful immunotherapies. These immunotherapies often act on a single step of the cancer–immunity cycle. In recent years, the discovery of new nanomaterials has dramatically expanded the functions and potential applications of nanomaterials. In addition to acting as drug-delivery platforms, some nanomaterials can induce the immunogenic cell death (ICD) of cancer cells or regulate the profile and strength of the immune response as immunomodulators. Based on their versatility, nanomaterials may serve as an integrated platform for multiple drugs or therapeutic strategies, simultaneously targeting several steps of the cancer–immunity cycle to enhance the outcome of anticancer immune response. To illustrate the critical roles of nanomaterials in cancer immunotherapies based on cancer–immunity cycle, this review will comprehensively describe the crosstalk between the immune system and cancer, and the current applications of nanomaterials, including drug carriers, ICD inducers, and immunomodulators. Moreover, this review will provide a detailed discussion of the knowledge regarding developing combinational cancer immunotherapies based on the cancer–immunity cycle, hoping to maximize the efficacy of these treatments assisted by nanomaterials.
Collapse
Affiliation(s)
- Qianqian Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Zhaoqing Shi
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Fan Zhang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Weiwei Zeng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
- Corresponding authors. Tel./fax: +86 20 84723750
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
- Corresponding authors. Tel./fax: +86 20 84723750
| |
Collapse
|
38
|
Systemic Inflammatory Markers as Predictors of Postoperative Complications and Survival in Patients With Advanced Head and Neck Squamous Cell Carcinoma Undergoing Free-Flap Reconstruction. J Oral Maxillofac Surg 2021; 80:744-755. [PMID: 35032441 DOI: 10.1016/j.joms.2021.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE The aim of this study was to determine the prognostic value of systemic inflammatory indices as factors for postoperative complications and survival in patients with advanced stages of p16-negative head and neck squamous cell carcinoma undergoing free-flap reconstruction. METHODS This was a retrospective cohort study. The primary predictor variables were inflammatory markers such as neutrophil, lymphocyte, monocyte, and platelet count, neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio, lymphocyte-monocyte ratio, derived NLR, systemic immune-inflammation, and systemic inflammatory marker index (SIM). Multivariate regression analyses were used to measure the associations between systemic inflammatory indices and overall and disease-free survival as a primary outcome and occurrence of postoperative complications as a secondary outcome measure. RESULTS The sample was composed of 69 male (76.67%) and 21 female (23.33%) patients, with an average age of 61.15 ± 9.79 years. The median follow-up time was 24 months, and 73 of 91 (66.43%) patients were alive during the median follow-up. Overall disease survival correlated with systemic immune-inflammation (P = .022, cutoff >1,005.3, sensitivity 67.1%, and specificity 70.6%) and SIM (P = .0001, cutoff >4.05, sensitivity 90.4%, and specificity 41.2%), preoperative platelets (P = .036, cutoff <194, sensitivity 28.8%, and specificity 94.1%), and postoperative lymphocytes (P = .012, cutoff <0.6, sensitivity 38%, and specificity 76.5%), whereas increased SIM (P = .042, cutoff >4.05, sensitivity 91.3%, and specificity 38.1%), NLR (P = .031, cutoff >13.2, sensitivity 56.9%, and specificity 60%), and preoperative platelets (P = .006, cutoff <244, sensitivity 52.3%, and specificity 76%) were associated with adverse disease-free survival. The cumulative postoperative complication rate was 34.5%, of which 13.3% accounted for major complications, whereas derived NLR (P = .013, DF 1, χ2 test 6.161, cutoff >2.3) and postoperative lymphocytes (P = .009, DF 1, χ2 test 6.756, cutoff <1) correlated with occurrence of complications. CONCLUSIONS Inflammatory indices as measures of inflammation-related systemic dysfunction may be associated with adverse survival in patients with head and neck squamous cell carcinoma and occurrence of postoperative complications and with specific cutoff values.
Collapse
|
39
|
Liu Z, Sun J, Gong T, Tang H, Shen Y, Liu C. The Prognostic and Immunological Value of Guanylate-Binding Proteins in Lower-Grade Glioma: Potential Markers or Not? Front Genet 2021; 12:651348. [PMID: 34759950 PMCID: PMC8573089 DOI: 10.3389/fgene.2021.651348] [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: 01/09/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
Seven guanylate-binding proteins (GBPs, GBP1–7), identified as a subfamily of interferon-γ-induced guanosine triphosphate hydrolases (GTPases), has been reported to be closely associated with tumor progression, metastasis, and prognosis of cancer patients in recent years. However, the expression patterns, prognostic value, immune infiltration relevance, and biological functions of GBPs in lower-grade glioma (LGG) remain elusive. In this study, by analysis and verification through multiple public data platforms, we found that GBP1, 2, 3, 4 were significantly upregulated in LGG tissues vs normal brain tissue. Analysis based on the Cox proportional hazard ratio and Kaplan–Meier plots demonstrated that the high expressions of GBP 1, 2, 3, 4 were significantly correlated with the poor prognosis of LGG patients. Correlation analysis of clinical parameters of LGG patients indicated that the expressions of GBP 1, 2, 3, 4 were significantly associated with the histological subtype and tumor histological grade of LGG. Furthermore, the correlation analysis of immune infiltration showed that the expressions of GBP1, 2, 3, 4 were significantly and positively correlated with the level of tumor immune-infiltrating cells. In particular, GBP1, 2, 3, 4 expressions were strongly correlated with the infiltration levels of monocyte, TAM, and M1/M2 macrophage, revealing their potential to regulate the polarity of macrophages. Finally, we used the GSEA method to explore the signaling pathways potentially regulated by GBP1, 2, 3, 4 and found that they were all closely associated with immune-related signaling pathways. Collectively, these findings suggested that GBP1, 2, 3, 4 were potent biomarkers to determine the prognosis and immune cell infiltration of LGG patients.
Collapse
Affiliation(s)
- Zhuang Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jifeng Sun
- Department of Radiation Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Ting Gong
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huixin Tang
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yanna Shen
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Chang Liu
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| |
Collapse
|
40
|
Wang W, Jing H, Liu J, Bu D, Zhang Y, Zhu T, Lu K, Xu Y, Cheng M, Liu J, Yao J, Huang S, Wang L. Correlation between schistosomiasis and CD8+ T cell and stromal PD-L1 as well as the different prognostic role of CD8+ T cell and PD-L1 in schistosomal-associated colorectal cancer and non-schistosomal-associated colorectal cancer. World J Surg Oncol 2021; 19:321. [PMID: 34743724 PMCID: PMC8573878 DOI: 10.1186/s12957-021-02433-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/24/2021] [Indexed: 02/08/2023] Open
Abstract
Background The effect of schistosomiasis on CD8+ T cells and then on PD-L1 expression was unknown, and the utility of CD8+ TILs as a biomarker for schistosomal-associated colorectal cancer (SCRC) rarely has been reported. Methods Three hundred thirty-eight patients with colorectal cancer (CRC) were enrolled. Immunohistochemical analysis was conducted to evaluate the expression of PD-L1 and the infiltration of CD8+ T cells. Results In the total cohort, the results showed that CD8+ TIL density was positively correlated with tumoral (p = 0.0001) and stromal PD-L1 expression (p = 0.0102). But there were no correlation between schistosomiasis and CD8+ TILs and PD-L1. Furthermore, CD8+ TIL density (p = 0.010), schistosomiasis (p = 0.042) were independent predictive factors for overall survival (OS). Stromal PD-L1 (sPD-L1) was correlated with OS (p = 0.046), but it was not an independent predictor. In patients without schistosomiasis, CD8 + T cells (p = 0.002) and sPD-L1 (p = 0.005) were associated with better OS. In patients with schistosomiasis, CD8 + T cells were independent prognosis factor (p = 0.045). Conclusions The study showed that CD8+ TILs was an independent predictive factor for OS in CRC and SCRC patients. The expression of PD-L1 was positively associated with CD8 + TILs density. There were no correlation between schistosomiasis and CD8 + TILs and PD-L1. Stromal PD-L1 but not tPD-L1 was significantly associated with OS, whereas it was not an independent prognostic factor. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-021-02433-w.
Collapse
Affiliation(s)
- Weixia Wang
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Hongyan Jing
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Jican Liu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Dacheng Bu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Yingyi Zhang
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Ting Zhu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Kui Lu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Yanchao Xu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Meihong Cheng
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Jing Liu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Junxia Yao
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Sinian Huang
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Limei Wang
- Department of Pathology, Qingpu Branch of Zhongshan Hospital, Fudan University, No. 1158 East Park Road, Qingpu District, Shanghai, 201700, People's Republic of China.
| |
Collapse
|
41
|
Shimada E, Endo M, Matsumoto Y, Tsuchihashi K, Ito M, Kusaba H, Nabeshima A, Nawata T, Maekawa A, Matsunobu T, Setsu N, Fujiwara T, Iida K, Nakagawa M, Hirose T, Kanahori M, Oyama R, Isobe T, Ariyama H, Kohashi K, Yamamoto H, Oda Y, Iwamoto Y, Akashi K, Baba E, Nakashima Y. Does the Use of Peripheral Immune-Related Markers Indicate Whether to Administer Pazopanib, Trabectedin, or Eribulin to Advanced Soft Tissue Sarcoma Patients? J Clin Med 2021; 10:jcm10214972. [PMID: 34768491 PMCID: PMC8584915 DOI: 10.3390/jcm10214972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Pazopanib, trabectedin, and eribulin are administered for the treatment of soft tissue sarcomas (STSs); however, there is little consensus on which agent should be preferentially used in a clinical setting. This study assessed whether peripheral immune-related markers served as a useful reference when selecting pazopanib, trabectedin, or eribulin. This study included 63 patients who were administered pazopanib, trabectedin, or eribulin for advanced STSs between March 2015 and December 2020. Patients were divided into three groups based on the first drug administered among these three drugs. Differences in overall survival (OS) or progression-free survival (PFS) among the three groups were analyzed. OS showed no significant differences among the drugs administered first. For patients with low neutrophil-to-lymphocyte ratio (NLR), the OS of patients administered pazopanib as the first choice was shorter than the others (hazard ratio [HR] = 9.53, 95% confidence interval [CI] = 1.94–18.13, p = 0.0018). In the low platelet-to-lymphocyte ratio (PLR) subgroup, the OS of the patients administered eribulin for the first choice was longer than that of the others (HR = 0.32, 95%CI = 0.10–0.98, p = 0.046). Therefore, NLR and PLR might be used as prognostic indicators to dictate whether STS patients receive pazopanib, trabectedin, or eribulin.
Collapse
Affiliation(s)
- Eijiro Shimada
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Makoto Endo
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
- Correspondence: ; Tel.: +81-92-642-5488
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Kenji Tsuchihashi
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.T.); (M.I.); (H.K.); (H.A.); (K.A.)
| | - Mamoru Ito
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.T.); (M.I.); (H.K.); (H.A.); (K.A.)
| | - Hitoshi Kusaba
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.T.); (M.I.); (H.K.); (H.A.); (K.A.)
| | - Akira Nabeshima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Tomoya Nawata
- Department of Orthopaedic Surgery, Kyushu Rosai Hospital, 1-1, Sonekita, Kokuraminami-ku, Kitakyushu, Fukuoka 800-0296, Japan; (T.N.); (A.M.); (T.M.); (Y.I.)
| | - Akira Maekawa
- Department of Orthopaedic Surgery, Kyushu Rosai Hospital, 1-1, Sonekita, Kokuraminami-ku, Kitakyushu, Fukuoka 800-0296, Japan; (T.N.); (A.M.); (T.M.); (Y.I.)
| | - Tomoya Matsunobu
- Department of Orthopaedic Surgery, Kyushu Rosai Hospital, 1-1, Sonekita, Kokuraminami-ku, Kitakyushu, Fukuoka 800-0296, Japan; (T.N.); (A.M.); (T.M.); (Y.I.)
| | - Nokitaka Setsu
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Toshifumi Fujiwara
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Keiichiro Iida
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Makoto Nakagawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Takeshi Hirose
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Masaya Kanahori
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Ryunosuke Oyama
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| | - Taichi Isobe
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (T.I.); (E.B.)
| | - Hiroshi Ariyama
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.T.); (M.I.); (H.K.); (H.A.); (K.A.)
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (H.Y.); (Y.O.)
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (H.Y.); (Y.O.)
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (H.Y.); (Y.O.)
| | - Yukihide Iwamoto
- Department of Orthopaedic Surgery, Kyushu Rosai Hospital, 1-1, Sonekita, Kokuraminami-ku, Kitakyushu, Fukuoka 800-0296, Japan; (T.N.); (A.M.); (T.M.); (Y.I.)
| | - Koichi Akashi
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.T.); (M.I.); (H.K.); (H.A.); (K.A.)
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (T.I.); (E.B.)
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (E.S.); (Y.M.); (A.N.); (N.S.); (T.F.); (K.I.); (M.N.); (T.H.); (M.K.); (R.O.); (Y.N.)
| |
Collapse
|
42
|
Shetty SS, Padam KSR, Hunter KD, Kudva A, Radhakrishnan R. Biological implications of the immune factors in the tumour microenvironment of oral cancer. Arch Oral Biol 2021; 133:105294. [PMID: 34735925 DOI: 10.1016/j.archoralbio.2021.105294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The objective of this review is to decipher the biological implications of the immune factors in the tumour microenvironment in oral cancer. The restoration of balance between tumour tolerance and tumour eradication by the host immune cells is critical to provide effective therapeutic strategies. DESIGN The specific role of the stromal and the immune components in oral cancer was reviewed with a tailored search strategy using relevant keywords. The articles were retrieved from bibliometric databases indexed in PubMed, Scopus, and Embase. An in silico analysis was performed to identify potential drug candidates for immunotherapy, by accessing the Drug-Gene Interactions Database (DGIdb) using the rDGIdb package. RESULTS There is compelling evidence for the role of the cellular and extracellular components of the tumour microenvironment in inducing immunosuppression and progression of oral cancer. The druggable candidates specifically targeting the immune system are a viable option in the treatment of oral cancer as they can regulate the tumour microenvironment. CONCLUSION A complex interaction between the tumour and the immunological microenvironment influences the disease outcome in oral cancer. Targeting specific components of the immune system might be relevant, as immunotherapy may become the new standard of care for oral cancer.
Collapse
Affiliation(s)
- Smitha Sammith Shetty
- Department of Oral Pathology, Faculty of Dentistry, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal 576104, India
| | - Kanaka Sai Ram Padam
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Keith D Hunter
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Adarsh Kudva
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
43
|
Cozzolino M, Celia G. The psychosocial genomics paradigm of hypnosis and mind-body integrated psychotherapy: Experimental evidence. AMERICAN JOURNAL OF CLINICAL HYPNOSIS 2021; 64:123-138. [PMID: 34723776 DOI: 10.1080/00029157.2021.1947767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The psychosocial genomics paradigm first proposed by Ernest Rossi established an epistemological shift in our application of hypnosis. We present original experimental research conducted within this paradigm that highlights the mind-gene relationship and, in particular, the positive health effects associated with hypnosis and mind-body integrated psychotherapy. We document that these approaches can stimulate epigenetic modifications and the expression of genes related to anti-inflammatory processes. These strategies strengthen the immune system and reduce oxidative stress both in normal and in oncological participants.
Collapse
|
44
|
Koh SA. An update on immunotherapy with PD-1 and PD-L1 blockade. Yeungnam Univ J Med 2021; 38:308-317. [PMID: 34496466 PMCID: PMC8688791 DOI: 10.12701/yujm.2021.01312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 11/06/2022] Open
Abstract
Cancer is the leading cause of death and is on the rise worldwide. Until 2010, the development of targeted treatment was mainly focused on the growth mechanisms of cancer. Since then, drugs with mechanisms related to tumor immunity, especially immune checkpoint inhibitors, have proven effective, and most pharmaceutical companies are striving to develop related drugs. Programmed cell death-1 and programmed cell death ligand-1 inhibitors have shown great success in various cancer types. They showed durable and sustainable responses and were approved by the U.S. Food and Drug Administration. However, the response to inhibitors showed low percentages of cancer patients; 15% to 20%. Therefore, combination strategies with immunotherapy and conventional treatments were used to overcome the low response rate. Studies on combination therapy have typically reported improvements in the response rate and efficacy in several cancers, including non-small cell lung cancer, small cell lung cancer, breast cancer, and urogenital cancers. The combination of chemotherapy or targeted agents with immunotherapy is one of the leading pathways for cancer treatment.
Collapse
Affiliation(s)
- Sung Ae Koh
- Department of Hematology-Oncology, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
45
|
Clinical importance of preoperative red-cell volume distribution width as a prognostic marker in patients undergoing radical surgery for pancreatic cancer. Surg Today 2021; 52:465-474. [PMID: 34524510 PMCID: PMC8873122 DOI: 10.1007/s00595-021-02374-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/15/2021] [Indexed: 12/18/2022]
Abstract
Background and purpose A new noninvasive biomarker is being sought to predict the prognosis of patients with pancreatic cancer. Red-cell volume distribution width (RDW), a descriptive parameter for erythrocyte variation, has been shown to have prognostic value for some tumor types. Our purpose was to assess the RDW value to predict the prognosis of patients with pancreatic cancer. Methods The subjects of this retrospective study were 792 patients who underwent radical surgery for pancreatic cancer, divided into high-RDW and low-RDW groups based on receiver operating characteristic (ROC) curve analysis (15.6%). The controlling nutritional status (CONUT) score was used to assess preoperative nutritional status. Statistical analysis was conducted to investigate the differences between the high and low RDW groups, and to explore the possibility of the RDW being used as prognostic predictor for patients with pancreatic cancer. Results The immune-nutritional status was worse in the high-RDW group than in the low-RDW group. The high-RDW group patients also had a poorer prognosis. Risk factor analysis showed that the RDW could be an independent risk factor for pancreatic cancer. Conclusions The RDW is associated with immune-nutritional status in pancreatic cancer patients and can be used as an independent prognostic factor for their postoperative survival. Supplementary Information The online version contains supplementary material available at 10.1007/s00595-021-02374-7.
Collapse
|
46
|
Xiao M, Du C, Zhang C, Zhang X, Li S, Zhang D, Jia W. Bioinformatics analysis of the prognostic value of NEK8 and its effects on immune cell infiltration in glioma. J Cell Mol Med 2021; 25:8748-8763. [PMID: 34374193 PMCID: PMC8435421 DOI: 10.1111/jcmm.16831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/28/2021] [Accepted: 07/15/2021] [Indexed: 12/26/2022] Open
Abstract
Glioma is the most common malignancy of the nervous system with high rates of recurrence and mortality, even after surgery. The 5‐year survival rate is only about 5%. NEK8 is involved in multiple biological processes in a variety of cancers; however, its role in glioma is still not clear. In the current study, we evaluated the prognostic value of NEK8, as well as its role in the pathogenesis of glioma. Using a bioinformatics approach and RNA‐seq data from public databases, we found that NEK8 expression is elevated in glioma tissues; we further verified this result by RT‐PCR, Western blotting and immunochemistry using clinical samples. Functional enrichment analyses of genes with correlated expression indicated that elevated NEK8 expression is associated with increased immune cell infiltration in glioma and may affect the tumour microenvironment via the regulation of DNA damage/repair. Survival analyses revealed that high levels of NEK8 are associated with a poorer prognosis; higher WHO grade, IDH status, 1p/19q codeletion, age and NEK8 were identified as an independent prognostic factor. These findings support the crucial role of NEK8 in the progression of glioma via effects on immune cell infiltration and suggest that it is a new prognostic biomarker.
Collapse
Affiliation(s)
- Meng Xiao
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China.,Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chaoyang Du
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Chuanbo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Xinzhong Zhang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Shaomin Li
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China.,Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dainan Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wang Jia
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
47
|
Zhou J, Lei M, Peng XL, Wei DX, Yan LK. Fenton Reaction Induced by Fe-Based Nanoparticles for Tumor Therapy. J Biomed Nanotechnol 2021; 17:1510-1524. [PMID: 34544529 DOI: 10.1166/jbn.2021.3130] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Fenton reaction, a typical inorganic reaction, is broadly utilized in the field of wastewater treatment. Recently In case of its ability to inhibit the growth of cancer cells, it has been frequently reported in cancer treatment. Using the unique tumor microenvironment in cancer cells, many iron-based nanoparticles have been developed to release iron ions in cancer cells to induce Fenton reaction. In this mini review, we outline several different types of iron-based nanoparticles and several main means to enhance Fenton reaction in cancer cells. Finally, we discussed the advantages and disadvantages of iron-based nanoparticles for cancer therapy, prospected the future development of iron-based nanoparticles. It is believed that iron-based nanoparticles can make certain contribution to the cause of human cancer in the future.
Collapse
Affiliation(s)
- Jian Zhou
- Polymer Materials & Engineering Department, School of Materials Science & Engineering, Chang'an University, Xi'an 710064, China
| | - Miao Lei
- Polymer Materials & Engineering Department, School of Materials Science & Engineering, Chang'an University, Xi'an 710064, China
| | - Xue-Liang Peng
- Electronics Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Dai-Xu Wei
- Electronics Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Lu-Ke Yan
- Polymer Materials & Engineering Department, School of Materials Science & Engineering, Chang'an University, Xi'an 710064, China
| |
Collapse
|
48
|
Schernberg A, Vernerey D, Goldstein D, Van Laethem JL, Glimelius B, van Houtte P, Bonnetain F, Louvet C, Hammel P, Huguet F. Predictive Value of Neutrophils Count for Local Tumor Control After Chemoradiotherapy in Patients With Locally Advanced Pancreatic Carcinoma. Int J Radiat Oncol Biol Phys 2021; 110:1022-1031. [PMID: 33548338 DOI: 10.1016/j.ijrobp.2021.01.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Baseline neutrophil count may predict overall survival (OS) in patients with locally advanced pancreatic cancer (LAPC). METHODS AND MATERIALS The international multicenter randomized LAP07 phase 3 trial has enrolled 442 patients with LAPC. We analyzed the prognostic value of both baseline neutrophilia (neutrophil count >7 g/L) and elevated or increasing neutrophil count as (1) neutrophilia or (2) increased absolute neutrophil count after induction chemotherapy versus baseline for OS, progression-free survival, and local control (LC). A Cox proportional hazard model was used to assess elevated or increasing neutrophil count status by randomly assigned treatment interactions for each endpoint. RESULTS Among the 442 patients, 47 patients (11%) with baseline neutrophilia had worse OS (median 8.9 vs 13.3 months; P = .01). After induction chemotherapy, among the 235 patients whose blood counts were available, 90 patients (38%) had elevated or increasing neutrophil count associated with poorer OS in univariate (median 14.4 vs 17.9 months; P = .001) and multivariate analysis (P = .004). Elevated or increasing neutrophil count was also predictive of a decreased benefit of chemoradiation therapy on LC. In 126 patients without elevated or increasing neutrophil count, 1-year LC was 80% in the chemoradiation arm versus 54% in the chemotherapy arm (P < .001; interaction test P = .015). CONCLUSIONS In this study, baseline neutrophilia and increased absolute neutrophil count were associated with worse OS in this large series of patients with LAPC. In addition, the counts were an independent prognosis factor and a strong predictive LC biomarker for chemoradiation therapy benefit. An assessment of neutrophils counts can help to improve the selection of patients who might benefit from chemoradiation therapy after induction chemotherapy.
Collapse
Affiliation(s)
| | - Dewi Vernerey
- Methodological and Quality of Life in Oncology Unit, INSERM UMR 1098, University Hospital of Besancon, France
| | - David Goldstein
- Kinghorn Cancer Centre and Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | | | - Bengt Glimelius
- Department of Oncology, University of Uppsala, Akademiska Sjukhuset, Sweden
| | | | - Franck Bonnetain
- Methodological and Quality of Life in Oncology Unit, INSERM UMR 1098, University Hospital of Besancon, France; Groupe Coopérateur Multidisciplinaire en Oncologie, Paris, France
| | - Christophe Louvet
- Groupe Coopérateur Multidisciplinaire en Oncologie, Paris, France; Département d'Oncologie Médicale, Institut Mutualiste Montsouris, Paris, France
| | - Pascal Hammel
- Groupe Coopérateur Multidisciplinaire en Oncologie, Paris, France; Service d'Oncologie Digestive, Hôpital Beaujon, Clichy, France
| | - Florence Huguet
- Service d'Oncologie Radiothérapie, Hôpital Tenon, Paris, France; Groupe Coopérateur Multidisciplinaire en Oncologie, Paris, France
| |
Collapse
|
49
|
Silva MO, Almeida BS, Sales NS, Diniz MO, Aps LRMM, Rodrigues KB, Silva JR, Moreno ACR, Porchia BFMM, Sulczewski FB, Boscardin SB, Ferreira LCS. Antigen Delivery to DEC205 + Dendritic Cells Induces Immunological Memory and Protective Therapeutic Effects against HPV-Associated Tumors at Different Anatomical Sites. Int J Biol Sci 2021; 17:2944-2956. [PMID: 34345218 PMCID: PMC8326119 DOI: 10.7150/ijbs.57038] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/02/2021] [Indexed: 12/27/2022] Open
Abstract
The generation of successful anticancer vaccines relies on the ability to induce efficient and long-lasting immune responses to tumor antigens. In this scenario, dendritic cells (DCs) are essential cellular components in the generation of antitumor immune responses. Thus, delivery of tumor antigens to specific DC populations represents a promising approach to enhance the efficiency of antitumor immunotherapies. In the present study, we employed antibody-antigen conjugates targeting a specific DC C-type lectin receptor. For that purpose, we genetically fused the anti-DEC205 monoclonal antibody to the type 16 human papillomavirus (HPV-16) E7 oncoprotein to create a therapeutic vaccine to treat HPV-associated tumors in syngeneic mouse tumor models. The therapeutic efficacy of the αDEC205-E7 mAb was investigated in three distinct anatomical tumor models (subcutaneous, lingual and intravaginal). The immunization regimen comprised two doses of the αDEC205-E7 mAb coadministered with a DC maturation stimulus (Polyinosinic:polycytidylic acid, poly (I:C)) as an adjuvant. The combined immunotherapy produced robust antitumor effects on both the subcutaneous and orthotopic tumor models, stimulating rapid tumor regression and long-term survival. These outcomes were related to the activation of tumor antigen-specific CD8+ T cells in both systemic compartments and lymphoid tissues. The αDEC205-E7 antibody plus poly (I:C) administration induced long-lasting immunity and controlled tumor relapses. Our results highlight that the delivery of HPV tumor antigens to DCs, particularly via the DEC205 surface receptor, is a promising therapeutic approach, providing new opportunities for the development of alternative immunotherapies for patients with HPV-associated tumors at different anatomical sites.
Collapse
Affiliation(s)
- Mariângela O Silva
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Bianca S Almeida
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Natiely S Sales
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mariana O Diniz
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luana R M M Aps
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Karine B Rodrigues
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jamile R Silva
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Ana C R Moreno
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Bruna F M M Porchia
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Fernando B Sulczewski
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Silvia B Boscardin
- Laboratory of Antigen Targeting to Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Luís C S Ferreira
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
50
|
Suzuki T, Ishibashi Y, Tsujimoto H, Nomura S, Kouzu K, Itazaki Y, Sugihara T, Harada M, Ito N, Sugasawa H, Kishi Y, Ueno H. A Novel Systemic Inflammatory Score Combined With Immunoinflammatory Markers Accurately Reflects Prognosis in Patients With Esophageal Cancer. In Vivo 2021; 34:3705-3711. [PMID: 33144487 DOI: 10.21873/invivo.12218] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022]
Abstract
AIM To establish a novel systemic inflammatory score (SIS) combined with neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and C-reactive protein/albumin ratio (CAR) and to validate its prognostic value and relation with serum cytokine levels in patients who underwent esophagectomy for esophageal cancer (EC). PATIENTS AND METHODS Preoperative NLR, PLR, and CAR were evaluated in 102 patients undergoing esophageal resection for EC from 2009 to 2014. Receiver operating characteristic (ROC) curves censored for 5-year survival were plotted to determine the cutoff values of each measure. Each measure was scored 1 if it was above the cutoff value (NLR >3.12, PLR >230, and CAR >0.085) and scored 0 if it was below that. The SIS was defined as the sum of these values and was divided into the two groups: High SIS (SIS=2-3) and low SIS (SIS=0-1). Univariate and multivariate analyses were used to determine the prognostic significance. The area under the ROCs (AUROC) was compared to verify the discriminative power of survival prediction. In addition, we analyzed the relationship between SIS and perioperative serum interleukin (IL)-6 and IL-10 levels. RESULTS In the clinicopathological findings, only tumor depth was significantly related to SIS (p=0.004). At 0.732, the AUROC of SIS was the highest (NLR=0.618, PLR=0.545), and CAR=0.712). The high-SIS group had a significantly poorer prognosis than the low-SIS group (p=0.011). SIS was identified as an independent prognostic factor in the multivariate analysis (hazard ratio=1.96, 95% confidence intervaI=1.11-3.41, p=0.020). The preoperative serum interleukin-6 level was significantly low (p=0.046) and postoperative serum interleukin-10 level was significantly high in the high-SIS group (p=0.047). CONCLUSION SIS was a superior predictor of prognosis compared with existing immunoinflammatory markers and closely reflected the fluctuation of peripheral inflammatory cytokines in patients with EC.
Collapse
Affiliation(s)
- Takafumi Suzuki
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yusuke Ishibashi
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | | | - Shinsuke Nomura
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Keita Kouzu
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yujiro Itazaki
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Takao Sugihara
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Manabu Harada
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Nozomi Ito
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Hidekazu Sugasawa
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Saitama, Japan
| |
Collapse
|