4501
|
Rodrigues R, Duarte D, Vale N. Drug Repurposing in Cancer Therapy: Influence of Patient’s Genetic Background in Breast Cancer Treatment. Int J Mol Sci 2022; 23:ijms23084280. [PMID: 35457144 PMCID: PMC9028365 DOI: 10.3390/ijms23084280] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer is among the leading causes of death worldwide and it is estimated that in 2040 more than 29 million people will be diagnosed with some type of cancer. The most prevalent type of cancer in women, worldwide, is breast cancer, a type of cancer associated with a huge death rate. This high mortality is mainly a consequence of the development of drug resistance, which is one of the major challenges to overcome in breast cancer treatment. As a result, research has been focused on finding novel therapeutical weapons, specifically ones that allow for a personalized treatment, based on patients’ characteristics. Although the scientific community has been concerned about guaranteeing the quality of life of cancer patients, researchers are also aware of the increasing costs related to cancer treatment, and efforts have been made to find alternatives to the development of new drugs. The development of new drugs presents some disadvantages as it is a multistep process that is time- and money-consuming, involving clinical trials that commonly fail in the initial phases. A strategy to overcome these disadvantages is drug repurposing. In this review, we focused on describing potential repurposed drugs in the therapy of breast cancer, considering their pharmacogenomic profile, to assess the relationship between patients’ genetic variations and their response to a certain therapy. This review supports the need for the development of further fundamental studies in this area, in order to investigate and expand the knowledge of the currently used and novel potential drugs to treat breast cancer. Future clinical trials should focus on developing strategies to group cancer patients according to their clinical and biological similarities and to discover new potential targets, to enable cancer therapy to be more effective and personalized.
Collapse
Affiliation(s)
- Rafaela Rodrigues
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (D.D.)
| | - Diana Duarte
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (D.D.)
- Faculty of Pharmacy of University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (D.D.)
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Associate Laboratory RISE–Health Research Network, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Correspondence: ; Tel.: +351-220426537
| |
Collapse
|
4502
|
Yu B, Dai W, Pang L, Sang Q, Li F, Yu J, Feng H, Li J, Hou J, Yan C, Su L, Zhu Z, Li YY, Liu B. The dynamic alteration of transcriptional regulation by crucial TFs during tumorigenesis of gastric cancer. Mol Med 2022; 28:41. [PMID: 35421923 PMCID: PMC9008954 DOI: 10.1186/s10020-022-00468-7] [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: 12/12/2021] [Accepted: 04/04/2022] [Indexed: 11/26/2022] Open
Abstract
Background The mechanisms of Gastric cancer (GC) initiation and progression are complicated, at least partly owing to the dynamic changes of gene regulation during carcinogenesis. Thus, investigations on the changes in regulatory networks can improve the understanding of cancer development and provide novel insights into the molecular mechanisms of cancer. Methods Differential co-expression analysis (DCEA), differential gene regulation network (GRN) modeling and differential regulation analysis (DRA) were integrated to detect differential transcriptional regulation events between gastric normal mucosa and cancer samples based on GSE54129 dataset. Cytological experiments and IHC staining assays were used to validate the dynamic changes of CREB1 regulated targets in different stages. Results A total of 1955 differentially regulated genes (DRGs) were identified and prioritized in a quantitative way. Among the top 1% DRGs, 14 out of 19 genes have been reported to be GC relevant. The four transcription factors (TFs) among the top 1% DRGs, including CREB1, BPTF, GATA6 and CEBPA, were regarded as crucial TFs relevant to GC progression. The differentially regulated links (DRLs) around the four crucial TFs were then prioritized to generate testable hypotheses on the differential regulation mechanisms of gastric carcinogenesis. To validate the dynamic alterations of gene regulation patterns of crucial TFs during GC progression, we took CREB1 as an example to screen its differentially regulated targets by using cytological and IHC staining assays. Eventually, TCEAL2 and MBNL1 were proved to be differentially regulated by CREB1 during tumorigenesis of gastric cancer. Conclusions By combining differential networking information and molecular cell experiments verification, testable hypotheses on the regulation mechanisms of GC around the core TFs and their top ranked DRLs were generated. Since TCEAL2 and MBNL1 have been reported to be potential therapeutic targets in SCLC and breast cancer respectively, their translation values in GC are worthy of further investigation. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00468-7.
Collapse
|
4503
|
Pan X, Wang J, Guo L, Na F, Du J, Chen X, Zhong A, Zhao L, Zhang L, Zhang M, Wan X, Wang M, Liu H, Dai S, Tan P, Chen J, Liu Y, Hu B, Chen C. Identifying a confused cell identity for esophageal squamous cell carcinoma. Signal Transduct Target Ther 2022; 7:122. [PMID: 35418165 PMCID: PMC9008022 DOI: 10.1038/s41392-022-00946-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 02/07/2023] Open
Abstract
The cell identity of malignant cells and how they acquire it are fundamental for our understanding of cancer. Here, we report that esophageal squamous cell carcinoma (ESCC) cells display molecular features equally similar but distinct to all three types of normal esophageal epithelial cells, which we term as confused cell identity (CCI). CCI is an independent prognostic marker associated with poor prognosis in ESCC. Further, we identify tropomyosin 4 (TPM4) as a critical CCI gene that promotes the aggressiveness of ESCC in vitro and in vivo. And TPM4 creates CCI through activating the Jak/STAT-SOX2 pathway. Thus, our study suggests an unrecognized feature of ESCC cells, which might be of value for clinic prognosis and potential interference.
Collapse
Affiliation(s)
- Xiangyu Pan
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Linjie Guo
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feifei Na
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiajia Du
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuelan Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ailing Zhong
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Zhao
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Zhang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mengsha Zhang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xudong Wan
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Manli Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongyu Liu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Siqi Dai
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Tan
- Dpartment of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyao Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Liu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Bing Hu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Chong Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
4504
|
Teng K, Jackson HW. Genetic perturbations go spatial. CELL GENOMICS 2022; 2:100120. [PMID: 36776528 PMCID: PMC9903767 DOI: 10.1016/j.xgen.2022.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Tissue-tumor interactivity is the culmination of cell intrinsic features and their extrinsic interactions with the environment. Recently in Cell, Dhainaut and Rose et al. established a strategy to track pooled CRISPR-modified cells in vivo using protein barcodes (Pro-Codes) and measure their impact on the tumor microenvironment through multiplexed imaging and spatial transcriptomics of intact tissues.1.
Collapse
Affiliation(s)
- Katie Teng
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Hartland Warren Jackson
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada,Ontario Institute for Cancer Research, Toronto, ON, Canada,Corresponding author
| |
Collapse
|
4505
|
Saltarella I, Apollonio B, Lamanuzzi A, Desantis V, Mariggiò MA, Desaphy JF, Vacca A, Frassanito MA. The Landscape of lncRNAs in Multiple Myeloma: Implications in the "Hallmarks of Cancer", Clinical Perspectives and Therapeutic Opportunities. Cancers (Basel) 2022; 14:cancers14081963. [PMID: 35454868 PMCID: PMC9032822 DOI: 10.3390/cancers14081963] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is an aggressive hematological neoplasia caused by the uncontrolled proliferation of aberrant plasmacells. Neoplastic transformation and progression are driven by a number of biological processes, called ‘hallmarks of cancer’, which are regulated by different molecules, including long non-coding RNAs. A deeper understanding of the mechanisms that regulate MM development and progression will help to improve patients stratification and management, and promote the identification of new therapeutic targets. Abstract Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nucleotides that are not translated into proteins. Nowadays, lncRNAs are gaining importance as key regulators of gene expression and, consequently, of several biological functions in physiological and pathological conditions, including cancer. Here, we point out the role of lncRNAs in the pathogenesis of multiple myeloma (MM). We focus on their ability to regulate the biological processes identified as “hallmarks of cancer” that enable malignant cell transformation, early tumor onset and progression. The aberrant expression of lncRNAs in MM suggests their potential use as clinical biomarkers for diagnosis, patient stratification, and clinical management. Moreover, they represent ideal candidates for therapeutic targeting.
Collapse
Affiliation(s)
- Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine “Guido Baccelli”, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy; (I.S.); (B.A.); (A.L.); (V.D.); (A.V.)
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy;
| | - Benedetta Apollonio
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine “Guido Baccelli”, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy; (I.S.); (B.A.); (A.L.); (V.D.); (A.V.)
| | - Aurelia Lamanuzzi
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine “Guido Baccelli”, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy; (I.S.); (B.A.); (A.L.); (V.D.); (A.V.)
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine “Guido Baccelli”, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy; (I.S.); (B.A.); (A.L.); (V.D.); (A.V.)
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy;
| | - Maria Addolorata Mariggiò
- Department of Biomedical Sciences and Human Oncology, Unit of General Pathology, University of Bari Aldo Moro, I-70124 Bari, Italy;
| | - Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy;
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine “Guido Baccelli”, University of Bari Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy; (I.S.); (B.A.); (A.L.); (V.D.); (A.V.)
| | - Maria Antonia Frassanito
- Department of Biomedical Sciences and Human Oncology, Unit of General Pathology, University of Bari Aldo Moro, I-70124 Bari, Italy;
- Correspondence:
| |
Collapse
|
4506
|
da Silva EL, Mesquita FP, de Sousa Portilho AJ, Bezerra ECA, Daniel JP, Aranha ESP, Farran S, de Vasconcellos MC, de Moraes MEA, Moreira-Nunes CA, Montenegro RC. Differences in glucose concentration shows new perspectives in gastric cancer metabolism. Toxicol In Vitro 2022; 82:105357. [PMID: 35427737 DOI: 10.1016/j.tiv.2022.105357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 12/06/2022]
Abstract
Gastric cancer (GC) is among the deadliest cancers worldwide despite available therapies, highlighting the need for novel therapies and pharmacological agents. Metabolic deregulation is a potential study area for new anticancer targets, but the in vitro metabolic studies are controversial, as different ranges of glucose used in the culture medium can influence results. In this study, we evaluated cellular viability, glucose uptake, and LDH activity in gastric cell lines when exposed to different glucose concentrations: high (HG, 25 mM), low (LG, 5.5 mM), and free (FG, 0 mM) glucose mediums. Moreover, we evaluated how glucose variations may influence cellular phenotype and the expression of genes related to epithelial-mesenchymal transition (EMT), metabolism, and cancer development in metastatic GC cells (AGP-01). Results showed that in the FG metastatic cells evidenced higher viability when compared with other cell lines and that when exposed to either LG or HG mediums most of the phenotypic assays did not differ. However, cells exposed to LG increased colony formation and mRNA levels of metabolic-related genes when compared to HG medium. Our results recommend LG medium to metabolic studies once glucose concentration is closer to physiological levels. These findings are important to point out new relevant targets in metabolic reprogramming that can be alternatives to current chemotherapies in patients with metastatic GC.
Collapse
Affiliation(s)
- Emerson Lucena da Silva
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Felipe Pantoja Mesquita
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Adrhyann Jullyanne de Sousa Portilho
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Emanuel Cintra Austregésilo Bezerra
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Julio Paulino Daniel
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Elenn Suzany Pereira Aranha
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Av. General Rodrigo Octavio Jordão Ramos, 1200 - Coroado, Manaus, Brazil
| | - Sarah Farran
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center - Riad El-Solh, Beirut, Lebanon
| | - Marne Carvalho de Vasconcellos
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Av. General Rodrigo Octavio Jordão Ramos, 1200 - Coroado, Manaus, Brazil
| | - Maria Elisabete Amaral de Moraes
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Caroline Aquino Moreira-Nunes
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Raquel Carvalho Montenegro
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil.
| |
Collapse
|
4507
|
Liao R, Shi YJ, Chuong MD, Cao J. Editorial: Gastrointestinal Tumor Heterogeneity and Related Anti-Cancer Strategies. Front Oncol 2022; 12:873240. [PMID: 35480121 PMCID: PMC9035924 DOI: 10.3389/fonc.2022.873240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rui Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu-Jun Shi
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission (NHC), West China Hospital, Sichuan University, Chengdu, China
| | - Michael D. Chuong
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, United States
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4508
|
Advances in aptamer-based nuclear imaging. Eur J Nucl Med Mol Imaging 2022; 49:2544-2559. [PMID: 35394153 DOI: 10.1007/s00259-022-05782-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/26/2022] [Indexed: 01/12/2023]
Abstract
Aptamers are short oligonucleotides that bind to specific target molecules. They have been extensively explored in biomedical applications, including biosensing, medical imaging, and disease treatment. Their adjustable affinity for specific biomarkers stimulates more translational efforts, such as nuclear imaging of tumors in preclinical and clinical settings. In this review, we present recent advances of aptamer-based nuclear imaging and compare aptamer tracers with other biogenic probes in forms of peptides, nanobodies, monoclonal antibodies, and antibody fragments. Fundamental properties of aptamer-based radiotracers are highlighted and potential directions to improve aptamer's imaging performance are discussed. Despite many translational obstacles to overcome, we envision aptamers to be a versatile tool for cancer nuclear imaging in the near future.
Collapse
|
4509
|
Impact of Lipid Metabolism on Antitumor Immune Response. Cancers (Basel) 2022; 14:cancers14071850. [PMID: 35406621 PMCID: PMC8997602 DOI: 10.3390/cancers14071850] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary One of the causes of failure of anticancer therapies is the reprogramming of lipid metabolism. Cells of innate and adaptive immunity present in the tumor microenvironment can be affected by this metabolic switch and thus present changes in their anti- or protumor phenotype. In this review, modifications induced by lipid metabolism will be described for innate immune cells, such as macrophages, dendritic cells and MDSCs, and also for adaptive immune cells, such as CD4+ and CD8+ T cells and B cells. Finally, antitumor therapeutic strategies targeting lipid metabolism will be presented. Abstract Over the past decade, metabolic reprogramming has been defined as a hallmark of cancer. More recently, a large number of studies have demonstrated that metabolic reprogramming can modulate the differentiation and functions of immune cells, and thus modify the antitumor response. Increasing evidence suggests that modified energy metabolism could be responsible for the failure of antitumor immunity. Indeed, tumor-infiltrating immune cells play a key role in cancer, and metabolic switching in these cells has been shown to help determine their phenotype: tumor suppressive or immune suppressive. Recent studies in the field of immunometabolism focus on metabolic reprogramming in the tumor microenvironment (TME) by targeting innate and adaptive immune cells and their associated anti- or protumor phenotypes. In this review, we discuss the lipid metabolism of immune cells in the TME as well as the effects of lipids; finally, we expose the link between therapies and lipid metabolism.
Collapse
|
4510
|
Wang Z, Zhao S, Lin X, Chen G, Kang J, Ma Z, Wang Y, Li Z, Xiao X, He A, Xiang D. Application of Organoids in Carcinogenesis Modeling and Tumor Vaccination. Front Oncol 2022; 12:855996. [PMID: 35371988 PMCID: PMC8968694 DOI: 10.3389/fonc.2022.855996] [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: 01/16/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Organoids well recapitulate organ-specific functions from their tissue of origin and remain fundamental aspects of organogenesis. Organoids are widely applied in biomedical research, drug discovery, and regenerative medicine. There are various cultivated organoid systems induced by adult stem cells and pluripotent stem cells, or directly derived from primary tissues. Researchers have drawn inspiration by combination of organoid technology and tissue engineering to produce organoids with more physiological relevance and suitable for translational medicine. This review describes the value of applying organoids for tumorigenesis modeling and tumor vaccination. We summarize the application of organoids in tumor precision medicine. Extant challenges that need to be conquered to make this technology be more feasible and precise are discussed.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shasha Zhao
- State Key Laboratory of Oncogenes and Related Genes, the Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaolin Lin
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglong Chen
- Department of General Surgery, Zhengzhou University, Affiliated Cancer Hospital (Henan Cancer Hospital), Zhengzhou, China
| | - Jiawei Kang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | | | - Yiming Wang
- Shanghai OneTar Biomedicine, Shanghai, China
| | - Zhi Li
- Department of General Surgery, Zhengzhou University, Affiliated Cancer Hospital (Henan Cancer Hospital), Zhengzhou, China
| | - Xiuying Xiao
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aina He
- Department of Oncology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dongxi Xiang
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, The Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
4511
|
Cerneckis J, Cui Q, He C, Yi C, Shi Y. Decoding pseudouridine: an emerging target for therapeutic development. Trends Pharmacol Sci 2022; 43:522-535. [DOI: 10.1016/j.tips.2022.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/12/2022] [Accepted: 03/22/2022] [Indexed: 01/18/2023]
|
4512
|
Phenotypic plasticity during metastatic colonization. Trends Cell Biol 2022; 32:854-867. [DOI: 10.1016/j.tcb.2022.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/20/2022]
|
4513
|
Thomas J, Sonpavde G. Molecularly Targeted Therapy towards Genetic Alterations in Advanced Bladder Cancer. Cancers (Basel) 2022; 14:1795. [PMID: 35406567 PMCID: PMC8997162 DOI: 10.3390/cancers14071795] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Despite the introduction of immune checkpoint inhibitors and antibody-drug conjugates to the management of advanced urothelial carcinoma, the disease is generally incurable. The increasing incorporation of next-generation sequencing of tumor tissue into the characterization of bladder cancer has led to a better understanding of the somatic genetic aberrations potentially involved in its pathogenesis. Genetic alterations have been observed in kinases, such as FGFRs, ErbBs, PI3K/Akt/mTOR, and Ras-MAPK, and genetic alterations in critical cellular processes, such as chromatin remodeling, cell cycle regulation, and DNA damage repair. However, activating mutations or fusions of FGFR2 and FGFR3 remains the only validated therapeutically actionable alteration, with erdafitinib as the only targeted agent currently approved for this group. Bladder cancer is characterized by genomic heterogeneity and a high tumor mutation burden. This review highlights the potential relevance of aberrations and discusses the current status of targeted therapies directed at them.
Collapse
Affiliation(s)
- Jonathan Thomas
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Guru Sonpavde
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
| |
Collapse
|
4514
|
Cascone T, Chelvanambi M, Wargo JA. Immunotherapy response-associated Akkermansia: canary in a coal mine? Trends Immunol 2022; 43:337-339. [DOI: 10.1016/j.it.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
|
4515
|
Maiorano BA, Lorusso D, Maiorano MFP, Ciardiello D, Parrella P, Petracca A, Cormio G, Maiello E. The Interplay between PARP Inhibitors and Immunotherapy in Ovarian Cancer: The Rationale behind a New Combination Therapy. Int J Mol Sci 2022; 23:3871. [PMID: 35409229 PMCID: PMC8998760 DOI: 10.3390/ijms23073871] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer (OC) has a high impact on morbidity and mortality in the female population. Survival is modest after platinum progression. Therefore, the search for new therapeutic strategies is of utmost importance. BRCA mutations and HR-deficiency occur in around 50% of OC, leading to increased response and survival after Poly (ADP-ribose) polymerase inhibitors (PARPis) administration. PARPis represent a breakthrough for OC therapy, with three different agents approved. On the contrary, immune checkpoint inhibitors (ICIs), another breakthrough therapy for many solid tumors, led to modest results in OC, without clinical approvals and even withdrawal of clinical trials. Therefore, combinations aiming to overcome resistance mechanisms have become of great interest. Recently, PARPis have been evidenced to modulate tumor microenvironment at the molecular and cellular level, potentially enhancing ICIs responsiveness. This represents the rationale for the combined administration of PARPis and ICIs. Our review ought to summarize the preclinical and translational features that support the contemporary administration of these two drug classes, the clinical trials conducted so far, and future directions with ongoing studies.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, Foundation Casa Sollievo Della Sofferenza IRCCS, San Giovanni Rotondo, 71013 Foggia, Italy; (D.C.); (E.M.)
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Domenica Lorusso
- Gynecologic Oncology Unit, Catholic University of the Sacred Heart, Scientific Directorate, Fondazione Policlinico “A. Gemelli” IRCCS, 00168 Rome, Italy;
| | - Mauro Francesco Pio Maiorano
- Division of Obstetrics and Gynecology, Biomedical and Human Oncological Science, University of Bari “Aldo Moro”, 70121 Bari, Italy; (M.F.P.M.); (G.C.)
| | - Davide Ciardiello
- Oncology Unit, Foundation Casa Sollievo Della Sofferenza IRCCS, San Giovanni Rotondo, 71013 Foggia, Italy; (D.C.); (E.M.)
- Oncology Unit, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Paola Parrella
- Oncology Laboratory, Foundation Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Antonio Petracca
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Gennaro Cormio
- Division of Obstetrics and Gynecology, Biomedical and Human Oncological Science, University of Bari “Aldo Moro”, 70121 Bari, Italy; (M.F.P.M.); (G.C.)
| | - Evaristo Maiello
- Oncology Unit, Foundation Casa Sollievo Della Sofferenza IRCCS, San Giovanni Rotondo, 71013 Foggia, Italy; (D.C.); (E.M.)
| |
Collapse
|
4516
|
Sharma A, Sinha S, Shrivastava N. Therapeutic Targeting Hypoxia-Inducible Factor (HIF-1) in Cancer: Cutting Gordian Knot of Cancer Cell Metabolism. Front Genet 2022; 13:849040. [PMID: 35432450 PMCID: PMC9008776 DOI: 10.3389/fgene.2022.849040] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/09/2022] [Indexed: 12/20/2022] Open
Abstract
Metabolic alterations are one of the hallmarks of cancer, which has recently gained great attention. Increased glucose absorption and lactate secretion in cancer cells are characterized by the Warburg effect, which is caused by the metabolic changes in the tumor tissue. Cancer cells switch from oxidative phosphorylation (OXPHOS) to aerobic glycolysis due to changes in glucose degradation mechanisms, a process known as “metabolic reprogramming”. As a result, proteins involved in mediating the altered metabolic pathways identified in cancer cells pose novel therapeutic targets. Hypoxic tumor microenvironment (HTM) is anticipated to trigger and promote metabolic alterations, oncogene activation, epithelial-mesenchymal transition, and drug resistance, all of which are hallmarks of aggressive cancer behaviour. Angiogenesis, erythropoiesis, glycolysis regulation, glucose transport, acidosis regulators have all been orchestrated through the activation and stability of a transcription factor termed hypoxia-inducible factor-1 (HIF-1), hence altering crucial Warburg effect activities. Therefore, targeting HIF-1 as a cancer therapy seems like an extremely rational approach as it is directly involved in the shift of cancer tissue. In this mini-review, we present a brief overview of the function of HIF-1 in hypoxic glycolysis with a particular focus on novel therapeutic strategies currently available.
Collapse
Affiliation(s)
- Abhilasha Sharma
- Department of Life Science, University School of Sciences, Gujarat University, Ahmedabad, India
| | | | - Neeta Shrivastava
- Shri B.V. Patel Education Trust, Ahmedabad, India
- *Correspondence: Neeta Shrivastava,
| |
Collapse
|
4517
|
Liu J, Hong M, Li Y, Chen D, Wu Y, Hu Y. Programmed Cell Death Tunes Tumor Immunity. Front Immunol 2022; 13:847345. [PMID: 35432318 PMCID: PMC9005769 DOI: 10.3389/fimmu.2022.847345] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/28/2022] [Indexed: 12/14/2022] Open
Abstract
The demise of cells in various ways enables the body to clear unwanted cells. Studies over the years revealed distinctive molecular mechanisms and functional consequences of several key cell death pathways. Currently, the most intensively investigated programmed cell death (PCD) includes apoptosis, necroptosis, pyroptosis, ferroptosis, PANoptosis, and autophagy, which has been discovered to play crucial roles in modulating the immunosuppressive tumor microenvironment (TME) and determining clinical outcomes of the cancer therapeutic approaches. PCD can play dual roles, either pro-tumor or anti-tumor, partly depending on the intracellular contents released during the process. PCD also regulates the enrichment of effector or regulatory immune cells, thus participating in fine-tuning the anti-tumor immunity in the TME. In this review, we focused primarily on apoptosis, necroptosis, pyroptosis, ferroptosis, PANoptosis, and autophagy, discussed the released molecular messengers participating in regulating their intricate crosstalk with the immune response in the TME, and explored the immunological consequence of PCD and its implications in future cancer therapy developments.
Collapse
Affiliation(s)
- Jing Liu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Minjing Hong
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yijia Li
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Dan Chen
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yangzhe Wu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yi Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, China
| |
Collapse
|
4518
|
Seguin L, Durandy M, Feral CC. Lung Adenocarcinoma Tumor Origin: A Guide for Personalized Medicine. Cancers (Basel) 2022; 14:cancers14071759. [PMID: 35406531 PMCID: PMC8996976 DOI: 10.3390/cancers14071759] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Lung cancer is the leading cause of cancer-related death worldwide, with an average 5-year survival rate of approximately 15%. Among the multiple histological type of lung cancer, adenocarcinoma is the most common. Adenocarcinoma is characterized by a high degree of heterogeneity at many levels, including histological, cellular, and molecular. Understanding the cell of origin of adenocarcinoma, and the molecular changes during tumor progression, will allow better therapeutic strategies. Abstract Lung adenocarcinoma, the major form of lung cancer, is the deadliest cancer worldwide, due to its late diagnosis and its high heterogeneity. Indeed, lung adenocarcinoma exhibits pronounced inter- and intra-tumor heterogeneity cofounding precision medicine. Tumor heterogeneity is a clinical challenge driving tumor progression and drug resistance. Several key pieces of evidence demonstrated that lung adenocarcinoma results from the transformation of progenitor cells that accumulate genetic abnormalities. Thus, a better understanding of the cell of origin of lung adenocarcinoma represents an opportunity to unveil new therapeutic alternatives and stratify patient tumors. While the lung is remarkably quiescent during homeostasis, it presents an extensive ability to respond to injury and regenerate lost or damaged cells. As the lung is constantly exposed to potential insult, its regenerative potential is assured by several stem and progenitor cells. These can be induced to proliferate in response to injury as well as differentiate into multiple cell types. A better understanding of how genetic alterations and perturbed microenvironments impact progenitor-mediated tumorigenesis and treatment response is of the utmost importance to develop new therapeutic opportunities.
Collapse
|
4519
|
Carroll V. The vascular outsiders. Br J Cancer 2022; 126:1509-1510. [PMID: 35352021 PMCID: PMC9130215 DOI: 10.1038/s41416-022-01795-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/04/2022] [Accepted: 03/16/2022] [Indexed: 01/22/2023] Open
Abstract
A recent perspective on vessel co-option and angiotropic extravascular migratory metastasis by Lugassy et al. suggests cancers use both mechanisms sequentially during tumour growth and spread.
Collapse
Affiliation(s)
- Veronica Carroll
- Senior Lecturer in Vascular Biology, Section of Cell Biology, St George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.
| |
Collapse
|
4520
|
The Role of Circulating Tumor Cells in the Prognosis of Metastatic Triple-Negative Breast Cancers: A Systematic Review of the Literature. Biomedicines 2022; 10:biomedicines10040769. [PMID: 35453519 PMCID: PMC9025371 DOI: 10.3390/biomedicines10040769] [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: 02/15/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is one of the leading causes of death in women worldwide. One subtype of breast cancer is the triple-negative, which accounts for 15% of total breast cancer cases and is known for its poor prognosis. The main cause of death is due to metastasis. Circulating tumor cells (CTCs) play a key role in the metastatic process. CTCs arise either by detaching from the primary tumor or from cancer stem cells undergoing an epithelial-to-mesenchymal transition (EMT). This review aims to present up-to-date data concerning the role of CTC numbers in relation to the prognostic and treatment response in metastatic triple-negative breast cancer (mTNBC) patients, and also to discuss the methods used for CTCs’ identification. A search in the MEDLINE database was performed. A total of 234 articles were identified. The results of the 24 eligible studies showed that positive CTC status is associated with shorter overall survival (OS) and progression-free survival (PFS) in mTNBC patients. Furthermore, a decrease in number of CTCs during therapy seems to be a favorable prognostic factor, making CTCs’ detection an important prognostic tool before and during therapy in mTNBC patients. The methods used for CTC detection are still developing and need further improvement.
Collapse
|
4521
|
Ma C, Ma RJ, Hu K, Zheng QM, Wang YP, Zhang N, Sun ZG. The molecular mechanism of METTL3 promoting the malignant progression of lung cancer. Cancer Cell Int 2022; 22:133. [PMID: 35331234 PMCID: PMC8944087 DOI: 10.1186/s12935-022-02539-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer remains one of the major causes of cancer-related death globally. Recent studies have shown that aberrant m6A levels caused by METTL3 are involved in the malignant progression of various tumors, including lung cancer. The m6A modification, the most abundant RNA chemical modification, regulates RNA stabilization, splicing, translation, decay, and nuclear export. The methyltransferase complex plays a key role in the occurrence and development of many tumors by installing m6A modification. In this complex, METTL3 is the first identified methyltransferase, which is also the major catalytic enzyme. Recent findings have revealed that METTL3 is remarkably associated with different aspects of lung cancer progression, influencing the prognosis of patients. In this review, we will focus on the underlying mechanism of METT3 in lung cancer and predict the future work and potential clinical application of targeting METTL3 for lung cancer therapy.
Collapse
Affiliation(s)
- Chao Ma
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, 250013 Shandong China
- School of Clinical Medicine, Weifang Medical University, Weifang, 261053 Shangdong China
| | - Rui-Jie Ma
- Cheeloo College of Medicine, Shandong University, Jinan, 250013 Shangdong China
| | - Kang Hu
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, 250013 Shandong China
- School of Clinical Medicine, Weifang Medical University, Weifang, 261053 Shangdong China
| | - Qi-Ming Zheng
- Cheeloo College of Medicine, Shandong University, Jinan, 250013 Shangdong China
| | - Ye-Peng Wang
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, 250013 Shandong China
| | - Nan Zhang
- Breast Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, 250013 Shandong China
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, 250013 Shandong China
| |
Collapse
|
4522
|
Shi Z, Zheng J, Tang W, Bai Y, Zhang L, Xuan Z, Sun H, Shao C. Multifunctional Nanomaterials for Ferroptotic Cancer Therapy. Front Chem 2022; 10:868630. [PMID: 35402376 PMCID: PMC8987283 DOI: 10.3389/fchem.2022.868630] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/02/2022] [Indexed: 01/03/2023] Open
Abstract
Patient outcomes from the current clinical cancer therapy remain still far from satisfactory. However, in recent years, several biomedical discoveries and nanotechnological innovations have been made, so there is an impetus to combine these with conventional treatments to improve patient experience and disease prognosis. Ferroptosis, a term first coined in 2012, is an iron-dependent regulated cell death (RCD) based on the production of reactive oxygen species (ROS) and the consequent oxidization of polyunsaturated fatty acids (PUFAs). Many nanomaterials that can induce ferroptosis have been explored for applications in cancer therapy. In this review, we summarize the recent developments in ferroptosis-based nanomaterials for cancer therapy and discuss the future of ferroptosis, nanomedicine, and cancer therapy.
Collapse
Affiliation(s)
- Zhiyuan Shi
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jianzhong Zheng
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenbin Tang
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Bai
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lei Zhang
- School of Public Health, Xiamen Univerisity, Xiamen, China
| | - Zuodong Xuan
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Huimin Sun
- Central Laboratory, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Huimin Sun, ; Chen Shao,
| | - Chen Shao
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Huimin Sun, ; Chen Shao,
| |
Collapse
|
4523
|
Li J, Chen D, Shen M. Tumor Microenvironment Shapes Colorectal Cancer Progression, Metastasis, and Treatment Responses. Front Med (Lausanne) 2022; 9:869010. [PMID: 35402443 PMCID: PMC8984105 DOI: 10.3389/fmed.2022.869010] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most devastating diseases that accounts for numerous deaths worldwide. Tumor cell-autonomous pathways, such as the oncogenic signaling activation, significantly contribute to CRC progression and metastasis. Recent accumulating evidence suggests that the CRC microenvironment also profoundly promotes or represses this process. As the roles of the tumor microenvironment (TME) in CRC progression and metastasis is gradually uncovered, the importance of these non-cell-autonomous signaling pathways is appreciated. However, we are still at the beginning of this TME function exploring process. In this review, we summarize the current understanding of the TME in CRC progression and metastasis by focusing on the gut microbiota and host cellular and non-cellular components. We also briefly discuss TME-remodeling therapies in CRC.
Collapse
Affiliation(s)
- Jun Li
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dawei Chen
- Wayne State University School of Medicine, Detroit, MI, United States
| | - Minhong Shen
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Oncology, Wayne State University School of Medicine and Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, United States
- *Correspondence: Minhong Shen,
| |
Collapse
|
4524
|
Varela L, Garcia-Rendueles MER. Oncogenic Pathways in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23063223. [PMID: 35328644 PMCID: PMC8952192 DOI: 10.3390/ijms23063223] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer and neurodegenerative diseases are two of the leading causes of premature death in modern societies. Their incidence continues to increase, and in the near future, it is believed that cancer will kill more than 20 million people per year, and neurodegenerative diseases, due to the aging of the world population, will double their prevalence. The onset and the progression of both diseases are defined by dysregulation of the same molecular signaling pathways. However, whereas in cancer, these alterations lead to cell survival and proliferation, neurodegenerative diseases trigger cell death and apoptosis. The study of the mechanisms underlying these opposite final responses to the same molecular trigger is key to providing a better understanding of the diseases and finding more accurate treatments. Here, we review the ten most common signaling pathways altered in cancer and analyze them in the context of different neurodegenerative diseases such as Alzheimer's (AD), Parkinson's (PD), and Huntington's (HD) diseases.
Collapse
Affiliation(s)
- Luis Varela
- Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, School of Medicine, Yale University, 310 Cedar St. BML 330, New Haven, CT 06520, USA
- Correspondence: (L.V.); (M.E.R.G.-R.)
| | - Maria E. R. Garcia-Rendueles
- Precision Nutrition and Cancer Program, IMDEA Food Institute, Campus Excelencia Internacional UAM+CSIC, 28049 Madrid, Spain
- Correspondence: (L.V.); (M.E.R.G.-R.)
| |
Collapse
|
4525
|
Kumar-Sinha C, Chinnaiyan AM. Defining cancer growth beyond the mitotic index. Nat Cell Biol 2022; 24:285-287. [PMID: 35292782 DOI: 10.1038/s41556-022-00862-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA. .,Department of Pathology, University of Michigan, Ann Arbor, MI, USA. .,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA. .,Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
4526
|
Malagraba G, Yarmohammadi M, Javed A, Barceló C, Rubio-Tomás T. The Role of LSD1 and LSD2 in Cancers of the Gastrointestinal System: An Update. Biomolecules 2022; 12:462. [PMID: 35327654 PMCID: PMC8946813 DOI: 10.3390/biom12030462] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023] Open
Abstract
Epigenetic mechanisms are known to play a key role in cancer progression. Specifically, histone methylation involves reversible post-translational modification of histones that govern chromatin structure remodelling, genomic imprinting, gene expression, DNA damage repair, and meiotic crossover recombination, among other chromatin-based activities. Demethylases are enzymes that catalyse the demethylation of their substrate using a flavin adenine dinucleotide-dependent amine oxidation process. Lysine-specific demethylase 1 (LSD1) and its homolog, lysine-specific demethylase 2 (LSD2), are overexpressed in a variety of human cancer types and, thus, regulate tumour progression. In this review, we focus on the literature from the last 5 years concerning the role of LSD1 and LSD2 in the main gastrointestinal cancers (i.e., gastric cancer, liver cancer, pancreatic cancer, and colorectal cancer).
Collapse
Affiliation(s)
- Gianluca Malagraba
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBA), 07120 Palma de Mallorca, Spain;
| | - Mahdieh Yarmohammadi
- Central Tehran Branch, Department of Biology, Faculty of Sciences, Islamic Azad University, Tehran 1955847881, Iran;
| | - Aadil Javed
- Cancer Biology Laboratory, Department of Bioengineering, Faculty of Engineering, Ege University, Izmir 35040, Turkey;
| | - Carles Barceló
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBA), 07120 Palma de Mallorca, Spain;
| | - Teresa Rubio-Tomás
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- School of Medicine, University of Crete, 70013 Herakleion, Crete, Greece
| |
Collapse
|
4527
|
Wu Y, Zhang T, Zhang X, Gao Q. Decoding the complexity of metastasis. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0031. [PMID: 35289156 PMCID: PMC8958888 DOI: 10.20892/j.issn.2095-3941.2022.0031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/21/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Yingcheng Wu
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Tiancheng Zhang
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaoming Zhang
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200433, China
| |
Collapse
|
4528
|
Gorostiola González M, Janssen APA, IJzerman AP, Heitman LH, van Westen GJP. Oncological drug discovery: AI meets structure-based computational research. Drug Discov Today 2022; 27:1661-1670. [PMID: 35301149 DOI: 10.1016/j.drudis.2022.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 01/22/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023]
Abstract
The integration of machine learning and structure-based methods has proven valuable in the past as a way to prioritize targets and compounds in early drug discovery. In oncological research, these methods can be highly beneficial in addressing the diversity of neoplastic diseases portrayed by the different hallmarks of cancer. Here, we review six use case scenarios for integrated computational methods, namely driver prediction, computational mutagenesis, (off)-target prediction, binding site prediction, virtual screening, and allosteric modulation analysis. We address the heterogeneity of integration approaches and individual methods, while acknowledging their current limitations and highlighting their potential to bring drugs for personalized oncological therapies to the market faster.
Collapse
Affiliation(s)
- Marina Gorostiola González
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Antonius P A Janssen
- Oncode Institute, Utrecht, The Netherlands; Molecular Physiology, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Gerard J P van Westen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands.
| |
Collapse
|
4529
|
Ansari RA, Abbas A. Editorial: Epigenetic and Transcriptional Dysregulations in Cancer and Therapeutic Opportunities. Front Genet 2022; 13:857380. [PMID: 35273642 PMCID: PMC8901498 DOI: 10.3389/fgene.2022.857380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rais A Ansari
- Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Ata Abbas
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
4530
|
Pasello G, Remon J, Felley-Bosco E. Editorial: Understanding the Interplay Between the Tumor Immune Microenvironment and Genetic Alterations in Thoracic Malignancies. Front Oncol 2022; 12:871544. [PMID: 35359422 PMCID: PMC8960239 DOI: 10.3389/fonc.2022.871544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Giulia Pasello
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 2, Istituto Oncologico Veneto Instituto di Ricerca e Cura a Carattere Scientifico (IOV IRCCS), Padua, Italy
- *Correspondence: Giulia Pasello, ;
| | - Jordi Remon
- Department of Medical Oncology, Centro Integral Oncológico Clara Campal (HM-CIOCC), Hospital HM Nou Delfos, HM Hospitales, Barcelona, Spain
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
4531
|
A comprehensive systematic review and network meta-analysis: the role of anti-angiogenic agents in advanced epithelial ovarian cancer. Sci Rep 2022; 12:3803. [PMID: 35264616 PMCID: PMC8907284 DOI: 10.1038/s41598-022-07731-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
The efficacy of anti-angiogenic agents (AAAs) in epithelial ovarian cancer (EOC) remains unclear. Therefore, we conducted a systematic review and network meta-analysis (NMA) to synthesize evidence of their comparative effectiveness for improving overall survival (OS) among EOC patients. We searched six databases for randomized controlled trials (RCTs) from their inception to February 2021. We performed an NMA with hazard ratios (HRs) and 95%-confidence intervals (CIs) to evaluate comparative effectiveness among different AAAs in chemotherapy-naïve and recurrent EOC. P-score was used to provide an effectiveness hierarchy ranking. Sensitivity NMA was carried out by focusing on studies that reported high-risk chemotherapy-naïve, platinum-resistant, and platinum-sensitive EOC. The primary outcome was OS. We identified 23 RCTs that assessed the effectiveness of AAAs. In recurrent EOC, concurrent use of trebananib (10 mg/kg) with chemotherapy was likely to be the best option (P-score: 0.88, HR 1.67, 95% CI 0.94; 2.94). The NMA indicated that bevacizumab plus chemotherapy followed by maintenance bevacizumab (P-score: 0.99) and pazopanib combined with chemotherapy (P-score: 0.79) both had the highest probability of being the best intervention for improving OS in high-risk chemotherapy-naïve and platinum-resistant EOC, respectively. AAAs may not play a significant clinical role in non-high-risk chemotherapy-naïve and platinum-sensitive EOC.
Collapse
|
4532
|
Cytokine-Induced Senescence in the Tumor Microenvironment and Its Effects on Anti-Tumor Immune Responses. Cancers (Basel) 2022; 14:cancers14061364. [PMID: 35326515 PMCID: PMC8946098 DOI: 10.3390/cancers14061364] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 02/04/2023] Open
Abstract
In contrast to surgical excision, chemotherapy or radiation therapy, immune checkpoint blockade therapies primarily influence cells in the tumor microenvironment, especially the tumor-associated lymphocytes and antigen-presenting cells. Besides complete remission of tumor lesions, in some patients, early tumor regression is followed by a consolidation phase where residing tumors remain dormant. Whereas the cytotoxic mechanisms of the regression phase (i.e., apoptosis, necrosis, necroptosis, and immune cell-mediated cell death) have been extensively described, the mechanisms underlying the dormant state are still a matter of debate. Here, we propose immune-mediated induction of senescence in cancers as one important player. Senescence can be achieved by tumor-associated antigen-specific T helper 1 cells, cytokines or antibodies targeting immune checkpoints. This concept differs from cytotoxic treatment, which often targets the genetic makeup of cancer cells. The immune system's ability to establish "defensive walls" around tumors also places the tumor microenvironment into the fight against cancer. Those "defensive walls" isolate the tumor cells instead of increasing the selective pressure. They also keep the tumor cells in a non-proliferating state, thereby correcting the derailed tissue homeostasis. In conclusion, strengthening the senescence surveillance of tumors by the immune cells of the microenvironment is a future goal to dampen this life-threatening disease.
Collapse
|
4533
|
Deciphering Tumour Heterogeneity: From Tissue to Liquid Biopsy. Cancers (Basel) 2022; 14:cancers14061384. [PMID: 35326534 PMCID: PMC8946040 DOI: 10.3390/cancers14061384] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Most malignant tumours are highly heterogeneous at molecular and phenotypic levels. Tumour variability poses challenges for the management of patients, as it arises between patients and even evolves in space and time within a single patient. Currently, treatment-decision making usually relies on the molecular characteristics of a limited tumour tissue sample at the time of diagnosis or disease progression but does not take into account the complexity of the bulk tumours and their constant evolution over time. In this review, we explore the extent of tumour heterogeneity and report the mechanisms that promote and sustain this diversity in cancers. We summarise the clinical strikes of tumour diversity in the management of patients with cancer. Finally, we discuss the current material and technological approaches that are relevant to adequately appreciate tumour heterogeneity. Abstract Human solid malignancies harbour a heterogeneous set of cells with distinct genotypes and phenotypes. This heterogeneity is installed at multiple levels. A biological diversity is commonly observed between tumours from different patients (inter-tumour heterogeneity) and cannot be fully captured by the current consensus molecular classifications for specific cancers. To extend the complexity in cancer, there are substantial differences from cell to cell within an individual tumour (intra-tumour heterogeneity, ITH) and the features of cancer cells evolve in space and time. Currently, treatment-decision making usually relies on the molecular characteristics of a limited tumour tissue sample at the time of diagnosis or disease progression but does not take into account the complexity of the bulk tumours and their constant evolution over time. In this review, we explore the extent of tumour heterogeneity with an emphasis on ITH and report the mechanisms that promote and sustain this diversity in cancers. We summarise the clinical strikes of ITH in the management of patients with cancer. Finally, we discuss the current material and technological approaches that are relevant to adequately appreciate ITH.
Collapse
|
4534
|
Hishida T, Vazquez-Ferrer E, Hishida-Nozaki Y, Takemoto Y, Hatanaka F, Yoshida K, Prieto J, Sahu SK, Takahashi Y, Reddy P, O’Keefe DD, Rodriguez Esteban C, Knoepfler PS, Nuñez Delicado E, Castells A, Campistol JM, Kato R, Nakagawa H, Izpisua Belmonte JC. Myc Supports Self-Renewal of Basal Cells in the Esophageal Epithelium. Front Cell Dev Biol 2022; 10:786031. [PMID: 35309931 PMCID: PMC8931341 DOI: 10.3389/fcell.2022.786031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/04/2022] [Indexed: 11/17/2022] Open
Abstract
It is widely believed that cellular senescence plays a critical role in both aging and cancer, and that senescence is a fundamental, permanent growth arrest that somatic cells cannot avoid. Here we show that Myc plays an important role in self-renewal of esophageal epithelial cells, contributing to their resistance to cellular senescence. Myc is homogeneously expressed in basal cells of the esophageal epithelium and Myc positively regulates their self-renewal by maintaining their undifferentiated state. Indeed, Myc knockout induced a loss of the undifferentiated state of esophageal epithelial cells resulting in cellular senescence while forced MYC expression promoted oncogenic cell proliferation. A superoxide scavenger counteracted Myc knockout-induced senescence, therefore suggesting that a mitochondrial superoxide takes part in inducing senescence. Taken together, these analyses reveal extremely low levels of cellular senescence and senescence-associated phenotypes in the esophageal epithelium, as well as a critical role for Myc in self-renewal of basal cells in this organ. This provides new avenues for studying and understanding the links between stemness and resistance to cellular senescence.
Collapse
Affiliation(s)
- Tomoaki Hishida
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
- Laboratory of Biological Chemistry, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Eric Vazquez-Ferrer
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Yuriko Hishida-Nozaki
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Yuto Takemoto
- Department of Basic Medical Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Fumiyuki Hatanaka
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Kei Yoshida
- Department of Basic Medical Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Javier Prieto
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Sanjeeb Kumar Sahu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Yuta Takahashi
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Pradeep Reddy
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - David D. O’Keefe
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
| | | | - Paul S. Knoepfler
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA, United States
| | | | - Antoni Castells
- Gastroenterology Department, Hospital Clinic, CIBEREHD, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Josep M. Campistol
- Gastroenterology Department, Hospital Clinic, CIBEREHD, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Ryuji Kato
- Department of Basic Medical Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Hiroshi Nakagawa
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, Philadelphia, PA, United States
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States
- *Correspondence: Juan Carlos Izpisua Belmonte,
| |
Collapse
|
4535
|
Plundrich D, Chikhladze S, Fichtner-Feigl S, Feuerstein R, Briquez PS. Molecular Mechanisms of Tumor Immunomodulation in the Microenvironment of Colorectal Cancer. Int J Mol Sci 2022; 23:2782. [PMID: 35269922 PMCID: PMC8910988 DOI: 10.3390/ijms23052782] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer remains one of the most important health challenges in our society. The development of cancer immunotherapies has fostered the need to better understand the anti-tumor immune mechanisms at play in the tumor microenvironment and the strategies by which the tumor escapes them. In this review, we provide an overview of the molecular interactions that regulate tumor inflammation. We particularly discuss immunomodulatory cell-cell interactions, cell-soluble factor interactions, cell-extracellular matrix interactions and cell-microbiome interactions. While doing so, we highlight relevant examples of tumor immunomodulation in colorectal cancer.
Collapse
Affiliation(s)
- Dorothea Plundrich
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sophia Chikhladze
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Department of Biomedical Sciences, Cedars-Sinai Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 900048, USA
- Department of Medicine, Cedars-Sinai Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 900048, USA
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Reinhild Feuerstein
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Priscilla S Briquez
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
4536
|
Solasonine Induces Apoptosis and Inhibits Proliferation of Bladder Cancer Cells by Suppressing NRP1 Expression. JOURNAL OF ONCOLOGY 2022; 2022:7261486. [PMID: 35281516 PMCID: PMC8906937 DOI: 10.1155/2022/7261486] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/11/2022] [Indexed: 12/16/2022]
Abstract
Solasonine, a steroidal alkaloid extracted from Solanum nigrum L., has been found to exert inhibitory effect on cancers. However, the underlying anticancer mechanisms of solasonine, particularly in urinary bladder cancer (BC), remain unclear. In this study, we identified the potential targets and biological functions associated with solasonine activity using a bioinformatics approach. Ingenuity pathway analysis revealed that neuropilin-1 (NRP1) and other signaling pathways, such as PI3K/AKT and ERK/MAPK pathways, were potentially involved in the therapeutic effects of solasonine. The ability of solasonine in inducing apoptosis and inhibiting proliferation in BC cells was confirmed experimentally, and the inhibition of ERK/MAPK, P38/MAPK, and PI3K/AKT pathways was validated by Western blot. Mechanistically, solasonine suppressed the expression of NRP1 protein, but not that of mRNA. Further results of molecular docking and molecular dynamics simulation analysis indicated that solasonine could directly bind to the b1 domain of NRP1 protein with a reasonable and stable docking conformation. We previously found that targeting NRP1 is a potential antitumor strategy. Combined with these findings, it can be speculated that the binding of solasonine with NRP1 on the cell membrane could prevent the formation of NRP1/VEGFA/VEGFR2 and NRP1/EGFR complexes, resulting in the inhibition of downstream signaling, including ERK/MAPK, P38/MAPK, and PI3K/AKT pathways. Additionally, intracellular solasonine could inhibit the membrane localization of NRP1 and provoke its cytoplasmic retention, facilitating the degradation of NRP1 protein in the cytoplasm. The dual effects induced by the binding of solasonine to NRP1 extracellularly and intracellularly could account for the antiproliferative and proapoptotic effects of solasonine on BC.
Collapse
|
4537
|
Abstract
In this interview, Professor Peter W Laird speaks with Storm Johnson, Commissioning Editor for Epigenomics, on his work to date in the field of cancer epigenetics. Dr Peter W Laird is a Professor at Van Andel Institute (VAI) in Grand Rapids, Michigan. He earned his B.S. and M.S., Cum Laude, from the University of Leiden, The Netherlands. He trained for his PhD with Dr Piet Borst, The Netherlands Cancer Institute, and as a postdoc with Dr Anton Berns, The Netherlands Cancer Institute, and with Dr Rudolf Jaenisch, at the Whitehead Institute for Biomedical Research in Cambridge, MA, USA. He joined the faculty at the University of Southern California in 1996, where he served as the Founding Director of the USC Epigenome Center and also as the Leader of the Epigenetics and Regulation Program of the Norris Comprehensive Cancer Center. In 2014, he relocated to VAI to join Dr Peter Jones in building an internationally acclaimed research center focused on Epigenetics. Dr Laird published the first demonstration of the causal role for DNA methylation in oncogenesis (Cell, 1995) [1]. He served as the Principal Investigator for all DNA methylation data production for the Cancer Genome Atlas (TCGA) and led many TCGA analysis efforts. He has been awarded 10 patents related to DNA methylation technology by the United States Patent and Trademark Office, one of which is the basis for the first US FDA-approved blood-based DNA methylation assay for cancer (Epi proColon). His research findings include the report of a close link between DNA methylation and BRAF mutation in colorectal cancer (Nature Genetics, 2006) [2], the discovery that embryonic stem cell polycomb repressor targets are predisposed to abnormal DNA methylation in cancer (Nature Genetics, 2007) [3], the identification of a novel epigenetic subtype of glioma (G-CIMP), tightly associated with IDH1 mutation (Cancer Cell, 2010) [4], and the connection between nuclear architecture, late replication, and domains of epigenetic instability (Nature Genetics, 2011) [5], later showing a link with mitotic cell division, thus providing a mechanistic explanation for the loss of DNA methylation in aging and cancer first described four decades ago (Nature Genetics, 2018) [6].
Collapse
|
4538
|
Castañeda C, Castillo M, Bernabe L, Suarez N, Fassan M, Sanchez J, Tello K, Alatrista R, Chavez I, Ruiz E, Bazan Y, Barreda F, Valdivia D, Meng W, Chakravarti A, Sanchez J, Taxa L, Montenegro P. The relationship between tumour infiltrating lymphocytes, Epstein-Barr virus and Helicobacter pylori infection in gastric cancer. Ecancermedicalscience 2022; 16:1362. [PMID: 35685959 PMCID: PMC9085163 DOI: 10.3332/ecancer.2022.1362] [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: 09/29/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Epstein-Barr virus (EBV) and Helicobacter pylori (HP) infections have been extensively recognised as gastric cancer (GC) triggers, and recent publications suggest they could behave as predictive markers for immune-modulating therapies. Tumour-infiltrating lymphocytes (TILs) have also been identified as a predictive biomarker for immunotherapy in different malignancies. This study aimed to investigate the association between EBV and HP infection with TIL levels in GC. METHODS TIL evaluation in haematoxylin-eosin was performed by a pathologist and density of CD3, CD8 and CD163 positive (immunohistochemistry staining) immune cells was calculated with the use of digital pathology software. EBV infection was detected by in situ hybridisation (ISH) and by quantitative polymerase chain reaction (qPCR). Methylation status of EBV-related genes was detected by PCR and a methylome analysis was performed by the Illumina Infinium MethylationEPIC BeadChip. HP status was detected by qPCR. RESULTS We included 98 resected GC Peruvian cases in our evaluation. Median TIL percentage was 30. The proportion of EBV+ detected by ISH was 24.1%, of EBV+ detected by qPCR was 41.8%, while 70% showed methylation of EBV-related genes, and 58.21% of cases were HP+. Younger age (p = 0.024), early stages (p = 0.001), HP+ (p = 0.036) and low CD8 density (p = 0.046) were associated with longer overall survival (OS). High TIL level was associated with intestinal subtype (p < 0.001), with grade 2 (p < 0.001), with EBV qPCR+ (p = 0.001), and with methylation of EBV-related genes (p = 0.007). Cases with high TIL level and cases that are EBV positive share eight genes with similarly methylated status in the metabolomic analysis. High CD8 density was associated with EBV PCR+ (p = 0.012) and HP- (0.005). CONCLUSION Lower CD8 density and HP+ predict longer OS. High TIL level is associated with EBV+ and methylation of EBV-related genes, while lower CD8 density is associated with HP+ GC.
Collapse
Affiliation(s)
- Carlos Castañeda
- Facultad de Ciencias de la Salud, Universidad Cientifica del Sur, Lima 15067, Peru
- Departamento de Oncologia Medica, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0001-6200-0856
| | - Miluska Castillo
- Departamento de Investigación, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-0111-3176
| | - Luis Bernabe
- Departamento de Investigación, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0003-1896-7060
| | - Nancy Suarez
- Departamento de Investigación, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0001-5955-3919
| | - Matteo Fassan
- Department of Medicine-DIMED, Surgical Pathology Unit, University of Padua, Padua 35122, Italy
- Veneto Institute of Oncology, IOV-IRCCS, Padua 35128, Italy
- https://orcid.org/0000-0001-6515-5482
| | - Joselyn Sanchez
- Departamento de Investigación, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-6764-4180
| | - Katherine Tello
- Departamento de Investigación, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-4981-3411
| | - Raul Alatrista
- Departamento de Investigación, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Ivan Chavez
- Departamento de Cirugía en Abdomen, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-3431-3262
| | - Eloy Ruiz
- Departamento de Cirugía en Abdomen, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-4644-0074
| | - Yaqueline Bazan
- Departamento de Cirugía en Abdomen, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-7337-2396
| | - Fernando Barreda
- Departamento de Especialidades Médicas, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-7923-6299
| | - Daniel Valdivia
- Departamento de Especialidades Médicas, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-5917-6452
| | - Wei Meng
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Juvenal Sanchez
- Departamento de Patologia, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- https://orcid.org/0000-0002-9825-8573
| | - Luis Taxa
- Departamento de Patologia, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Paola Montenegro
- Departamento de Oncologia Medica, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| |
Collapse
|
4539
|
Abstract
The innate lymphoid cell (ILC) family is composed of natural killer (NK) cells, ILC1, ILC2 and ILC3, which participate in immune responses to virus, bacteria, parasites and transformed cells. ILC1, ILC2 and ILC3 subsets are mostly tissue-resident, and are profoundly imprinted by their organ of residence. They exhibit pleiotropic effects, driving seemingly paradoxical responses such as tissue repair and, alternatively, immunopathology toward allergens and promotion of tumorigenesis. Despite this, a trickle of studies now suggests that non-NK ILCs may not be overwhelmingly tumorigenic and could potentially be harnessed to drive anti-tumor responses. Here, we examine the pleiotropic behavior of ILCs in cancer and begin to unravel the gap in our knowledge that exposes a new horizon for thinking about modifying ILCs and targeting them for immunotherapy.
Collapse
|
4540
|
Nenu I, Stefanescu H, Procopet B, Sparchez Z, Minciuna I, Mocan T, Leucuta D, Morar C, Grigorescu M, Filip GA, Socaciu C. Navigating through the Lipid Metabolism Maze: Diagnosis and Prognosis Metabolites of Hepatocellular Carcinoma versus Compensated Cirrhosis. J Clin Med 2022; 11:1292. [PMID: 35268381 PMCID: PMC8910918 DOI: 10.3390/jcm11051292] [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: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: The pursuit of finding biomarkers for the diagnosis and prognosis of hepatocellular carcinoma (HCC) has never been so paramount in the days of personalized medicine. The main objective of our study is to identify new biomarkers for diagnosing HCC, and to identify which patients are at risk of developing tumor recurrence, decompensation, or even possesses the risk of cancer-related death. (2) Methods: We have conducted an untargeted metabolomics study from the serum of 69 European patients—32 compensated cirrhotic patients without HCC (controls), and 37 cirrhotic patients with HCC with compensated underlying liver disease (cases), that underwent curative treatment (surgery or ablation), performing ultra-high-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry (UHPLC-QTOF- (ESI+)-MS) with an emphasis on lipid metabolites. (3) Results: 1,25-dihydroxy cholesterol (m/z = 419.281), myristyl palmitate (m/z = 453.165), 25-hydroxy vitamin D2 (m/z = 413.265), 12-ketodeoxycholic acid (m/z = 391.283), lysoPC (21:4) (m/z = 558.291), and lysoPE (22:2) (m/z = 534.286) represent notable biomarkers that differentiate compensated cirrhosis from early HCC, and ceramide species are depleted in the serum of HCC patients. Regarding prognosis, no metabolite identified in our study could determine tumor relapse. To distinguish between the HCC patients that survived curative treatment and those at risk that developed tumor burden, we have identified two notable phosphocholines (PC (30:2); PC (30:1)) with AUROCs of 0.820 and 0.807, respectively, that seem to increase when patients are at risk. In a univariate analysis, arachidonic acid was the only metabolite to predict decompensation (OR = 0.1, 95% CI: 0−0.16, p < 0.005), while in the multivariate analysis, dismally, no variable was associated with decompensation. Furthermore, in the multivariate analysis, we have found out for the first time that the increased expression of 1,25-dihydroxy cholesterol, myristyl palmitate, 12-keto deoxycholic acid, lysoPC (21:4), and lysoPE (22:2) are independent markers of survival. (4) Conclusions: Our study reveals that lipids play a crucial role in discriminating compensated cirrhosis and early hepatocellular carcinoma, and might represent markers of survival and prognosis in personalized and minimally invasive medicine.
Collapse
Affiliation(s)
- Iuliana Nenu
- 3rd Medical Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (B.P.); (Z.S.); (I.M.); (T.M.); (M.G.)
- Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Horia Stefanescu
- Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Bogdan Procopet
- 3rd Medical Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (B.P.); (Z.S.); (I.M.); (T.M.); (M.G.)
- Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Zeno Sparchez
- 3rd Medical Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (B.P.); (Z.S.); (I.M.); (T.M.); (M.G.)
- Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Iulia Minciuna
- 3rd Medical Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (B.P.); (Z.S.); (I.M.); (T.M.); (M.G.)
- Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Tudor Mocan
- 3rd Medical Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (B.P.); (Z.S.); (I.M.); (T.M.); (M.G.)
- Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Daniel Leucuta
- Department of Medical Statistics, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Corina Morar
- Center for Applied Biotechnology BIODIATECH, SC Proplanta, 400478 Cluj-Napoca, Romania; (C.M.); (C.S.)
| | - Mircea Grigorescu
- 3rd Medical Department, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (B.P.); (Z.S.); (I.M.); (T.M.); (M.G.)
- Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Gabriela Adriana Filip
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Carmen Socaciu
- Center for Applied Biotechnology BIODIATECH, SC Proplanta, 400478 Cluj-Napoca, Romania; (C.M.); (C.S.)
| |
Collapse
|
4541
|
Morale MG, Tamura RE, Rubio IGS. Metformin and Cancer Hallmarks: Molecular Mechanisms in Thyroid, Prostate and Head and Neck Cancer Models. Biomolecules 2022; 12:357. [PMID: 35327549 PMCID: PMC8945547 DOI: 10.3390/biom12030357] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
Metformin is the most used drug for type 2 diabetes (T2DM). Its antitumor activity has been described by clinical studies showing reduced risk of cancer development in T2DM patients, as well as management of T2DM compared with those receiving other glucose-lowering drugs. Metformin has a plethora of molecular actions in cancer cells. This review focused on in vitro data on the action mechanisms of metformin on thyroid, prostate and head and neck cancer. AMPK activation regulating specific downstream targets is a constant antineoplastic activity in different types of cancer; however, AMPK-independent mechanisms are also relevant. In vitro evidence makes it clear that depending on the type of tumor, metformin has different actions; its effects may be modulated by different cell conditions (for instance, presence of HPV infection), or it may regulate tissue-specific factors, such as the Na+/I- symporter (NIS) and androgen receptors. The hallmarks of cancer are a set of functional features acquired by the cell during malignant development. In vitro studies show that metformin regulates almost all the hallmarks of cancer. Interestingly, metformin is one of these therapeutic agents with the potential to synergize with other chemotherapeutic agents, with low cost, low side effects and high positive consequences. Some questions are still challenging: Are metformin in vitro data able to translate from bench to bedside? Does metformin affect drug resistance? Can metformin be used as a generic anticancer drug for all types of tumors? Which are the specific actions of metformin on the peculiarities of each type of cancer? Several clinical trials are in progress or have been concluded for repurposing metformin as an anticancer drug. The continuous efforts in the field and future in vitro studies will be essential to corroborate clinical trials results and to elucidate the raised questions.
Collapse
Affiliation(s)
- Mirian Galliote Morale
- Department of Biological Sciences, Federal University of São Paulo, Diadema, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil; (M.G.M.); (R.E.T.)
- Laboratory of Cancer Molecular Biology, Federal University of São Paulo, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil
| | - Rodrigo Esaki Tamura
- Department of Biological Sciences, Federal University of São Paulo, Diadema, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil; (M.G.M.); (R.E.T.)
- Laboratory of Cancer Molecular Biology, Federal University of São Paulo, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil
| | - Ileana Gabriela Sanchez Rubio
- Department of Biological Sciences, Federal University of São Paulo, Diadema, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil; (M.G.M.); (R.E.T.)
- Laboratory of Cancer Molecular Biology, Federal University of São Paulo, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil
- Thyroid Molecular Sciences Laboratory, Federal University of São Paulo, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil
| |
Collapse
|
4542
|
Stikbakke E, Wilsgaard T, Haugnes HS, Pedersen MI, Knutsen T, Støyten M, Giovannucci E, Eggen AE, Thune I, Richardsen E. Expression of miR-24-1-5p in Tumor Tissue Influences Prostate Cancer Recurrence: The PROCA- life Study. Cancers (Basel) 2022; 14:cancers14051142. [PMID: 35267449 PMCID: PMC8909269 DOI: 10.3390/cancers14051142] [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: 12/26/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/22/2022] Open
Abstract
The role of miR-24-1-5p and its prognostic implications associated with prostate cancer are mainly unknown. In a population-based cohort, the Prostate Cancer Study throughout life (PROCA-life), all men had a general health examination at study entry and were followed between 1994 and 2016. Patients with available tissue samples after a prostatectomy with curative intent were identified (n = 189). The tissue expression of miR-24-1-5p in prostate cancer was examined by in situ hybridization (ISH) in tissue microarray (TMA) blocks by semi-quantitative scoring by two independent investigators. Multivariable Cox regression models were used to study the associations between miR-24-1-5p expression and prostate cancer recurrence. The prostate cancer patients had a median age of 65.0 years (range 47−75 years). The Cancer of the Prostate Risk Assessment Postsurgical Score, International Society of Urological Pathology grade group, and European Association of Urology Risk group were all significant prognostic factors for five-year recurrence-free survival (p < 0.001). Prostate cancer patients with a high miR-24-1-5p expression (≥1.57) in the tissue had a doubled risk of recurrence compared to patients with low expression (HR 1.99, 95% CI 1.13−3.51). Our study suggests that a high expression of miR-24-1-5p is associated with an increased risk of recurrence of prostate cancer after radical prostatectomy, which points to the potential diagnostic and therapeutic value of detecting miR-24-1-5p in prostate cancer cases.
Collapse
Affiliation(s)
- Einar Stikbakke
- Department of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (H.S.H.); (T.K.); (M.S.); (I.T.)
- Department of Oncology, University Hospital of North Norway, 9038 Tromsø, Norway
- Correspondence:
| | - Tom Wilsgaard
- Department of Community Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (T.W.); (A.E.E.)
| | - Hege Sagstuen Haugnes
- Department of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (H.S.H.); (T.K.); (M.S.); (I.T.)
- Department of Oncology, University Hospital of North Norway, 9038 Tromsø, Norway
| | - Mona Irene Pedersen
- Translational Cancer Research Group, Institute of Clinical Medicine, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (M.I.P.); (E.R.)
| | - Tore Knutsen
- Department of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (H.S.H.); (T.K.); (M.S.); (I.T.)
- Department of Urology, University Hospital of North Norway, 9038 Tromsø, Norway
| | - Martin Støyten
- Department of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (H.S.H.); (T.K.); (M.S.); (I.T.)
- Department of Oncology, University Hospital of North Norway, 9038 Tromsø, Norway
| | - Edward Giovannucci
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
- Departments of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Anne Elise Eggen
- Department of Community Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (T.W.); (A.E.E.)
| | - Inger Thune
- Department of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (H.S.H.); (T.K.); (M.S.); (I.T.)
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0316 Oslo, Norway
- Department of Oncology, The Cancer Centre, Oslo University Hospital, 0424 Oslo, Norway
| | - Elin Richardsen
- Translational Cancer Research Group, Institute of Clinical Medicine, UiT, The Arctic University of Norway, 9037 Tromsø, Norway; (M.I.P.); (E.R.)
- Department of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9037 Tromsø, Norway
- Department of Clinical Pathology, University Hospital of North Norway, 9038 Tromsø, Norway
| |
Collapse
|
4543
|
Merali N, Chouari T, Kayani K, Rayner CJ, Jiménez JI, Krell J, Giovannetti E, Bagwan I, Relph K, Rockall TA, Dhillon T, Pandha H, Annels NE, Frampton AE. A Comprehensive Review of the Current and Future Role of the Microbiome in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:1020. [PMID: 35205769 PMCID: PMC8870349 DOI: 10.3390/cancers14041020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second most common cause of cancer death in the USA by 2030, yet progress continues to lag behind that of other cancers, with only 9% of patients surviving beyond 5 years. Long-term survivorship of PDAC and improving survival has, until recently, escaped our understanding. One recent frontier in the cancer field is the microbiome. The microbiome collectively refers to the extensive community of bacteria and fungi that colonise us. It is estimated that there is one to ten prokaryotic cells for each human somatic cell, yet, the significance of this community in health and disease has, until recently, been overlooked. This review examines the role of the microbiome in PDAC and how it may alter survival outcomes. We evaluate the possibility of employing microbiomic signatures as biomarkers of PDAC. Ultimately this review analyses whether the microbiome may be amenable to targeting and consequently altering the natural history of PDAC.
Collapse
Affiliation(s)
- Nabeel Merali
- Minimal Access Therapy Training Unit (MATTU), Leggett Building, University of Surrey, Daphne Jackson Road, Guildford GU2 7WG, UK; (N.M.); (T.A.R.)
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital, Egerton Road, Guildford GU2 7XX, UK; (T.C.); (K.K.); (C.J.R.)
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
| | - Tarak Chouari
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital, Egerton Road, Guildford GU2 7XX, UK; (T.C.); (K.K.); (C.J.R.)
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
| | - Kayani Kayani
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital, Egerton Road, Guildford GU2 7XX, UK; (T.C.); (K.K.); (C.J.R.)
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Charles J. Rayner
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital, Egerton Road, Guildford GU2 7XX, UK; (T.C.); (K.K.); (C.J.R.)
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
| | - José I. Jiménez
- Department of Life Sciences, South Kensington Campus, Imperial College London, London SW7 2AZ, UK;
| | - Jonathan Krell
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK;
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Fondazione Pisa per la Scienza, 56017 San Giuliano, Italy
| | - Izhar Bagwan
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
| | - Kate Relph
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
| | - Timothy A. Rockall
- Minimal Access Therapy Training Unit (MATTU), Leggett Building, University of Surrey, Daphne Jackson Road, Guildford GU2 7WG, UK; (N.M.); (T.A.R.)
| | - Tony Dhillon
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
| | - Hardev Pandha
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
| | - Nicola E. Annels
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
| | - Adam E. Frampton
- Minimal Access Therapy Training Unit (MATTU), Leggett Building, University of Surrey, Daphne Jackson Road, Guildford GU2 7WG, UK; (N.M.); (T.A.R.)
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital, Egerton Road, Guildford GU2 7XX, UK; (T.C.); (K.K.); (C.J.R.)
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK; (I.B.); (K.R.); (T.D.); (H.P.); (N.E.A.)
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK;
| |
Collapse
|
4544
|
Ramírez R, Ceprian N, Figuer A, Valera G, Bodega G, Alique M, Carracedo J. Endothelial Senescence and the Chronic Vascular Diseases: Challenges and Therapeutic Opportunities in Atherosclerosis. J Pers Med 2022; 12:jpm12020215. [PMID: 35207703 PMCID: PMC8874678 DOI: 10.3390/jpm12020215] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is probably one of the paradigms of disease linked to aging. Underlying the physiopathology of atherosclerosis are cellular senescence, oxidative stress, and inflammation. These factors are increased in the elderly and from chronic disease patients. Elevated levels of oxidative stress affect cellular function and metabolism, inducing senescence. This senescence modifies the cell phenotype into a senescent secretory phenotype. This phenotype activates immune cells, leading to chronic systemic inflammation. Moreover, due to their secretory phenotype, senescence cells present an increased release of highlighted extracellular vesicles that will change nearby/neighborhood cells and paracrine signaling. For this reason, searching for specific senescent cell biomarkers and therapies against the development/killing of senescent cells has become relevant. Recently, senomorphic and senolityc drugs have become relevant in slowing down or eliminating senescence cells. However, even though they have shown promising results in experimental studies, their clinical use is still yet to be determined.
Collapse
Affiliation(s)
- Rafael Ramírez
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.R.); (A.F.)
| | - Noemi Ceprian
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain; (N.C.); (G.V.)
| | - Andrea Figuer
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.R.); (A.F.)
| | - Gemma Valera
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain; (N.C.); (G.V.)
| | - Guillermo Bodega
- Departamento de Biomedicina y Biotecnología, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain;
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (R.R.); (A.F.)
- Correspondence: (M.A.); (J.C.)
| | - Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain; (N.C.); (G.V.)
- Correspondence: (M.A.); (J.C.)
| |
Collapse
|
4545
|
Brady DC, Hmeljak J, Dar AC. Understanding and drugging RAS: 40 years to break the tip of the iceberg. Dis Model Mech 2022; 15:274631. [PMID: 35244677 PMCID: PMC8905715 DOI: 10.1242/dmm.049519] [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] [Indexed: 11/20/2022] Open
Abstract
Several cancers and rare genetic diseases are caused by dysregulation in the RAS signaling pathway. RAS proteins serve as molecular switches that regulate pathways involved in cellular growth, differentiation and survival. These pathways have been an intense area of investigation for four decades, since the initial identification of somatic RAS mutations linked to human cancers. In the past few years, inhibitors against several RAS effectors, as well as direct inhibitors of the K-RAS mutant G12C, have been developed. This Special Issue in DMM includes original Research articles on RAS-driven cancers and RASopathies. The articles provide insights into mechanisms and biomarkers, and evaluate therapeutic targets. Several articles also present new disease models, whereas others describe technologies or approaches to evaluate the function of RAS in vivo. The collection also includes a series of Review articles on RAS biology and translational aspects of defining and treating RAS-driven diseases. In this Editorial, we summarize this collection and discuss the potential impact of the articles within this evolving area of research. We also identify areas of growth and possible future developments. Summary: This Editorial introduces DMM’s new Special Issue on the RAS pathway. The Guest Editors reflect on the impact of the featured articles on the landscape of the RAS field.
Collapse
Affiliation(s)
- Donita C Brady
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julija Hmeljak
- The Company of Biologists, Bidder Building, Station Road, Histon, Cambridge CB24 9LF, UK
| | - Arvin C Dar
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, Center for Therapeutic Discovery, Mount Sinai, New York, NY 10029-5674, USA
| |
Collapse
|
4546
|
Ramadan R, van Driel MS, Vermeulen L, van Neerven SM. Intestinal stem cell dynamics in homeostasis and cancer. Trends Cancer 2022; 8:416-425. [DOI: 10.1016/j.trecan.2022.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 12/31/2022]
|
4547
|
Treatment-driven tumour heterogeneity and drug resistance: lessons from solid tumours. Cancer Treat Rev 2022; 104:102340. [DOI: 10.1016/j.ctrv.2022.102340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
4548
|
Antiangiogenic Phytochemicals Constituent of Diet as Promising Candidates for Chemoprevention of Cancer. Antioxidants (Basel) 2022; 11:antiox11020302. [PMID: 35204185 PMCID: PMC8868078 DOI: 10.3390/antiox11020302] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/04/2022] Open
Abstract
Despite the extensive knowledge on cancer nature acquired over the last years, the high incidence of this disease evidences a need for new approaches that complement the clinical intervention of tumors. Interestingly, many types of cancer are closely related to dietary habits associated with the Western lifestyle, such as low fruit and vegetable intake. Recent advances around the old-conceived term of chemoprevention highlight the important role of phytochemicals as good candidates for the prevention or treatment of cancer. The potential to inhibit angiogenesis exhibited by many natural compounds constituent of plant foods makes them especially interesting for their use as chemopreventive agents. Here, we review the antitumoral potential, with a focus on the antiangiogenic effects, of phenolic and polyphenolic compounds, such as quercetin or myricetin; terpenoids, such as ursolic acid or kahweol; and anthraquinones from Aloe vera, in different in vitro and in vivo assays, and the available clinical data. Although clinical trials have failed to assess the preventive role of many of these compounds, encouraging preclinical data support the efficacy of phytochemicals constituent of diet in the prevention and treatment of cancer, but a deeper understanding of their mechanisms of action and better designed clinical trials are urgently needed.
Collapse
|
4549
|
Identification, Culture and Targeting of Cancer Stem Cells. Life (Basel) 2022; 12:life12020184. [PMID: 35207472 PMCID: PMC8879966 DOI: 10.3390/life12020184] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Chemoresistance, tumor progression, and metastasis are features that are frequently seen in cancer that have been associated with cancer stem cells (CSCs). These cells are a promising target in the future of cancer therapy but remain largely unknown. Deregulation of pathways that govern stemness in non-tumorigenic stem cells (SCs), such as Notch, Wnt, and Hedgehog pathways, has been described in CSC pathogenesis, but it is necessary to conduct further studies to discover potential new therapeutic targets. In addition, some markers for the identification and characterization of CSCs have been suggested, but the search for specific CSC markers in many cancer types is still under development. In addition, methods for CSC cultivation are also under development, with great heterogeneity existing in the protocols used. This review focuses on the most recent aspects of the identification, characterization, cultivation, and targeting of human CSCs, highlighting the advances achieved in the clinical implementation of therapies targeting CSCs and remarking those potential areas where more research is still required.
Collapse
|
4550
|
Campos-Silva C, López-Borrego S, Felgueres MJ, Esteso G, Vales-Gomez M. NKG2D Ligands in Liquid Biopsy: The Importance of Soluble and Vesicle-Bound Proteins for Immune Modulation. Crit Rev Immunol 2022; 42:21-40. [PMID: 36374819 DOI: 10.1615/critrevimmunol.2022045263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The identification of biomarkers allowing diagnostics, prognostics and patient classification is still a challenge in oncological research for patient management. Improvements in patient survival achieved with immunotherapies substantiate that biomarker studies rely not only on cellular pathways contributing to the pathology, but also on the immune competence of the patient. If these immune molecules can be studied in a non-invasive manner, the benefit for patients and clinicians is obvious. The immune receptor Natural Killer Group 2 Member D (NKG2D) represents one of the main systems involved in direct recognition of tumor cells by effector lymphocytes (T and Natural Killer cells), and in immune evasion. The biology of NKG2D and its ligands comprises a complex network of cellular pathways leading to the expression of these tumor-associated ligands on the cell surface or to their release either as soluble proteins, or in extracellular vesicles that potently inhibit NKG2D-mediated responses. Increased levels of NKG2D-ligands in patient serum correlate with tumor progression and poor prognosis; however, most studies did not test the biochemical form of these molecules. Here we review the biology of the NKG2D receptor and ligands, their role in cancer and in patient response to immunotherapies, as well as the changes provoked in this system by non-immune cancer therapies. Further, we discuss the use of NKG2D-L in liquid biopsy, including methods to analyse vesicle-associated proteins. We propose that the evaluation in cancer patients of the whole NKG2D system can provide crucial information about patient immune competence and risk of tumor progression.
Collapse
Affiliation(s)
- Carmen Campos-Silva
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Silvia López-Borrego
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - María José Felgueres
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Gloria Esteso
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Mar Vales-Gomez
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| |
Collapse
|