1
|
Liao X, Huang H, Qiu B, Chen J, Zhang A, Liang H, Huang C, Mei F, Mao J, Liu F, Jin M, Peng X, Ma H, Ding W, Qi S, Bao Y. Effects of recombinant human growth hormone in severe neurosurgical patients: A single center, retrospective study. PLoS One 2025; 20:e0317219. [PMID: 39792837 PMCID: PMC11723630 DOI: 10.1371/journal.pone.0317219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
PURPOSE To explore the effects of recombinant human growth hormone (r-hGH) on inflammatory mediators, immune cells and prognosis in severe neurosurgical patients. METHODS From August 2020 to June 2021, a total of 236 patients who admitted to the neurosurgical intensive care unit (NSICU) were retrospectively analyzed. The patients were divided into GH group (97 cases) and nGH group (139 cases) according to whether they received r-hGH treatment. Parameters including CD4+ T cell counts, inflammatory mediators and prognosis were recorded and assessed. RESULTS The results showed that the cure time of pneumonia and intracranial infection in GH group patients was significantly shorter than in the nGH group (24.25 ± 4.89 days and 21.33 ± 1.53 days versus 29.13 ± 7.43 days and 25.17 ± 2.32 days, respectively). However, there was no significant difference in GOS scores between two groups (31.96% ≤ 3 and 68.04% > 3 vs 39.57% ≤ 3 and 60.43% > 3) (P = 0.232). Furthermore, the number of CD4+ T cells and CD8+ T cells in the GH group showed a significant upward trend. Last but not least, significant differences were also observed in IL-6 and IL-10 levels between two groups at days 1, 3, and 7. CONCLUSION The application of r-hGH in severe neurosurgical patients was effective in increasing the number of CD4+ T cells, down-regulating inflammatory mediators, shortening the cure time of pneumonia, intracranial infections and urinary tract infections, and improving patients' prognosis.
Collapse
Affiliation(s)
- Xixian Liao
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Haorun Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Binghui Qiu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaping Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - An Zhang
- Department of Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Haoxin Liang
- Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuanping Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fen Mei
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Mao
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fan Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ming Jin
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Xiaojie Peng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Haidie Ma
- First Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenjie Ding
- First Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yun Bao
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Sun Y, Yin Z, Li S, Wu L, Zhang Y, Zhao Y, Gomes Dos Santos IL, Subudhi S, Lei P, Muzikansky A, Yuan Z, Rueda BR, Jain RK, Xu L. Losartan rewires the tumor-immune microenvironment and suppresses IGF-1 to overcome resistance to chemo-immunotherapy in ovarian cancer. Br J Cancer 2024; 131:1683-1693. [PMID: 39369055 PMCID: PMC11554678 DOI: 10.1038/s41416-024-02863-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Ovarian cancer (OvCa) is the most lethal of the gynecologic malignancies. Immune checkpoint inhibitors, which have revolutionized the treatment of multiple malignancies, have had limited efficacy in OvCa patients. This lack of effectiveness is partly due to the abnormal ovarian tumor microenvironment (TME), displaying a desmoplastic, highly fibrotic extracellular matrix. High extracellular matrix deposition leads to a buildup of compressive forces that cause tumor blood vessel collapse, reduced vessel perfusion, poor delivery of drugs, and compromised trafficking of cytotoxic T-cells to these tumors. METHODS Using two syngeneic OvCa models, we tested the effect of losartan, a widely prescribed anti-hypertensive drug, on reprogramming the TME and chemosensitizing the cancer cells. RESULTS Losartan treatment (i) reprograms the TME leading to increased vascular perfusion, and thus enhances drug delivery and immune effector cell intratumoral infiltration and function; and (ii) rewires the OvCa cells by suppressing the IGF-1 signaling, resulting in enhanced chemosensitivity. As a result of the combined tumor and stromal effects, losartan treatment enhances the efficacy of chemo-immunotherapy in OvCa models. CONCLUSION The safety and low cost ( < $1-2/day) of losartan warrant rapid translation of our findings to patients with OvCa.
Collapse
Affiliation(s)
- Yao Sun
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Clinical Research for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Zhenzhen Yin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Shuang Li
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Limeng Wu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yanling Zhang
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yanxia Zhao
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430023, China
| | - Igor L Gomes Dos Santos
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Sonu Subudhi
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Pinji Lei
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Alona Muzikansky
- Division of Biostatistics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhiyong Yuan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430023, China
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Clinical Research for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Lei Xu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
3
|
Mokhtari N, Ahmadi N, Moradi S, Farmani S, Kheyrani E, Dolatabadi NF. Experimental and in silico analysis of LINC01279 expression in tumor of patients with breast cancer. J Appl Genet 2024:10.1007/s13353-024-00908-6. [PMID: 39465460 DOI: 10.1007/s13353-024-00908-6] [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: 09/08/2023] [Revised: 08/18/2024] [Accepted: 09/22/2024] [Indexed: 10/29/2024]
Abstract
Breast cancer (BC) is characterized by the increase of malignant cells in the breast. The malignant cells begin in the lining of the breast milk glands or ducts (ductal epithelium). BC is the most frequent cancer in women, but it may also occur in males. Long non-coding RNAs (lncRNA) have been demonstrated to control the development and incidence of cancer. However, some lncRNAs experience potential changes in BC, but their role has not been well studied. LINC01279 is known as a valuable biomarker in gastric cancer but has not yet been studied in BC. Changes in LINC01279 expression levels in BC samples were investigated by microarray. Q-PCR was also used to evaluate the expression of LINC01279 in the tumor and normal adjacent samples of 30 BC patients. The LINC01279 co-expressed gene module was discovered using weighted gene correlation network analysis (WGCNA) on the relevant dataset. The top ten hub genes were determined using gene ontology (GO) functional enrichments on the co-expressed gene module. The results of the bioinformatics study showed an increase in LINC01279 expression levels (log2FC = 3.228749561, adj.P.Val = 1.69E - 12) in tumor samples compared to normal marginal tissue. Q-PCR results also showed a significant increase in LINC01279 expression (P-value = 0.0005) in tumor samples. WGCNA analysis identified that the black module is the LINC01279 co-expressed module, and functional annotation analysis of black module genes enriched in significant cancer-related pathways and processes, including cell growth and/or maintenance, regulation of immune response, regulation of cell proliferation, and epithelial-to-mesenchymal transition (EMT). Regarding the real-time PCR results, the analysis of expression patterns has illuminated a distinct association between the heightened expression levels of LINC01279, and the stages of cancer progression as well as the metastatic potential of tumors. However, intriguingly, our observations have failed to reveal any statistically significant correlations between the relative expression of LINC01279 and tumor grade classification, or the presence of ER, PR, and HER2 biomarkers. The present study could provide a new perspective on the molecular regulatory. Processes associated with BC pathogenic mechanisms are linked to the LINC01279, although further research is needed on the possible role of this lncRNA in BC.
Collapse
Affiliation(s)
- Negar Mokhtari
- Department of Cellular and Molecular Biology, Islamic Azad University, Najafabad Branch, Isfahan, Iran
| | - Najmeh Ahmadi
- Departmant of Medical Laboratory Sciences, School of Paramedical Sciences, Gerash University of Medical Sciences, Gerash, Iran
| | - Sahar Moradi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Isfahan, Iran
| | - Shiva Farmani
- Department of Biology, Faculty of Basic Sciences, Yazd University, Yazd, Iran
| | | | | |
Collapse
|
4
|
Somri-Gannam L, Meisel-Sharon S, Hantisteanu S, Bar-Noy T, Sigal E, Groisman G, Hallak M, Werner H, Bruchim I. IGF1R inhibition and PD-1 blockade improve anti-tumor immune response in epithelial ovarian cancer. Front Oncol 2024; 14:1410447. [PMID: 39450263 PMCID: PMC11499063 DOI: 10.3389/fonc.2024.1410447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/10/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction The insulin-like growth factor (IGF) system plays a key role in regulating growth and invasiveness in epithelial ovarian cancer (EOC) and is considered a promising therapeutic target. EOC is an immunosuppressive disease, although there are limited data about the involvement of the IGF1R system in the anti-tumor immune response in the EOC microenvironment. Methods In the current study, we hypothesized that IGF 1 receptor (IGF1R) involvement in the maturation of dendritic cells (DC) with the co-inhibition of IGF1R and PD-1 would affect the EOC microenvironment. Results We found that DC pretreated with IGF1R inhibitor resulted in fewer EOC cells. Moreover, in vivo experiments conducted with an EOC mouse model, with anti-PD-1/IGF1R combined, resulted in lower tumor weight compared to individual treatments. Additionally, anti-PD-1/IGF1R treatment increased DC by 34% compared with AEW-541 and 40% with anti-PD-1. The combined treatment increased CD8+ T-cell levels compared to AEW-541 alone. RNA-seq data analysis indicated that anti-PD-1/IGF1R led to a more potent immune response, as reflected by altered gene expression levels related to anti-tumor immune response, compared with either treatment alone. Discussion These findings provide novel evidence that IGF1R axis inhibition combined with PD-1 blockade may be an effective therapeutic strategy for selected EOC patient populations.
Collapse
Affiliation(s)
- Lina Somri-Gannam
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shilhav Meisel-Sharon
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Shay Hantisteanu
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Tomer Bar-Noy
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Emiliya Sigal
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Gabriel Groisman
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Mordechai Hallak
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Bruchim
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| |
Collapse
|
5
|
Liang Y, Diao W, Yang X, Tao Y, Hong L, Li W. Regulator of calcineurin 3 as a novel predictor of diagnosis and prognosis in pan-cancer. Croat Med J 2024; 65:356-372. [PMID: 39219199 PMCID: PMC11399725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
AIM To assess diagnostic and prognostic value of regulator of calcineurin 3 (RCAN3) in various malignancies. METHODS RCAN3 expression levels were assessed across pan-cancer data sets including various molecular and immune subtypes. Receiver operating characteristic (ROC) and Kaplan-Meier curves were employed to determine the diagnostic and prognostic value of RCAN3 in pan-cancer, respectively. Enrichment analyses were used to identify RCAN3-associated terms and pathways. A special focus was placed on cervical squamous cell carcinoma and endocervical adenocarcinoma cervical cancer (CESC); we assessed the prognostic value of RCAN expression within distinct clinical subgroups and its effect on m6A modifications and immune infiltration. RESULTS RCAN3 expression varied not only in different cancer types but also in different molecular and immune subtypes of cancers. RCAN3 displayed high accuracy in diagnosing and predicting cancers, and RCAN3 expression level was associated with the prognosis of certain cancers. CESC patients with a high RCAN3 level had a worse overall survival, disease-specific survival, and progression-free interval. RCAN3 expression was related to multiple m6A modifier genes and immune cells. CONCLUSION In general, RCAN3 can serve as a novel biomarker for the diagnosis and prognosis in pan-cancer, especially in CESC. It may represent a promising molecular target for developing new treatments. However, our analysis is limited to bioinformatic predictions, and further biological experiments are necessary to verify our results.
Collapse
Affiliation(s)
| | | | | | | | | | - Wenle Li
- Wenle Li, Department of Gynecology, Affiliated Hospital of Guangdong Medical University, No. 57, Renmin Avenue South, Xiashan District, Zhanjiang, Guangdong, 524000, China,
| |
Collapse
|
6
|
Gan Q, Mao L, Shi R, Chang L, Wang G, Cheng J, Chen R. Prognostic Value and Immune Infiltration of HPV-Related Genes in the Immune Microenvironment of Cervical Squamous Cell Carcinoma and Endocervical Adenocarcinoma. Cancers (Basel) 2023; 15:1419. [PMID: 36900213 PMCID: PMC10000937 DOI: 10.3390/cancers15051419] [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: 12/25/2022] [Revised: 02/06/2023] [Accepted: 02/11/2023] [Indexed: 03/12/2023] Open
Abstract
Mounting evidence has highlighted the immune environment as a critical feature in the development of cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC). However, the relationship between the clinical characteristics of the immune environment and CESC remain unclear. Therefore, the aim of this study was to further characterize the relationship between the tumor and immune microenvironment and the clinical features of CESC using a variety of bioinformatic methods. Expression profiles (303 CESCs and three control samples) and relevant clinical data were obtained from The Cancer Genome Atlas. We divided CESC cases into different subtypes and performed a differential gene expression analysis. In addition, gene ontology (GO) and gene set enrichment analysis (GSEA) were performed to identify potential molecular mechanisms. Furthermore, data from 115 CESC patients from East Hospital were used to help identify the relationship between the protein expressions of key genes and disease-free survival using tissue microarray technology. Cases of CESC (n = 303) were divided into five subtypes (C1-C5) based on their expression profiles. A total of 69 cross-validated differentially expressed immune-related genes were identified. Subtype C4 demonstrated a downregulation of the immune profile, lower tumor immune/stroma scores, and worse prognosis. In contrast, the C1 subtype showed an upregulation of the immune profile, higher tumor immune/stroma scores, and better prognosis. A GO analysis suggested that changes in CESC were primarily enriched nuclear division, chromatin binding, and condensed chromosomes. In addition, GSEA demonstrated that cellular senescence, the p53 signaling pathway, and viral carcinogenesis are critical features of CESC. Moreover, high FOXO3 and low IGF-1 protein expression were closely correlated with decreased clinical prognosis. In summary, our findings provide novel insight into the relationship between the immune microenvironment and CESC. As such, our results may provide guidance for developing potential immunotherapeutic targets and biomarkers for CESC.
Collapse
Affiliation(s)
- Qiyu Gan
- Department of Gynecology and Obstetrics, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Luning Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100021, China
| | - Rui Shi
- Department of Gynecology and Obstetrics, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Linlin Chang
- Department of Gynecology and Obstetrics, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Guozeng Wang
- Department of Gynecology and Obstetrics, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Jingxin Cheng
- Department of Gynecology and Obstetrics, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Rui Chen
- Department of Gynecology, Shanghai United Family Hospital, Shanghai 200120, China
| |
Collapse
|
7
|
Interactions between Platelets and Tumor Microenvironment Components in Ovarian Cancer and Their Implications for Treatment and Clinical Outcomes. Cancers (Basel) 2023; 15:cancers15041282. [PMID: 36831623 PMCID: PMC9953912 DOI: 10.3390/cancers15041282] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Platelets, the primary operatives of hemostasis that contribute to blood coagulation and wound healing after blood vessel injury, are also involved in pathological conditions, including cancer. Malignancy-associated thrombosis is common in ovarian cancer patients and is associated with poor clinical outcomes. Platelets extravasate into the tumor microenvironment in ovarian cancer and interact with cancer cells and non-cancerous elements. Ovarian cancer cells also activate platelets. The communication between activated platelets, cancer cells, and the tumor microenvironment is via various platelet membrane proteins or mediators released through degranulation or the secretion of microvesicles from platelets. These interactions trigger signaling cascades in tumors that promote ovarian cancer progression, metastasis, and neoangiogenesis. This review discusses how interactions between platelets, cancer cells, cancer stem cells, stromal cells, and the extracellular matrix in the tumor microenvironment influence ovarian cancer progression. It also presents novel potential therapeutic approaches toward this gynecological cancer.
Collapse
|
8
|
de Billy E, Pellegrino M, Orlando D, Pericoli G, Ferretti R, Businaro P, Ajmone-Cat MA, Rossi S, Petrilli LL, Maestro N, Diomedi-Camassei F, Pezzullo M, De Stefanis C, Bencivenga P, Palma A, Rota R, Del Bufalo F, Massimi L, Weber G, Jones C, Carai A, Caruso S, De Angelis B, Caruana I, Quintarelli C, Mastronuzzi A, Locatelli F, Vinci M. Dual IGF1R/IR inhibitors in combination with GD2-CAR T-cells display a potent anti-tumor activity in diffuse midline glioma H3K27M-mutant. Neuro Oncol 2022; 24:1150-1163. [PMID: 34964902 PMCID: PMC9248389 DOI: 10.1093/neuonc/noab300] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Diffuse midline gliomas (DMG) H3K27M-mutant, including diffuse intrinsic pontine glioma (DIPG), are pediatric brain tumors associated with grim prognosis. Although GD2-CAR T-cells demonstrated significant anti-tumor activity against DMG H3K27M-mutant in vivo, a multimodal approach may be needed to more effectively treat patients. We investigated GD2 expression in DMG/DIPG and other pediatric high-grade gliomas (pHGG) and sought to identify chemical compounds that would enhance GD2-CAR T-cell anti-tumor efficacy. METHODS Immunohistochemistry in tumor tissue samples and immunofluorescence in primary patient-derived cell lines were performed to study GD2 expression. We developed a high-throughput cell-based assay to screen 42 kinase inhibitors in combination with GD2-CAR T-cells. Cell viability, western blots, flow-cytometry, real time PCR experiments, DIPG 3D culture models, and orthotopic xenograft model were applied to investigate the effect of selected compounds on DIPG cell death and CAR T-cell function. RESULTS GD2 was heterogeneously, but widely, expressed in the tissue tested, while its expression was homogeneous and restricted to DMG/DIPG H3K27M-mutant cell lines. We identified dual IGF1R/IR antagonists, BMS-754807 and linsitinib, able to inhibit tumor cell viability at concentrations that do not affect CAR T-cells. Linsitinib, but not BMS-754807, decreases activation/exhaustion of GD2-CAR T-cells and increases their central memory profile. The enhanced anti-tumor activity of linsitinib/GD2-CAR T-cell combination was confirmed in DIPG models in vitro, ex vivo, and in vivo. CONCLUSION Our study supports the development of IGF1R/IR inhibitors to be used in combination with GD2-CAR T-cells for treating patients affected by DMG/DIPG and, potentially, by pHGG.
Collapse
Affiliation(s)
- Emmanuel de Billy
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Marsha Pellegrino
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Domenico Orlando
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Giulia Pericoli
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Roberta Ferretti
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Pietro Businaro
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | | | - Sabrina Rossi
- Department of Laboratories-Pathology Unit, Bambino Gesù Children’s
Hospital-IRCCS, Rome, Italy
| | - Lucia Lisa Petrilli
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Nicola Maestro
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | | | - Marco Pezzullo
- Research Laboratories, Bambino Gesù Children’s
Hospital-IRCCS, Rome, Italy
| | | | - Paola Bencivenga
- Research Laboratories, Bambino Gesù Children’s
Hospital-IRCCS, Rome, Italy
| | - Alessia Palma
- Research Laboratories, Bambino Gesù Children’s
Hospital-IRCCS, Rome, Italy
| | - Rossella Rota
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Francesca Del Bufalo
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Luca Massimi
- Department of Pediatric Neurosurgery, Catholic University Medical
School, Rome, Italy
| | - Gerrit Weber
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer
Research, Sutton, UK
| | - Andrea Carai
- Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children’s
Hospital-IRCCS, Rome, Italy
| | - Simona Caruso
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Biagio De Angelis
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Ignazio Caruana
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Concetta Quintarelli
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico
II, Naples, Italy
| | - Angela Mastronuzzi
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
- Department of Pediatrics, Sapienza University of Rome,
Rome, Italy
| | - Maria Vinci
- Department of Onco-hematology, Gene and Cell Therapy, Bambino Gesù
Children’s Hospital–IRCCS, Rome, Italy
| |
Collapse
|
9
|
Franson A, Koschmann C. Enhancing GD2 CAR T-cell therapy with IGF-1R blockade: Are DIPG CAR T cells ready for combinatorial therapy? Neuro Oncol 2022; 24:1164-1165. [PMID: 35323961 PMCID: PMC9248381 DOI: 10.1093/neuonc/noac073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Andrea Franson
- Department of Pediatrics, University of Michigan Medical
School, Ann Arbor, Michigan, USA
| | - Carl Koschmann
- Corresponding Author: Carl Koschmann, MD, Department of Pediatrics,
University of Michigan Medical School, 3520D MSRB I, 1150 W Medical Center Drive, Ann
Arbor MI, 48109, USA ()
| |
Collapse
|
10
|
Badr H, Blutrich R, Chan K, Tong J, Taylor P, Zhang W, Kafri R, Röst HL, Tsao MS, Moran MF. Proteomic characterization of a candidate polygenic driver of metabolism in non-small cell lung cancer. J Mol Biol 2022; 434:167636. [PMID: 35595168 DOI: 10.1016/j.jmb.2022.167636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/03/2022] [Accepted: 05/08/2022] [Indexed: 11/18/2022]
Abstract
Proteome analysis revealed signatures of co-expressed upregulated metabolism proteins highly conserved between primary and non-small cell lung cancer (NSCLC) patient-derived xenograft tumors (Li et al. 2014, Nat. Communications 5:5469). The C10 signature is encoded by seven genes (ADSS, ATP2A2, CTPS1, IMPDH2, PKM2, PTGES3, SGPL1) and DNA alterations in C10-encoding genes are associated with longer survival in a subset of NSCLC. To explore the C10 signature as an oncogenic driver and address potential mechanisms of action, C10 protein expression and protein-protein interactions were determined. In independent NSCLC cohorts, the coordinated expression of C10 proteins was significant and mutations in C10 genes were associated with better outcome. Affinity purification-mass spectrometry and in vivo proximity-based biotin identification defined a C10 interactome involving 667 proteins including candidate drug targets and clusters associated with glycolysis, calcium homeostasis, and nucleotide and sphingolipid metabolism. DNA alterations in genes encoding C10 interactome components were also found to be associated with better survival. These data support the notion that the coordinated upregulation of the C10 signature impinges metabolic processes that collectively function as an oncogenic driver in NSCLC.
Collapse
Affiliation(s)
- Heba Badr
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Ron Blutrich
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Kaitlin Chan
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Jiefei Tong
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Paul Taylor
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; SPARC BioCentre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Wen Zhang
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Ran Kafri
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Hannes L Röst
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; Departments of Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Michael F Moran
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada; SPARC BioCentre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| |
Collapse
|
11
|
Cela V, Daniele S, Obino MER, Ruggiero M, Zappelli E, Ceccarelli L, Papini F, Marzi I, Scarfò G, Tosi F, Franzoni F, Martini C, Artini PG. Endometrial Dysbiosis Is Related to Inflammatory Factors in Women with Repeated Implantation Failure: A Pilot Study. J Clin Med 2022; 11:2481. [PMID: 35566605 PMCID: PMC9101226 DOI: 10.3390/jcm11092481] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
An abnormal endometrial microbiota has been suggested to impair the process of embryo implantation, thus leading to repeated implantation failure (RIF) in women undergoing in vitro fertilization (IVF). However, the molecular mechanisms linking uterine microbiota and IVF out-comes are still an open question. The aim of this cohort study was to outline the relationship between endometrial microbiota, inflammation and IVF outcomes. To this purpose, endometrial microbiota and selected components of the “cytokine network” were analyzed in women presenting RIF and divided between eubiosis and dysbiosis groups, according to the percentage of endometrial lactobacilli (≥90% or <90%, respectively). The Dysbiosis group presented significantly higher tissue concentrations of the inflammatory markers (IL-6, IL-1β, HIF-1α and COX-2) and significantly lower levels of the anti-inflammatory/well-being factors, IL-10 and IGF-1, with respect to women with eubiosis. Moreover, the Lactobacillus percentage was negatively related to the concentrations of the inflammatory molecules and positively related to IL-10/IGF-1. Interestingly, the number of IVF attempts was directly related to the levels of the inflammatory factors COX-2, IL-1β and HIF-1α in the eubiosis group. Overall, endometrial dysbiosis was demonstrated to be associated with inflammation-related endometrial changes affecting the process of embryo implantation, underlining the importance of assessing uterine microbiota in patients undergoing IVF.
Collapse
Affiliation(s)
- Vito Cela
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (V.C.); (M.E.R.O.); (F.P.); (I.M.)
- San Rossore Clinic Care, 56100 Pisa, Italy; (M.R.); (F.T.)
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy; (E.Z.); (L.C.); (C.M.)
| | - Maria Elena Rosa Obino
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (V.C.); (M.E.R.O.); (F.P.); (I.M.)
| | - Maria Ruggiero
- San Rossore Clinic Care, 56100 Pisa, Italy; (M.R.); (F.T.)
| | - Elisa Zappelli
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy; (E.Z.); (L.C.); (C.M.)
| | - Lorenzo Ceccarelli
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy; (E.Z.); (L.C.); (C.M.)
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Francesca Papini
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (V.C.); (M.E.R.O.); (F.P.); (I.M.)
| | - Ilaria Marzi
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (V.C.); (M.E.R.O.); (F.P.); (I.M.)
| | - Giorgia Scarfò
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (G.S.); (F.F.)
| | - Fulvia Tosi
- San Rossore Clinic Care, 56100 Pisa, Italy; (M.R.); (F.T.)
| | - Ferdinando Franzoni
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (G.S.); (F.F.)
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy; (E.Z.); (L.C.); (C.M.)
| | - Paolo Giovanni Artini
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (V.C.); (M.E.R.O.); (F.P.); (I.M.)
- San Rossore Clinic Care, 56100 Pisa, Italy; (M.R.); (F.T.)
| |
Collapse
|
12
|
Simonov M, Vlizlo V, Stybel V, Peleno R, Salata V, Matviishyn T, Кhimych M, Gorobei O. Levels of insulin-like growth factor in bovine, goat, and sheep milk in different lactation periods: The etiological factor of cancer in humans. INTERNATIONAL JOURNAL OF ONE HEALTH 2021. [DOI: 10.14202/ijoh.2021.246-250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Despite the positive aspects of consuming dairy products, there are also some reservations. Recently, a large number of publications have demonstrated convincing evidence of a relationship between the intake of dairy products and the development of cancer in humans. This study aimed to determine the level of insulin-like growth factor [IGF], which can cause cancer in consumers, in the milk of farm animals (cows, goats, and sheep) obtained at different stages of lactation.
Materials and Methods: Enzyme-linked immunosorbent assay and statistical method were used for data processing. The IGF level in milk was determined using an enzyme-linked immunosorbent assay. Milk was collected from cows of Holstein and Ukrainian Black-and-White dairy breed, goats of Alpine and Saanen breed, and sheep of Tsigai and East Friesian breed. Milk samples in cows were collected at the beginning, on the peak, and at the end of lactation.
Results: The obtained results showed that the highest IGF level was detected in milk obtained at the beginning of lactation, but its level depended on the animal species. More specifically, the highest level of the hormone was typical for goat's milk and for cow's milk of Holstein breed. Slightly lower values were revealed in the milk of cows of Black-and-White Ukrainian breed and sheep. During lactation, the absolute content of IGF in the milk of all studied animal species decreased. It should be noted that the most pronounced decrease in the IGF level in milk during lactation was noted in sheep (25-40 times), and minor changes were observed in goats (12-14 times). The results also showed that there were individual variations in the IGF level in milk in farm animals.
Conclusion: The highest IGF level was noted in goat's milk, and the lowest level was observed in sheep's milk. Cow's milk, in this regard, had an intermediate IGF level. The level of this particular hormone depends on the period of lactation and the individual characteristics of the animal. It is advisable to include IGF as one of the assessment factors of dairy products' safety because this hormone can cause cancer in consumers of milk and dairy products.
Collapse
Affiliation(s)
- Marian Simonov
- Faculty of Social Development and Health, Stepan Gzhytskyi National University of Veterinary Medicine and Biotechnologies Lviv, Lviv, Ukraine
| | - Vasyl Vlizlo
- Faculty of Veterinary Medicine, Stepan Gzhytskyi National University of Veterinary Medicine and Biotechnologies Lviv, Lviv, Ukraine
| | - Volodymyr Stybel
- Faculty of Veterinary Medicine, Stepan Gzhytskyi National University of Veterinary Medicine and Biotechnologies Lviv, Lviv, Ukraine
| | - Ruslan Peleno
- Faculty of Veterinary Medicine, Stepan Gzhytskyi National University of Veterinary Medicine and Biotechnologies Lviv, Lviv, Ukraine
| | - Volodymyr Salata
- Faculty of Social Development and Health, Stepan Gzhytskyi National University of Veterinary Medicine and Biotechnologies Lviv, Lviv, Ukraine
| | - Taras Matviishyn
- Faculty of Social Development and Health, Stepan Gzhytskyi National University of Veterinary Medicine and Biotechnologies Lviv, Lviv, Ukraine
| | - Mariia Кhimych
- Faculty of Veterinary Medicine, Odesa State Agrarian University, Odesa, Ukraine
| | - Oleksii Gorobei
- Faculty of Veterinary Medicine, Odesa State Agrarian University, Odesa, Ukraine
| |
Collapse
|
13
|
Ha JH, Jayaraman M, Yan M, Dhanasekaran P, Isidoro C, Song YS, Dhanasekaran DN. Identification of GNA12-driven gene signatures and key signaling networks in ovarian cancer. Oncol Lett 2021; 22:719. [PMID: 34429759 PMCID: PMC8371953 DOI: 10.3892/ol.2021.12980] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
With the focus on defining the oncogenic network stimulated by lysophosphatidic acid (LPA) in ovarian cancer, the present study sought to interrogate the oncotranscriptome regulated by the LPA-mediated signaling pathway. LPA, LPA-receptor (LPAR) and LPAR-activated G protein 12 α-subunit, encoded by G protein subunit α 12 (GNA12), all serve an important role in ovarian cancer progression. While the general signaling mechanism regulated by LPA/LPAR/GNA12 has previously been characterized, the global transcriptomic network regulated by GNA12 in ovarian cancer pathophysiology remains largely unknown. To define the LPA/LPAR/GNA12-orchestrated oncogenic networks in ovarian cancer, transcriptomic and bioinformatical analyses were conducted using SKOV3 cells, in which the expression of GNA12 was silenced. Array analysis was performed in Agilent SurePrint G3 Human Comparative Genomic Hybridization 8×60 microarray platform. The array results were validated using Kuramochi cells. Gene and functional enrichment analyses were performed using Database for Annotation, Visualization and Integrated Discovery, Search Tool for Retrieval of Interacting Genes and Cytoscape algorithms. The results indicated a paradigm in which GNA12 drove ovarian cancer progression by upregulating a pro-tumorigenic network with AKT1, VEGFA, TGFB1, BCL2L1, STAT3, insulin-like growth factor 1 and growth hormone releasing hormone as critical hub and/or bottleneck nodes. Moreover, GNA12 downregulated a growth-suppressive network involving proteasome 20S subunit (PSM) β6, PSM α6, PSM ATPase 5, ubiquitin conjugating enzyme E2 E1, PSM non-ATPase 10, NDUFA4 mitochondrial complex-associated, NADH:ubiquinone oxidoreductase subunit B8 and anaphase promoting complex subunit 1 as hub or bottleneck nodes. In addition to providing novel insights into the LPA/LPAR/GNA12-regulated oncogenic networks in ovarian cancer, the present study identified several potential nodes in this network that could be assessed for targeted therapy.
Collapse
Affiliation(s)
- Ji-Hee Ha
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA.,Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Muralidharan Jayaraman
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA.,Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Mingda Yan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Padmaja Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Ciro Isidoro
- Laboratory of Molecular Pathology and NanoBioImaging, Department of Health Sciences, University of Eastern Piedmont, I-17-28100 Novara, Italy
| | - Yong-Sang Song
- Department of Obstetrics and Gynecology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul 151-921, Republic of Korea
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA.,Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| |
Collapse
|
14
|
The IGF-1 Signaling Pathway in Viral Infections. Viruses 2021; 13:v13081488. [PMID: 34452353 PMCID: PMC8402757 DOI: 10.3390/v13081488] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/12/2021] [Accepted: 07/21/2021] [Indexed: 01/29/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) and the IGF-1 receptor (IGF-1R) belong to the insulin-like growth factor family, and IGF-1 activates intracellular signaling pathways by binding specifically to IGF-1R. The interaction between IGF-1 and IGF-1R transmits a signal through a number of intracellular substrates, including the insulin receptor substrate (IRS) and the Src homology collagen (Shc) proteins, which activate two major intracellular signaling pathways: the phosphatidylinositol 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK) pathways, specifically the extracellular signal-regulated kinase (ERK) pathways. The PI3K/AKT kinase pathway regulates a variety of cellular processes, including cell proliferation and apoptosis. IGF1/IGF-1R signaling also promotes cell differentiation and proliferation via the Ras/MAPK pathway. Moreover, upon IGF-1R activation of the IRS and Shc adaptor proteins, Shc stimulates Raf through the GTPase Ras to activate the MAPKs ERK1 and ERK2, phosphorylate and several other proteins, and to stimulate cell proliferation. The IGF-1 signaling pathway is required for certain viral effects in oncogenic progression and may be induced as an effect of viral infection. The mechanisms of IGF signaling in animal viral infections need to be clarified, mainly because they are involved in multifactorial signaling pathways. The aim of this review is to summarize the current data obtained from virological studies and to increase our understanding of the complex role of the IGF-1 signaling axis in animal virus infections.
Collapse
|
15
|
Qiu Y, Su M, Liu L, Tang Y, Pan Y, Sun J. Clinical Application of Cytokines in Cancer Immunotherapy. Drug Des Devel Ther 2021; 15:2269-2287. [PMID: 34079226 PMCID: PMC8166316 DOI: 10.2147/dddt.s308578] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cytokines are key components of the immune system and play pivotal roles in anticancer immune response. Cytokines as either therapeutic agents or targets hold clinical promise for cancer precise treatment. Here, we provide an overview of the various roles of cytokines in the cancer immunity cycle, with a particular focus on the clinical researches of cytokine-based drugs in cancer therapy. We review 27 cytokines in 2630 cancer clinical trials registered with ClinicalTrials.gov that had completed recruitment up to January 2021 while summarizing important cases for each cytokine. We also discuss recent progress in methods for improving the delivery efficiency, stability, biocompatibility, and availability of cytokines in therapeutic applications.
Collapse
Affiliation(s)
- Yi Qiu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Mengxi Su
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Leyi Liu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yiqi Tang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yuan Pan
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Jianbo Sun
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| |
Collapse
|
16
|
Immune-Related Four-lncRNA Signature for Patients with Cervical Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3641231. [PMID: 33274204 PMCID: PMC7683128 DOI: 10.1155/2020/3641231] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 12/24/2022]
Abstract
Cervical cancer (CC) is a common gynecological malignancy for which prognostic and therapeutic biomarkers are urgently needed. The signature based on immune-related lncRNAs (IRLs) of CC has never been reported. This study is aimed at establishing an IRL signature for patients with CC. A cohort of 326 CC and 21 normal tissue samples with corresponding clinical information was included in this study. Twenty-eight IRLs were collected according to the Pearson correlation analysis between the immune score and lncRNA expression (p < 0.01). Four IRLs (BZRAP1-AS1, EMX2OS, ZNF667-AS1, and CTC-429P9.1) with the most significant prognostic values (p < 0.05) were identified which demonstrated an ability to stratify patients into the low-risk and high-risk groups by developing a risk score model. It was observed that patients in the low-risk group showed longer overall survival (OS) than those in the high-risk group in the training set, valid set, and total set. The area under the curve (AUC) of the receiver operating characteristic curve (ROC curve) for the four-IRL signature in predicting the one-, two-, and three-year survival rates was larger than 0.65. In addition, the low-risk and high-risk groups displayed different immune statuses in GSEA. These IRLs were also significantly correlated with immune cell infiltration. Our results showed that the IRL signature had a prognostic value for CC. Meanwhile, the specific mechanisms of the four IRLs in the development of CC were ascertained preliminarily.
Collapse
|
17
|
Wei CJ, Gu YH, Wang W, Ren JY, Cui XW, Lian X, Liu J, Wang HJ, Gu B, Li QF, Wang ZC. A narrative review of the role of fibroblasts in the growth and development of neurogenic tumors. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1462. [PMID: 33313207 PMCID: PMC7723559 DOI: 10.21037/atm-20-3218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neurogenic tumors, a group of tumors arising from neurogenic elements, could theoretically appear in every region of human bodies wherever nerves exist. Patients with these tumors suffer from both physical and psychological problems. However, as a relatively rare tumor type, therapies are relatively scarce for these tumors due to the limited understanding of the underlying mechanisms. Recently, a tailored tumor microenvironment containing multiple types of nonneoplastic cells has been considered to play an essential role in tumor survival, growth, and metastasis. Fibroblasts are a crucial constituent of the tumor microenvironment and have been found to promote tumor growth via multiple mechanisms. However, the understanding of the pivotal role of fibroblasts in the tumorigenesis and development of the neurogenic tumors is still incomplete, and studies in this area show differences in rates of progression among different neurogenic tumor subtypes. Nevertheless, all these neural crest-originated neoplasms show some similarities in the tumor microenvironment, indicating that studies of one subtype of neurogenic tumor might assist in clarifying the underlying mechanisms of other subtypes. This review aims to provide current studies showing the impacts of fibroblasts on major benign/malignant subtypes of neurogenic tumors, including neurofibromatosis type 1, neuroblastomas, pheochromocytomas, and malignant peripheral nerve sheath tumors. Multiple related mechanisms such as the fibroblasts regulating the tumor inflammation, angiogenesis, metabolism, and microenvironment establishment have been studied up to present. Consistently, we focus on how studies on various subtypes of these neurogenic tumors contribute to the establishment of potential future directions for further studies in this area. Clarifying the underlying mechanisms by which fibroblasts promote the growth and metastasis of neurogenic tumors will indicate new therapeutic targets for further clinical treatment.
Collapse
Affiliation(s)
- Cheng-Jiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Hui Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie-Yi Ren
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi-Wei Cui
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Lian
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui-Jing Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Wang G, Wang D, Sun M, Liu X, Yang Q. Identification of prognostic and immune-related gene signatures in the tumor microenvironment of endometrial cancer. Int Immunopharmacol 2020; 88:106931. [PMID: 32889237 DOI: 10.1016/j.intimp.2020.106931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/23/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022]
Abstract
Uterine corpus endometrial cancer (UCEC) is one of the most prevalent female malignancies in clinical practice. Due to the lack of effective biomarkers and personalized treatments, the prognosis of advanced-stage EC remains unfavorable. Modulation of the immune microenvironment is closely related to the onset and development of endometrial cancer. In the present study, we attempt to systematically analyze the characteristics of the immune microenvironment of endometrial cancer and investigate its association with clinical features by applying bioinformatics. RNA-Seq in TCGA (The Cancer Genome Atlas) and clinical follow-up information of patents were used for analysis. The Tumor Microenvironment (TME) score infiltration patterns of 523 endometrial cancer patients were evaluated using CIBERSORT. Random forest, multivariable cox analysis were used to build the TME score. Fisher's exact test was used to compare the genes that show significant differences in the frequency of mutations between groups. Two TME phenotypes were defined. There is a significant relationship between the TME score and grade. High TME score samples are highly expressed in immune activation, TGF pathway activation and immune checkpoint genes, and low TME score samples have high frequency mutations of PTEN, CSE1L and ITGB3. Therefore, describing the comprehensive landscape of UCEC's TME characteristics may help explain patients' response to immunotherapy and provide new strategies for cancer treatment.
Collapse
Affiliation(s)
- Guangwei Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Dandan Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Meige Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaofei Liu
- Department of Obstetrics and Gynecology, Shenyang Women's and Children's Hospital, Shenyang 110014, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
19
|
Zhao Z, Zhang N, Li A, Zhou B, Chen Y, Chen S, Huang M, Wu F, Zhang L. Insulin-like growth factor-1 receptor induces immunosuppression in lung cancer by upregulating B7-H4 expression through the MEK/ERK signaling pathway. Cancer Lett 2020; 485:14-26. [PMID: 32417396 DOI: 10.1016/j.canlet.2020.04.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 04/04/2020] [Accepted: 04/10/2020] [Indexed: 12/24/2022]
Abstract
The Insulin-like growth factor-1/Insulin-like growth factor-1 receptor (IGF1/IGF1R) axis contributes to immunosuppression during tumor progression; however, the underlying mechanism remains unclear. In the present study, we found that IGF1 stimulation or IGF1R overexpression (IGF1R-OE) could upregulate the expression of B7-H4, while IGF1R inhibition downregulated B7-H4 in both A549 and SPC-A-1 lung cancer cell lines. IGF1R-OE conferred the inhibition of CD8+ T cells by cancer cells in vitro, and induction of B7-H4 expression was mediated by the activation of the MEK/ERK1/2 signaling pathway. The in vitro findings were further confirmed in vivo using a Lewis lung cancer mouse model. IGF1R-OE promoted tumor growth and inhibited tumor infiltration by CD8+ T cells in the mouse model. However, this effect was suppressed when B7-H4 was knocked down in IGF1R-OE cells. Our findings suggest that IGF1R could induce immunosuppression in lung cancer by upregulating the expression of B7-H4 through the MEK/ERK pathway. B7-H4 may therefore be a potential therapeutic target for lung cancer immunotherapy.
Collapse
Affiliation(s)
- Zhiming Zhao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Ningyue Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Anqi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Bin Zhou
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu, China; Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, Soochow University, Suzhou, Jiangsu, China
| | - Yali Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Shaomu Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Moli Huang
- Department of Bioinformatics, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Fengying Wu
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Liang Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China; Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu, China; Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
20
|
Somri-Gannam L, Meisel-Sharon S, Hantisteanu S, Groisman G, Limonad O, Hallak M, Bruchim I. IGF1R Axis Inhibition Restores Dendritic Cell Antitumor Response in Ovarian Cancer. Transl Oncol 2020; 13:100790. [PMID: 32428851 PMCID: PMC7232112 DOI: 10.1016/j.tranon.2020.100790] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/24/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy. The insulin-like growth factor (IGF) system plays a key role in regulating growth and invasiveness in several malignancies, including ovarian cancer. IGF1R targeting showed antiproliferative activity of EOC cells. However, clinical studies failed to show significant benefit. EOC cells suppress antitumor immune responses by inducing dendritic cell (DC) dysfunction. The IGF1 axis can regulate DC maturation. The current study evaluated involvement of the IGF1 axis in DC differentiation in EOC. Studies were conducted on EOC and on a human monocyte cell line. Tissue microarray analysis (TMA) was performed on 36 paraffin blocks from EOC patients. Expression of IGF1R, p53, Ki67, BRCA1, and DC markers was evaluated using immunohistochemistry. Co-culture of EOC cells with DC pretreated with IGF1R inhibitor blocked cancer cell migration. TMA demonstrated higher rate of IGF1R protein expression in patients with advanced (76.9%) as compared to early (40%) EOC. A negative correlation between IGF1R protein expression and the CD1c marker was found. These findings provide evidence that IGF1R axis inhibition could be a therapeutic strategy for ovarian cancer by restoring DC-mediated antitumor immunity.
Collapse
Affiliation(s)
- Lina Somri-Gannam
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| | - Shilhav Meisel-Sharon
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel
| | - Shay Hantisteanu
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel
| | - Gabriel Groisman
- Institute of Pathology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Ofer Limonad
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel; Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Mordechai Hallak
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel; Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Ilan Bruchim
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel; Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| |
Collapse
|
21
|
Ma Z, Cai Y, Zhang L, Tian C, Lyu L. LINC00319 Promotes Cervical Cancer Progression Via Targeting miR-147a/IGF1R Pathway. Cancer Biother Radiopharm 2020:cbr.2020.3722. [PMID: 32644822 DOI: 10.1089/cbr.2020.3722] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background: Cervical cancer is identified as the fourth most common female malignancy worldwide. Recently, Linc00319 was reported to play an important role in the development and progression of cervical cancer. However, little is known about the molecular mechanism and clinical significance of Linc00319 in the carcinogenesis of cervical cancer. This study aims to reveal the biological function and molecular mechanisms of Linc00319 in cell proliferation, invasion, and migration of cervical cancer. Materials and Methods: In the current study, gene expression levels of Linc00319, miR-147a, and IFG1R were detected by quantitative real-time PCR in clinical tissue samples and cervical cancer cell lines. Protein levels were also determined by western blot assay in cervical cancer cells. CCK-8, transwell, and wound healing assays were used to test the proliferation, invasion, and migration of cervical cancer cell lines in vitro. Target genes were predicted through bioinformatics methods and then verified by gene engineering technology. Results: The authors' results showed that Linc00319 was upregulated in cervical cancer tissues and cell lines, while Linc00319silencing could inhibit cervical cancer cell proliferation, invasion, and migration. Further investigations showed that Linc00319 interacted with miR-147a and inhibited its expression, unregulated IGF1R to induce progression of cervical cancer. Conclusions: Their research indicated that Linc00319 might play an oncogenic role in cervical cancer and regulate the progression of cervical tumor growth by inhibiting the expression of miR-147a and activating IGF1R-related pathway. The findings suggest a novel molecular biomarker and therapeutic target for cervical tumor and may provide a novel therapeutic strategy for preventing the metastasis of cervical cancer.
Collapse
Affiliation(s)
- Zhe Ma
- Department of Obstetrics and Gynecology, Affiliated Hospital of Beihua University, Jilin City, China
| | - Yufei Cai
- Department of Obstetrics and Gynecology, Affiliated Hospital of Beihua University, Jilin City, China
| | - Limei Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Beihua University, Jilin City, China
| | - Chenchen Tian
- Department of Obstetrics and Gynecology, Affiliated Hospital of Beihua University, Jilin City, China
| | - Lin Lyu
- Department of Gynecology, The First Affiliated Hospital of Chengdu Medical College, Chengdu City, China
| |
Collapse
|