101
|
Chen X, Cao M, Wang P, Chu S, Li M, Hou P, Zheng J, Li Z, Bai J. The emerging roles of TRIM21 in coordinating cancer metabolism, immunity and cancer treatment. Front Immunol 2022; 13:968755. [PMID: 36159815 PMCID: PMC9506679 DOI: 10.3389/fimmu.2022.968755] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Tripartite motif containing-21 (TRIM21), an E3 ubiquitin ligase, was initially found to be involved in antiviral responses and autoimmune diseases. Recently studies have reported that TRIM21 plays a dual role in cancer promoting and suppressing in the occurrence and development of various cancers. Despite the fact that TRIM21 has effects on multiple metabolic processes, inflammatory responses and the efficacy of tumor therapy, there has been no systematic review of these topics. Herein, we discuss the emerging role and function of TRIM21 in cancer metabolism, immunity, especially the immune response to inflammation associated with tumorigenesis, and also the cancer treatment, hoping to shine a light on the great potential of targeting TRIM21 as a therapeutic target.
Collapse
Affiliation(s)
- Xintian Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Menghan Cao
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Pengfei Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Pingfu Hou
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Jin Bai, ; Zhongwei Li, ; Junnian Zheng,
| | - Zhongwei Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Jin Bai, ; Zhongwei Li, ; Junnian Zheng,
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Jin Bai, ; Zhongwei Li, ; Junnian Zheng,
| |
Collapse
|
102
|
Alizamir A, Dehghan Azad S, Pirdehghan A, Moradi A. Preoperative Neutrophil: Lymphocyte Ratio, Platelet: Lymphocyte Ratio, and C-Reactive Protein Levels Predictive Value in Determining the Severity of Breast Mass. IRANIAN JOURNAL OF PATHOLOGY 2022; 17:413-418. [PMID: 36532648 PMCID: PMC9745750 DOI: 10.30699/ijp.2022.539042.2727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 07/20/2022] [Indexed: 03/25/2025]
Abstract
BACKGROUND & OBJECTIVE Female breast cancer is one of the most prevalent malignancies among women. The critical step in management of breast cancer is an accurate diagnosis. Hence, peripheral blood-based tests would be one of the most favorable and less invasive methods to study. Recent studies have investigated the inflammatory parameters such as neutrophil: lymphocyte ratio (NLR), the platelet: lymphocyte ratio (PLR), and the C-reactive protein (CRP) levels. The elevation in mentioned parameters was proposed as a key factor in cancer progression. The main goal of this study was to investigate the association of NLR, PLR, and CRP levels in patients with breast lesions. METHODS The NLR, PLR, and CRP levels were calculated from 200 female patients presenting with either benign or malignant lesions. RESULTS The cut-off values of NLR, PLR, and CRP were 1.24, 96, and 10.36 mg/L, respectively. A significant difference in NLR (P<0.001), PLR (P<0.001), and CRP levels (P<0.001) were observed between the two major studied cohorts. CONCLUSION Elevated NLR, PLR, and CRP levels could predict the presence of malignancy. In addition to the low cost and properties of the mentioned methods, utilization of this data could facilitate and improve clinical decision-making for treatment.
Collapse
Affiliation(s)
- Aida Alizamir
- Department of Pathology, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Azar Pirdehghan
- Department of Community Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arash Moradi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
103
|
Chen J, Kang Z, Li S, Wang C, Zheng X, Cai Z, Pan L, Chen F, Li W. Molecular profile reveals immune-associated markers of medulloblastoma for different subtypes. Front Immunol 2022; 13:911260. [PMID: 35967388 PMCID: PMC9367478 DOI: 10.3389/fimmu.2022.911260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/04/2022] [Indexed: 11/22/2022] Open
Abstract
Medulloblastoma, a common pediatric malignant tumor, has been recognized to have four molecular subgroups [wingless (WNT), sonic hedgehog (SHH), group 3, group 4], which are defined by the characteristic gene transcriptomic and DNA methylomic profiles, and has distinct clinical features within each subgroup. The tumor immune microenvironment is integral in tumor initiation and progression and might be associated with therapeutic responses. However, to date, the immune infiltrative landscape of medulloblastoma has not yet been elucidated. Thus, we proposed MethylCIBERSORT to estimate the degree of immune cell infiltration and weighted correlation network analysis (WGCNA) to find modules of highly correlated genes. Synthesizing the hub genes in the protein–protein interaction (PPI) network and modules of the co-expression network, we identify three candidate biomarkers [GRB2-associated-binding protein 1 (GAB1), Abelson 1 (ABL1), and CXC motif chemokine receptor type 4 (CXCR4)] via the molecular profiles of medulloblastoma. Given this, we investigated the correlation between these three immune hub genes and immune checkpoint blockade response and the potential of drug prediction further. In addition, this study demonstrated a higher presence of endothelial cells and infiltrating immune cells in Group 3 tumor bulk. The above results will be conducive to better comprehending the immune-related pathogenesis and treatment of medulloblastoma.
Collapse
Affiliation(s)
- Jinyi Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuang Kang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shenglan Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Can Wang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaohong Zheng
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zehao Cai
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lexin Pan
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, China
| | - Feng Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
104
|
Zaitsev A, Chelushkin M, Dyikanov D, Cheremushkin I, Shpak B, Nomie K, Zyrin V, Nuzhdina E, Lozinsky Y, Zotova A, Degryse S, Kotlov N, Baisangurov A, Shatsky V, Afenteva D, Kuznetsov A, Paul SR, Davies DL, Reeves PM, Lanuti M, Goldberg MF, Tazearslan C, Chasse M, Wang I, Abdou M, Aslanian SM, Andrewes S, Hsieh JJ, Ramachandran A, Lyu Y, Galkin I, Svekolkin V, Cerchietti L, Poznansky MC, Ataullakhanov R, Fowler N, Bagaev A. Precise reconstruction of the TME using bulk RNA-seq and a machine learning algorithm trained on artificial transcriptomes. Cancer Cell 2022; 40:879-894.e16. [PMID: 35944503 DOI: 10.1016/j.ccell.2022.07.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/10/2022] [Accepted: 07/12/2022] [Indexed: 12/21/2022]
Abstract
Cellular deconvolution algorithms virtually reconstruct tissue composition by analyzing the gene expression of complex tissues. We present the decision tree machine learning algorithm, Kassandra, trained on a broad collection of >9,400 tissue and blood sorted cell RNA profiles incorporated into millions of artificial transcriptomes to accurately reconstruct the tumor microenvironment (TME). Bioinformatics correction for technical and biological variability, aberrant cancer cell expression inclusion, and accurate quantification and normalization of transcript expression increased Kassandra stability and robustness. Performance was validated on 4,000 H&E slides and 1,000 tissues by comparison with cytometric, immunohistochemical, or single-cell RNA-seq measurements. Kassandra accurately deconvolved TME elements, showing the role of these populations in tumor pathogenesis and other biological processes. Digital TME reconstruction revealed that the presence of PD-1-positive CD8+ T cells strongly correlated with immunotherapy response and increased the predictive potential of established biomarkers, indicating that Kassandra could potentially be utilized in future clinical applications.
Collapse
Affiliation(s)
| | | | | | | | - Boris Shpak
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | - Krystle Nomie
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | - Vladimir Zyrin
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | | | | | | | | | - Nikita Kotlov
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | | | | | - Daria Afenteva
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | | | - Susan Raju Paul
- The Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - Diane L Davies
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick M Reeves
- The Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Lanuti
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Madison Chasse
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | - Iris Wang
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | - Mary Abdou
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | | | | | - James J Hsieh
- Molecular Oncology, Division of Oncology, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Akshaya Ramachandran
- Molecular Oncology, Division of Oncology, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Yang Lyu
- Molecular Oncology, Division of Oncology, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Ilia Galkin
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA
| | | | - Leandro Cerchietti
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Mark C Poznansky
- The Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Nathan Fowler
- BostonGene, Corp., 95 Sawyer Road, Waltham, MA 02453, USA; Department of Lymphoma and Myeloma, MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 429, Houston, TX 77030, USA.
| | | |
Collapse
|
105
|
Hervás-Salcedo R, Martín-Antonio B. A Journey through the Inter-Cellular Interactions in the Bone Marrow in Multiple Myeloma: Implications for the Next Generation of Treatments. Cancers (Basel) 2022; 14:3796. [PMID: 35954459 PMCID: PMC9367481 DOI: 10.3390/cancers14153796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 02/05/2023] Open
Abstract
Tumors are composed of a plethora of extracellular matrix, tumor and non-tumor cells that form a tumor microenvironment (TME) that nurtures the tumor cells and creates a favorable environment where tumor cells grow and proliferate. In multiple myeloma (MM), the TME is the bone marrow (BM). Non-tumor cells can belong either to the non-hematological compartment that secretes soluble mediators to create a favorable environment for MM cells to grow, or to the immune cell compartment that perform an anti-MM activity in healthy conditions. Indeed, marrow-infiltrating lymphocytes (MILs) are associated with a good prognosis in MM patients and have served as the basis for developing different immunotherapy strategies. However, MM cells and other cells in the BM can polarize their phenotype and activity, creating an immunosuppressive environment where immune cells do not perform their cytotoxic activity properly, promoting tumor progression. Understanding cell-cell interactions in the BM and their impact on MM proliferation and the performance of tumor surveillance will help in designing efficient anti-MM therapies. Here, we take a journey through the BM, describing the interactions of MM cells with cells of the non-hematological and hematological compartment to highlight their impact on MM progression and the development of novel MM treatments.
Collapse
Affiliation(s)
| | - Beatriz Martín-Antonio
- Department of Experimental Hematology, Instituto de Investigación Sanitaria-Fundación Jiménez Diaz (IIS-FJD), University Autonomous of Madrid (UAM), 28040 Madrid, Spain
| |
Collapse
|
106
|
Hassanian H, Asadzadeh Z, Baghbanzadeh A, Derakhshani A, Dufour A, Rostami Khosroshahi N, Najafi S, Brunetti O, Silvestris N, Baradaran B. The expression pattern of Immune checkpoints after chemo/radiotherapy in the tumor microenvironment. Front Immunol 2022; 13:938063. [PMID: 35967381 PMCID: PMC9367471 DOI: 10.3389/fimmu.2022.938063] [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: 05/06/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
As a disease with the highest disease-associated burden worldwide, cancer has been the main subject of a considerable proportion of medical research in recent years, intending to find more effective therapeutic approaches with fewer side effects. Combining conventional methods with newer biologically based treatments such as immunotherapy can be a promising approach to treating different tumors. The concept of "cancer immunoediting" that occurs in the field of the tumor microenvironment (TME) is the aspect of cancer therapy that has not been at the center of attention. One group of the role players of the so-called immunoediting process are the immune checkpoint molecules that exert either co-stimulatory or co-inhibitory effects in the anti-tumor immunity of the host. It involves alterations in a wide variety of immunologic pathways. Recent studies have proven that conventional cancer therapies, such as chemotherapy, radiotherapy, or a combination of them, i.e., chemoradiotherapy, alter the "immune compartment" of the TME. The mentioned changes encompass a wide range of variations, including the changes in the density and immunologic type of the tumor-infiltrating lymphocytes (TILs) and the alterations in the expression patterns of the different immune checkpoints. These rearrangements can have either anti-tumor immunity empowering or immune attenuating sequels. Thus, recognizing the consequences of various chemo(radio)therapeutic regimens in the TME seems to be of great significance in the evolution of therapeutic approaches. Therefore, the present review intends to summarize how chemo(radio)therapy affects the TME and specifically some of the most important, well-known immune checkpoints' expressions according to the recent studies in this field.
Collapse
Affiliation(s)
- Hamidreza Hassanian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Derakhshani
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- McCaig Insitute, Hotchkiss Brain Institute, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- McCaig Insitute, Hotchkiss Brain Institute, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Departments of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | | | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G. Barresi” University of Messina, Messina, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
107
|
Wan Y, Wang Z, Yang N, Liu F. Treatment of Multiple Primary Malignancies With PD-1 Inhibitor Camrelizumab: A Case Report and Brief Literature Review. Front Oncol 2022; 12:911961. [PMID: 35865468 PMCID: PMC9294358 DOI: 10.3389/fonc.2022.911961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background With significant advances in the diagnostic tools and treatment modalities of cancer, the incidence of multiple primary malignancies (MPMs) has increased in the last decades. The therapeutic option changed with the arising of immune checkpoint inhibitors (ICIs), which have improved the survival of a broad spectrum of tumors. However, little information is available when it comes to the efficacy, resistance, and underlying mechanisms of ICIs. Case Presentation A 67-year-old woman was diagnosed with pulmonary sarcomatoid carcinoma (PSC) with a history of hepatocellular carcinoma (HCC) and viral hepatitis B. Following the lack of response to systemic chemotherapy, she was treated with camrelizumab, an anti-programmed cell death protein 1 monoclonal antibody, in combination with chemotherapy, and a partial response was obtained both in PSC and HCC. After a course of 9-month treatment, the PSC lesion shrank still, while HCC was evaluated as a progressive disease with an increase in the diameter of liver neoplasm, elevated alpha-fetoprotein, and enlarged abdominal lymph nodes. Then, with the addition of radiotherapy for abdominal metastasis, the lung lesion was continuously shrinking. In the meantime, the liver neoplasm and abdominal lymph nodes showed no significant enlargement. Conclusion Camrelizumab combination therapy could consistently benefit the MPM patients with PSC and HCC, which may be a promising option for patients with MPMs.
Collapse
Affiliation(s)
- Yuchen Wan
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- The First Faculty of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhixue Wang
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ning Yang
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fenye Liu
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Fenye Liu,
| |
Collapse
|
108
|
Survival-related indicators ALOX12B and SPRR1A are associated with DNA damage repair and tumor microenvironment status in HPV 16-negative head and neck squamous cell carcinoma patients. BMC Cancer 2022; 22:714. [PMID: 35768785 PMCID: PMC9241267 DOI: 10.1186/s12885-022-09722-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES To investigate prognostic-related gene signature based on DNA damage repair and tumor microenvironment statue in human papillomavirus 16 negative (HPV16-) head and neck squamous cell carcinoma (HNSCC). METHODS For the RNA-sequence matrix in HPV16- HNSCC in the Cancer Genome Atlas (TCGA) cohort, the DNA damage response (DDR) and tumor microenvironment (TM) status of each patient sample was estimated by using the ssGSEA algorithm. Through bioinformatics analysis in DDR_high/TM_high (n = 311) and DDR_high/TM_low (n = 53) groups, a survival-related gene signature was selected in the TCGA cohort. Two independent external validation cohorts (GSE65858 (n = 210) and GSE41613 (n = 97)) with HPV16- HNSCC patients validated the gene signature. Correlations among the clinical-related hub differentially expressed genes (DEGs) and infiltrated immunocytes were explored with the TIMER2.0 server. Drug screening based on hub DEGs was performed using the CellMiner and GSCALite databases. The loss-of-function studies were used to evaluate the effect of screened survival-related gene on the motility of HPV- HNSCC cells in vitro. RESULTS A high DDR level (P = 0.025) and low TM score (P = 0.012) were independent risk factors for HPV16- HNSCC. Downregulated expression of ALOX12B or SPRR1A was associated with poor survival rate and advanced cancer stages. The pathway enrichment analysis showed the DDR_high/TM_low samples were enriched in glycosphingolipid biosynthesis-lacto and neolacto series, glutathione metabolism, platinum drug resistance, and ferroptosis pathways, while the DDR_high/TM_low samples were enriched in Th17 cell differentiation, Neutrophil extracellular trap formation, PD - L1 expression and PD - 1 checkpoint pathway in cancer. Notably, the expression of ALOX12B and SPRR1A were negatively correlated with cancer-associated fibroblasts (CAFs) infiltration and CAFs downstream effectors. Sensitivity to specific chemotherapy regimens can be derived from gene expressions. In addition, ALOX12B and SPRR1A expression was associated with the mRNA expression of insulin like growth factor 1 receptor (IGF1R), AKT serine/threonine kinase 1 (AKT1), mammalian target of rapamycin (MTOR), and eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1) in HPV negative HNSCC. Down-regulation of ALOX12B promoted HPV- HNSCC cells migration and invasion in vitro. CONCLUSIONS ALOX12B and SPRR1A served as a gene signature for overall survival in HPV16- HNSCC patients, and correlated with the amount of infiltrated CAFs. The specific drug pattern was determined by the gene signature.
Collapse
|
109
|
Franzén AS, Raftery MJ, Pecher G. Implications for Immunotherapy of Breast Cancer by Understanding the Microenvironment of a Solid Tumor. Cancers (Basel) 2022; 14:3178. [PMID: 35804950 PMCID: PMC9264853 DOI: 10.3390/cancers14133178] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/17/2022] [Accepted: 06/25/2022] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is poorly immunogenic due to immunosuppressive mechanisms produced in part by the tumor microenvironment (TME). The TME is a peritumoral area containing significant quantities of (1) cancer-associated fibroblasts (CAF), (2) tumor-infiltrating lymphocytes (TIL) and (3) tumor-associated macrophages (TAM). This combination protects the tumor from effective immune responses. How these protective cell types are generated and how the changes in the developing tumor relate to these subsets is only partially understood. Immunotherapies targeting solid tumors have proven ineffective largely due to this protective TME barrier. Therefore, a better understanding of the interplay between the tumor, the tumor microenvironment and immune cells would both advance immunotherapeutic research and lead to more effective immunotherapies. This review will summarize the current understanding of the microenvironment of breast cancer giving implications for future immunotherapeutic strategies.
Collapse
Affiliation(s)
| | | | - Gabriele Pecher
- Competence Center of Immuno-Oncology and Translational Cell Therapy, Department of Hematology, Oncology and Tumorimmunology, CCM, Charité-Universitätsmedizin Berlin, Berlin Institute of Health @ Charité, 10117 Berlin, Germany; (A.S.F.); (M.J.R.)
| |
Collapse
|
110
|
Kraus FBT, Topalov NE, Deuster E, Hysenaj I, Mayr D, Chelariu-Raicu A, Beyer S, Kolben T, Burges A, Mahner S, Trillsch F, Jeschke U, Czogalla B. Expression pattern and prognostic potential of histamine receptors in epithelial ovarian cancer. J Cancer Res Clin Oncol 2022; 149:2501-2511. [PMID: 35751684 PMCID: PMC10129941 DOI: 10.1007/s00432-022-04114-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/06/2022] [Indexed: 10/17/2022]
Abstract
PURPOSE Despite recent advances in the treatment of ovarian cancer (OC), long-term remissions remain scarce. For a targeted approach, prognostic markers are indispensable for predicting survival and treatment response. Given their association with multiple hallmarks of cancer, histamine receptors (HR) are emerging as promising candidates. Here, we investigate their expression pattern and prognostic value in OC. METHODS Specimens of 156 epithelial OC patients were collected during cytoreductive surgery at the Department of Obstetrics and Gynecology, LMU, between 1990 and 2002 and combined in a tissue microarray. Immunohistochemical staining of the HR H1, H2, H3 and H4 was quantified by an immunoreactive score and linked with clinico-pathological data by Spearman's correlation. Via ROC curve analysis, optimal cut-off values for potential prognostic markers were defined. Overall survival (OS) was visualized in Kaplan-Maier curves and significances determined by log-rank testing. A Cox regression model was applied for multivariate analysis. RESULTS HR H3 and H4 expression was restricted to the cytosol of OC cells, while H1 was also present in the nucleus. A significant association between HR H1, H3 and H4 expression with several clinico-pathological parameters was revealed. In addition, HR H1 and H3 expression correlated positively, HR H4 expression negatively with OS. In addition, HR H3 was identified as independent prognostic marker for OS. HR H2 expression had no prognostic value. CONCLUSIONS HR H1, H3 and H4 could serve as potential predictors for OS of OC patients. Further research is warranted to elucidate their pathophysiologic role and their predictive and therapeutic potential in OC.
Collapse
Affiliation(s)
- Fabian B T Kraus
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany.
| | - Nicole E Topalov
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - E Deuster
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - I Hysenaj
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - D Mayr
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - A Chelariu-Raicu
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - S Beyer
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - T Kolben
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - A Burges
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - S Mahner
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - F Trillsch
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| | - U Jeschke
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany.,Department of Obstetrics and Gynecology, University Hospital Augsburg, Augsburg, Germany
| | - B Czogalla
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
111
|
Nadukkandy AS, Ganjoo E, Singh A, Dinesh Kumar L. Tracing New Landscapes in the Arena of Nanoparticle-Based Cancer Immunotherapy. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.911063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Over the past two decades, unique and comprehensive cancer treatment has ushered new hope in the holistic management of the disease. Cancer immunotherapy, which harnesses the immune system of the patient to attack the cancer cells in a targeted manner, scores over others by being less debilitating compared to the existing treatment strategies. Significant advancements in the knowledge of immune surveillance in the last few decades have led to the development of several types of immune therapy like monoclonal antibodies, cancer vaccines, immune checkpoint inhibitors, T-cell transfer therapy or adoptive cell therapy (ACT) and immune system modulators. Intensive research has established cancer immunotherapy to be a safe and effective method for improving survival and the quality of a patient’s life. However, numerous issues with respect to site-specific delivery, resistance to immunotherapy, and escape of cancer cells from immune responses, need to be addressed for expanding and utilizing this therapy as a regular mode in the clinical treatment. Development in the field of nanotechnology has augmented the therapeutic efficiency of treatment modalities of immunotherapy. Nanocarriers could be used as vehicles because of their advantages such as increased surface areas, targeted delivery, controlled surface and release chemistry, enhanced permeation and retention effect, etc. They could enhance the function of immune cells by incorporating immunomodulatory agents that influence the tumor microenvironment, thus enabling antitumor immunity. Robust validation of the combined effect of nanotechnology and immunotherapy techniques in the clinics has paved the way for a better treatment option for cancer than the already existing procedures such as chemotherapy and radiotherapy. In this review, we discuss the current applications of nanoparticles in the development of ‘smart’ cancer immunotherapeutic agents like ACT, cancer vaccines, monoclonal antibodies, their site-specific delivery, and modulation of other endogenous immune cells. We also highlight the immense possibilities of using nanotechnology to accomplish leveraging the coordinated and adaptive immune system of a patient to tackle the complexity of treating unique disease conditions and provide future prospects in the field of cancer immunotherapy.
Collapse
|
112
|
Su K, Peng Y, Yu H. Development of a Prognostic Model Based on Pyroptosis-Related Genes in Pancreatic Adenocarcinoma. DISEASE MARKERS 2022; 2022:9141117. [PMID: 35677632 PMCID: PMC9169203 DOI: 10.1155/2022/9141117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022]
Abstract
Background The importance of pyroptosis in tumorigenesis and cancer progression is becoming increasingly apparent. However, the efficacy of using pyroptosis-related genes (PRGs) in predicting the prognosis of pancreatic adenocarcinoma (PAAD) patients is unknown. Methods This investigation used two databases to obtain expression data for PAAD patients. Differentially expressed PRGs (DEPRGs) were identified between PAAD and control samples. Several bioinformatic approaches were used to analyze the biological functions of DEPRGs and to identify prognostic DERPGs. A miRNA-prognostic DEPRG-transcription factor (TF) regulatory network was created via the miRNet online tool. A risk score model was created after each patient's risk score was calculated. The microenvironments of the low- and high-risk groups were assessed using xCell, the expression of immune checkpoints was determined, and gene set variation analysis (GSVA) was performed. Finally, the efficacy of certain potential drugs was predicted using the pRRophetic algorithm, and the results in the high- and low-risk groups were compared. Results A total of 13 DEPRGs were identified between PAAD and control samples. Functional enrichment analysis showed that the DEPRGs had a close relationship with inflammation. In univariate and multivariate Cox regression analyses, GSDMC, IRF1, and PLCG1 were identified as prognostic biomarkers in PAAD. The results of the miRNA-prognostic DEPRG-TF regulatory network showed that GSDMC, IRF1, and PLCG1 were regulated by both specific and common miRNAs and TFs. Based on the risk score and other independent prognostic indicators, a nomogram with a good ability to predict the survival of PAAD patients was developed. By evaluating the tumor microenvironment, we observed that the immune and metabolic microenvironments of the two groups were substantially different. In addition, individuals in the low-risk group were more susceptible to axitinib and camptothecin, whereas lapatinib might be preferred for patients in the high-risk group. Conclusion Our study revealed the prognostic value of PRGs in PAAD and created a reliable model for predicting the prognosis of PAAD patients. Our findings will benefit the prognostication and treatment of PAAD patients.
Collapse
Affiliation(s)
- Kaifeng Su
- Medical Faculty of Ludwig-Maximilians-University of Munich, University Hospital of LMU Munich, Munich, Germany
| | - Yang Peng
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haochen Yu
- Medical Faculty of Ludwig-Maximilians-University of Munich, University Hospital of LMU Munich, Munich, Germany
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
113
|
Huang S, Liao M, Chen S, Zhang P, Xu F, Zhang H. Immune signatures of CD4 and CD68 predicts disease progression in cutaneous T cell lymphoma. Am J Transl Res 2022; 14:3037-3051. [PMID: 35702080 PMCID: PMC9185059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/12/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Cutaneous T-cell lymphoma (CTCL) is highly heterogeneous, and its prognosis is closely related to the disease stage. The tumor microenvironment (TME) is an important component of tumor tissue, driving cancer cell growth, progression, and metastasis. However, the diagnostic value of TME in CTCL has not yet been studied in-depth. To date, no study has performed a comprehensive evaluation of the significance of the TME in CTCL. METHODS Using xCell methods based on bulk RNA sequencing data, we inferred immune cell fraction in the TME in 126 patients and assessed the prognostic importance of immune cells. Consensus clustering was performed to determine the TME subtypes and characterize the transcriptome of each subtype. Based on the TME subtypes, we established the disease progression model using random forest algorithms and logistic regression. The efficacy of the model was examined using an additional 49-patient cohort. Finally, we validated our finding at the protein level using immunochemistry in a 16-patient cohort. RESULTS Patients with advanced CTCL presented with a more active immunity overall than those with early stage. Random forest algorithms revealed that the immune cells CD4, macrophages, and dendritic cells (DCs) were the most effective prognosis predictors. Therefore, we constructed a risk model using logistic regression based on these immune cells. The TME score could be used to effectively predict disease conditions in three datasets with the AUC of 0.9414, 0.7912, and 0.7665, respectively. Immunochemistry at the protein level revealed that helper T cells and the macrophage markers CD4 and CD68 could successfully distinguish different CTCL stages in patients, whereas the DC marker langerin showed no change with disease progression. CONCLUSION We found advanced-stage CTCL was associated with an active immune microenvironment, and the immune signatures CD4 and CD68 showed a relatively high accuracy in predicting CTCL disease progression.
Collapse
Affiliation(s)
- Sanling Huang
- Department of Hematology, Peking University Shenzhen HospitalShenzhen 518000, Guangdong, P. R. China
| | - Mengying Liao
- Department of Pathology, Peking University Shenzhen HospitalShenzhen 518000, Guangdong, P. R. China
| | - Siliang Chen
- Department of Hematology, Peking University Shenzhen HospitalShenzhen 518000, Guangdong, P. R. China
| | - Ping Zhang
- Department of Hematology, Peking University Shenzhen HospitalShenzhen 518000, Guangdong, P. R. China
| | - Fangzhou Xu
- Department of Hematology, Peking University Shenzhen HospitalShenzhen 518000, Guangdong, P. R. China
- The Clinical Trail Institute, Peking University Shenzhen HospitalShenzhen 518000, Guangdong, P. R. China
| | - Hongyu Zhang
- Department of Hematology, Peking University Shenzhen HospitalShenzhen 518000, Guangdong, P. R. China
- The Clinical Trail Institute, Peking University Shenzhen HospitalShenzhen 518000, Guangdong, P. R. China
| |
Collapse
|
114
|
Salerno S, Ståhlberg A, Holdfeldt A, Bexe Lindskog E, Landberg G. 5-fluorouracil treatment of patient-derived scaffolds from colorectal cancer reveal clinically critical information. J Transl Med 2022; 20:209. [PMID: 35562738 PMCID: PMC9102939 DOI: 10.1186/s12967-022-03423-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Colorectal cancer is a commonly diagnosed cancer worldwide. Unfortunately, many patients do not respond to standard chemotherapy treatments and develop disease relapse and metastases. Besides cancer cell specific genetic changes, heterogeneity in the tumor microenvironment contribute to the clinical presentation of the disease and can potentially also influence drug resistance. By using a recently developed patient-derived scaffold method monitoring how a standardized reporter cancer cell line adapts to various microenvironments treated with chemotherapy, we wanted to clarify how individual patient specific microenvironments influence the chemotherapy response in colorectal cancer. METHODS Surgically resected colorectal cancer specimens from 89 patients were decellularized to produce patient-derived scaffold, which were seeded with HT29 cells, cultured for 3 weeks, and treated with 5-fluorouracil. Gene expression changes of adapted and treated HT29 cells were monitored by qPCR and compared with clinical parameters including disease-free survival. RESULTS The effects of 5-fluorouracil treatment varied between different patient-derived scaffold, but generally induced a reduced expression of proliferation genes and increased expression of pluripotency and epithelial-to-mesenchymal transition genes. Interestingly, patient-derived scaffold cultures obtained from patients with disease recurrences showed a significantly less pronounced anti-proliferative effect of 5-fluorouracil and more pronounced increase of pluripotency, with MKI67 and POU5F1 being among the most significant genes linked to disease relapse in colorectal cancer. CONCLUSIONS Colorectal patient-derived scaffold can decode clinically relevant tumor microenvironmental influence of 5-fluorouracil treatment effects opening up for optimized precision medicine in colorectal cancer treatment.
Collapse
Affiliation(s)
- Simona Salerno
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - André Holdfeldt
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Elinor Bexe Lindskog
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Surgical Oncology Laboratory, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Göran Landberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden. .,Department of Pathology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
115
|
Pece R, Tavella S, Costa D, Varesano S, Camodeca C, Cuffaro D, Nuti E, Rossello A, Alfano M, D'Arrigo C, Galante D, Ravetti JL, Gobbi M, Tosetti F, Poggi A, Zocchi MR. Inhibitors of ADAM10 reduce Hodgkin lymphoma cell growth in 3D microenvironments and enhance brentuximab-vedotin effect. Haematologica 2022; 107:909-920. [PMID: 34109776 PMCID: PMC8968898 DOI: 10.3324/haematol.2021.278469] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/28/2021] [Indexed: 11/13/2022] Open
Abstract
Shedding of ADAM10 substrates, like TNFa or CD30, can affect both anti-tumor immune response and antibody-drug-conjugate (ADC)-based immunotherapy. We have published two new ADAM10 inhibitors, LT4 and MN8 able to prevent such shedding in Hodgkin lymphoma (HL). Since tumor tissue architecture deeply influences the outcome of anti-cancer treatments, we set up a new threedimensional (3D) culture systems to verify whether ADAM10 inhibitors can contribute to, or enhance, the anti-lymphoma effects of the ADC brentuximab-vedotin (BtxVed). In order to recapitulate some aspects of lymphoma structure and architecture, we assembled two 3D culture models: mixed spheroids made of HL lymph node (LN) mesenchymal stromal cells (MSC) and Reed Sternberg/Hodgkin lymphoma cells (HL cells) or collagen scaffolds repopulated with LN-MSC and HL cells. In these 3D systems we found that: i) the ADAM10 inhibitors LT4 and MN8 reduce ATP content or glucose consumption, related to cell proliferation, increasing lactate dehydrogenase release as a cell damage hallmark; ii) these events are paralleled by mixed spheroids size reduction and inhibition of CD30 and TNFa shedding; iii) the effects observed can be reproduced in repopulated HL LN-derived matrix or collagen scaffolds; iv) ADAM10 inhibitors enhance the anti-lymphoma effect of the anti-CD30 ADC BtxVed both in conventional cultures and in repopulated scaffolds. Thus, we provide evidence for a direct and combined antilymphoma effect of ADAM10 inhibitors with BtxVed, leading to the improvement of ADC effects; this is documented in 3D models recapitulating features of the LN microenvironment, that can be proposed as a reliable tool for anti-lymphoma drug testing.
Collapse
Affiliation(s)
- Roberta Pece
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino and Department of Experimental Medicine, University of Genoa
| | - Sara Tavella
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino and Department of Experimental Medicine, University of Genoa
| | - Delfina Costa
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino
| | - Serena Varesano
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino
| | | | | | - Elisa Nuti
- Department of Pharmacy, University of Pisa
| | | | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele
| | | | | | | | - Marco Gobbi
- Clinical Oncohematology, University of Genoa
| | - Francesca Tosetti
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, IRCCS San Raffaele Scientific Institute.
| |
Collapse
|
116
|
Zhang X, Zheng S, Hu C, Li G, Lin H, Xia R, Ye Y, He R, Li Z, Lin Q, Chen R, Zhou Q. Cancer-associated fibroblast-induced lncRNA UPK1A-AS1 confers platinum resistance in pancreatic cancer via efficient double-strand break repair. Oncogene 2022; 41:2372-2389. [PMID: 35264742 PMCID: PMC9010302 DOI: 10.1038/s41388-022-02253-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/06/2022] [Accepted: 02/16/2022] [Indexed: 12/27/2022]
Abstract
The tumor stroma of pancreatic ductal adenocarcinoma (PDAC) is characterized by an abundant and heterogeneous population of cancer-associated fibroblasts (CAFs), which are critically involved in chemoresistance. However, the underlying mechanism of CAFs in chemoresistance is unclear. Here, we show that CAFR, a CAF subset derived from platinum-resistant PDAC patients, assumes an iCAF phenotype and produces more IL8 than CAFS isolated from platinum-sensitive PDAC patients. CAFR-derived IL8 promotes oxaliplatin chemoresistance in PDAC. Based on long noncoding RNA (lncRNA) profiling in tumor cells incubated with CAF-CM, we found that UPK1A-AS1, whose expression is directly induced by IL8/NF-kappa B signaling, functions as a chemoresistance-promoting lncRNA and is critical for active IL8-induced oxaliplatin resistance. Impressively, blocking the activation of UPK1A-AS1 expression increases the oxaliplatin sensitivity of tumor cells in vivo. Mechanistically, UPK1A-AS1 strengthens the interaction between Ku70 and Ku80 to facilitate nonhomologous end joining (NHEJ), thereby enhancing DNA double-strand break (DSB) repair. Clinically, UPK1A-AS1 expression is positively correlated with IL8 expression, a poor chemotherapeutic response and a shorter progression-free survival (PFS) time in advanced PDAC patients. Collectively, our study reveals a lncRNA-mediated mechanism of CAF-derived paracrine IL8-dependent oxaliplatin resistance and highlights UPK1A-AS1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Xiang Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, People's Republic of China
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Shangyou Zheng
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Chonghui Hu
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China
- Guangdong cardiovascular Institute, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Guolin Li
- Department of Hepatobiliary, Pancreatic and Splenic surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, Guangdong, People's Republic of China
| | - Hongcao Lin
- General Surgery of Shenshan Medical Center, Memorial Hospital of Sun Yat-sen University, Shanwei, 516600, Guangdong, People's Republic of China
| | - Renpeng Xia
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China
- Department of Neonatal/General Surgery, Hunan Children's Hospital, Changsha, 410007, Hunan, People's Republic of China
| | - Yuancheng Ye
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Rihua He
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Zhihua Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, People's Republic of China
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Qing Lin
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
- School of medicine, South China University of Technology, Guangzhou, 510006, Guangdong, People's Republic of China.
| | - Rufu Chen
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
- Guangdong cardiovascular Institute, Guangzhou, 510080, Guangdong, People's Republic of China.
- School of medicine, South China University of Technology, Guangzhou, 510006, Guangdong, People's Republic of China.
| | - Quanbo Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, People's Republic of China.
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China.
| |
Collapse
|
117
|
Peña-Romero AC, Orenes-Piñero E. Dual Effect of Immune Cells within Tumour Microenvironment: Pro- and Anti-Tumour Effects and Their Triggers. Cancers (Basel) 2022; 14:1681. [PMID: 35406451 PMCID: PMC8996887 DOI: 10.3390/cancers14071681] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Our body is constantly exposed to pathogens or external threats, but with the immune response that our body can develop, we can fight off and defeat possible attacks or infections. Nevertheless, sometimes this threat comes from an internal factor. Situations such as the existence of a tumour also cause our immune system (IS) to be put on alert. Indeed, the link between immunology and cancer is evident these days, with IS being used as one of the important targets for treating cancer. Our IS is able to eliminate those abnormal or damaged cells found in our body, preventing the uncontrolled proliferation of tumour cells that can lead to cancer. However, in several cases, tumour cells can escape from the IS. It has been observed that immune cells, the extracellular matrix, blood vessels, fat cells and various molecules could support tumour growth and development. Thus, the developing tumour receives structural support, irrigation and energy, among other resources, making its survival and progression possible. All these components that accompany and help the tumour to survive and to grow are called the tumour microenvironment (TME). Given the importance of its presence in the tumour development process, this review will focus on one of the components of the TME: immune cells. Immune cells can support anti-tumour immune response protecting us against tumour cells; nevertheless, they can also behave as pro-tumoural cells, thus promoting tumour progression and survival. In this review, the anti-tumour and pro-tumour immunity of several immune cells will be discussed. In addition, the TME influence on this dual effect will be also analysed.
Collapse
Affiliation(s)
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, 30120 Murcia, Spain;
| |
Collapse
|
118
|
Dacheux MA, Lee SC, Shin Y, Norman DD, Lin KH, E S, Yue J, Benyó Z, Tigyi GJ. Prometastatic Effect of ATX Derived from Alveolar Type II Pneumocytes and B16-F10 Melanoma Cells. Cancers (Basel) 2022; 14:cancers14061586. [PMID: 35326737 PMCID: PMC8946623 DOI: 10.3390/cancers14061586] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/07/2022] [Accepted: 03/17/2022] [Indexed: 01/27/2023] Open
Abstract
Although metastases are the principal cause of cancer-related deaths, the molecular aspects of the role of stromal cells in the establishment of the metastatic niche remain poorly understood. One of the most prevalent sites for cancer metastasis is the lungs. According to recent research, lung stromal cells such as bronchial epithelial cells and resident macrophages secrete autotaxin (ATX), an enzyme with lysophospholipase D activity that promotes cancer progression. In fact, several studies have shown that many cell types in the lung stroma could provide a rich source of ATX in diseases. In the present study, we sought to determine whether ATX derived from alveolar type II epithelial (ATII) pneumocytes could modulate the progression of lung metastasis, which has not been evaluated previously. To accomplish this, we used the B16-F10 syngeneic melanoma model, which readily metastasizes to the lungs when injected intravenously. Because B16-F10 cells express high levels of ATX, we used the CRISPR-Cas9 technology to knock out the ATX gene in B16-F10 cells, eliminating the contribution of tumor-derived ATX in lung metastasis. Next, we used the inducible Cre/loxP system (Sftpc-CreERT2/Enpp2fl/fl) to generate conditional knockout (KO) mice in which ATX is specifically deleted in ATII cells (i.e., Sftpc-KO). Injection of ATX-KO B16-F10 cells into Sftpc-KO or Sftpc-WT control littermates allowed us to investigate the specific contribution of ATII-derived ATX in lung metastasis. We found that targeted KO of ATX in ATII cells significantly reduced the metastatic burden of ATX-KO B16-F10 cells by 30% (unpaired t-test, p = 0.028) compared to Sftpc-WT control mice, suggesting that ATX derived from ATII cells could affect the metastatic progression. We detected upregulated levels of cytokines such as IFNγ (unpaired t-test, p < 0.0001) and TNFα (unpaired t-test, p = 0.0003), which could favor the increase in infiltrating CD8+ T cells observed in the tumor regions of Sftpc-KO mice. Taken together, our results highlight the contribution of host ATII cells as a stromal source of ATX in the progression of melanoma lung metastasis.
Collapse
Affiliation(s)
- Mélanie A. Dacheux
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; (M.A.D.); (S.C.L.); (Y.S.); (D.D.N.); (K.-H.L.)
| | - Sue Chin Lee
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; (M.A.D.); (S.C.L.); (Y.S.); (D.D.N.); (K.-H.L.)
| | - Yoojin Shin
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; (M.A.D.); (S.C.L.); (Y.S.); (D.D.N.); (K.-H.L.)
| | - Derek D. Norman
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; (M.A.D.); (S.C.L.); (Y.S.); (D.D.N.); (K.-H.L.)
| | - Kuan-Hung Lin
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; (M.A.D.); (S.C.L.); (Y.S.); (D.D.N.); (K.-H.L.)
| | - Shuyu E
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; (S.E.); (J.Y.)
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; (S.E.); (J.Y.)
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, H-1428 Budapest, Hungary;
| | - Gábor J. Tigyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; (M.A.D.); (S.C.L.); (Y.S.); (D.D.N.); (K.-H.L.)
- Correspondence: ; Tel.: +1-901-448-4793
| |
Collapse
|
119
|
Qi C, Lei L, Hu J, Wang G, Liu J, Ou S. Identification of a five-gene signature deriving from the vacuolar ATPase (V-ATPase) sub-classifies gliomas and decides prognoses and immune microenvironment alterations. Cell Cycle 2022; 21:1294-1315. [PMID: 35266851 PMCID: PMC9132400 DOI: 10.1080/15384101.2022.2049157] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aberrant expression of coding genes of the V-ATPase subunits has been reported in glioma patients that can activate oncogenic pathways and result in worse prognosis. However, the predictive effect of a single gene is not specific or sensitive enough. In this study, by using a series of bioinformatics analyses, we identified five coding genes (ATP6V1C2, ATP6V1G2, TCIRG1, ATP6AP1 and ATP6AP2) of the V-ATPase that were related to glioma patient prognosis. Based on the expression of these genes, glioma patients were sub-classified into different prognosis clusters, of which C1 cluster performed better prognosis; however, C2 cluster showed more malignant phenotypes with oncogenic and immune-related pathway activation. The single-cell RNA-seq data revealed that ATP6AP1, ATP6AP2, ATP6V1G2 and TCIRG1 might be cell-type potential markers. Copy number variation and DNA promoter methylation potentially regulate these five gene expressions. A risk score model consisted of these five genes effectively predicted glioma prognosis and was fully validated by six independent datasets. The risk scores also showed a positive correlation with immune checkpoint expression. Importantly, glioma patients with high-risk scores presented resistance to traditional treatment. We also revealed that more inhibitory immune cells infiltration and higher rates of “non-response” to immune checkpoint blockade (ICB) treatment in the high-risk score group. In conclusion, our study identified a five-gene signature from the V-ATPase that could sub-classify gliomas into different phenotypes and their abnormal expression was regulated by distinct mechanisms and accompanied with immune microenvironment alterations potentially act as a biomarker for ICB treatment.
Collapse
Affiliation(s)
- Chunxiao Qi
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Lei Lei
- Department of Rheumatology and Immunology, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, Liaoning, China
| | - Jinqu Hu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Gang Wang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiyuan Liu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shaowu Ou
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
120
|
Shahzad MH, Feng L, Su X, Brassard A, Dhoparee-Doomah I, Ferri LE, Spicer JD, Cools-Lartigue JJ. Neutrophil Extracellular Traps in Cancer Therapy Resistance. Cancers (Basel) 2022; 14:1359. [PMID: 35267667 PMCID: PMC8909607 DOI: 10.3390/cancers14051359] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 02/04/2023] Open
Abstract
Neutrophils and their products are increasingly recognized to have a key influence on cancer progression and response to therapy. Their involvement has been shown in nearly every aspect of cancer pathophysiology with growing evidence now supporting their role in resistance to a variety of cancer therapies. Recently, the role of neutrophils in cancer progression and therapy resistance has been further complicated with the discovery of neutrophil extracellular traps (NETs). NETs are web-like structures of chromatin decorated with a variety of microbicidal proteins. They are released by neutrophils in a process called NETosis. NET-dependent mechanisms of cancer pathology are beginning to be appreciated, particularly with respect to tumor response to chemo-, immuno-, and radiation therapy. Several studies support the functional role of NETs in cancer therapy resistance, involving T-cell exhaustion, drug detoxification, angiogenesis, the epithelial-to-mesenchymal transition, and extracellular matrix remodeling mechanisms, among others. Given this, new and promising data suggests NETs provide a microenvironment conducive to limited therapeutic response across a variety of neoplasms. As such, this paper aims to give a comprehensive overview of evidence on NETs in cancer therapy resistance with a focus on clinical applicability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jonathan J. Cools-Lartigue
- Department of Surgery, Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, Montreal, QC H3G 1A4, Canada; (M.H.S.); (L.F.); (X.S.); (A.B.); (I.D.-D.); (L.E.F.); (J.D.S.)
| |
Collapse
|
121
|
Zhu L, Retana D, García‐Gómez P, Álvaro‐Espinosa L, Priego N, Masmudi‐Martín M, Yebra N, Miarka L, Hernández‐Encinas E, Blanco‐Aparicio C, Martínez S, Sobrino C, Ajenjo N, Artiga M, Ortega‐Paino E, Torres‐Ruiz R, Rodríguez‐Perales S, Soffietti R, Bertero L, Cassoni P, Weiss T, Muñoz J, Sepúlveda JM, González‐León P, Jiménez‐Roldán L, Moreno LM, Esteban O, Pérez‐Núñez Á, Hernández‐Laín A, Toldos O, Ruano Y, Alcázar L, Blasco G, Fernández‐Alén J, Caleiras E, Lafarga M, Megías D, Graña‐Castro O, Nör C, Taylor MD, Young LS, Varešlija D, Cosgrove N, Couch FJ, Cussó L, Desco M, Mouron S, Quintela‐Fandino M, Weller M, Pastor J, Valiente M. A clinically compatible drug-screening platform based on organotypic cultures identifies vulnerabilities to prevent and treat brain metastasis. EMBO Mol Med 2022; 14:e14552. [PMID: 35174975 PMCID: PMC8899920 DOI: 10.15252/emmm.202114552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 12/22/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
We report a medium-throughput drug-screening platform (METPlatform) based on organotypic cultures that allows to evaluate inhibitors against metastases growing in situ. By applying this approach to the unmet clinical need of brain metastasis, we identified several vulnerabilities. Among them, a blood-brain barrier permeable HSP90 inhibitor showed high potency against mouse and human brain metastases at clinically relevant stages of the disease, including a novel model of local relapse after neurosurgery. Furthermore, in situ proteomic analysis applied to metastases treated with the chaperone inhibitor uncovered a novel molecular program in brain metastasis, which includes biomarkers of poor prognosis and actionable mechanisms of resistance. Our work validates METPlatform as a potent resource for metastasis research integrating drug-screening and unbiased omic approaches that is compatible with human samples. Thus, this clinically relevant strategy is aimed to personalize the management of metastatic disease in the brain and elsewhere.
Collapse
Affiliation(s)
- Lucía Zhu
- Brain Metastasis GroupCNIOMadridSpain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Raúl Torres‐Ruiz
- Molecular Cytogenetics UnitCNIOMadridSpain,Division of Hematopoietic Innovative TherapiesCentro de Investigaciones EnergeticasMedioambientales y Tecnologicas (CIEMAT)MadridSpain
| | | | | | - Riccardo Soffietti
- Department of Neuro‐OncologyUniversity and City of Health and Science HospitalTurinItaly
| | - Luca Bertero
- Department of Medical SciencesUniversity of TurinTurinItaly
| | - Paola Cassoni
- Department of Medical SciencesUniversity of TurinTurinItaly
| | - Tobias Weiss
- Department of NeurologyClinical Neuroscience CenterUniversity Hospital Zurich and University of ZurichZurichSwitzerland
| | - Javier Muñoz
- Proteomics UnitProteoRedISCIIICNIOMadridSpain,Present address:
Cell Signaling and Clinical Proteomics GroupBiocruces Bizkaia Health Research InstituteBarakaldoSpain,Present address:
IkerbasqueBasque Foundation for ScienceBilbaoSpain
| | | | | | - Luis Jiménez‐Roldán
- Neurosurgery UnitHospital Universitario 12 de OctubreMadridSpain,Department of SurgeryUniversidad Complutense de MadridMadridSpain,Neuropathology UnitInstituto i+12, Hospital Universitario 12 de OctubreMadridSpain
| | | | - Olga Esteban
- Neurosurgery UnitHospital Universitario 12 de OctubreMadridSpain
| | - Ángel Pérez‐Núñez
- Neurosurgery UnitHospital Universitario 12 de OctubreMadridSpain,Department of SurgeryUniversidad Complutense de MadridMadridSpain,Neuro‐Oncology GroupResearch Institute Hospital 12 de Octubre (i+12)MadridSpain
| | | | - Oscar Toldos
- Neuropathology UnitInstituto i+12, Hospital Universitario 12 de OctubreMadridSpain
| | - Yolanda Ruano
- Pathology DepartmentInstituto i+12, Hospital Universitario 12 de OctubreMadridSpain,Universidad Francisco de VitoriaMadridSpain
| | - Lucía Alcázar
- Neurosurgery DepartmentHospital Universitario de La PrincesaMadridSpain
| | - Guillermo Blasco
- Neurosurgery DepartmentHospital Universitario de La PrincesaMadridSpain
| | | | | | - Miguel Lafarga
- Department of Anatomy and Cell Biology and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)University of Cantabria‐IDIVALSantanderSpain
| | | | | | - Carolina Nör
- Developmental and Stem Cell Biology Program and The Arthur and Sonia Labatt Brain Tumour Research CentreThe Hospital for Sick ChildrenTorontoONCanada
| | - Michael D Taylor
- Developmental and Stem Cell Biology Program and The Arthur and Sonia Labatt Brain Tumour Research CentreThe Hospital for Sick ChildrenTorontoONCanada
| | - Leonie S Young
- Endocrine Oncology Research GroupDepartment of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Damir Varešlija
- Endocrine Oncology Research GroupDepartment of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Nicola Cosgrove
- Endocrine Oncology Research GroupDepartment of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Fergus J Couch
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | - Lorena Cussó
- Departamento de Bioingeniería e Ingeniería AeroespacialUniversidad Carlos III de MadridMadridSpain,Instituto de Investigación Sanitaria Gregorio MarañónMadridSpain,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain,Unidad de Imagen AvanzadaCentro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Manuel Desco
- Departamento de Bioingeniería e Ingeniería AeroespacialUniversidad Carlos III de MadridMadridSpain,Instituto de Investigación Sanitaria Gregorio MarañónMadridSpain,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)MadridSpain,Unidad de Imagen AvanzadaCentro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | | | | | - Michael Weller
- Department of NeurologyClinical Neuroscience CenterUniversity Hospital Zurich and University of ZurichZurichSwitzerland
| | | | | |
Collapse
|
122
|
Nemes K, Johann PD, Tüchert S, Melchior P, Vokuhl C, Siebert R, Furtwängler R, Frühwald MC. Current and Emerging Therapeutic Approaches for Extracranial Malignant Rhabdoid Tumors. Cancer Manag Res 2022; 14:479-498. [PMID: 35173482 PMCID: PMC8841298 DOI: 10.2147/cmar.s289544] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Extracranial malignant rhabdoid tumors (extracranial MRT) are rare, highly aggressive malignancies affecting mainly infants and children younger than 3 years. Common anatomic sites comprise the kidneys (RTK – rhabdoid tumor of kidney) and other soft tissues (eMRT – extracranial, extrarenal malignant rhabdoid tumor). The genetic origin of these diseases is linked to biallelic pathogenic variants in the genes SMARCB1, or rarely SMARCA4, encoding subunits of the SWI/SNF chromatin-remodeling complex. Even if extracranial MRT seem to be quite homogeneous, recent epigenome analyses reveal a certain degree of epigenetic heterogeneity. Use of intensified therapies has modestly improved survival for extracranial MRT. Patients at standard risk profit from conventional therapies; most high-risk patients still experience a dismal course and often therapy resistance. Discoveries of clinical and molecular hallmarks and the exploration of experimental therapeutic approaches open exciting perspectives for clinical and molecularly stratified experimental treatment approaches. To ultimately improve the outcome of patients with extracranial MRTs, they need to be characterized and stratified clinically and molecularly. High-risk patients need novel therapeutic approaches including selective experimental agents in phase I/II clinical trials.
Collapse
Affiliation(s)
- Karolina Nemes
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany
| | - Pascal D Johann
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany.,Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefanie Tüchert
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Augsburg, Germany
| | - Patrick Melchior
- Department of Radiation Oncology, University of Saarland, Homburg, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm, Germany
| | - Rhoikos Furtwängler
- Department of Pediatric Hematology and Oncology, University of Saarland, Homburg, Germany
| | - Michael C Frühwald
- Paediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg, Germany
| |
Collapse
|
123
|
Qin M, Ma Y, Wang Z, Fang D, Wei J. Using immune-related lncRNAs to construct novel biomarkers and investigate the immune landscape of breast cancer. Transl Cancer Res 2022; 10:2991-3003. [PMID: 35116607 PMCID: PMC8799245 DOI: 10.21037/tcr-21-783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/26/2021] [Indexed: 01/10/2023]
Abstract
Background The role of immune-related long noncoding RNAs (irlncRNAs) in breast cancer (BRCA) is still unclear. Recently, studies have performed analyses based on the expression of irlncRNAs, however, in the present study, we used a novel method that did not require the specific expression levels of lncRNAs of BRCA patients. Methods We downloaded transcriptome and clinical data of BRCA patients from The Cancer Genome Atlas (TCGA), obtained immune genes from the Immport database, and extracted immune genes and lncRNAs for correlation analysis. Then, the differential expression of irlncRNA pairs (IRLPs) was determined and the prognostic signature was established by the IRLPs. The immune cell abundance of the TCGA-BRCA cohort was downloaded from the Tumor IMmune Estimation Resource (TIMER) database, and the relationship between the risk score of the IRLP signature and immune cell abundance was analyzed. Finally, we explored the relationship between risk scores and drug sensitivity based on the R package pRRophetic. Results Univariate cox regression results showed that 33 IRLPs had significant effects on the overall survival (OS) of BRCA patients. Then 22 IRLPs were obtained via lasso regression for further analysis. Multivariate regression analysis obtained 12 IRLPs to establish the IRLP prognostic signature. The model showed that this IRLP signature could act as a prognostic biomarker for BRCA patients. Kaplan-Meier (KM) survival analysis indicated that low-risk patients of IRLP’s signature had a better OS (P<0.001). Advanced status BRCA patients may have higher risk scores, and univariate and multivariate cox regression analyses showed that risk scores were independent prognostic factors of clinical features (P<0.001). The results of the relationship between risk scores and immune infiltration showed that M1 macrophages were higher in the low-risk group (P=0.00015), while M2 macrophages were higher in the high-risk group (P=0.0015). The high-risk group had a greater sensitivity to chemotherapeutic agents such as cisplatin, docetaxel, doxorubicin, and gemcitabine. Conclusions In present study, we used a novel method that did not require the specific expression levels of lncRNAs of BRCA patients, which can be used as a novel model for predicting the prognosis of BRCA patients.
Collapse
Affiliation(s)
- Muping Qin
- Department of Hematology, Baise People's Hospital, Baise, China.,Department of Oncology, Wuzhou Red Cross Hospital, Wuzhou, China
| | - Yanfei Ma
- Department of Breast and Thyroid Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zifan Wang
- Department of Breast and Thyroid Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.,Department of Burn Plastic and Wound Repair, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Dalang Fang
- Department of Breast and Thyroid Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jie Wei
- Department of Hematology, Baise People's Hospital, Baise, China
| |
Collapse
|
124
|
Chen H, Yang Y, Deng Y, Wei F, Zhao Q, Liu Y, Liu Z, Yu B, Huang Z. Delivery of CD47 blocker SIRPα-Fc by CAR-T cells enhances antitumor efficacy. J Immunother Cancer 2022; 10:jitc-2021-003737. [PMID: 35110357 PMCID: PMC8811602 DOI: 10.1136/jitc-2021-003737] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2022] [Indexed: 12/14/2022] Open
Abstract
Background Chimeric antigen receptor (CAR) T cell therapy has been successfully applied in treating lymphoma malignancies, but not in solid tumors. CD47 is highly expressed on tumor cells and its overexpression is believed to inhibit phagocytosis by macrophages and dendritic cells. Given the antitumor activity against preclinical model of CD47-blocking to induce the innate and adaptive immune system in the tumor microenvironment, here we developed a CAR-T cell secreting CD47 blocker signal regulatory protein α (SIRPα)-Fc fusion protein (Sirf CAR-T) to boost CAR-T cell therapeutic effect in solid tumor therapy. Methods Murine T cells were transduced to express a conventional anti-Trop2 CAR and Sirf CAR. The expression of SIRPα-Fc fusion protein in the supernatant of CAR-T cells and its effect on macrophage phagocytosis were tested in vitro. In vivo antitumor efficacy of CAR-T cells was evaluated in immunocompetent mice and analysis of the tumor microenvironment in the tumor-bearing mice was performed. Results We found that Sirf CAR-T cells dramatically decreased tumor burden and significantly prolonged survival in several syngeneic immunocompetent tumor models. Furthermore, we found that Sirf CAR-T cells induced more central memory T cells (TCM) and improved the persistence of CAR-T cells in tumor tissue, as well as decreased PD-1 expression on the CAR-T cell surface. In addition, we demonstrated that Sirf CAR-T cells could modulate the tumor microenvironment by decreasing myeloid-derived stem cells as well as increasing CD11c+ dendritic cells and M1-type macrophages in tumor tissue. Conclusions In summary, our findings indicate that CD47 blocker SIRPα-Fc enhances the antitumor efficacy of CAR-T cells and propose to block CD47/SIRPα signaling effect on CAR-T cells function, which could provide a new strategy for successful cancer immunotherapy by rationalizing combination of CD47 blocker and CAR-T cell therapy.
Collapse
Affiliation(s)
- Huanpeng Chen
- BioResource Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Institute of Human Virology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Yuying Yang
- Institute of Human Virology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, Guangdong, China.,Key Laboratory of Tropical Disease Control, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuqing Deng
- Faculty of Science, Monash University, Clayton, Victoria, Australia
| | - Fengjiao Wei
- Institute of Human Virology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, Guangdong, China.,Key Laboratory of Tropical Disease Control, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qingyu Zhao
- ICU, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yongqi Liu
- Laboratory Animal Center, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zhonghua Liu
- Laboratory Animal Center, South China Agricultural University, Guangzhou, Guangdong, China
| | - Bolan Yu
- BioResource Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaofeng Huang
- Institute of Human Virology, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, Guangdong, China .,Key Laboratory of Tropical Disease Control, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
125
|
Application of nanogels as drug delivery systems in multicellular spheroid tumor model. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
126
|
Identify potential prognostic indicators and tumor-infiltrating immune cells in pancreatic adenocarcinoma. Biosci Rep 2022; 42:230704. [PMID: 35083488 PMCID: PMC8859426 DOI: 10.1042/bsr20212523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Pancreatic adenocarcinoma (PAAD) is a kind of highly malignant tumor and lacks early diagnosis method and effective treatment. Tumor microenvironment (TME) is of great importance for the occurrence and development of PAAD. Thus, a comprehensive overview of genes and tumor-infiltrating immune cells (TICs) related to TME dynamic changes conduce to develop novel therapeutic targets and prognostic indicators. Methods: We used MAlignant Tumors using Expression data (ESTIMATE) algorithm to analyze the transcriptome RNA-seq data of 182 PAAD cases on The Cancer Genome Atlas (TCGA) platform. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), protein–protein interaction (PPI) network, COX regression analysis and gene set enrichment analysis (GSEA) were carried out to get the hub genes related to the prognosis of PAAD patients. These core genes were validated in GEPIA. CXCL10 expression as a poor prognostic indicator was validated in GEO database. Finally, CIBERSORT algorithm was applied to understand the status of TICs. Results: A total of 715 up-regulated differential expression genes (DEGs) and 57 down-regulated DEGs were found simultaneously in stromal and immune groups. These DEGs were mainly enriched in immune recognition, activation and response processes. CD4, CXCL12, CXCL10, CCL5 and CXCL9 were the top five core genes. Then, the validation of these genes showed that CD4, CXCL10, CXCL5, CXCL9 were up-regulated in PAAD. Among the core genes, CXCL10 had a negative correlation with the survival time of PAAD patients. CD8+ T cells, CD4+ T cells memory activated, macrophages M1 had positive correlation of CXCL10 expression, whereas regulatory T cells (Tregs), macrophages M0 and B cells memory had negative correlation. Conclusion: We generated a series of genes related to TME with prognostic implications and TICs in PAAD, which have the potential to be novel immunotherapy targets and prognostic markers. The data showed that CXCL10 was favorable as a poor prognostic indicator in PAAD patients.
Collapse
|
127
|
López Mendoza CM, Alcántara Quintana LE. Smart Drug Delivery Strategies for Cancer Therapy. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2021.753766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy is one of the most widely used strategies to fight cancer, although it has disadvantages such as accumulation in healthy organs and lack of specificity by cancer cells (non-targeted molecules), among others, resulting in adverse effects on patients that limit the dose or follow-up with the same. However, the treatment can also fail due to the resistance mechanisms that cancer cells have to these agents. Because of these limitations, smart drug delivery strategies have been developed to overcome treatment challenges. These smart drug strategies are made with the aim of passively or actively releasing the drug into the tumor environment, increasing the uptake of the chemotherapeutic agent by the cancer cells, thus reducing the adverse effects on other vital organs. Also, these strategies can be guided with molecules on their surface that interact with the tumor microenvironment or with specific receptors on the cancer cell membrane, thus conferring high affinity. This mini review summarizes advances in the development of drug delivery techniques for cancer treatment, including different smart nanocarriers with single or multifunctional stimuli responsiveness. At the same time, we highlight the toxicity and delivery of these strategies in in vivo models. Despite innovation in smart delivery techniques, there are still biodistribution and customization challenges to be overcome in future research.
Collapse
|
128
|
Charalampakis N, Tsakatikas S, Schizas D, Kykalos S, Tolia M, Fioretzaki R, Papageorgiou G, Katsaros I, Abdelhakeem AAF, Sewastjanow-Silva M, Rogers JE, Ajani JA. Trimodality treatment in gastric and gastroesophageal junction cancers: Current approach and future perspectives. World J Gastrointest Oncol 2022; 14:181-202. [PMID: 35116110 PMCID: PMC8790425 DOI: 10.4251/wjgo.v14.i1.181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/28/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric and gastroesophageal junction (GEJ) cancers represent an aggressive group of malignancies with poor prognosis even when diagnosed in relatively early stage, with an increasing incidence both in Asia and in Western countries. These cancers are characterized by heterogeneity as a result of different pathogenetic mechanisms as shown in recent molecular analyses. Accordingly, the understanding of phenotypic and genotypic correlations/classifications has been improved. Current therapeutic strategies have also advanced and moved beyond surgical extirpation alone, with the incorporation of other treatment modalities, such as radiation and chemotherapy (including biologics). Chemoradiotherapy has been used as postoperative treatment after suboptimal gastrectomy to ensure local disease control but also improvement in survival. Preoperative chemoradiotherapy/chemotherapy has been employed to increase the chance of a successful R0 resection and pathologic complete response rate, which is associated with improved long-term outcomes. Several studies have defined various chemotherapy regimens to accompany radiation (before and after surgery). Recently, addition of immunotherapy after trimodality of gastroesophageal cancer has produced an advantage in disease-free interval. Targeted agents used in the metastatic setting are being investigated in the early setting with mixed results. The aim of this review is to summarize the existing data on trimodality approaches for gastric and GEJ cancers, highlight the remaining questions and present the current research effort addressing them.
Collapse
Affiliation(s)
- Nikolaos Charalampakis
- Department of Medical Oncology, Metaxa Cancer Hospital of Piraeus, Piraeus 18537, Greece
| | - Sergios Tsakatikas
- Department of Medical Oncology, Metaxa Cancer Hospital of Piraeus, Piraeus 18537, Greece
| | - Dimitrios Schizas
- TheFirst Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, Athens 11527, Greece
| | - Stylianos Kykalos
- TheSecond Propedeutic Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, Athens 11527, Greece
| | - Maria Tolia
- Department of Radiation Oncology, University Hospital of Crete, Heraklion 71110, Greece
| | - Rodanthi Fioretzaki
- Department of Medical Oncology, Metaxa Cancer Hospital of Piraeus, Piraeus 18537, Greece
| | - Georgios Papageorgiou
- Department of Medical Oncology, Metaxa Cancer Hospital of Piraeus, Piraeus 18537, Greece
| | - Ioannis Katsaros
- Department of General Surgery, Metaxa Cancer Hospital of Piraeus, Piraeus 18537, Greece
| | - Ahmed Adel Fouad Abdelhakeem
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Matheus Sewastjanow-Silva
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Jane E Rogers
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
129
|
Wang YC, Zheng WL, Yu W, Quan RL, Zhao YJ. Erythropoietin-producing hepatocyte kinase receptor A1 facilitating the prgression of SGC-7901 cells and its transplanted tumor by increasing the expression of interleukin-6 and vascular endothelial growth factor in tumor microenvironment. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221125612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objectives Many researches showed that Erythropoietin-producing hepatocyte kinase receptor A1 (EphA1) can promote the occurrence and development of malignant tumors and may be related to tumor microenvironment. But most of them are phenomenon studies, and there are few in-depth and complete mechanism studies. This study aims to understand how EphA1 promotes the progression of malignant tumors by regulating tumor microenvironment (focusing on Interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF)) from two experimental dimensions of in vitro and in vivo by using genetic engineering technology. Material and Methods We used genetic engineering technology to enhance and knock down EphA1 gene expression in SGC-7901 cells, respectively, and analyzed its influence on cell function and the expression levels of VEGF and IL-6 in cells. Subsequently, we constructed human EphA1 gene overexpression, EphA1 gene silencing, and normal expression of human EphA1 gene subcutaneous transplanted tumor models of SGC-7901 cells nude mice, and analyzed the differences in tumor development and the changes in the expression levels of VEGF and ILl-6 in tumor tissues. Results After EphA1 gene expression was enhanced, the proliferation, invasion and migration of SGC-7901 cells were enhanced, and apoptosis was weakened, and the expression levels of VEGF and IL-6 were increased. While the opposite results were found when EphA1 gene expression were knocked down. Meanwhile, tumor formation time and growth rate of subcutaneous transplantation in nude mice were advanced and the expression levels of VEGF and IL-6 in tumor tissues were increased when EphA1 gene expression were overexpressed by genetic engineering technology. Similarly, the opposite effect occurred in transplanted tumor model when EphA1 gene was silenced. Conclusion Our study showed that EphA1 can up-regulating VEGF and IL-6 expression, thereby enhancing the inflammatory environment and angiogenesis in the tumor microenvironment, and this helps to promote the progression of SGC-7901 cells and its transplanted tumor.
Collapse
Affiliation(s)
- Yong-Cang Wang
- Department of Gastrointestinal Oncology Surgery, Anhui Provincial Cancer Hospital (West District of The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, P.R. China
| | - Wen-Lin Zheng
- Department of Gastrointestinal Oncology Surgery, Anhui Provincial Cancer Hospital (West District of The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, P.R. China
| | - Wei Yu
- Department of Gastrointestinal Oncology Surgery, Anhui Provincial Cancer Hospital (West District of The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, P.R. China
| | - Rui-Liang Quan
- Department of Gastrointestinal Oncology Surgery, Anhui Provincial Cancer Hospital (West District of The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, P.R. China
| | - Ya-Jun Zhao
- Department of Gastrointestinal Oncology Surgery, Anhui Provincial Cancer Hospital (West District of The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China), Hefei, P.R. China
| |
Collapse
|
130
|
Gitto SB, George E, Medvedev S, Simpkins F, Powell DJ. Humanized Patient-Derived Xenograft Models of Ovarian Cancer. Methods Mol Biol 2022; 2424:255-274. [PMID: 34918300 DOI: 10.1007/978-1-0716-1956-8_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In vivo modeling of cancer is a critical step in testing novel therapeutic strategies to advance patient care. Here we describe how to develop a humanized patient-derived xenograft (PDX) model of ovarian cancer that uses orthotopically transplanted patient ovarian tumors with autologous transfer of expanded tumor infiltrating T cells (TILs) as a model that can be utilized to test immunomodulating therapeutics in vivo.
Collapse
Affiliation(s)
- Sarah B Gitto
- Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Erin George
- Ovarian Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sergey Medvedev
- Ovarian Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fiona Simpkins
- Ovarian Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Powell
- Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
131
|
Loss-of-function of the hippo transducer TAZ reduces mammary tumor growth through a myeloid-derived suppressor cell-dependent mechanism. Cancer Gene Ther 2022; 29:1791-1800. [PMID: 35840667 PMCID: PMC9663307 DOI: 10.1038/s41417-022-00502-0] [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: 01/05/2022] [Revised: 06/01/2022] [Accepted: 06/23/2022] [Indexed: 02/04/2023]
Abstract
TAZ, one of the key effectors in the Hippo pathway, is often dysregulated in breast cancer, leading to cancer stemness, survival, and metastasis. However, the mechanistic bases of these tumor outcomes are incompletely understood and even less is known about the potential role played by the non-malignant cellular constituents of the tumor microenvironment (TME). Here, we revealed an inverse correlation between TAZ expression and survival in triple-negative breast cancer (TNBC), but not other subtypes of breast cancer. We found that TAZ knockdown in two murine TNBC tumor cell line models significantly inhibited tumor growth and metastasis in immune competent but not immune deficient hosts. RNA-seq analyses identified substantial alterations in immune components in TAZ knockdown tumors. Using mass cytometry analysis, we found that TAZ-deficiency altered the immune landscape of the TME leading to significant reductions in immune suppressive populations, namely myeloid-derived suppressor cells (MDSCs) and macrophages accompanied by elevated CD8+ T cell/myeloid cell ratios. Mechanistic studies demonstrated that TAZ-mediated tumor growth was MDSC-dependent in that MDSC depletion led to reduced tumor growth in control, but not TAZ-knockdown tumor cells. Altogether, we identified a novel non-cancer cell-autonomous mechanism by which tumor-intrinsic TAZ expression aids tumor progression. Thus, our findings advance an understanding of the crosstalk between tumor-derived TAZ expression and the immune contexture within the TME, which may lead to new therapeutic interventions for TNBC or other TAZ-driven cancers.
Collapse
|
132
|
Bou Malhab LJ, Abdel-Rahman WM. Obesity and Inflammation: Colorectal Cancer Engines. Curr Mol Pharmacol 2022; 15:620-646. [PMID: 34488607 DOI: 10.2174/1874467214666210906122054] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022]
Abstract
The prevalence of obesity continues to increase to the extent that it became a worldwide pandemic. An accumulating body of evidence has associated obesity with the development of different types of cancer, including colorectal cancer, which is a notorious disease with a high mortality rate. At the molecular level, colorectal cancer is a heterogenous disease characterized by a myriad of genetic and epigenetic alterations associated with various forms of genomic instability (detailed in Supplementary Materials). Recently, the microenvironment has emerged as a major factor in carcinogenesis. Our aim is to define the different molecular alterations leading to the development of colorectal cancer in obese patients with a focus on the role of the microenvironment in carcinogenesis. We also highlight all existent molecules in clinical trials that target the activated pathways in obesity-associated colorectal cancer, whether used as single treatments or in combination. Obesity predisposes to colorectal cancer via creating a state of chronic inflammation with dysregulated adipokines, inflammatory mediators, and other factors such as immune cell infiltration. A unifying theme in obesity-mediated colorectal cancer is the activation of the PI3K/AKT, mTOR/MAPK, and STAT3 signaling pathways. Different inhibitory molecules towards these pathways exist, increasing the therapeutic choice of obesity-associated colon cancer. However, obese patients are more likely to suffer from chemotherapy overdosing. Preventing obesity through maintaining a healthy and active lifestyle remains to be the best remedy.
Collapse
Affiliation(s)
- Lara J Bou Malhab
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Wael M Abdel-Rahman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
133
|
Gris-Cárdenas I, Rábano M, Vivanco MDM. Patient-Derived Explant Cultures of Normal and Tumor Human Breast Tissue. Methods Mol Biol 2022; 2471:301-307. [PMID: 35175605 DOI: 10.1007/978-1-0716-2193-6_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tissue culture has evolved considerably over the last few years, including cell culture in three dimensions, organoids, cocultures of different cell types and the use of diverse types of matrices in an attempt to mimic conditions that more closely resemble those found in the original tissue or organ. In this chapter, we describe how patient-derived breast tissue can be cultured on sponges for several days, maintaining their original architecture and with the capacity to respond to treatments. This protocol facilitates the study of the tissue responses without the need for extensive tissue manipulation, cell digestion or use of a biomaterial as scaffold, while maintaining the stroma and extracellular matrix organization. This method has the potential to improve preclinical testing by contributing to provide more accurate data reflecting cell-cell and cell-matrix interactions, tumor microenvironment, drug effects or stem cell function in normal- and pathophysiology of the breast.
Collapse
Affiliation(s)
- Isabel Gris-Cárdenas
- Cancer Heterogeneity Laboratory, Centre for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Miriam Rábano
- Cancer Heterogeneity Laboratory, Centre for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Maria D M Vivanco
- Cancer Heterogeneity Laboratory, Centre for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain.
| |
Collapse
|
134
|
Barbosa MAG, Xavier CPR, Pereira RF, Petrikaitė V, Vasconcelos MH. 3D Cell Culture Models as Recapitulators of the Tumor Microenvironment for the Screening of Anti-Cancer Drugs. Cancers (Basel) 2021; 14:190. [PMID: 35008353 PMCID: PMC8749977 DOI: 10.3390/cancers14010190] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Today, innovative three-dimensional (3D) cell culture models have been proposed as viable and biomimetic alternatives for initial drug screening, allowing the improvement of the efficiency of drug development. These models are gaining popularity, given their ability to reproduce key aspects of the tumor microenvironment, concerning the 3D tumor architecture as well as the interactions of tumor cells with the extracellular matrix and surrounding non-tumor cells. The development of accurate 3D models may become beneficial to decrease the use of laboratory animals in scientific research, in accordance with the European Union's regulation on the 3R rule (Replacement, Reduction, Refinement). This review focuses on the impact of 3D cell culture models on cancer research, discussing their advantages, limitations, and compatibility with high-throughput screenings and automated systems. An insight is also given on the adequacy of the available readouts for the interpretation of the data obtained from the 3D cell culture models. Importantly, we also emphasize the need for the incorporation of additional and complementary microenvironment elements on the design of 3D cell culture models, towards improved predictive value of drug efficacy.
Collapse
Affiliation(s)
- Mélanie A. G. Barbosa
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Biofabrication Group, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickevičiaus g 9, LT-44307 Kaunas, Lithuania;
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
135
|
Han ZJ, Li YB, Yang LX, Cheng HJ, Liu X, Chen H. Roles of the CXCL8-CXCR1/2 Axis in the Tumor Microenvironment and Immunotherapy. MOLECULES (BASEL, SWITZERLAND) 2021; 27:molecules27010137. [PMID: 35011369 PMCID: PMC8746913 DOI: 10.3390/molecules27010137] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/12/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
In humans, Interleukin-8 (IL-8 or CXCL8) is a granulocytic chemokine with multiple roles within the tumor microenvironment (TME), such as recruiting immunosuppressive cells to the tumor, increasing tumor angiogenesis, and promoting epithelial-to-mesenchymal transition (EMT). All of these effects of CXCL8 on individual cell types can result in cascading alterations to the TME. The changes in the TME components such as the cancer-associated fibroblasts (CAFs), the immune cells, the extracellular matrix, the blood vessels, or the lymphatic vessels further influence tumor progression and therapeutic resistance. Emerging roles of the microbiome in tumorigenesis or tumor progression revealed the intricate interactions between inflammatory response, dysbiosis, metabolites, CXCL8, immune cells, and the TME. Studies have shown that CXCL8 directly contributes to TME remodeling, cancer plasticity, and the development of resistance to both chemotherapy and immunotherapy. Further, clinical data demonstrate that CXCL8 could be an easily measurable prognostic biomarker in patients receiving immune checkpoint inhibitors. The blockade of the CXCL8-CXCR1/2 axis alone or in combination with other immunotherapy will be a promising strategy to improve antitumor efficacy. Herein, we review recent advances focusing on identifying the mechanisms between TME components and the CXCL8-CXCR1/2 axis for novel immunotherapy strategies.
Collapse
Affiliation(s)
- Zhi-Jian Han
- The Key Laboratory of the Digestive System Tumors of Gansu Province, Tumor Center, Lanzhou University Second Hospital, Lanzhou 730000, China; (Y.-B.L.); (L.-X.Y.); (H.-J.C.)
- Correspondence: (Z.-J.H.); (H.C.); Tel.: +86-186-9310-9388 (Z.-J.H.); +86-150-0946-7790 (H.C.)
| | - Yang-Bing Li
- The Key Laboratory of the Digestive System Tumors of Gansu Province, Tumor Center, Lanzhou University Second Hospital, Lanzhou 730000, China; (Y.-B.L.); (L.-X.Y.); (H.-J.C.)
| | - Lu-Xi Yang
- The Key Laboratory of the Digestive System Tumors of Gansu Province, Tumor Center, Lanzhou University Second Hospital, Lanzhou 730000, China; (Y.-B.L.); (L.-X.Y.); (H.-J.C.)
| | - Hui-Juan Cheng
- The Key Laboratory of the Digestive System Tumors of Gansu Province, Tumor Center, Lanzhou University Second Hospital, Lanzhou 730000, China; (Y.-B.L.); (L.-X.Y.); (H.-J.C.)
| | - Xin Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China;
| | - Hao Chen
- The Key Laboratory of the Digestive System Tumors of Gansu Province, Tumor Center, Lanzhou University Second Hospital, Lanzhou 730000, China; (Y.-B.L.); (L.-X.Y.); (H.-J.C.)
- Correspondence: (Z.-J.H.); (H.C.); Tel.: +86-186-9310-9388 (Z.-J.H.); +86-150-0946-7790 (H.C.)
| |
Collapse
|
136
|
Liu C, Cong X, Wang Y, Guo Q, Xie Y, Geng F, Guo J, Dong L, Zhou Y, Wu H, Yu B, Wu J, Zhang H, Yu X, Kong W. Fast DNA Vaccination Strategy Elicits a Stronger Immune Response Dependent on CD8 +CD11c + Cell Accumulation. Front Oncol 2021; 11:752444. [PMID: 34950581 PMCID: PMC8691261 DOI: 10.3389/fonc.2021.752444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Conventional DNA vaccine strategies usually employ a regimen of immunizations at 2-week or longer intervals to induce effective memory cell-dependent immune responses. Clinical cancer treatment requires a faster immunization strategy to contend with tumor progression. In this study, a novel fast immunization strategy was established, wherein a DNA vaccine was intramuscularly administered on days 0, 2, and 5 in a murine lung cancer model. Effector cells peaked 7 to 10 days after the last vaccination. Compared with traditional 2-week-interval immunization strategies, antigen-specific cytolysis and INF-γ secretion were significantly enhanced under the fast vaccination approach. As a result, the rapidly administered DNA vaccine elicited stronger and more prompt antitumor effects. The probable underlying mechanism of fast immunization was the accumulation of CD8+CD11c+ antigen-presenting cells at the injection site, which enhanced subsequent antigen presentation. In conclusion, the fast DNA vaccination strategy shortened vaccination time to 5 days and elicited a stronger antitumor immune response.
Collapse
Affiliation(s)
- Chenlu Liu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Xianling Cong
- Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Yuqian Wang
- Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Qianqian Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Yu Xie
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Fei Geng
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Jie Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Ling Dong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Yi Zhou
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| |
Collapse
|
137
|
Taeb S, Ashrafizadeh M, Zarrabi A, Rezapoor S, Musa AE, Farhood B, Najafi M. Role of Tumor Microenvironment in Cancer Stem Cells Resistance to Radiotherapy. Curr Cancer Drug Targets 2021; 22:18-30. [PMID: 34951575 DOI: 10.2174/1568009622666211224154952] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/29/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
Cancer is a chronic disorder that involves several elements of both the tumor and the host stromal cells. At present, the complex relationship between the various factors presents in the tumor microenvironment (TME) and tumor cells, as well as immune cells located within the TME, is still poorly known. Within the TME, the crosstalk of these factors and immune cells essentially determines how a tumor reacts to the treatment and how the tumor can ultimately be destroyed, remain dormant, or develop and metastasize. Also, in TME, reciprocal crosstalk between cancer-associated fibroblasts (CAFs), extracellular matrix (ECM), hypoxia-inducible factor (HIF) intensifies the proliferation capacity of cancer stem cells (CSCs). CSCs are subpopulation of cells that reside within the tumor bulk and have the capacity to self-renew, differentiate, and repair DNA damage. These characteristics make CSCs develop resistance to a variety of treatments, such as radiotherapy (RT). RT is a frequent and often curative treatment for local cancer which mediates tumor elimination by cytotoxic actions. Also, cytokines and growth factors that are released into TME, have been involved in the activation of tumor radioresistance and the induction of different immune cells, altering local immune responses. In this review, we discuss the pivotal role of TME in resistance of CSCs to RT.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 , Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Turkey
| | - Saeed Rezapoor
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences., Iraq
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Iran
| |
Collapse
|
138
|
Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010146. [PMID: 35008569 PMCID: PMC8745566 DOI: 10.3390/ijms23010146] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer progression with uncontrolled tumor growth, local invasion, and metastasis depends largely on the proteolytic activity of numerous matrix metalloproteinases (MMPs), which affect tissue integrity, immune cell recruitment, and tissue turnover by degrading extracellular matrix (ECM) components and by releasing matrikines, cell surface-bound cytokines, growth factors, or their receptors. Among the MMPs, MMP-14 is the driving force behind extracellular matrix and tissue destruction during cancer invasion and metastasis. MMP-14 also influences both intercellular as well as cell-matrix communication by regulating the activity of many plasma membrane-anchored and extracellular proteins. Cancer cells and other cells of the tumor stroma, embedded in a common extracellular matrix, interact with their matrix by means of various adhesive structures, of which particularly invadopodia are capable to remodel the matrix through spatially and temporally finely tuned proteolysis. As a deeper understanding of the underlying functional mechanisms is beneficial for the development of new prognostic and predictive markers and for targeted therapies, this review examined the current knowledge of the interplay of the various MMPs in the cancer context on the protein, subcellular, and cellular level with a focus on MMP14.
Collapse
|
139
|
Yao J, Duan L, Huang X, Liu J, Fan X, Xiao Z, Yan R, Liu H, An G, Hu B, Ge Y. Development and Validation of a Prognostic Gene Signature Correlated With M2 Macrophage Infiltration in Esophageal Squamous Cell Carcinoma. Front Oncol 2021; 11:769727. [PMID: 34926275 PMCID: PMC8677679 DOI: 10.3389/fonc.2021.769727] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is the most common type of esophageal cancer and the seventh most prevalent cause of cancer-related death worldwide. Tumor microenvironment (TME) has been confirmed to play an crucial role in ESCC progression, prognosis, and the response to immunotherapy. There is a need for predictive biomarkers of TME-related processes to better prognosticate ESCC outcomes. Aim To identify a novel gene signature linked with the TME to predict the prognosis of ESCC. Methods We calculated the immune/stromal scores of 95 ESCC samples from The Cancer Genome Atlas (TCGA) using the ESTIMATE algorithm, and identified differentially expressed genes (DEGs) between high and low immune/stromal score patients. The key prognostic genes were further analyzed by the intersection of protein–protein interaction (PPI) networks and univariate Cox regression analysis. Finally, a risk score model was constructed using multivariate Cox regression analysis. We evaluated the associations between the risk score model and immune infiltration via the CIBERSORT algorithm. Moreover, we validated the signature using the Gene Expression Omnibus (GEO) database. Within the ten gene signature, five rarely reported genes were further validated with quantitative real time polymerase chain reaction (qRT-PCR) using an ESCC tissue cDNA microarray. Results A total of 133 up-regulated genes were identified as DEGs. Ten prognostic genes were selected based on intersection analysis of univariate COX regression analysis and PPI, and consisted of C1QA, C1QB, C1QC, CD86, C3AR1, CSF1R, ITGB2, LCP2, SPI1, and TYROBP (HR>1, p<0.05). The expression of 9 of these genes in the tumor samples were significantly higher compared to matched adjacent normal tissue based on the GEO database (p<0.05). Next, we assessed the ability of the ten-gene signature to predict the overall survival of ESCC patients, and found that the high-risk group had significantly poorer outcomes compared to the low-risk group using univariate and multivariate analyses in the TCGA and GEO cohorts (HR=2.104, 95% confidence interval:1.343-3.295, p=0.001; HR=1.6915, 95% confidence interval:1.053-2.717, p=0.0297). Additionally, receiver operating characteristic (ROC) curve analysis demonstrated a relatively sensitive and specific profile for the signature (1-, 2-, 3-year AUC=0.672, 0.854, 0.81). To identify the basis for these differences in the TME, we performed correlation analyses and found a significant positive correlation with M1 and M2 macrophages and CD8+ T cells, as well as a strong correlation to M2 macrophage surface markers. A nomogram based on the risk score and select clinicopathologic characteristics was constructed to predict overall survival of ESCC patients. For validation, qRT-PCR of an ESCC patient cDNA microarray was performed, and demonstrated that C1QA, C3AR1, LCP2, SPI1, and TYROBP were up-regulated in tumor samples and predict poor prognosis. Conclusion This study established and validated a novel 10-gene signature linked with M2 macrophages and poor prognosis in ESCC patients. Importantly, we identified C1QA, C3AR1, LCP2, SPI1, and TYROBP as novel M2 macrophage-correlated survival biomarkers. These findings may identify potential targets for therapy in ESCC patients.
Collapse
Affiliation(s)
- Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ling Duan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xuying Huang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jian Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaona Fan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zeru Xiao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Rui Yan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Heshu Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yang Ge
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
140
|
Sclafani F, Corrò C, Koessler T. Debating Pros and Cons of Total Neoadjuvant Therapy in Rectal Cancer. Cancers (Basel) 2021; 13:cancers13246361. [PMID: 34944980 PMCID: PMC8699289 DOI: 10.3390/cancers13246361] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Rectal cancers represent one third of all colorectal tumours. Patients diagnosed with localised colon cancer undergo surgery upfront, likely followed by adjuvant chemotherapy. Those diagnosed with localised rectal cancer, however, frequently benefit from neoadjuvant treatments with either radiotherapy or chemoradiotherapy before undergoing surgery. On the other hand, the benefit of adjuvant chemotherapy in this setting is more controversial. The main challenges in treating patients affected by rectal cancer encompass: decreasing the risks of local relapse and distant metastases, preserving the sphincter and minimising treatment-associated functional sequelae, and improving overall survival. Some of these fuelled the concept of total neoadjuvant therapy, namely giving all available treatments including radiotherapy and systemic chemotherapy before surgery. Here, we critically review the pros and cons of such a treatment strategy, but also discuss the biological rational to support neoadjuvant treatment intensification. Abstract Recently, two large, randomised phase III clinical trials of total neoadjuvant therapy (TNT) in locally advanced rectal cancer were published (RAPIDO and PRODIGE 23). These two trials compared short-course radiotherapy (SCRT) followed by chemotherapy with standard chemoradiotherapy (CRT) and chemotherapy followed by CRT with standard CRT, respectively. They showed improvement in some of the outcomes such as distant recurrence and pathological complete response (pCR). No improvement, however, was observed in local disease control or the de-escalation of surgical procedures. Although it seems lawful to integrate TNT within the treatment algorithm of localised stage II and III rectal cancer, many questions remain unanswered, including which are the optimal criteria to identify patients who are most likely to benefit from this intensive treatment. Instead of providing a sterile summary of trial results, we put these in perspective in a pros and cons manner. Moreover, we discuss some biological aspects of rectal cancer, which may provide some insights into the current decision-making process, and represent the basis for the future development of alternative, more effective treatment strategies.
Collapse
Affiliation(s)
- Francesco Sclafani
- Department of Medical Oncology, Institut Jules Bordet, Rue Meylemeersch 90, 1070 Anderlecht, Belgium;
- Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| | - Claudia Corrò
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland;
- Swiss Cancer Center Léman, Geneva and Lausanne, 1005 Lausanne, Switzerland
| | - Thibaud Koessler
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland;
- Swiss Cancer Center Léman, Geneva and Lausanne, 1005 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
- Correspondence:
| |
Collapse
|
141
|
Lafuente-Gómez N, Latorre A, Milán-Rois P, Rodriguez Diaz C, Somoza Á. Stimuli-responsive nanomaterials for cancer treatment: boundaries, opportunities and applications. Chem Commun (Camb) 2021; 57:13662-13677. [PMID: 34874370 DOI: 10.1039/d1cc05056g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Small molecule drugs, including most chemotherapies, are rapidly degraded and/or eliminated from the body, which is why high doses of these drugs are necessary, potentially producing toxic effects. Several types of nanoparticles loaded with anti-cancer drugs have been designed to overcome the disadvantages of conventional therapies. Modified nanoparticles can circulate for a long time, thus improving the solubility and biodistribution of drugs. Furthermore, they also allow the controlled release of the payload once its target tissue has been reached. These mechanisms can reduce the exposure of healthy tissues to chemotherapeutics, since the drugs are only released in the presence of specific tumour stimuli. Overall, these properties can improve the effectiveness of treatments while reducing undesirable side effects. In this article, we review the recent advances in stimuli-responsive albumin, gold and magnetic nanostructures for controlled anti-cancer drug delivery. These nanostructures were designed to release drugs in response to different internal and external stimuli of the cellular environment, including pH, redox, light and magnetic fields. We also describe various examples of applications of these nanomaterials. Overall, we shed light on the properties, potential clinical translation and limitations of stimuli-responsive nanoparticles for cancer treatment.
Collapse
Affiliation(s)
- Nuria Lafuente-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Ana Latorre
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Paula Milán-Rois
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Ciro Rodriguez Diaz
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain. .,Unidad Asociada al Centro Nacional de Biotecnología (CSIC), 28049 Madrid, Spain
| |
Collapse
|
142
|
Rodríguez CM, Velásquez-Berrío M, Rúa C, Viana M, Abrahams VM, Cadavid AP, Alvarez AM. Antiphospholipid Antibodies From Women With Pregnancy Morbidity and Vascular Thrombosis Induce Endothelial Mitochondrial Dysfunction, mTOR Activation, and Autophagy. Front Physiol 2021; 12:706743. [PMID: 34912234 PMCID: PMC8667788 DOI: 10.3389/fphys.2021.706743] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 10/18/2021] [Indexed: 01/09/2023] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by thrombosis and pregnancy morbidity (PM) obstetric events together with persistent high titers of circulating antiphospholipid antibodies (aPL). Several mechanisms that explain the development of thrombosis and PM in APS include the association of aPL with alterations in the coagulation cascade and inflammatory events. Other mechanisms disturbing cellular homeostases, such as mitochondrial dysfunction, autophagy, and cell proliferation, have been described in other autoimmune diseases. Therefore, the objective of this study was to investigate the impact of aPL from different patient populations on endothelial cell mitochondrial function, activation of the mammalian target of rapamycin (mTOR) and autophagy pathways, and cellular growth. Using an in vitro model, human umbilical vein endothelial cells (HUVECs) were treated with polyclonal immunoglobulin G (IgG) purified from the serum of women with both PM and vascular thrombosis (PM/VT), with VT only (VT), or with PM and non-criteria aPL (seronegative-obstetric APS, SN-OAPS). We included IgG from women with PM without aPL (PM/aPL-) and healthy women with previous uncomplicated pregnancies (normal human serum, NHS) as control groups. Mitochondrial function, mTOR activation, autophagy, and cell proliferation were evaluated by Western blotting, flow cytometry, and functional assays. IgG from women with PM/VT increased HUVEC mitochondrial hyperpolarization and activation of the mTOR and autophagic pathways, while IgG from patients with VT induced endothelial autophagy and cell proliferation in the absence of elevated mTOR activity or mitochondrial dysfunction. IgG from the SN-OAPS patient group had no effect on any of these HUVEC responses. In conclusion, aPL from women with PM and vascular events induce cellular stress evidenced by mitochondrial hyperpolarization and increased activation of the mTOR and autophagic pathways which may play a role in the pathogenesis of obstetric APS.
Collapse
Affiliation(s)
- Carlos M. Rodríguez
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Manuela Velásquez-Berrío
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Carolina Rúa
- Grupo de Investigación en Trombosis, Facultad de Medicina, Universidad de Antioquia (UdeA), Medellín, Colombia
| | - Marta Viana
- Grupo de Metabolismo y Función Vascular, Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo (RIVATREM), Chillán, Chile
| | - Vikki M. Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Angela P. Cadavid
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
- Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo (RIVATREM), Chillán, Chile
| | - Angela M. Alvarez
- Grupo Reproducción, Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad de Antioquia (UdeA), Medellín, Colombia
| |
Collapse
|
143
|
Jang SD, Song J, Kim HA, Im CN, Khawar IA, Park JK, Kuh HJ. Anti-Cancer Activity Profiling of Chemotherapeutic Agents in 3D Co-Cultures of Pancreatic Tumor Spheroids with Cancer-Associated Fibroblasts and Macrophages. Cancers (Basel) 2021; 13:5955. [PMID: 34885065 PMCID: PMC8656537 DOI: 10.3390/cancers13235955] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/29/2022] Open
Abstract
Activated pancreatic stellate cells (aPSCs) and M2 macrophages modulate tumor progression and therapeutic efficacy in pancreatic ductal adenocarcinoma (PDAC) via epithelial-mesenchymal transition (EMT). Here, our aim was to analyze the anti-invasion effects of anti-cancer agents where EMT-inducing cancer-stroma interaction occurs under three-dimensional (3D) culture conditions. We used microfluidic channel chips to co-culture pancreatic tumor spheroids (TSs) with aPSCs and THP-1-derived M2 macrophages (M2 THP-1 cells) embedded in type I collagen. Under stromal cell co-culture conditions, PANC-1 TSs displayed elevated expression of EMT-related proteins and increased invasion and migration. When PANC-1 TSs were exposed to gemcitabine, 5-fluorouracil, oxaliplatin, or paclitaxel, 30-50% cells were found unaffected, with no significant changes in the dose-response profiles under stromal cell co-culture conditions. This indicated intrinsic resistance to these drugs and no further induction of drug resistance by stromal cells. Paclitaxel had a significant anti-invasion effect; in contrast, oxaliplatin did not show such effect despite its specific cytotoxicity in M2 THP-1 cells. Overall, our findings demonstrate that the TS-stroma co-culture model of PDAC is useful for activity profiling of anti-cancer agents against cancer and stromal cells, and analyzing the relationship between anti-stromal activity and anti-invasion effects.
Collapse
Affiliation(s)
- So-Dam Jang
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Jeeyeun Song
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Hyun-Ah Kim
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Chang-Nim Im
- Graduate Program for Future Medical Research Leaders, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Iftikhar Ali Khawar
- Graduate Program for Future Medical Research Leaders, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jong Kook Park
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
144
|
Receptor-Tyrosine Kinase Inhibitor Ponatinib Inhibits Meningioma Growth In Vitro and In Vivo. Cancers (Basel) 2021; 13:cancers13235898. [PMID: 34885009 PMCID: PMC8657092 DOI: 10.3390/cancers13235898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
To date, there is no standard-of-care systemic therapy for the treatment of aggressive meningiomas. Receptor tyrosine kinases (RTK) are frequently expressed in aggressive meningiomas and are associated with poor survival. Ponatinib is a FDA- and EMA-approved RTK inhibitor and its efficacy in meningioma has not been studied so far. Therefore, we investigated ponatinib as a potential drug candidate against meningioma. Cell viability and cell proliferation of ponatinib-treated meningioma cells were assessed using crystal violet assay, manual counting and BrdU assay. Treated meningioma cell lines were subjected to flow cytometry to evaluate the effects on cell cycle and apoptosis. Meningioma-bearing mice were treated with ponatinib to examine antitumor effects in vivo. qPCR was performed to assess the mRNA levels of tyrosine kinase receptors after ponatinib treatment. Full-length cDNA sequencing was carried out to assess differential gene expression. IC50 values of ponatinib were between 171.2 and 341.9 nM in three meningioma cell lines. Ponatinib induced G0/G1 cell cycle arrest and subsequently led to an accumulation of cells in the subG1-phase. A significant induction of apoptosis was observed in vitro. In vivo, ponatinib inhibited meningioma growth by 72.6%. Mechanistically, this was associated with downregulation of PDGFRA/B and FLT3 mRNA levels, and mitochondrial dysfunction. Taken together, ponatinib is a promising candidate for targeted therapy in the treatment of aggressive meningioma.
Collapse
|
145
|
The Upregulation of PLXDC2 Correlates with Immune Microenvironment Characteristics and Predicts Prognosis in Gastric Cancer. DISEASE MARKERS 2021; 2021:5669635. [PMID: 34777633 PMCID: PMC8589478 DOI: 10.1155/2021/5669635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 02/08/2023]
Abstract
Tumor microenvironment (TME) has been demonstrated to exhibit a regulatory effect on the progressions of gastric cancer (GC). However, the related functions of stromal and immune components (TME-associated genes) in TME remain largely unclear. From the TCGA dataset, we downloaded the clinical data of 375 GC cases and then estimated the percentage of tumor-infiltrating immunocytes (TICs) and the levels of immune and stromal constituents by the use of CIBERSORT and ESTIMATE tolls. Univariate assays were applied to study the differentially expressed genes. The associations between the clinical information of GC patients and the expressions of the specific genes were analyzed based on the TCGA datasets. The effect of Plexin domain containing 2 (PLXDC2) expression on TICs was conducted. We observed that PLXDC2 expression was distinctly upregulated in GC specimens compared with nontumor gastric specimens. Its upregulation was associated with advanced clinical stages and predicted a shorter overall survival of GC patients. The genes in the group of higher expressing PLXDC2 were primarily enriched in immunity-associated events. By the use of CIBERSORT, we observed that PLXDC2 expressions were related to the proportion of dendritic cells resting, T cell CD4 memory resting, eosinophils, mastocyte resting, mononuclear cells, plasma cells, T cell follicle helper, macrophage M2, and dendritic cells activated. Overall, our discoveries revealed that the expression of PLXDC2 was remarkable in GC, might be a possible biomarker for GC, and provided novel contents regarding immune infiltrates, offering novel insight for treatments of GC.
Collapse
|
146
|
Pharmaco-proteogenomic profiling of pediatric diffuse midline glioma to inform future treatment strategies. Oncogene 2021; 41:461-475. [PMID: 34759345 PMCID: PMC8782719 DOI: 10.1038/s41388-021-02102-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022]
Abstract
Diffuse midline glioma (DMG) is a deadly pediatric and adolescent central nervous system (CNS) tumor localized along the midline structures of the brain atop the spinal cord. With a median overall survival (OS) of just 9–11-months, DMG is characterized by global hypomethylation of histone H3 at lysine 27 (H3K27me3), driven by recurring somatic mutations in H3 genes including, HIST1H3B/C (H3.1K27M) or H3F3A (H3.3K27M), or through overexpression of EZHIP in patients harboring wildtype H3. The recent World Health Organization’s 5th Classification of CNS Tumors now designates DMG as, ‘H3 K27-altered’, suggesting that global H3K27me3 hypomethylation is a ubiquitous feature of DMG and drives devastating transcriptional programs for which there are no treatments. H3-alterations co-segregate with various other somatic driver mutations, highlighting the high-level of intertumoral heterogeneity of DMG. Furthermore, DMG is also characterized by very high-level intratumoral diversity with tumors harboring multiple subclones within each primary tumor. Each subclone contains their own combinations of driver and passenger lesions that continually evolve, making precision-based medicine challenging to successful execute. Whilst the intertumoral heterogeneity of DMG has been extensively investigated, this is yet to translate to an increase in patient survival. Conversely, our understanding of the non-genomic factors that drive the rapid growth and fatal nature of DMG, including endogenous and exogenous microenvironmental influences, neurological cues, and the posttranscriptional and posttranslational architecture of DMG remains enigmatic or at best, immature. However, these factors are likely to play a significant role in the complex biological sequelae that drives the disease. Here we summarize the heterogeneity of DMG and emphasize how analysis of the posttranslational architecture may improve treatment paradigms. We describe factors that contribute to treatment response and disease progression, as well as highlight the potential for pharmaco-proteogenomics (i.e., the integration of genomics, proteomics and pharmacology) in the management of this uniformly fatal cancer.
Collapse
|
147
|
Abdelghany L, Zhang X, Kawabata T, Goto S, El-Mahdy N, Jingu K, Li TS. Nicaraven prevents the fast growth of inflamed tumors by an anti-inflammatory mechanism. Med Oncol 2021; 39:7. [PMID: 34761342 DOI: 10.1007/s12032-021-01602-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/21/2021] [Indexed: 11/25/2022]
Abstract
Inflammatory microenvironment is known to accelerate the progression of malignant tumors. We investigated the possible anti-inflammatory effect of nicaraven on slowing tumor growth. Tumor-bearing mice randomly received nicaraven injection (50 mg/kg daily, i.p, n = 8) or placebo treatment (n = 8) for 10 days, and then sacrificed for evaluations. Nicaraven administration effectively inhibited the fast growth of tumor, as a large tumor (> 1.0 g) developed finally in three of the eight mice received placebo treatment. Cytokines/chemokines array indicated that nicaraven reduced the levels of CXCL10 and SDF-1 in the tumor as well as the levels of IL-2 and MIP-2 in serum. Immunofluorescence staining showed that nicaraven significantly reduced the recruitment of macrophages and neutrophils in the tumor. Interestingly, western blot indicated that the expression of CD86, CD206, and NIMP-R14 was especially enhanced in the three large-size tumors, suggesting the potential role of nicaraven in preventing the hyper-inflammatory tumor microenvironment. Moreover, the expression of PARP-1 was downregulated, but the expression of phospho-p38 MAPK, phospho-MKK-3/6, and phospho-MSK-1 was upregulated in the large-size tumors, suggesting the involvement of p38 MAPK pathway in the anti-inflammatory effect of nicaraven. Taken together, our study suggests that nicaraven may effectively prevent the fast growth of inflamed tumors by an anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Lina Abdelghany
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Xu Zhang
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Tsuyoshi Kawabata
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Nageh El-Mahdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Keiichi Jingu
- Department of Radiation Oncology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| |
Collapse
|
148
|
Jiang H, Li Y, Xiang X, Tang Z, Liu K, Su Q, Zhang X, Li L. Chaetocin: A review of its anticancer potentials and mechanisms. Eur J Pharmacol 2021; 910:174459. [PMID: 34464601 DOI: 10.1016/j.ejphar.2021.174459] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/17/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Chaetocin is a natural metabolite product with various biological activities and pharmacological functions isolated from Chaetomium species fungi belonging to the thiodiketopyrazines. Numerous studies have demonstrated a wide range of antitumor activities of chaetocin in vitro and in vivo. Several studies have demonstrated that chaetocin suppresses the growth and proliferation of various tumour cells by regulating multiple signalling pathways related to tumour initiation and progression, inducing cancer cell apoptosis (intrinsic and extrinsic), enhancing autophagy, inducing cell cycle arrest, and inhibiting tumour angiogenesis, invasion, and migration. The antitumor effects and molecular mechanisms of chaetocin are reviewed and analysed in this paper, and the prospective applications of chaetocin in cancer prevention and therapy are also discussed. This review aimed to summarize the recent advances in the antitumor activity of chaetocin and to provide a rationale for further exploring the potential application of chaetocin in overcoming cancer in the future.
Collapse
Affiliation(s)
- Hangyu Jiang
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Yuqi Li
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Xiaocong Xiang
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Zhili Tang
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Qiang Su
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Xiaofen Zhang
- Department of Urology, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China.
| | - Lin Li
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; College of Bioengineering, Chongqing University, Chongqing, China.
| |
Collapse
|
149
|
Margarit DH, González NS, Romanelli LM, Fendrik AJ, Scagliotti AF, Reale MV. An integrative model of cancer cell differentiation with immunotherapy . Phys Biol 2021; 18. [PMID: 34633296 DOI: 10.1088/1478-3975/ac2e72] [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: 05/24/2021] [Accepted: 10/11/2021] [Indexed: 11/11/2022]
Abstract
In order to improve cancer treatments, cancer cell differentiation and immunotherapy are the subjects of several studies in different branches of interdisciplinary sciences. In this work, we develop a new population model that integrates other complementary ones, thus emphasizing the relationship between cancer cells at different differentiation stages and the main immune system cells. For this new system, specific ranges were found where transdifferentiation of differentiated cancer cells can occur. In addition, a specific therapy against cancer stem cells was analysed by simulating cytotoxic cell vaccines. In reference to the latter, the different combinations of parameters that optimize it were studied.
Collapse
Affiliation(s)
- David H Margarit
- Instituto de Ciencias, Universidad Nacional de General Sarmiento (UNGS), J M Gutiérrez 1150, Los Polvorines (B1613), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Nadia S González
- Instituto de Ciencias, Universidad Nacional de General Sarmiento (UNGS), J M Gutiérrez 1150, Los Polvorines (B1613), Buenos Aires, Argentina
| | - Lilia M Romanelli
- Instituto de Ciencias, Universidad Nacional de General Sarmiento (UNGS), J M Gutiérrez 1150, Los Polvorines (B1613), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Alejandro J Fendrik
- Instituto de Ciencias, Universidad Nacional de General Sarmiento (UNGS), J M Gutiérrez 1150, Los Polvorines (B1613), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Ariel F Scagliotti
- Instituto de Ciencias, Universidad Nacional de General Sarmiento (UNGS), J M Gutiérrez 1150, Los Polvorines (B1613), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Marcela V Reale
- Instituto de Ciencias, Universidad Nacional de General Sarmiento (UNGS), J M Gutiérrez 1150, Los Polvorines (B1613), Buenos Aires, Argentina.,Departamento de Ingeniería e Investigaciones Tecnológicas, Universidad Nacional de La Matanza (UNLaM), Florencio Varela 1903, San Justo (B1754), Buenos Aires, Argentina
| |
Collapse
|
150
|
Li N, Yu K, Lin Z, Zeng D. Identifying immune subtypes of uterine corpus endometrial carcinoma and a four-paired-lncRNA signature with immune-related lncRNAs. Exp Biol Med (Maywood) 2021; 247:221-236. [PMID: 34704492 DOI: 10.1177/15353702211053588] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Uterine corpus endometrial carcinoma (UCEC) is the third most frequent gynecological malignancies in the female reproductive system. Long non-coding RNAs (lncRNAs) are closely involved in tumor progression. This study aimed to develop an immune subtyping system and a prognostic model based on lncRNAs for UCEC. Paired lncRNAs and non-negative matrix factorization were applied to identify immune subtypes. Enrichment analysis was conducted to assess functional pathways, immune-related genes, and cells. Univariate and multivariate Cox regression analysis were performed to analyze the relation between lncRNAs and overall survival (OS). A prognostic model was constructed and optimized by least absolute shrinkage and selection operator (LASSO) and Akaike information criterion (AIC). Two immune subtypes (C1 and C2) and four paired-prognostic lncRNAs closely associated with overall survival were identified. Some immune features, sensitivity of chemotherapy and immunotherapy, and the relation with immune escape showed variations between two subtypes. A nomogram established based on prognostic model and clinical features was effective in OS prediction. The immune subtyping system based on lncRNAs and the four-paired-lncRNA signature was predictive of UCEC prognosis and can facilitate personalized therapies such as immunotherapy or RNA-based therapy for UCEC patients.
Collapse
Affiliation(s)
- Nan Li
- Liuzhou Maternity and Child Healthcare Hospital, Liuzhou 545001, China
| | - Kai Yu
- Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou 545001, China
| | - Zhong Lin
- Guangxi Health Commission Key Laboratory of Birth Cohort Study in Pregnant Women of Advanced Age, Liuzhou 545001, China
| | - Dingyuan Zeng
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| |
Collapse
|