1
|
Wroblewski TH, Karabacak M, Seah C, Yong RL, Margetis K. Radiomic Consensus Clustering in Glioblastoma and Association with Gene Expression Profiles. Cancers (Basel) 2024; 16:4256. [PMID: 39766155 PMCID: PMC11674874 DOI: 10.3390/cancers16244256] [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: 11/19/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Glioblastoma (GBM) is the most common malignant primary central nervous system tumor with extremely poor prognosis and survival outcomes. Non-invasive methods like radiomic feature extraction, which assess sub-visual imaging features, provide a potentially powerful tool for distinguishing molecular profiles across groups of patients with GBM. Using consensus clustering of MRI-based radiomic features, this study aims to investigate differential gene expression profiles based on radiomic clusters. METHODS Patients from the TCGA and CPTAC datasets (n = 114) were included in this study. Radiomic features including T1, T1 with contrast, T2, and FLAIR MRI sequences were extracted using PyRadiomics. Selected radiomic features were then clustered using ConsensusClusterPlus (k-means base algorithm and Euclidean distance), which iteratively subsamples and clusters 80% of the data to identify stable clusters by calculating the frequency in which each patient is a member of a cluster across iterations. Gene expression data (available for n = 69 patients) was analyzed using differential gene expression (DEG) and gene set enrichment (GSEA) approaches, after batch correction using ComBat-seq. RESULTS Three distinct clusters were identified based on the relative consensus matrix and cumulative distribution plots (Cluster 1, n = 25; Cluster 2, n = 46; Cluster 3, n = 43). No significant differences in patient demographic characteristics, MGMT methylation status, tumor location, or overall survival were identified across clusters. Differentially expressed genes were identified in Cluster 1, which have been previously associated with GBM prognosis, recurrence, and treatment sensitivity. GSEA of Cluster 1 showed an enrichment of genes upregulated for immune-related and DNA metabolism pathways and genes downregulated in pathways associated with protein and histone deacetylation. Clusters 2 and 3 exhibited fewer DEGs which failed to reach significance after multiple testing corrections. CONCLUSIONS Consensus clustering of radiomic features revealed unique gene expression profiles in the GBM cohort which likely represent subtle differences in tumor biology and radiosensitivity that are not visually discernible, underscoring the potential of radiomics to serve as a non-invasive alternative for identifying GBM molecular heterogeneity. Further investigation is still required to validate these findings and their clinical implications.
Collapse
Affiliation(s)
- Tadeusz H. Wroblewski
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA;
- MD-PhD Program, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Mert Karabacak
- Department of Neurosurgery, Mount Sinai Health System, New York, NY 10029, USA; (M.K.); (R.L.Y.)
| | - Carina Seah
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Raymund L. Yong
- Department of Neurosurgery, Mount Sinai Health System, New York, NY 10029, USA; (M.K.); (R.L.Y.)
| | - Konstantinos Margetis
- Department of Neurosurgery, Mount Sinai Health System, New York, NY 10029, USA; (M.K.); (R.L.Y.)
| |
Collapse
|
2
|
Jia D, Wu K, Luo J, Xu X, Pan W, Zhao M, Li S, Gong J, Gong J. Wogonin Alleviates DCD Liver Ischemia/Reperfusion Injury by Regulating ALOX15/iNOS-mediated Ferroptosis. Transplantation 2024; 108:2374-2385. [PMID: 38946036 DOI: 10.1097/tp.0000000000005123] [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] [Indexed: 07/02/2024]
Abstract
BACKGROUND Donation after circulatory death livers are more susceptible to ischemia/reperfusion injury (IRI) because of a longer period of warm ischemia. Growing evidence now suggests that ferroptosis plays a key regulatory role in the development of IRI, so targeting ferroptosis may be an effective strategy to alleviate IRI in liver transplantation (LT). METHODS Using donation after circulatory death LT models in rats and oxygen-glucose deprivation/reoxygenation (OGD/R) models in BRL-3A cells, we tested the effect of the Chinese medicine monomer wogonin on liver IRI and explored the specific mechanism. RESULTS Wogonin attenuated liver IRI and increased the survival rate of rats by inhibiting lipid peroxidation and ferroptosis. Mechanistically, arachidonic acid 15-lipoxygenase-1 (ALOX15) and inducible nitric oxide synthase (iNOS) were identified as potential targets of baicalein through bioinformatics analysis combined with in vivo and in vitro experiments. This result was further confirmed by molecular docking and cellular thermal shift assays. Finally, we silenced ALOX15 and iNOS in the OGD/R cell model and found that silencing ALOX15 and iNOS could reproduce the regulatory effect of wogonin and abrogate the regulatory effect of wogonin. CONCLUSIONS In brief, this study emphasizes that wogonin exerts a protective effect in liver IRI through the regulation of ALOX15- and iNOS-mediated ferroptosis. ALOX15 and iNOS are potential targets for intervention in IRI induced by LT, and wogonin is a drug candidate for LT patients.
Collapse
Affiliation(s)
- Degong Jia
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Dadario NB, Boyett DM, Teasley DE, Chabot PJ, Winans NJ, Argenziano MG, Sperring CP, Canoll P, Bruce JN. Unveiling the Inflammatory Landscape of Recurrent Glioblastoma through Histological-Based Assessments. Cancers (Basel) 2024; 16:3283. [PMID: 39409905 PMCID: PMC11476027 DOI: 10.3390/cancers16193283] [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: 08/21/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
The glioblastoma (GBM) tumor microenvironment consists of a heterogeneous mixture of neoplastic and non-neoplastic cells, including immune cells. Tumor recurrence following standard-of-care therapy results in a rich landscape of inflammatory cells throughout the glioma-infiltrated cortex. Immune cells consisting of glioma-associated macrophages and microglia (GAMMs) overwhelmingly constitute the bulk of the recurrent glioblastoma (rGBM) microenvironment, in comparison to the highly cellular and proliferative tumor microenvironment characteristic of primary GBM. These immune cells dynamically interact within the tumor microenvironment and can contribute to disease progression and therapy resistance while also providing novel targets for emerging immunotherapies. Within these varying contexts, histological-based assessments of immune cells in rGBM, including immunohistochemistry (IHC) and immunofluorescence (IF), offer a critical way to visualize and examine the inflammatory landscape. Here, we exhaustively review the available body of literature on the inflammatory landscape in rGBM as identified through histological-based assessments. We highlight the heterogeneity of immune cells throughout the glioma-infiltrated cortex with a focus on microglia and macrophages, drawing insights from canonical and novel immune-cell histological markers to estimate cell phenotypes and function. Lastly, we discuss opportunities for immunomodulatory treatments aiming to harness the inflammatory landscape in rGBM.
Collapse
Affiliation(s)
- Nicholas B. Dadario
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Deborah M. Boyett
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Damian E. Teasley
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Peter J. Chabot
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Nathan J. Winans
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Michael G. Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Colin P. Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Peter Canoll
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA
| | - Jeffrey N. Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| |
Collapse
|
4
|
Chen S, Hu T, Zhao J, Zhu Q, Wang J, Huang Z, Xiang C, Zhao R, Zhu C, Lu S, Han Y. Novel molecular subtypes of METex14 non-small cell lung cancer with distinct biological and clinical significance. NPJ Precis Oncol 2024; 8:159. [PMID: 39060379 PMCID: PMC11282101 DOI: 10.1038/s41698-024-00642-6] [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: 12/29/2023] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Not all MET exon 14 skipping (METex14) NSCLC patients benefited from MET inhibitors. We hypothesized an inter-tumoral heterogeneity in METex14 NSCLC. Investigations at genomic and transcriptomic level were conducted in METex14 NSCLC samples from stage I-III and recurrent/metastatic patients as discovery and validation cohort. Four molecular subtypes were discovered. MET-Driven subtype, with the worst prognosis, displayed MET overexpression, enrichment of MET-related pathways, and higher infiltration of fibroblast and regulatory T cells. Immune-Activated subtype having the most idea long-term survival, had higher tertiary lymphoid structures, spatial co-option of PD-L1+ cancer cells, and GZMK+ CD8+ T cell. FGFR- and Bypass-Activated subtypes displayed FGFR2 overexpression and enrichments of multiple oncogenic pathways respectively. In the validation cohort, patients with MET-Driven subtype had better response to MET inhibitors than those with MET overexpression. Thus, molecular subtypes of METex14 NSCLC with distinct biological and clinical significance may indicate more precise therapeutic strategies for METex14 NSCLC patients.
Collapse
Affiliation(s)
- Shengnan Chen
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tao Hu
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Jikai Zhao
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qian Zhu
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jin Wang
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Zhan Huang
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Chan Xiang
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ruiying Zhao
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Changbin Zhu
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China.
| | - Shun Lu
- Department of Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
5
|
Gong S, Xiang K, Chen L, Zhuang H, Song Y, Chen J. Integrated bioinformatics analysis identified leucine rich repeat containing 15 and secreted phosphoprotein 1 as hub genes for calcific aortic valve disease and osteoarthritis. IET Syst Biol 2024; 18:77-91. [PMID: 38566328 PMCID: PMC11179158 DOI: 10.1049/syb2.12091] [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: 06/05/2023] [Revised: 01/05/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Calcific aortic valve disease (CAVD) and osteoarthritis (OA) are common diseases in the ageing population and share similar pathogenesis, especially in inflammation. This study aims to discover potential diagnostic and therapeutic targets in patients with CAVD and OA. Three CAVD datasets and one OA dataset were obtained from the Gene Expression Omnibus database. We used bioinformatics methods to search for key genes and immune infiltration, and established a ceRNA network. Immunohistochemical staining was performed to verify the expression of candidate genes in human and mice aortic valve tissues. Two key genes obtained, leucine rich repeat containing 15 (LRRC15) and secreted phosphoprotein 1 (SPP1), were further screened using machine learning and verified in human and mice aortic valve tissues. Compared to normal tissues, the infiltration of immune cells in CAVD tissues was significantly higher, and the expressions of LRRC15 and SPP1 were positively correlated with immune cells infiltration. Moreover, the ceRNA network showed extensive regulatory interactions based on LRRC15 and SPP1. The authors' findings identified LRRC15 and SPP1 as hub genes in immunological mechanisms during CAVD and OA initiation and progression, as well as potential targets for drug development.
Collapse
Affiliation(s)
- Shuji Gong
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Kun Xiang
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Le Chen
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Huanwei Zhuang
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Yaning Song
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Jinlan Chen
- Department of Cardiovascular SurgeryThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| |
Collapse
|
6
|
Montemurro N, Pahwa B, Tayal A, Shukla A, De Jesus Encarnacion M, Ramirez I, Nurmukhametov R, Chavda V, De Carlo A. Macrophages in Recurrent Glioblastoma as a Prognostic Factor in the Synergistic System of the Tumor Microenvironment. Neurol Int 2023; 15:595-608. [PMID: 37218976 PMCID: PMC10204554 DOI: 10.3390/neurolint15020037] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM) is a common and highly malignant primary tumor of the central nervous system in adults. Ever more recent papers are focusing on understanding the role of the tumor microenvironment (TME) in affecting tumorigenesis and the subsequent prognosis. We assessed the impact of macrophages in the TME on the prognosis in patients with recurrent GBM. A PubMed, MEDLINE and Scopus review was conducted to identify all studies dealing with macrophages in the GBM microenvironment from January 2016 to December 2022. Glioma-associated macrophages (GAMs) act critically in enhancing tumor progression and can alter drug resistance, promoting resistance to radiotherapy and establishing an immunosuppressive environment. M1 macrophages are characterized by increased secretion of proinflammatory cytokines, such as IL-1ß, tumor necrosis factor (TNF), IL-27, matrix metalloproteinase (MMPs), CCL2, and VEGF (vascular endothelial growth factor), IGF1, that can lead to the destruction of the tissue. In contrast, M2 is supposed to participate in immunosuppression and tumor progression, which is formed after being exposed to the macrophage M-CSF, IL-10, IL-35 and the transforming growth factor-ß (TGF-β). Because there is currently no standard of care in recurrent GBM, novel identified targeted therapies based on the complex signaling and interactions between the glioma stem cells (GSCs) and the TME, especially resident microglia and bone-marrow-derived macrophages, may be helpful in improving the overall survival of these patients in the near future.
Collapse
Affiliation(s)
- Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| | - Bhavya Pahwa
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anish Tayal
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | - Anushruti Shukla
- University College of Medical Sciences and GTB Hospital, New Delhi 110095, India
| | | | - Issael Ramirez
- Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Renat Nurmukhametov
- Department of Spinal Surgery, Central Clinical Hospital of the Russian Academy of Sciences, 121359 Moscow, Russia
| | - Vishal Chavda
- Department of Pathology, Stanford of School of Medicine, Stanford University Medical Centre, Palo Alto, CA 94305, USA
| | - Antonella De Carlo
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), University of Pisa, 56100 Pisa, Italy
| |
Collapse
|
7
|
LRRC superfamily expression in stromal cells predicts the clinical prognosis and platinum resistance of ovarian cancer. BMC Med Genomics 2023; 16:10. [PMID: 36653841 PMCID: PMC9850808 DOI: 10.1186/s12920-023-01435-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Leucine-rich repeat sequence domains are known to mediate protein‒protein interactions. Recently, some studies showed that members of the leucine rich repeat containing (LRRC) protein superfamily may become new targets for the diagnosis and treatment of tumours. However, it is not known whether any of the LRRC superfamily genes is expressed in the stroma of ovarian cancer (OC) and is associated with prognosis. METHODS The clinical data and transcriptional profiles of OC patients from the public databases TCGA (n = 427), GTEx (n = 88) and GEO (GSE40266 and GSE40595) were analysed by R software. A nomogram model was also generated through R. An online public database was used for auxiliary analysis of prognosis, immune infiltration and protein‒protein interaction (PPI) networks. Immunohistochemistry and qPCR were performed to determine the protein and mRNA levels of genes in high-grade serous ovarian cancer (HGSC) tissues of participants and the MRC-5 cell line induced by TGF-β. RESULTS LRRC15 and LRRC32 were identified as differentially expressed genes from the LRRC superfamily by GEO transcriptome analysis. PPI network analysis suggested that they were most enriched in TGF-β signalling. The TCGA-GTEx analysis results showed that only LRRC15 was highly expressed in both cancer-associated fibroblasts (CAFs) and the tumour stroma of OC and was related to clinical prognosis. Based on this, we developed a nomogram model to predict the incidence of adverse outcomes in OC. Moreover, LRRC15 was positively correlated with CAF infiltration and negatively correlated with CD8 + T-cell infiltration. As a single indicator, LRRC15 had the highest accuracy (AUC = 0.920) in predicting the outcome of primary platinum resistance. CONCLUSIONS The LRRC superfamily is related to the TGF-β pathway in the microenvironment of OC. LRRC15, as a stromal biomarker, can predict the clinical prognosis of HGSC and promote the immunosuppressive microenvironment. LRRC15 may be a potential therapeutic target for reversing primary resistance in OC.
Collapse
|
8
|
Baurand PE, Balland J, Reynas C, Ramseyer M, Vivier D, Bellaye PS, Collin B, Paul C, Denat F, Asgarov K, Pallandre JR, Ringenbach L. Development of Anti-LRRC15 Small Fragments for Imaging Purposes Using a Phage-Display ScFv Approach. Int J Mol Sci 2022; 23:ijms232012677. [PMID: 36293532 PMCID: PMC9604383 DOI: 10.3390/ijms232012677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
The human leucine-rich repeat-containing protein 15 (LRRC15) is a membrane protein identified as a marker of CAF (cancer-associated fibroblast) cells whose overexpression is positively correlated with cancer grade and outcome. Nuclear molecular imaging (i.e., SPECT and PET) to track LRRC15 expression could be very useful in guiding further therapeutic strategies. In this study, we developed an ScFv mouse phage-display library to obtain small fragment antibodies against human LRRC15 for molecular imaging purposes. Mice were immunized with recombinant human LRRC15 (hLRRC15), and lymph node cells were harvested for ScFv (single-chain variable fragment) phage-display analysis. The built library was used for panning on cell lines with constitutive or induced expression after transfection. The choice of best candidates was performed by screening various other cell lines, using flow cytometry. The selected candidates were reformatted into Cys-ScFv or Cys-diabody by addition of cysteine, and cloned in mammalian expression vectors to obtain batches of small fragments that were further used in site-specific radiolabeling tests. The obtained library was 1.2 × 107 cfu/µg with an insertion rate >95%. The two panning rounds performed on cells permittedenrichment of 2 × 10−3. Screening with flow cytometry allowed us to identify 28 specific hLRRC15 candidates. Among these, two also recognized murine LRCC15 and were reformatted into Cys-ScFv and Cys-diabody. They were expressed transiently in a mammalian system to obtain 1.0 to 4.5 mg of Cys fragments ready for bioconjugation and radiolabeling. Thus, in this paper, we demonstrate the relevance of the phage-display ScFv library approach for the fast-track development of small antibodies for imaging and/or immunotherapy purposes.
Collapse
Affiliation(s)
- Pierre-Emmanuel Baurand
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
- Correspondence:
| | - Jérémy Balland
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
| | - Chloé Reynas
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
| | - Mélanie Ramseyer
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
| | - Delphine Vivier
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS 6302, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Pierre-Simon Bellaye
- Plateforme D’imagerie et de Radiothérapie Précliniques (PIRP), Service de Médecine Nucléaire, Centre Georges-François Leclerc, 1 Rue du Pr Marion, 21000 Dijon, France
| | - Bertrand Collin
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS 6302, Université de Bourgogne Franche-Comté, 21000 Dijon, France
- Plateforme D’imagerie et de Radiothérapie Précliniques (PIRP), Service de Médecine Nucléaire, Centre Georges-François Leclerc, 1 Rue du Pr Marion, 21000 Dijon, France
| | - Catherine Paul
- Laboratoire d’Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
- LIIC, EA7269, Université de Bourgogne Franche Comté, 21000 Dijon, France
| | - Franck Denat
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS 6302, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Kamal Asgarov
- INSERM, EFS BFC, UMR1098, RIGHT, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, 25000 Besançon, France
- Clinical Investigation Center in Biotherapy, INSERM CIC-BT1431, Besançon University Hospital, 25000 Besançon, France
- ITAC Platform, University of Bourgogne Franche-Comté, 25000 Besançon, France
| | - Jean-René Pallandre
- INSERM, EFS BFC, UMR1098, RIGHT, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, 25000 Besançon, France
| | - Laurence Ringenbach
- Diaclone SAS-Part of Medix Biochemica Group, 6 Rue Dr Jean-François-Xavier Girod, BP 1985, 25000 Besançon, France
| |
Collapse
|
9
|
Jiang Y, Wang X, Li L, He J, Jin Q, Long D, Liu C, Zhou W, Liu K. A systematic analysis of C5ORF46 in gastrointestinal tumors as a potential prognostic and immunological biomarker. Front Genet 2022; 13:926943. [PMID: 35991552 PMCID: PMC9389054 DOI: 10.3389/fgene.2022.926943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Chromosome 5 open reading frame 46 (C5ORF46), also known as antimicrobial peptide with 64 amino acid residues (AP-64) and skin and saliva-secreted protein 1 (SSSP1), belongs to the family of open reading frame genes and encodes a small exosomal protein. C5ORF46 has been implicated in antibacterial activity and associated with patient prognosis in pancreatic cancer, colorectal cancer, and stomach cancer. These findings highlight the importance of C5ORF46 in gastrointestinal (GI) tumor inception and development. However, the prognostic and immunological value of C5ORF46 in human GI tumors remains largely unknown. In this study, we sought to explore the potential value of C5ORF46 in GI tumor prognosis and immunology.Method: RNA sequencing (RNA-seq) was performed on the tumor and tumor-adjacent normal samples we collected to identify potential target genes for GI tumors. Apart from our RNA-seq data, all original data were downloaded from The Cancer Genome Atlas (TCGA) database and integrated via Strawberry Perl (v 5.32.0) and R (v 4.1.1). The differential expression of C5ORF46 was examined with Oncomine, Tumor Immune Estimation Resource (TIMER), Gene Expression Profiling Interactive Analysis (GEPIA), Cancer Cell Line Encyclopedia (CCLE), the Human Protein Atlas (HPA) and TCGA databases. The c-BioPortal database was used to investigate the genomic alterations of C5ORF46. The effect of C5ORF46 on prognosis and clinical phenotypes was explored via bioinformatics analyses on the TCGA and GEPIA databases. We used the bioinformatics analyses based on the TCGA database to analyze tumor mutational burden (TMB), microsatellite instability (MSI), tumor immune cell infiltration, and the correlations between C5ORF46 expression and several immune-related genes. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was carried out via the DAVID website and presented as bubble charts using ShengXinRen online tools. Gene set enrichment analysis (GSEA) was performed using R scripts based on data downloaded from the GSEA website. Immunohistochemistry (IHC) was used to validate the expression of C5ORF46 in GI tumors.Results: The results of our RNA-seq data indicated a critical role for C5ORF46 in colon carcinogenesis. Consistently, we demonstrated that C5ORF46 was highly expressed in tumor tissues compared to normal tissues in human GI tumors. Moreover, a strong correlation was observed between C5ORF46 expression levels and patient prognosis, staging, TMB, MSI, and immune cell infiltration. Further, C5ORF46 presented as an important regulator in the tumor microenvironment (TME) and was active in the regulation of cancer immune functions. C5ORF46 is significantly correlated with genes regulating inflammation and immune responses.Conclusion:C5ORF46 may serve as a biomarker for GI tumor prognosis and immunology. C5ORF46 could be a novel target for GI tumor immunotherapy.
Collapse
Affiliation(s)
- Yuhong Jiang
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaobo Wang
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lun Li
- Department of Breast-Thyroid Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jun He
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qianqian Jin
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dongju Long
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chao Liu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Weihan Zhou
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Kuijie Liu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- *Correspondence: Kuijie Liu,
| |
Collapse
|
10
|
Menna G, Mattogno PP, Donzelli CM, Lisi L, Olivi A, Della Pepa GM. Glioma-Associated Microglia Characterization in the Glioblastoma Microenvironment through a 'Seed-and Soil' Approach: A Systematic Review. Brain Sci 2022; 12:718. [PMID: 35741603 PMCID: PMC9220868 DOI: 10.3390/brainsci12060718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 12/04/2022] Open
Abstract
Background and aim: Ever since the discovery of tumor-associated immune cells, there has been growing interest in the understanding of the mechanisms underlying the crosstalk between these cells and tumor cells. A "seed and soil" approach has been recently introduced to describe the glioblastoma (GBM) landscape: tumor microenvironments act as fertile "soil" and interact with the "seed" (glial and stem cells compartment). In the following article, we provide a systematic review of the current evidence pertaining to the characterization of glioma-associated macrophages and microglia (GAMs) and microglia and macrophage cells in the glioma tumor microenvironment (TME). Methods: An online literature search was launched on PubMed Medline and Scopus using the following research string: "((Glioma associated macrophages OR GAM OR Microglia) AND (glioblastoma tumor microenvironment OR TME))". The last search for articles pertinent to the topic was conducted in February 2022. Results: The search of the literature yielded a total of 349 results. A total of 235 studies were found to be relevant to our research question and were assessed for eligibility. Upon a full-text review, 58 articles were included in the review. The reviewed papers were further divided into three categories based on their focus: (1) Microglia maintenance of immunological homeostasis and protection against autoimmunity; (2) Microglia crosstalk with dedifferentiated and stem-like glioblastoma cells; (3) Microglia migratory behavior and its activation pattern. Conclusions: Aggressive growth, inevitable recurrence, and scarce response to immunotherapies are driving the necessity to focus on the GBM TME from a different perspective to possibly disentangle its role as a fertile 'soil' for tumor progression and identify within it feasible therapeutic targets. Against this background, our systematic review confirmed microglia to play a paramount role in promoting GBM progression and relapse after treatments. The correct and extensive understanding of microglia-glioma crosstalk could help in understanding the physiopathology of this complex disease, possibly opening scenarios for improvement of treatments.
Collapse
Affiliation(s)
- Grazia Menna
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Pier Paolo Mattogno
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Carlo Maria Donzelli
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Lucia Lisi
- Institute of Pharmacology, Catholic University of Rome, 00168 Rome, Italy;
| | - Alessandro Olivi
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| | - Giuseppe Maria Della Pepa
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.M.); (P.P.M.); (C.M.D.); (A.O.)
| |
Collapse
|
11
|
Xu WX, Wang DD, Zhao ZQ, Zhang HD, Yang SJ, Zhang Q, Li L, Zhang J. Exosomal microRNAs shuttling between tumor cells and macrophages: cellular interactions and novel therapeutic strategies. Cancer Cell Int 2022; 22:190. [PMID: 35578228 PMCID: PMC9109313 DOI: 10.1186/s12935-022-02594-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/18/2022] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles secreted by tumor microenvironment (TME) cells are vital players in tumor progression through transferring nucleic acids and proteins. Macrophages are the main immune cells in TME and tumor associated macrophages (TAM) express M2 phenotype, which induce tumor proliferation, angiogenesis, invasion, metastasis and immune elimination, resulting in the subsequent evolution of malignancies. There are a high number of studies confirmed that tumor cells and TAM interact with each other through extracellular vesicles in various cancers, like pancreatic ductal adenocarcinoma, gastric cancer, breast cancer, ovarian cancer, colon cancer, glioblastoma, hepatocellular cancer, and lung cancer. Herein, this review summarizes the current knowledge on mechanisms of communications between tumor cells and TAM via extracellular vesicles, mainly about microRNAs, and targeting these events might represent a novel approach in the clinical implications of this knowledge into successful anti-cancer strategies.
Collapse
Affiliation(s)
- Wen-Xiu Xu
- Department of General Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guanzhou Road, Nanjing, 210029, China
| | - Dan-Dan Wang
- Department of General Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guanzhou Road, Nanjing, 210029, China
| | - Zhi-Qiang Zhao
- The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, No. 62, Huaihai Road (S.), Huaian, 223002, China
| | - He-Da Zhang
- Department of General Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guanzhou Road, Nanjing, 210029, China
| | - Su-Jin Yang
- Department of General Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guanzhou Road, Nanjing, 210029, China
| | - Qian Zhang
- Department of General Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guanzhou Road, Nanjing, 210029, China
| | - Lei Li
- Department of General Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guanzhou Road, Nanjing, 210029, China.
| | - Jian Zhang
- Department of General Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guanzhou Road, Nanjing, 210029, China.
| |
Collapse
|
12
|
Tang H, Liu W, Xu Z, Zhao J, Wang W, Yu Z, Wei M. Integrated microenvironment-associated genomic profiles identify LRRC15 mediating recurrent glioblastoma-associated macrophages infiltration. J Cell Mol Med 2021; 25:5534-5546. [PMID: 33960636 PMCID: PMC8184692 DOI: 10.1111/jcmm.16563] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/22/2021] [Accepted: 04/08/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant intracranial tumour with intrinsic infiltrative characteristics, which could lead to most patients eventually relapse. The prognosis of recurrent GBM patients remains unsatisfactory. Cancer cell infiltration and their interaction with the tumour microenvironment (TME) could promote tumour recurrence and treatment resistance. In our study, we aimed to identify potential tumour target correlated with rGBM microenvironment based on the gene expression profiles and clinical information of rGBM patients from The Cancer Genome Atlas (TCGA) database. LRRC15 gene with prognostic value was screened by univariate and multivariate analysis, and the correlation between macrophages and LRRC15 was identified as well. Furthermore, the prognosis correlation and immune characteristics of LRRC15 were validated using the Chinese Glioma Genome Atlas (CGGA) database and our clinical tissues by immunochemistry assay. Additionally, we utilized the transwell assay and carboxy fluorescein succinimidyl ester (CFSE) tracking to further confirm the effects of LRRC15 on attracting microglia/macrophages and tumour cell proliferation in the TME. Gene profiles‐based rGBM microenvironment identified that LRRC15 could act in collusion with microglia/macrophages in the rGBM microenvironment to promote the poor prognosis, especially in mesenchymal subtype, indicating the strategies of targeting LRRC15 to improve macrophages‐based immunosuppressive effects could be promising for rGBM treatments.
Collapse
Affiliation(s)
- Haichao Tang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, China
| | - Wensi Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, China
| | - Zhaoxu Xu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, China
| | - Jianhang Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, China
| | - Weitao Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|