1
|
Shi H, Sun X, Wu Y, Cui Q, Sun S, Ji N, Liu Y. Targeting the tumor microenvironment in primary central nervous system lymphoma: Implications for prognosis. J Clin Neurosci 2024; 124:36-46. [PMID: 38642434 DOI: 10.1016/j.jocn.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/06/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare extranodal non-Hodgkin lymphoma, and there is limited research on its tumor microenvironment (TME). Nevertheless, more and more studies have evidence that TME has essential effects on tumor cell proliferation, immune escape, and drug resistance. Thus, it is critical to elucidate the role of TME in PCNSL. The understanding of the PCNSL TME is gradually unfolding, including factors that distinguish it from systemic diffuse large B-cell lymphoma (DLBCL). The TME in PCNSL exhibits both transcriptional and spatial intratumor heterogeneity. Cellular interactions between tumor cells and stroma cells reveal immune evasion signaling. The comparative analysis between PCNSL and DLBCL suggests that PCNSL is more likely to be an immunologically deficient tumor. In PCNSL, T cell exhaustion and downregulation of macrophage immune function are accompanied by suppressive microenvironmental factors such as M2 polarized macrophages, endothelin B receptor, HLA depletion, PD-L1, and TIM-3. MMP-9, Integrin-β1, and ICAM-1/LFA-1 play crucial roles in transendothelial migration towards the CNS, while CXCL13/CXCR5, CD44, MAG, and IL-8 are essential for brain parenchymal invasion. Further, macrophages, YKL-40, CD31, CD105, PD-1/PD-L1 axis, osteopontin, galectin-3, aggregative perivascular tumor cells, and HLA deletion may contribute to poor outcomes in patients with PCNSL. This article reviews the effect of various components of TME on the progression and prognosis of PCNSL patients to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Han Shi
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Xuefei Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Yuchen Wu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Qu Cui
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Shengjun Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Nan Ji
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China
| | - Yuanbo Liu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, 100070 Beijing, China.
| |
Collapse
|
2
|
Zhou W, Nie J, Zhang D. Comprehensive Analysis of Key Endoplasmic Reticulum Stress-Related Genes and Immune Infiltrates in Stanford Type A Aortic Dissection. Anatol J Cardiol 2024; 28:236-244. [PMID: 38445624 PMCID: PMC11059230 DOI: 10.14744/anatoljcardiol.2024.4251] [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: 01/09/2024] [Accepted: 02/07/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Type A aortic dissection is a fatal disease. However, the role of endoplasmic reticulum stress-related genes (ERSRGs) in type A aortic dissection has not yet been fully clarified. METHODS Differentially expressed genes in the aorta of type A aortic dissection patients were analyzed based on the GSE52093 database. The ERSRGs were downloaded from the GeneCards website. Functional enrichment analysis and protein-protein interaction analysis were performed on the acquired differentially expressed ERSRGs. The mRNA expression of the 5 top key differentially expressed ERSRGs was further explored in GSE153434 and clinical samples. Immune infiltration correlation analysis was performed on the validated key genes. Finally, we constructed regulatory networks of transcription factors, miRNAs, and chemicals. RESULTS Twelve differentially expressed ERSRGs were identified, of which 8 genes were downregulated and 4 genes were upregulated. GeneMANIA was adopted to analyze these genes and their interacting proteins, and the results showed that the main function was calcium ion transport. Four key genes, ACTC1, CASQ2, SPP1, and REEP1, were validated in GSE153434 and clinical samples. The area under the ROC curve values for ACTC1, CASQ2, SPP1, and REEP1 were 0.92, 0.96, 0.89, and 1.00, respectively. ACTC1 and REEP1 correlated with multiple immune cells. Many transcription factors, microRNAs, and chemicals were identified with the potential to regulate these 4 key genes. CONCLUSION In this study, we identified 12 differentially expressed ERSRGs by analyzing the Gene Expression Omnibus database. Four key genes may influence the development of type A aortic dissection by regulating endoplasmic reticulum stress. These results expand our understanding of type A aortic dissection, and the 4 key genes are expected to be diagnostic markers and potential therapeutic targets.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Jun Nie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Dafa Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
3
|
The Intracellular and Secreted Sides of Osteopontin and Their Putative Physiopathological Roles. Int J Mol Sci 2023; 24:ijms24032942. [PMID: 36769264 PMCID: PMC9917417 DOI: 10.3390/ijms24032942] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/21/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Classically, osteopontin (OPN) has been described as a secreted glycophosprotein. Indeed, most data concerning its physiological and pathological roles are mainly related to the secreted OPN (sOPN). However, there are several instances in which intracellular OPN (iOPN) has been described, presenting some specific roles in distinct experimental models, such as in the immune system, cancer cells, and neurological disorders. We herein aimed to highlight and discuss some of these secreted and intracellular roles of OPN and their putative clinical and biological impacts. Moreover, by consolidating data from the OPN protein database, we also analyzed the occurrence of signal peptide (SP) sequences and putative subcellular localization, especially concerning currently known OPN splicing variants (OPN-SV). Comprehending the roles of OPN in its distinct cellular and tissue environments may provide data regarding the additional applications of this protein as biomarkers and targets for therapeutic purposes, besides further describing its pleiotropic roles.
Collapse
|
4
|
Rizzello C, Cancila V, Sangaletti S, Botti L, Ratti C, Milani M, Dugo M, Bertoni F, Tripodo C, Chiodoni C, Colombo MP. Intracellular osteopontin protects from autoimmunity-driven lymphoma development inhibiting TLR9-MYD88-STAT3 signaling. Mol Cancer 2022; 21:215. [PMID: 36503430 PMCID: PMC9743519 DOI: 10.1186/s12943-022-01687-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Autoimmune disorders, including Systemic Lupus Erythematosus (SLE), are associated with increased incidence of hematological malignancies. The matricellular protein osteopontin (OPN) has been linked to SLE pathogenesis, as SLE patients show increased serum levels of OPN and often polymorphisms in its gene. Although widely studied for its pro-tumorigenic role in different solid tumours, the role of OPN in autoimmunity-driven lymphomagenesis has not been investigated yet. METHODS To test the role of OPN in the SLE-associated lymphomagenesis, the SLE-like prone Faslpr/lpr mutation was transferred onto an OPN-deficient background. Spleen from Faslpr/lpr and OPN-/-Faslpr/lpr mice, as well as purified B cells, were analysed by histopathology, flow cytometry, Western Blot, immunohistochemistry, immunofluorescence and gene expression profile to define lymphoma characteristics and investigate the molecular mechanisms behind the observed phenotype. OPN cellular localization in primary splenic B cells and mouse and human DLBCL cell lines was assessed by confocal microscopy. Finally, gain of function experiments, by stable over-expression of the secreted (sOPN) and intracellular OPN (iOPN) in OPN-/-Faslpr/lpr -derived DLBCL cell lines, were performed for further validation experiments. RESULTS Despite reduced autoimmunity signs, OPN-/-Faslpr/lpr mice developed splenic lymphomas with higher incidence than Faslpr/lpr counterparts. In situ and ex vivo analysis featured such tumours as activated type of diffuse large B cell lymphoma (ABC-DLBCL), expressing BCL2 and c-MYC, but not BCL6, with activated STAT3 signaling. OPN-/-Faslpr/lpr B lymphocytes showed an enhanced TLR9-MYD88 signaling pathway, either at baseline or after stimulation with CpG oligonucleotides, which mimic dsDNA circulating in autoimmune conditions. B cells from Faslpr/lpr mice were found to express the intracellular form of OPN. Accordingly, gene transfer-mediated re-expression of iOPN, but not of its secreted isoform, into ABC-DLBCL cell lines established from OPN-/-Faslpr/lpr mice, prevented CpG-mediated activation of STAT3, suggesting that the intracellular form of OPN may represent a brake to TLR9 signaling pathway activation. CONCLUSION These data indicate that, in the setting of SLE-like syndrome in which double strand-DNA chronically circulates and activates TLRs, B cell intracellular OPN exerts a protective role in autoimmunity-driven DLBCL development, mainly acting as a brake in the TLR9-MYD88-STAT3 signaling pathway.
Collapse
Affiliation(s)
- Celeste Rizzello
- grid.417893.00000 0001 0807 2568Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Valeria Cancila
- grid.10776.370000 0004 1762 5517Tumor Immunology Unit, Department of Health Science, University of Palermo School of Medicine, Palermo, Italy
| | - Sabina Sangaletti
- grid.417893.00000 0001 0807 2568Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Laura Botti
- grid.417893.00000 0001 0807 2568Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Chiara Ratti
- grid.417893.00000 0001 0807 2568Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Matteo Milani
- grid.417893.00000 0001 0807 2568Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Matteo Dugo
- grid.417893.00000 0001 0807 2568Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy ,grid.18887.3e0000000417581884Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan, Italy
| | - Francesco Bertoni
- grid.29078.340000 0001 2203 2861Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Via F. Chiesa 5, 6500 Bellinzona, Switzerland ,grid.419922.5Oncology Institute of Southern Switzerland, Ente Ospedialiero Cantonale, Via A. Gallino 12, 6500 Bellinzona, Switzerland
| | - Claudio Tripodo
- grid.10776.370000 0004 1762 5517Tumor Immunology Unit, Department of Health Science, University of Palermo School of Medicine, Palermo, Italy ,grid.7678.e0000 0004 1757 7797FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Claudia Chiodoni
- grid.417893.00000 0001 0807 2568Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Mario P. Colombo
- grid.417893.00000 0001 0807 2568Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| |
Collapse
|
5
|
Pandey V, Fleming-Martinez A, Bastea L, Doeppler HR, Eisenhauer J, Le T, Edenfield B, Storz P. CXCL10/CXCR3 signaling contributes to an inflammatory microenvironment and its blockade enhances progression of murine pancreatic precancerous lesions. eLife 2021; 10:60646. [PMID: 34328416 PMCID: PMC8360647 DOI: 10.7554/elife.60646] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/29/2021] [Indexed: 01/18/2023] Open
Abstract
The development of pancreatic cancer requires recruitment and activation of different macrophage populations. However, little is known about how macrophages are attracted to the pancreas after injury or an oncogenic event, and how they crosstalk with lesion cells or other cells of the lesion microenvironment. Here, we delineate the importance of CXCL10/CXCR3 signaling during the early phase of murine pancreatic cancer. We show that CXCL10 is produced by pancreatic precancerous lesion cells in response to IFNγ signaling and that inflammatory macrophages are recipients for this chemokine. CXCL10/CXCR3 signaling in macrophages mediates their chemoattraction to the pancreas, enhances their proliferation, and maintains their inflammatory identity. Blocking of CXCL10/CXCR3 signaling in vivo shifts macrophage populations to a tumor-promoting (Ym1+, Fizz+, Arg1+) phenotype, increases fibrosis, and mediates progression of lesions, highlighting the importance of this pathway in PDA development. This is reversed when CXCL10 is overexpressed in PanIN cells.
Collapse
Affiliation(s)
- Veethika Pandey
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Alicia Fleming-Martinez
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Ligia Bastea
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Heike R Doeppler
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Jillian Eisenhauer
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Tam Le
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Brandy Edenfield
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, United States
| |
Collapse
|
6
|
Fuhr V, Vafadarnejad E, Dietrich O, Arampatzi P, Riedel A, Saliba AE, Rosenwald A, Rauert-Wunderlich H. Time-Resolved scRNA-Seq Tracks the Adaptation of a Sensitive MCL Cell Line to Ibrutinib Treatment. Int J Mol Sci 2021; 22:ijms22052276. [PMID: 33668876 PMCID: PMC7956352 DOI: 10.3390/ijms22052276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/16/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Since the approval of ibrutinib for relapsed/refractory mantle cell lymphoma (MCL), the treatment of this rare mature B-cell neoplasm has taken a great leap forward. Despite promising efficacy of the Bruton tyrosine kinase inhibitor, resistance arises inevitably and the underlying mechanisms remain to be elucidated. Here, we aimed to decipher the response of a sensitive MCL cell line treated with ibrutinib using time-resolved single-cell RNA sequencing. The analysis uncovered five subpopulations and their individual responses to the treatment. The effects on the B cell receptor pathway, cell cycle, surface antigen expression, and metabolism were revealed by the computational analysis and were validated by molecular biological methods. The observed upregulation of B cell receptor signaling, crosstalk with the microenvironment, upregulation of CD52, and metabolic reprogramming towards dependence on oxidative phosphorylation favor resistance to ibrutinib treatment. Targeting these cellular responses provide new therapy options in MCL.
Collapse
Affiliation(s)
- Viktoria Fuhr
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken, 97080 Würzburg, Germany; (V.F.); (A.R.)
| | - Ehsan Vafadarnejad
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97080 Würzburg, Germany; (E.V.); (O.D.); (A.-E.S.)
| | - Oliver Dietrich
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97080 Würzburg, Germany; (E.V.); (O.D.); (A.-E.S.)
| | - Panagiota Arampatzi
- Core Unit Systems Medicine, University of Würzburg, 97080 Würzburg, Germany;
| | - Angela Riedel
- Mildred Scheel Early Career Center (MSNZ), University Hospital of Würzburg, 97080 Würzburg, Germany;
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97080 Würzburg, Germany; (E.V.); (O.D.); (A.-E.S.)
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken, 97080 Würzburg, Germany; (V.F.); (A.R.)
| | - Hilka Rauert-Wunderlich
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken, 97080 Würzburg, Germany; (V.F.); (A.R.)
- Correspondence:
| |
Collapse
|
7
|
Lovisa F, Garbin A, Crotti S, Di Battista P, Gallingani I, Damanti CC, Tosato A, Carraro E, Pillon M, Mafakheri E, Romanato F, Gaffo E, Biffi A, Bortoluzzi S, Agostini M, Mussolin L. Increased Tenascin C, Osteopontin and HSP90 Levels in Plasmatic Small Extracellular Vesicles of Pediatric ALK-Positive Anaplastic Large Cell Lymphoma: New Prognostic Biomarkers? Diagnostics (Basel) 2021; 11:diagnostics11020253. [PMID: 33562105 PMCID: PMC7915848 DOI: 10.3390/diagnostics11020253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past 15 years, several biological and pathological characteristics proved their significance in pediatric anaplastic lymphoma kinase (ALK)-positive anaplastic large-cell lymphoma (ALCL) prognostic stratification. However, the identification of new non-invasive disease biomarkers, relying on the most important disease mechanisms, is still necessary. In recent years, plasmatic circulating small extracellular vesicles (S-EVs) gathered great importance both as stable biomarker carriers and active players in tumorigenesis. In the present work, we performed a comprehensive study on the proteomic composition of plasmatic S-EVs of pediatric ALCL patients compared to healthy donors (HDs). By using a mass spectrometry-based proteomics approach, we identified 50 proteins significantly overrepresented in S-EVs of ALCL patients. Gene Ontology enrichment analysis disclosed cellular components and molecular functions connected with S-EV origin and vesicular trafficking, whereas cell adhesion, glycosaminoglycan metabolic process, extracellular matrix organization, collagen fibril organization and acute phase response were the most enriched biological processes. Of importance, consistently with the presence of nucleophosmin (NPM)-ALK fusion protein in ALCL cells, a topological enrichment analysis based on Reactome- and Kyoto Encyclopedia of Genes and Genomes (KEGG)-derived networks highlighted a dramatic increase in proteins of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway in ALCL S-EVs, which included heat shock protein 90-kDa isoform alpha 1 (HSP90AA1), osteopontin (SPP1/OPN) and tenascin C (TNC). These results were validated by Western blotting analysis on a panel of ALCL and HD cases. Further research is warranted to better define the role of these S-EV proteins as diagnostic and, possibly, prognostic parameters at diagnosis and for ALCL disease monitoring.
Collapse
Affiliation(s)
- Federica Lovisa
- Maternal and Child Health Department, Padova University, 35128 Padova, Italy; (F.L.); (A.G.); (P.D.B.); (I.G.); (C.C.D.); (A.T.); (A.B.)
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
| | - Anna Garbin
- Maternal and Child Health Department, Padova University, 35128 Padova, Italy; (F.L.); (A.G.); (P.D.B.); (I.G.); (C.C.D.); (A.T.); (A.B.)
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
| | - Sara Crotti
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
| | - Piero Di Battista
- Maternal and Child Health Department, Padova University, 35128 Padova, Italy; (F.L.); (A.G.); (P.D.B.); (I.G.); (C.C.D.); (A.T.); (A.B.)
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
| | - Ilaria Gallingani
- Maternal and Child Health Department, Padova University, 35128 Padova, Italy; (F.L.); (A.G.); (P.D.B.); (I.G.); (C.C.D.); (A.T.); (A.B.)
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
| | - Carlotta Caterina Damanti
- Maternal and Child Health Department, Padova University, 35128 Padova, Italy; (F.L.); (A.G.); (P.D.B.); (I.G.); (C.C.D.); (A.T.); (A.B.)
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
| | - Anna Tosato
- Maternal and Child Health Department, Padova University, 35128 Padova, Italy; (F.L.); (A.G.); (P.D.B.); (I.G.); (C.C.D.); (A.T.); (A.B.)
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
| | - Elisa Carraro
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padova University Hospital, 35128 Padova, Italy; (E.C.); (M.P.)
| | - Marta Pillon
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padova University Hospital, 35128 Padova, Italy; (E.C.); (M.P.)
| | - Erfan Mafakheri
- Department of Physics and Astronomy, Padova University, 35131 Padova, Italy; (E.M.); (F.R.)
| | - Filippo Romanato
- Department of Physics and Astronomy, Padova University, 35131 Padova, Italy; (E.M.); (F.R.)
- IOM-CNR, S.S. 14 km 163,5, 34149 Trieste, Italy
| | - Enrico Gaffo
- Department of Molecular Medicine, Padova University, 35121 Padova, Italy; (E.G.); (S.B.)
| | - Alessandra Biffi
- Maternal and Child Health Department, Padova University, 35128 Padova, Italy; (F.L.); (A.G.); (P.D.B.); (I.G.); (C.C.D.); (A.T.); (A.B.)
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
| | - Stefania Bortoluzzi
- Department of Molecular Medicine, Padova University, 35121 Padova, Italy; (E.G.); (S.B.)
- CRIBI Interdepartmental Research Center for Innovative Biotechnologies (CRIBI), Padova University, 35121 Padova, Italy
| | - Marco Agostini
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
- First Surgical Clinic Section, Department of Surgical, Oncological and Gastroenterological Sciences, Padova University, 35128 Padova, Italy
| | - Lara Mussolin
- Maternal and Child Health Department, Padova University, 35128 Padova, Italy; (F.L.); (A.G.); (P.D.B.); (I.G.); (C.C.D.); (A.T.); (A.B.)
- Istituto di Ricerca Pediatrica Città della Speranza, 35127 Padova, Italy; (S.C.); (M.A.)
- Correspondence:
| |
Collapse
|
8
|
The extracellular matrix: A key player in the pathogenesis of hematologic malignancies. Blood Rev 2020; 48:100787. [PMID: 33317863 DOI: 10.1016/j.blre.2020.100787] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/10/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022]
Abstract
Hematopoietic stem and progenitor cells located in the bone marrow lay the foundation for multiple lineages of mature hematologic cells. Bone marrow niches are architecturally complex with specific cellular, physiochemical, and biomechanical factors. Increasing evidence suggests that the bone marrow microenvironment contributes to the pathogenesis of hematological neoplasms. Numerous studies have deciphered the role of genetic mutations and chromosomal translocations in the development hematologic malignancies. Significant progress has also been made in understanding how the cellular components and cytokine interactions within the bone marrow microenvironment promote the evolution of hematologic cancers. Although the extracellular matrix is known to be a key player in the pathogenesis of various diseases, it's role in the progression of hematologic malignancies is less understood. In this review, we discuss the interactions between the extracellular matrix and malignant cells, and provide an overview of the role of extracellular matrix remodeling in sustaining hematologic malignancies.
Collapse
|
9
|
Jiménez I, Carabia J, Bobillo S, Palacio C, Abrisqueta P, Pagès C, Nieto JC, Castellví J, Martínez-Ricarte F, Escoda L, Perla C, Céspedes Torrez DH, Boix J, Purroy N, Puigdefàbregas L, Seoane J, Bosch F, Crespo M. Repolarization of tumor infiltrating macrophages and increased survival in mouse primary CNS lymphomas after XPO1 and BTK inhibition. J Neurooncol 2020; 149:13-25. [PMID: 32691208 PMCID: PMC7452938 DOI: 10.1007/s11060-020-03580-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Patients diagnosed with primary central nervous system lymphoma (PCNSL) often face dismal outcomes due to the limited availability of therapeutic options. PCNSL cells frequently have deregulated B-cell receptor (BCR) signaling, but clinical responses to its inhibition using ibrutinib have been brief. In this regard, blocking nuclear export by using selinexor, which covalently binds to XPO1, can also inhibit BCR signaling. Selinexor crosses the blood-brain barrier and was recently shown to have clinical activity in a patient with refractory diffuse large B-cell lymphoma in the CNS. We studied selinexor alone or in combination with ibrutinib in pre-clinical mouse models of PCNSL. METHODS Orthotopic xenograft models were established by injecting lymphoma cells into the brain parenchyma of athymic mice. Tumor growth was monitored by bioluminescence. Malignant cells and macrophages were studied by immunohistochemistry and flow cytometry. RESULTS Selinexor blocked tumor growth and prolonged survival in a bioluminescent mouse model, while its combination with ibrutinib further increased survival. CNS lymphoma in mice was infiltrated by tumor-promoting M2-like macrophages expressing PD-1 and SIRPα. Interestingly, treatment with selinexor and ibrutinib favored an anti-tumoral immune response by shifting polarization toward inflammatory M1-like and diminishing PD-1 and SIRPα expression in the remaining tumor-promoting M2-like macrophages. CONCLUSIONS These data highlight the pathogenic role of the innate immune microenvironment in PCNSL and provide pre-clinical evidence for the development of selinexor and ibrutinib as a new promising therapeutic option with cytotoxic and immunomodulatory potential.
Collapse
Affiliation(s)
- Isabel Jiménez
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, Barcelona, 08035, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Júlia Carabia
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, Barcelona, 08035, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Sabela Bobillo
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
- Department of Hematology, Experimental Hematology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Carles Palacio
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
- Department of Hematology, Experimental Hematology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Pau Abrisqueta
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
- Department of Hematology, Experimental Hematology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Carlota Pagès
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, Barcelona, 08035, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Juan C Nieto
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, Barcelona, 08035, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Josep Castellví
- Department of Pathology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Francisco Martínez-Ricarte
- Department of Neurosurgery, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Lourdes Escoda
- Department of Hematology, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Cristóbal Perla
- Department of Neurosurgery, Hospital Universitari Joan XXIII, Tarragona, Spain
| | | | - Joan Boix
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, Barcelona, 08035, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Noelia Purroy
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, Barcelona, 08035, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts and Broad Institute of MIT and Harvard, Cambridge, MA, UK
| | - Lluís Puigdefàbregas
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, Barcelona, 08035, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
| | - Joan Seoane
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
- Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, 08035, Barcelona, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), CIBERONC, Barcelona, Spain
| | - Francesc Bosch
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain
- Department of Hematology, Experimental Hematology, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Marta Crespo
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret, 115-117, Barcelona, 08035, Spain.
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain.
| |
Collapse
|
10
|
You H, Baluszek S, Kaminska B. Supportive roles of brain macrophages in CNS metastases and assessment of new approaches targeting their functions. Am J Cancer Res 2020; 10:2949-2964. [PMID: 32194848 PMCID: PMC7053204 DOI: 10.7150/thno.40783] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/22/2020] [Indexed: 12/17/2022] Open
Abstract
Metastases to the central nervous system (CNS) occur frequently in adults and their frequency increases with the prolonged survival of cancer patients. Patients with CNS metastases have short survival, and modern therapeutics, while effective for extra-cranial cancers, do not reduce metastatic burden. Tumor cells attract and reprogram stromal cells, including tumor-associated macrophages that support cancer growth by promoting tissue remodeling, invasion, immunosuppression and metastasis. Specific roles of brain resident and infiltrating macrophages in creating a pre-metastatic niche for CNS invading cancer cells are less known. There are populations of CNS resident innate immune cells such as: parenchymal microglia and non-parenchymal, CNS border-associated macrophages that colonize CNS in early development and sustain its homeostasis. In this study we summarize available data on potential roles of different brain macrophages in most common brain metastases. We hypothesize that metastatic cancer cells exploit CNS macrophages and their cytoprotective mechanisms to create a pre-metastatic niche and facilitate metastatic growth. We assess current pharmacological strategies to manipulate functions of brain macrophages and hypothesize on their potential use in a therapy of CNS metastases. We conclude that the current data strongly support a notion that microglia, as well as non-parenchymal macrophages and peripheral infiltrating macrophages, are involved in multiple stages of CNS metastases. Understanding their contribution will lead to development of new therapeutic strategies.
Collapse
|
11
|
Barranco G, Fernández E, Rivas S, Quezada R, Nava D, Aguilar J, García A, Astudillo H, Lome C, Ruiz E. Osteopontin expression and its relationship with prognostic factors in diffuse large B-cell lymphoma. Hematol Rep 2019; 11:7964. [PMID: 31579151 PMCID: PMC6761465 DOI: 10.4081/hr.2019.7964] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 07/05/2019] [Indexed: 01/20/2023] Open
Abstract
The aim of this study is to explore the expression of osteopontin (OPN) and its relationship with prognostic factors and survival in diffuse large B cell lymphoma (DLBCL). A tissue microarray was performed for immunohistochemical evaluation. Contingency tables were analyzed for trends; chi-square test was used to determine differences between groups. Univariate and multivariate Cox proportional hazards-regression analyses were performed to evaluate the impact of prognostic factors on survival. Expression of OPN was observed in 28%. It was different in non-germinal center DLBCL (P=0.04). The mean overall survival (OS) was lower in patients with positive OPN expression (19.762; CI 95% 14.269-25.255) it was not significant (P=0.123). It is not possible to establish a clear relationship between the expression by immunohistochemistry of osteopontin and a poor prognosis but it would be important to complement the analysis with other techniques as PCR or NGS that allow us to assess the influence of the isoforms and post-translational modifications of OPN on the biological behavior of DLBCL. Our findings indicate that OPN expression could be associated with a more aggressive variant of lymphoma: non-germinal center DLBCL.
Collapse
Affiliation(s)
| | - Edith Fernández
- Translational Medicine, National Institute of Cancerology, Mexico City.,Computational Genomics, National Institute of Genomic Medicine, Mexico City
| | - Silvia Rivas
- Department of Hematology, National Institute of Cancerology, Mexico City
| | - Roxana Quezada
- Department of Pathology, National Institute of Cancerology, Mexico City
| | - Dolores Nava
- Department of Pathology, National Institute of Cancerology, Mexico City
| | - José Aguilar
- Department of Internal Medicine, National Institute of Cancerology, Mexico City
| | - Abelardo García
- Translational Medicine, National Institute of Cancerology, Mexico City
| | - Horacio Astudillo
- Laboratory of Translational Cancer Research and Cellular Therapy, National Medical Center Siglo XXI, Mexico City, Mexico
| | - Carmen Lome
- Department of Pathology, National Institute of Cancerology, Mexico City
| | - Erika Ruiz
- Translational Medicine, National Institute of Cancerology, Mexico City
| |
Collapse
|
12
|
You H, Baluszek S, Kaminska B. Immune Microenvironment of Brain Metastases-Are Microglia and Other Brain Macrophages Little Helpers? Front Immunol 2019; 10:1941. [PMID: 31481958 PMCID: PMC6710386 DOI: 10.3389/fimmu.2019.01941] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022] Open
Abstract
Brain metastases are common intracranial neoplasms and their frequency increases with prolonged survival of cancer patients. New pharmaceuticals targeting oncogenic kinases and immune checkpoint inhibitors augment both overall and progression-free survival in patients with brain metastases, but are not fully successful in reducing metastatic burden and still a majority of oncologic patients die due to dissemination of the disease. Despite therapy advancements, median survival of patients with brain metastases is several months, although it may vary in different types or subtypes of cancer. Contribution of the innate immune system to cancer progression is well established. Tumor-associated macrophages (TAMs), instead of launching antitumor responses, promote extracellular matrix degradation, secrete immunosuppressive cytokines, promote neoangiogenesis and tumor growth. While their roles as pro-tumorigenic cells facilitating tissue remodeling, invasion and metastasis is well documented, much less is known about the immune microenvironment of brain metastases and roles of specific immune cells in those processes. The central nervous system (CNS) is armed in resident myeloid cells: microglia and perivascular macrophages which colonize CNS in early development and maintain homeostasis in brain parenchyma and at brain-blood vessels interfaces. In this study we discuss available data on the immune composition of most common brain metastases, focusing on interactions between metastatic cancer cells and microglia, perivascular and meningeal macrophages. Cancer cells ‘highjack’ several CNS protective mechanisms and may employ microglia and CNS-border associated macrophages into helping cancer cells to colonize a pre-metastatic niche. We describe emerging molecular insights into mechanisms governing communication between microglia and metastatic cancer cells that culminate in activation of CNS resident microglia and trafficking of monocytic cells from the periphery. We present mechanisms controlling those processes in brain metastases and hypothesize on potential therapeutic approaches. In summary, microglia and non-parenchymal brain macrophages are involved in multiple stages of a metastatic disease and, unlike tumor cells, are genetically stable and predictable, which makes them an attractive target for anticancer therapies.
Collapse
Affiliation(s)
- Hua You
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.,School of Laboratory Medicine, YouJiang Medical University for Nationalities, Baise, China.,Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Szymon Baluszek
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.,Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
13
|
Bastos ACSF, Blunck CB, Emerenciano M, Gimba ERP. Osteopontin and their roles in hematological malignancies: Splice variants on the new avenues. Cancer Lett 2017; 408:138-143. [PMID: 28844708 DOI: 10.1016/j.canlet.2017.08.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/13/2017] [Accepted: 08/16/2017] [Indexed: 10/24/2022]
Abstract
Osteopontin (OPN) is a protein expressed in several tissues, including bone marrow, in which it performs distinct roles, such as modulating hematopoietic stem cell niche and bone remodeling. Most data in hematological malignancies (HMs) refers to total OPN (tOPN), comprehending the sum of distinct OPN splicing isoforms (OPN-SI), while reports describing the expression and roles of each OPN-SI are scarce. This review aims to summarize tOPN roles in HMs and provide evidence that OPN-SIs can also modulate specific functions in HMs biology. We summarize that upregulated tOPN can modulate HMs (leukemia, lymphoma and myeloma) progression, inducing cell adhesion, invasion, angiogenesis, cell differentiation and extramedullary and/or central nervous system infiltration. Based on this expression pattern, tOPN has been pointed out as a biomarker in those HMs, thus providing potential targets for therapeutic approaches. Our group found that OPN-SIs are expressed in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL) cell lines (unpublished data), providing early evidence that OPN-SIs are also expressed in BCP-ALL. Further studies should investigate whether these OPN-SIs can differently modulate HMs biology and their putative application as auxiliary biomarkers for HMs.
Collapse
Affiliation(s)
| | - Caroline Barbieri Blunck
- Instituto Nacional de Câncer, Coordenação de Pesquisa, Programa de Hematologia-Oncologia Pediátrico, Rio de Janeiro, Brazil
| | - Mariana Emerenciano
- Instituto Nacional de Câncer, Coordenação de Pesquisa, Programa de Hematologia-Oncologia Pediátrico, Rio de Janeiro, Brazil
| | - Etel Rodrigues Pereira Gimba
- Instituto Nacional de Câncer, Coordenação de Pesquisa, Programa de Oncobiologia Celular e Molecular, Rio de Janeiro, Brazil; Universidade Federal Fluminense, Instituto de Humanidades e Saúde (IHS), Departamento de Ciências da Natureza (RCN), Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Osteopontin at the Crossroads of Inflammation and Tumor Progression. Mediators Inflamm 2017; 2017:4049098. [PMID: 28769537 PMCID: PMC5523273 DOI: 10.1155/2017/4049098] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/04/2017] [Indexed: 12/13/2022] Open
Abstract
Complex interactions between tumor and host cells regulate systemic tumor dissemination, a process that begins early at the primary tumor site and goes on until tumor cells detach themselves from the tumor mass and start migrating into the blood or lymphatic vessels. Metastatic cells colonize the target organs and are capable of surviving and growing at distant sites. In this context, osteopontin (OPN) appears to be a key determinant of the crosstalk between cancer cells and the host microenvironment, which in turn modulates immune evasion. OPN is overexpressed in several human carcinomas and has been implicated in inflammation, tumor progression, and metastasis. Thus, it represents one of the most attracting targets for cancer therapy. Within the tumor mass, OPN is secreted in various forms either by the tumor itself or by stroma cells, and it can exert either pro- or antitumorigenic effects according to the cell type and tumor microenvironment. Thus, targeting OPN for therapeutic purposes needs to take into account the heterogeneous functions of the multiple OPN forms with regard to cancer formation and progression. In this review, we will describe the role of systemic, tumor-derived, and stroma-derived OPN, highlighting its pivotal role at the crossroads of inflammation and tumor progression.
Collapse
|
15
|
Hao C, Cui Y, Owen S, Li W, Cheng S, Jiang WG. Human osteopontin: Potential clinical applications in cancer (Review). Int J Mol Med 2017; 39:1327-1337. [PMID: 28440483 PMCID: PMC5428945 DOI: 10.3892/ijmm.2017.2964] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022] Open
Abstract
Human osteopontin (OPN) is a glycosylated phosphoprotein which is expressed in a variety of tissues in the body. In recent years, accumulating evidence has indicated that the aberrant expression of OPN is closely associated with tumourigensis, progression and most prominently with metastasis in several tumour types. In this review, we present the current knowledge on the expression profiles of OPN and its main splice variants in human cancers, as well as the potential implications in patient outcome. We also discuss its putative clinical application as a cancer biomarker and as a therapeutic target.
Collapse
Affiliation(s)
- Chengcheng Hao
- Department of Biochemistry and Molecular Biology
- Beijing Key Laboratory of Cancer and Metastasis Research, Capital Medical University, Beijing 100069, P.R. China
| | - Yuxin Cui
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Sionen Owen
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Wenbin Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Shan Cheng
- Department of Biochemistry and Molecular Biology
- Beijing Key Laboratory of Cancer and Metastasis Research, Capital Medical University, Beijing 100069, P.R. China
| | - Wen G. Jiang
- Correspondence to: Professor Wen G. Jiang, Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Henry Wellcome Building, Heath Park Way, Cardiff CF14 4XN, UK, E-mail:
| |
Collapse
|