201
|
Poschenrieder A, Schottelius M, Schwaiger M, Wester HJ. Preclinical evaluation of [(68)Ga]NOTA-pentixafor for PET imaging of CXCR4 expression in vivo - a comparison to [(68)Ga]pentixafor. EJNMMI Res 2016; 6:70. [PMID: 27655427 PMCID: PMC5031577 DOI: 10.1186/s13550-016-0227-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/17/2016] [Indexed: 01/20/2023] Open
Abstract
Background Due to its overexpression in a variety of tumor types, the chemokine receptor 4 (CXCR4) represents a highly relevant diagnostic and therapeutic target in nuclear oncology. Recently, [68Ga]pentixafor has emerged as an excellent imaging agent for positron emission tomography (PET) of CXCR4 expression in vivo. In this study, the corresponding [68Ga]-1,4,7-triazacyclononane-triacetic acid (NOTA) analog was preclinically evaluated and compared to the 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) parent compound [68Ga]pentixafor. Methods NOTA-pentixafor was synthesized by combining solid and solution-phase peptide synthesis. The CXCR4 receptor affinities of [68Ga]pentixafor and [68Ga]NOTA-pentixafor were determined in competitive binding assays using the leukemic CXCR4-expressing Jurkat T-cell line and [125I]FC131 as the radioligand. Internalization and cell efflux assays were performed using CXCR4-transfected Chem-1 cells. Small-animal PET and biodistribution studies were carried out using Daudi-tumor bearing SCID mice. Results [68Ga]NOTA-pentixafor showed a 1.4-fold improved affinity towards CXCR4 (IC50). However, internalization efficiency into CXCR4+-Chem-1 cells was substantially decreased compared to [68Ga]pentixafor. Accordingly, small-animal PET imaging and biodistribution studies revealed a 9.5-fold decreased uptake of [68Ga]NOTA-pentixafor in Daudi lymphoma xenografts (1.7 ± 0.4 % vs 16.2 ± 3.8 % ID/g at 90 min p.i.) and higher levels of non-specific accumulation, primarily in the excretory organs such as the liver, intestines, and kidneys (2.3 ± 0.9 % vs 2.0 ± 0.3 % ID/g, 1.9 ± 0.8 % vs 0.7 ± 0.2 % ID/g, and 2.7 ± 1.1 % vs 1.7 ± 0.9 % ID/g, respectively). Conclusions Despite enhanced CXCR4-affinity in vitro, the [68Ga]NOTA-analog of pentixafor showed reduced CXCR4 targeting efficiency in vivo. In combination with enhanced background accumulation, this resulted in significantly inferior PET imaging contrast, and thus, [68Ga]NOTA-pentixafor offers no advantages over [68Ga]pentixafor.
Collapse
Affiliation(s)
- Andreas Poschenrieder
- Pharmaceutical Radiochemistry, Technische Universität München, Walther-Meißner-Str.3, 85748, Garching, Germany.
| | - Margret Schottelius
- Pharmaceutical Radiochemistry, Technische Universität München, Walther-Meißner-Str.3, 85748, Garching, Germany
| | - Markus Schwaiger
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, 81675, München, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, Walther-Meißner-Str.3, 85748, Garching, Germany
| |
Collapse
|
202
|
Atretkhany KSN, Drutskaya MS, Nedospasov SA, Grivennikov SI, Kuprash DV. Chemokines, cytokines and exosomes help tumors to shape inflammatory microenvironment. Pharmacol Ther 2016; 168:98-112. [PMID: 27613100 DOI: 10.1016/j.pharmthera.2016.09.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Relationship between inflammation and cancer is now well-established and represents a paradigm that our immune response does not necessarily serves solely to protect us from infections and cancer. Many specific mechanisms that link chronic inflammation to cancer promotion and metastasis have been uncovered in the recent years. Here we are focusing on the effects that tumors may exert on inflammatory cascades, tuning the immune system ability to cause tumor promotion or regression. In particular, we discuss the contributions of chemokines, cytokines and exosomes to the processes such as induction of inflammation and tumorigenesis. Overall, tumor-elicited inflammation is a key driver of tumor progression and an essential component of tumor microenvironment.
Collapse
Affiliation(s)
- K-S N Atretkhany
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Biological Faculty, Lomonosov Moscow State University, 119234, Moscow, Russia
| | - M S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Biological Faculty, Lomonosov Moscow State University, 119234, Moscow, Russia
| | - S A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Biological Faculty, Lomonosov Moscow State University, 119234, Moscow, Russia; German Rheumatology Research Center (DRFZ), Berlin, Germany
| | - S I Grivennikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Fox Chase Cancer Center, Cancer Prevention and Control Program, Philadelphia, PA, USA.
| | - D V Kuprash
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Vavilova Str. 32, Russia; Biological Faculty, Lomonosov Moscow State University, 119234, Moscow, Russia.
| |
Collapse
|
203
|
Guo Q, Gao BL, Zhang XJ, Liu GC, Xu F, Fan QY, Zhang SJ, Yang B, Wu XH. CXCL12-CXCR4 Axis Promotes Proliferation, Migration, Invasion, and Metastasis of Ovarian Cancer. Oncol Res 2016; 22:247-58. [PMID: 26629936 PMCID: PMC7842602 DOI: 10.3727/096504015x14343704124430] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The CXCL12-CXCR4 chemokine axis may play a very important role in ovarian cancer cells proliferation, migration, invasion, and peritoneal metastasis in vitro and in vivo. In this study, transfected SKOV3-CXCR4, transfected vector SKOV3-negative, nontransfected SKOV3 ovarian cancer cells, and human peritoneal mesothelial cells (HPMCs) were cultivated in vitro, and the proliferation, migration, and invasion of these ovarian cancer cells were investigated with or without the influence of the CXCL12-CXCR4 axis. Nude mice models of ovarian cancer were created by injection of ovarian cancer cells into the peritoneal cavity for investigation of ovarian cancer cells metastasis. Our results demonstrated that in the SKOV3-CXCR4 group, the cell number of proliferation, migration, or penetration through the Matrigel membrane treated with CXCL12 was significantly (p < 0.05) greater than those treated with CXCR4 antibody or CXCR4 antagonist AMD 3100 in a concentration-dependent manner. In the SKOV3-negative and the nontransfected SKOV3 groups, no significant (p > 0.05) differences existed in the cell number of proliferation, migration, or penetration. Coculture of HPMCs and SKOV3-CXCR4 had significantly (p < 0.05) higher migration and invasion rates than the SKOV3-CXCR4-only group. In nude mice seeded with ovarian cancer cells, the tumor weight in the nude mice injected with SKOV3-CXCR4 cells was significantly (p < 0.05) greater than in the group injected with the SKOV3-negative or nontransfected SKOV3 cells. Taken together, our results show that the CXCL12-CXCR4 chemokine axis can significantly promote the proliferation, migration, invasion, and peritoneal metastasis of ovarian cancer cells, and interference with this axis may serve as a new therapeutic target in treating ovarian cancers.
Collapse
Affiliation(s)
- Qing Guo
- Department of Obstetrics and Gynecology, Shijiazhuang First Hospital, Hebei Medical University, Shijiazhuang, China
| | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Liu T, Li X, You S, Bhuyan SS, Dong L. Effectiveness of AMD3100 in treatment of leukemia and solid tumors: from original discovery to use in current clinical practice. Exp Hematol Oncol 2016; 5:19. [PMID: 27429863 PMCID: PMC4947283 DOI: 10.1186/s40164-016-0050-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/08/2016] [Indexed: 12/16/2022] Open
Abstract
AMD3100, also known as plerixafor, was originally developed as an anti-human immunodeficiency virus (HIV) drug, and later characterized as a C-X-C chemokine receptor type 4 (CXCR4) antagonist. Previous reviews have focused on the application of AMD3100 in the treatment of HIV, but a comprehensive evaluation of AMD3100 in the treatment of leukemia, solid tumor, and diagnosis is lacking. In this review, we broadly describe AMD3100, including the background, functional mechanism and clinical applications. Until the late 1990s, CXCR4 was known as a crucial factor for hematopoietic stem and progenitor cell (HSPC) retention in bone marrow. Subsequently, the action and synergy of plerixafor with Granulocyte-colony stimulating factor (G-CSF) led to the clinical approval of plerixafor as the first compound for mobilization of HSPCs. The amount of HSPC mobilization and the rapid kinetics promoted additional clinical uses. Recently, CXCR4/CXCL12 (C-X-C motif chemokine 12) axis was found to be involved in a variety of roles in tumors, including leukemic stem cell (LSC) homing and signaling transduction. Thus, CXCR4 targeting has been a treatment strategy against leukemia and solid tumors. Understanding this mechanism will help shed light on therapeutic potential for HIV infection, inflammatory diseases, stem-cell mobilization, leukemia, and solid tumors. Clarifying the CXCR4/CXCL12 axis and role of AMD3100 will help remove malignant cells from the bone marrow niche, rendering them more accessible to targeted therapeutic agents.
Collapse
Affiliation(s)
- Tao Liu
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA.,Department of Oncology, The Affiliated Jiangyin Hospital of Southeast University Medical College, Wuxi, 214400 Jiangsu People's Republic of China
| | - Xiaobo Li
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA.,Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193 China
| | - Shuo You
- Department of Neurosurgery, Winship Cancer Institute, Emory University, Atlanta, GA 30322 USA
| | - Soumitra S Bhuyan
- School of Public Health, Division of Health Systems, Management, and Policy, The University of Memphis, Memphis, TN 38152 USA
| | - Lei Dong
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA
| |
Collapse
|
205
|
Pal J, Patil V, Mondal B, Shukla S, Hegde AS, Arivazhagan A, Santosh V, Somasundaram K. Epigenetically silenced GNG4 inhibits SDF1α/CXCR4 signaling in mesenchymal glioblastoma. Genes Cancer 2016; 7:136-47. [PMID: 27382437 PMCID: PMC4918951 DOI: 10.18632/genesandcancer.105] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The most common and aggressive form of primary brain tumor in adults is glioblastoma (GBM). From the global DNA methylation profiling study, previously published from our laboratory, we identified Guanine Nucleotide binding-protein Gamma subunit 4 (GNG4) to be one of the most hyper methylated and down regulated genes in GBM. GBM derived cell lines showed reduced GNG4 transcript levels, which could be reversed by methylation inhibitor treatment. Bisulphite sequencing confirmed the methylation status in glioblastoma tumor tissue and GBM derived cell lines. Overexpression of GNG4 was found to inhibit proliferation and colony formation of GBM cell lines and in vitro transformation of immortalized human astrocytes, thus suggesting a potential tumor suppressor role of GNG4 in GBM. Correlation of GNG4 transcript levels with that of all GPCRs from TCGA data revealed chemokine receptors as the potential target of GNG4. Furthermore, exogenous over expression of GNG4 inhibited SDF1α/CXCR4-dependent chemokine signaling as seen by reduced pERK and pJNK and GBM cell migration. The inhibitory association between GNG4 and SDF1α/CXCR4 was more evident in mesenchymal subtype of GBM. Thus, this study identifies GNG4 as an inhibitor of SDF1α/CXCR4-dependent signaling and emphasizes the significance of epigenetic inactivation of GNG4 in glioblastoma, especially in mesenchymal subtype.
Collapse
Affiliation(s)
- Jagriti Pal
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Vikas Patil
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Baisakhi Mondal
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Sudhanshu Shukla
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Alangar S Hegde
- Sri Satya Sai Institute of Higher Medical Sciences, Bangalore, India
| | - Arimappamagan Arivazhagan
- Departments of Neuropathology and Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore, India
| | - Vani Santosh
- Departments of Neuropathology and Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore, India
| | - Kumaravel Somasundaram
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| |
Collapse
|
206
|
Li X, Kuang Y, Huang X, Zou L, Huang L, Yang T, Li W, Yang Y. Preparation and characterization of a new monoclonal antibody against CXCR4 using lentivirus vector. Int Immunopharmacol 2016; 36:100-105. [DOI: 10.1016/j.intimp.2016.04.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 11/27/2022]
|
207
|
Liebick M, Schläger C, Oppermann M. Analysis of Chemokine Receptor Trafficking by Site-Specific Biotinylation. PLoS One 2016; 11:e0157502. [PMID: 27310579 PMCID: PMC4911081 DOI: 10.1371/journal.pone.0157502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/31/2016] [Indexed: 12/17/2022] Open
Abstract
Chemokine receptors undergo internalization and desensitization in response to ligand activation. Internalized receptors are either preferentially directed towards recycling pathways (e.g. CCR5) or sorted for proteasomal degradation (e.g. CXCR4). Here we describe a method for the analysis of receptor internalization and recycling based on specific Bir A-mediated biotinylation of an acceptor peptide coupled to the receptor, which allows a more detailed analysis of receptor trafficking compared to classical antibody-based detection methods. Studies on constitutive internalization of the chemokine receptors CXCR4 (12.1% ± 0.99% receptor internalization/h) and CCR5 (13.7% ± 0.68%/h) reveals modulation of these processes by inverse (TAK779; 10.9% ± 0.95%/h) or partial agonists (Met-CCL5; 15.6% ± 0.5%/h). These results suggest an actively driven internalization process. We also demonstrate the advantages of specific biotinylation compared to classical antibody detection during agonist-induced receptor internalization, which may be used for immunofluorescence analysis as well. Site-specific biotinylation may be applicable to studies on trafficking of transmembrane proteins, in general.
Collapse
MESH Headings
- Amides/pharmacology
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/isolation & purification
- Basophils/cytology
- Basophils/drug effects
- Basophils/metabolism
- Biotin/chemistry
- Biotin/metabolism
- Biotinylation
- CCR5 Receptor Antagonists/pharmacology
- Carbon-Nitrogen Ligases/genetics
- Carbon-Nitrogen Ligases/metabolism
- Cell Line, Tumor
- Chemokine CCL5/pharmacology
- Escherichia coli Proteins/genetics
- Escherichia coli Proteins/metabolism
- Gene Expression
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Mice
- Protein Transport/drug effects
- Quaternary Ammonium Compounds/pharmacology
- Rats
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Receptors, CXCR5/antagonists & inhibitors
- Receptors, CXCR5/genetics
- Receptors, CXCR5/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Transfection
Collapse
Affiliation(s)
- Marcel Liebick
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| | - Christian Schläger
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| | - Martin Oppermann
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| |
Collapse
|
208
|
Broussas M, Boute N, Akla B, Berger S, Beau-Larvor C, Champion T, Robert A, Beck A, Haeuw JF, Goetsch L, Bailly C, Dumontet C, Matthes T, Corvaia N, Klinguer-Hamour C. A New Anti-CXCR4 Antibody That Blocks the CXCR4/SDF-1 Axis and Mobilizes Effector Cells. Mol Cancer Ther 2016; 15:1890-9. [PMID: 27297868 DOI: 10.1158/1535-7163.mct-16-0041] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/29/2016] [Indexed: 11/16/2022]
Abstract
The type IV C-X-C-motif chemokine receptor (CXCR4) is expressed in a large variety of human cancers, including hematologic malignancies, and this receptor and its ligand, stromal cell-derived factor-1 (SDF-1), play a crucial role in cancer progression. We generated a humanized immunoglobulin G1 mAb, hz515H7, which binds human CXCR4, efficiently competes for SDF-1 binding, and induces a conformational change in CXCR4 homodimers. Furthermore, it inhibits both CXCR4 receptor-mediated G-protein activation and β-arrestin-2 recruitment following CXCR4 activation. The binding of the hz515H7 antibody to CXCR4 inhibits the SDF-1-induced signaling pathway, resulting in reduced phosphorylation of downstream effectors, such as Akt, Erk1/2, p38, and GSK3β. Hz515H7 also strongly inhibits cell migration and proliferation and, while preserving normal blood cells, induces both antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity against neoplastic cells. In mouse xenograft models, hz515H7 displays antitumor activities with multiple hematologic tumor cell lines, with its Fc-mediated effector functions proving essential in this context. Furthermore, hz515H7 binds to primary tumor cells from acute myeloid leukemia and multiple myeloma patients. Collectively, our results demonstrate two major mechanisms of action, making hz515H7 unique in this regard. Its potential as a best-in-class molecule is currently under investigation in a phase I clinical trial. Mol Cancer Ther; 15(8); 1890-9. ©2016 AACR.
Collapse
Affiliation(s)
- Matthieu Broussas
- Department of Experimental Oncology, Centre d'Immunologie Pierre Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Nicolas Boute
- Unit of Molecular and Cellular Biology, CIPF, Saint-Julien-en-Genevois, France
| | - Barbara Akla
- Department of Experimental Oncology, Centre d'Immunologie Pierre Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Sven Berger
- Unit of Molecular and Cellular Biology, CIPF, Saint-Julien-en-Genevois, France
| | - Charlotte Beau-Larvor
- Department of Experimental Oncology, Centre d'Immunologie Pierre Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Thierry Champion
- Department of Physico-chemistry, CIPF, Saint-Julien-en-Genevois, France
| | - Alain Robert
- Unit of Molecular and Cellular Biology, CIPF, Saint-Julien-en-Genevois, France
| | - Alain Beck
- Department of Physico-chemistry, CIPF, Saint-Julien-en-Genevois, France
| | | | - Liliane Goetsch
- Department of Experimental Oncology, Centre d'Immunologie Pierre Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Christian Bailly
- Contract Development and Manufacturing Organization, Toulouse, France
| | | | - Thomas Matthes
- Hematology Service and Service of Clinical Pathology, University Hospital Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
209
|
Kim B, Kim YC, Park B. Pomolic acid inhibits metastasis of HER2 overexpressing breast cancer cells through inactivation of the ERK pathway. Int J Oncol 2016; 49:744-52. [DOI: 10.3892/ijo.2016.3568] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/01/2016] [Indexed: 11/05/2022] Open
|
210
|
Conway EM, Pikor LA, Kung SHY, Hamilton MJ, Lam S, Lam WL, Bennewith KL. Macrophages, Inflammation, and Lung Cancer. Am J Respir Crit Care Med 2016; 193:116-30. [PMID: 26583808 DOI: 10.1164/rccm.201508-1545ci] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer mortality worldwide, and at only 18%, it has one of the lowest 5-year survival rates of all malignancies. With its highly complex mutational landscape, treatment strategies against lung cancer have proved largely ineffective. However with the recent success of immunotherapy trials in lung cancer, there is renewed enthusiasm in targeting the immune component of tumors. Macrophages make up the majority of the immune infiltrate in tumors and are a key cell type linking inflammation and cancer. Although the mechanisms through which inflammation promotes cancer are not fully understood, two connected hypotheses have emerged: an intrinsic pathway, driven by genetic alterations that lead to neoplasia and inflammation, and an extrinsic pathway, driven by inflammatory conditions that increase cancer risk. Here, we discuss the contribution of macrophages to these pathways and subsequently their roles in established tumors. We highlight studies investigating the association of macrophages with lung cancer prognosis and discuss emerging therapeutic strategies for targeting macrophages in the tumor microenvironment.
Collapse
Affiliation(s)
- Emma M Conway
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Larissa A Pikor
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Sonia H Y Kung
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Melisa J Hamilton
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Stephen Lam
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Wan L Lam
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Kevin L Bennewith
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
211
|
Morimoto M, Matsuo Y, Koide S, Tsuboi K, Shamoto T, Sato T, Saito K, Takahashi H, Takeyama H. Enhancement of the CXCL12/CXCR4 axis due to acquisition of gemcitabine resistance in pancreatic cancer: effect of CXCR4 antagonists. BMC Cancer 2016; 16:305. [PMID: 27175473 PMCID: PMC4866076 DOI: 10.1186/s12885-016-2340-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 05/08/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The CXCL12-CXCR4 signaling axis in malignant tumor biology has increased in importance, and these peptides are implicated in tumor growth, invasion and metastasis. The aim of our study was to examine the important role of the axis in pancreatic cancer (PaCa) cells' relationship with stromal cells in gemcitabine-resistant (GEM-R) tumors and to confirm the effectiveness of CXCR4 antagonists for the treatment of GEM-R PaCa cells. METHODS We established two GEM-R PaCa cell lines using MIA PaCa-2 and AsPC-1 cells. The expression of CXCR4 mRNA in PaCa cells and the expression of CXCL12 mRNA in fibroblasts were examined by reverse transcription polymerase chain reaction (RT-PCR). The expression of CXCR4 protein in PaCa cells was examined by immunosorbent assay (ELISA) and immunocytochemistry. Using Matrigel invasion assays and animal studies, we then examined the effects of two CXCR4 antagonists, AMD11070 and KRH3955, on the invasiveness and tumorigenicity of GEM-R PaCa cells stimulated by CXCL12. RESULTS We found that the expression of CXCR4 in GEM-R PaCa cells was significantly enhanced by GEM but not in normal GEM-sensitive (GEM-S) PaCa cells. In RT-PCR and ELISA assays, the production and secretion of CXCL12 from fibroblasts was significantly enhanced by co-culturing with GEM-R PaCa cells treated with GEM. In Matrigel invasion assays, the invasiveness of GEM-R PaCa cells treated with GEM was significantly activated by fibroblast-derived CXCL12 and was significantly inhibited by CXCR4 antagonists, AMD11070 and KRH3955. In vivo, the tumorigenicity of GEM-R PaCa cells was activated by GEM, and it was significantly inhibited by the addition with CXCR4 antagonists. CONCLUSIONS Our findings demonstrate that the CXCL12-CXCR4 signaling axis plays an important role in PaCa cells' resistance to GEM. CXCR4 expression was significantly enhanced by the exposure to GEM in GEM-R PaCa cells but not in GEM-S PaCa cells. Furthermore, CXCR4 antagonists can inhibit the growth and invasion of GEM-R PaCa cells. These agents may be useful as second-line chemotherapy for GEM-R PaCa in the future.
Collapse
Affiliation(s)
- Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan.
| | - Shuji Koide
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan
| | - Ken Tsuboi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan
| | - Tomoya Shamoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan
| | - Takafumi Sato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan
| | - Hiromitsu Takeyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Kawasumi 1, Mizuho-cho, Mizuhoku, Nagoya, 467-8601, Japan
| |
Collapse
|
212
|
Kim B, Yoon J, Yoon SW, Park B. Onbaekwon Suppresses Colon Cancer Cell Invasion by Inhibiting Expression of the CXC Chemokine Receptor 4. Integr Cancer Ther 2016; 16:244-251. [PMID: 27160279 PMCID: PMC5739121 DOI: 10.1177/1534735416645182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cysteine X cysteine (CXC) chemokine receptor 4 (CXCR4) and C-X-C motif chemokine 12 (CXCL12) were originally identified as chemoattractants between immune cells and sites of inflammation. Since studies have validated an increased level of CXCL12 and its receptor in patients with colorectal cancers, CXCL12/CXCR4 axis has been considered as a valuable marker of cancer metastasis. Therefore, identification of CXCR4 inhibitors has great potential to abrogate tumor metastasis. Onbaekwon (OBW) is a complex herbal formula that is derived from the literature of traditional Korean medicine Dongeuibogam. In this study, we demonstrated that OBW suppressed CXCR4 expression in various cancer cell types in a concentration- and time-dependent manner. Both proteasomal and lysosomal inhibitors had no effect to prevent the OBW-induced suppression of CXCR4, suggesting that the inhibitory effect of OBW was not due to proteolytic degradation but occurred at the transcriptional level. Electrophoretic mobility shift assay further confirmed that OBW could block endogenous activation of nuclear factor kappa B, a key transcription factor that regulates the expression of CXCR4 in colon cancer cells. Consistent with the aforementioned molecular basis, OBW abolished cell invasion induced by CXCL12 in colon cancer cells. Together, our results suggest that OBW, as a novel inhibitor of CXCR4, could be a promising therapeutic agent contributing to cancer treatment.
Collapse
Affiliation(s)
- Buyun Kim
- 1 Keimyung University, Daegu, South Korea
| | | | - Seong Woo Yoon
- 2 Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | | |
Collapse
|
213
|
Hainsworth JD, Reeves JA, Mace JR, Crane EJ, Hamid O, Stille JR, Flynt A, Roberson S, Polzer J, Arrowsmith ER. A Randomized, Open-Label Phase 2 Study of the CXCR4 Inhibitor LY2510924 in Combination with Sunitinib Versus Sunitinib Alone in Patients with Metastatic Renal Cell Carcinoma (RCC). Target Oncol 2016; 11:643-653. [DOI: 10.1007/s11523-016-0434-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
214
|
Cheng ZH, Shi YX, Yuan M, Xiong D, Zheng JH, Zhang ZY. Chemokines and their receptors in lung cancer progression and metastasis. J Zhejiang Univ Sci B 2016; 17:342-351. [PMID: 27143261 PMCID: PMC4868824 DOI: 10.1631/jzus.b1500258] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/05/2016] [Indexed: 01/10/2023]
Abstract
Lung cancer is the leading cause of cancer-related mortality around the world. Despite advancements in diagnosis, surgical techniques, and neoadjuvant chemoradiotherapy over the last decade, the mortality rate is still high and the 5-year survival is a dismal 15%. Fortunately, early detection by low-dose computed tomography (LDCT) scans has reduced mortality by 20%; yet, overall, 5-year-survival remains low at less than 20%. Therefore, in order to ameliorate this situation, a thorough understanding of the underlying molecular mechanisms is urgently needed. Chemokines and their receptors, crucial microenvironmental factors, play important roles in lung tumor genesis, progression, and metastasis, and exploring the mechanisms of this might bring new insights into early diagnosis and precisely targeted treatment. Consequently, this review will mainly focus on recent advancements on the axes of chemokines and their receptors of lung cancer.
Collapse
Affiliation(s)
- Zeng-hui Cheng
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
- Department of Radiology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai 201700, China
| | - Yu-xin Shi
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Min Yuan
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Dan Xiong
- Department of Clinical Laboratory, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Jiang-hua Zheng
- Department of Clinical Laboratory, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zhi-yong Zhang
- Department of Radiology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| |
Collapse
|
215
|
Bunn PA, Minna JD, Augustyn A, Gazdar AF, Ouadah Y, Krasnow MA, Berns A, Brambilla E, Rekhtman N, Massion PP, Niederst M, Peifer M, Yokota J, Govindan R, Poirier JT, Byers LA, Wynes MW, McFadden DG, MacPherson D, Hann CL, Farago AF, Dive C, Teicher BA, Peacock CD, Johnson JE, Cobb MH, Wendel HG, Spigel D, Sage J, Yang P, Pietanza MC, Krug LM, Heymach J, Ujhazy P, Zhou C, Goto K, Dowlati A, Christensen CL, Park K, Einhorn LH, Edelman MJ, Giaccone G, Gerber DE, Salgia R, Owonikoko T, Malik S, Karachaliou N, Gandara DR, Slotman BJ, Blackhall F, Goss G, Thomas R, Rudin CM, Hirsch FR. Small Cell Lung Cancer: Can Recent Advances in Biology and Molecular Biology Be Translated into Improved Outcomes? J Thorac Oncol 2016; 11:453-74. [PMID: 26829312 PMCID: PMC4836290 DOI: 10.1016/j.jtho.2016.01.012] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Paul A Bunn
- University of Colorado Cancer Center, Aurora, Colorado
| | - John D Minna
- University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Adi F Gazdar
- University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | - Anton Berns
- Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | - Jun Yokota
- Institute of Predictive and Personalized Medicine of Cancer, Barcelona, Spain; National Cancer Center Research Institute, Tokyo, Japan
| | | | - John T Poirier
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lauren A Byers
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Murry W Wynes
- International Association for the Study of Lung Cancer, Aurora, Colorado
| | | | | | | | - Anna F Farago
- Massachusetts General Hospital, Boston, Massachusetts
| | - Caroline Dive
- Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | | | | | - Jane E Johnson
- University of Texas Southwestern Medical Center, Dallas, Texas
| | - Melanie H Cobb
- University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - David Spigel
- Sara Cannon Research Institute, Nashville, Tennessee
| | | | - Ping Yang
- Mayo Clinic Cancer Center, Rochester, Minnesota
| | | | - Lee M Krug
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - John Heymach
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Caicun Zhou
- Cancer Institute of Tongji University Medical School, Shanghai, China
| | - Koichi Goto
- National Cancer Center Hospital East, Chiba, Japan
| | - Afshin Dowlati
- Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio
| | | | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | - Martin J Edelman
- University of Maryland, Greenebaum Cancer Center, Baltimore, Maryland
| | | | - David E Gerber
- University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | | | | | - David R Gandara
- University of California Davis Comprehensive Cancer Center, Davis, California
| | - Ben J Slotman
- Vrije Universiteit Medical Center, Amsterdam, Netherlands
| | | | | | | | | | - Fred R Hirsch
- University of Colorado Cancer Center, Aurora, Colorado.
| |
Collapse
|
216
|
Zirafi O, Hermann PC, Münch J. Proteolytic processing of human serum albumin generates EPI-X4, an endogenous antagonist of CXCR4. J Leukoc Biol 2016; 99:863-8. [PMID: 26965637 DOI: 10.1189/jlb.2mr1115-521rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/11/2016] [Indexed: 12/14/2022] Open
Abstract
The chemokine receptor CXCR4 is an important G protein-coupled receptor. Signaling via CXCL12 regulates a number of important biologic processes, including immune responses, organogenesis, or hematopoiesis. Dysregulation of CXCR4 signaling is associated with a variety of diseases, such as cancer development and metastasis, immunodeficiencies, or chronic inflammation. Here, we review our findings on endogenous peptide inhibitor of CXCR4 as a novel antagonist of CXCR4. This peptide is a 16-residue fragment of human serum albumin and was isolated as an inhibitor of CXCR4-tropic human immunodeficiency virus type 1 from a blood-derived peptide library. Endogenous peptide inhibitor of CXCR4 binds the second extracellular loop of CXCR4, thereby preventing engagement of CXCL12 and antagonizing the receptor. Consequently, endogenous peptide inhibitor of CXCR4 inhibits CXCL12-mediated migration of CXCR4-expressing cells in vitro, mobilizes hematopoietic stem cells, and suppresses inflammatory responses in vivo. We discuss the generation of endogenous peptide inhibitor of CXCR4, its relevance as biomarker for disease, and its role in human immunodeficiency virus/acquired immunodeficiency syndrome pathogenesis and cancer. Furthermore, we discuss why optimized endogenous peptide inhibitor of CXCR4 derivatives might have advantages over other CXCR4 antagonists.
Collapse
Affiliation(s)
- Onofrio Zirafi
- Institute of Molecular Virology, University of Ulm, Ulm, Germany
| | - Patrick C Hermann
- Department of Internal Medicine I, University of Ulm, Ulm, Germany; and
| | - Jan Münch
- Institute of Molecular Virology, University of Ulm, Ulm, Germany; Ulm Peptide Pharmaceuticals, University of Ulm, Ulm, Germany
| |
Collapse
|
217
|
Wang HC, Li TY, Chao YJ, Hou YC, Hsueh YS, Hsu KH, Shan YS. KIT Exon 11 Codons 557-558 Deletion Mutation Promotes Liver Metastasis Through the CXCL12/CXCR4 Axis in Gastrointestinal Stromal Tumors. Clin Cancer Res 2016; 22:3477-87. [PMID: 26936919 DOI: 10.1158/1078-0432.ccr-15-2748] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/15/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE KIT mutations, the most prevalent genetic event in gastrointestinal stromal tumors (GIST), are associated with malignant features and poor prognosis. Aggressive GISTs possess a high propensity to spread to the liver. This study aimed to explore the role of KIT mutations in GIST liver metastasis. EXPERIMENTAL DESIGN A total of 170 GISTs were used to determine the association between KIT mutations and liver metastasis. Immunohistochemistry was performed to assess the correlation of KIT mutations with CXCR4 and ETV1 expression. Genetic and pharmacologic methods were used to study the regulation of CXCR4 and ETV1 by KIT mutations. RESULTS Codons 557 and 558 in KIT exon 11 were deletion hot spots in GISTs. KIT exon 11 deletions involving codons 557-558 were highly associated with liver metastasis. Overexpression of mutant KIT with exon 11 codons 557-558 deletion (KIT Δ557-558) increased GIST cell motility and liver metastasis. Mechanistically, overexpression of KIT Δ557-558 in GIST cells increased ETV1 and CXCR4 expression. CXCR4 knockdown counteracted KIT Δ557-558-mediated cell migration. Moreover, KIT Δ557-558-induced CXCR4 expression could be abolished by silencing ETV1. The chromatin immunoprecipitation assay showed that ETV1 directly bound to the CXCR4 promoter. After ERK inhibitor PD325901 treatment, the upregulation of ETV1 by KIT Δ557-558 was prevented. In addition, KIT exon 11 codons 557-558 deletion enhanced CXCL12-mediated GIST cell migration and invasion. CONCLUSIONS KIT exon 11 557-558 deletion upregulates CXCR4 through increased binding of ETV1 to the CXCR4 promoter in GIST cells, which thus promotes liver metastasis. These findings highlighted the potential therapeutic targets for metastatic GISTs. Clin Cancer Res; 22(14); 3477-87. ©2016 AACR.
Collapse
Affiliation(s)
- Hao-Chen Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Ying Li
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Jui Chao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yuan-Shuo Hsueh
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Kai-Hsi Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. Department of Surgery, Tainan Hospital, Department of Health, Executive Yuan, Tainan, Taiwan.
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan.
| |
Collapse
|
218
|
Es-Haghi M, Bassami M, Dehghani H. Construction and Quantitative Validation of Chicken CXCR4 Expression Reporter. Mol Biotechnol 2016; 58:202-11. [PMID: 26809356 DOI: 10.1007/s12033-016-9917-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Site directional migration is an important biological event and an essential behavior for latent migratory cells. A migratory cell maintains its motility, survival, and proliferation abilities by a network of signaling pathways where CXCR4/SDF signaling route plays crucial role for directed homing of a polarized cell. The chicken embryo due to its specific vasculature modality has been used as a valuable model for organogenesis, migration, cancer, and metastasis. In this research, the regulatory regions of chicken CXCR4 gene have been characterized in a chicken hematopoietic lymphoblast cell line (MSB1). A region extending from -2000 bp upstream of CXCR4 gene to +68 after its transcriptional start site, in addition to two other mutant fragments were constructed and cloned in a promoter-less reporter vector. Promoter activity was analyzed by quantitative real-time RT-PCR and flow cytometry techniques. Our findings show that the full sequence from -2000 to +68 bp of CXCR4 regulatory region is required for maximum promoter functionality, while the mutant CXCR4 promoter fragments show a partial promoter activity. The chicken CXCR4 promoter validated in this study could be used for characterization of directed migratory cells in chicken development and disease models.
Collapse
Affiliation(s)
- Masoumeh Es-Haghi
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammadreza Bassami
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hesam Dehghani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.
- Embryonic and Stem Cell Biology and Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
219
|
Kashyap MK, Kumar D, Jones H, Amaya-Chanaga CI, Choi MY, Melo-Cardenas J, Ale-Ali A, Kuhne MR, Sabbatini P, Cohen LJ, Shelat SG, Rassenti LZ, Kipps TJ, Cardarelli PM, Castro JE. Ulocuplumab (BMS-936564 / MDX1338): a fully human anti-CXCR4 antibody induces cell death in chronic lymphocytic leukemia mediated through a reactive oxygen species-dependent pathway. Oncotarget 2016; 7:2809-2822. [PMID: 26646452 PMCID: PMC4823073 DOI: 10.18632/oncotarget.6465] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
The CXCR4 receptor (Chemokine C-X-C motif receptor 4) is highly expressed in different hematological malignancies including chronic lymphocytic leukemia (CLL). The CXCR4 ligand (CXCL12) stimulates CXCR4 promoting cell survival and proliferation, and may contribute to the tropism of leukemia cells towards lymphoid tissues. Therefore, strategies targeting CXCR4 may constitute an effective therapeutic approach for CLL. To address that question, we studied the effect of Ulocuplumab (BMS-936564), a fully human IgG4 anti-CXCR4 antibody, using a stroma--CLL cells co-culture model. We found that Ulocuplumab (BMS-936564) inhibited CXCL12 mediated CXCR4 activation-migration of CLL cells at nanomolar concentrations. This effect was comparable to AMD3100 (Plerixafor--Mozobil), a small molecule CXCR4 inhibitor. However, Ulocuplumab (BMS-936564) but not AMD3100 induced apoptosis in CLL at nanomolar concentrations in the presence or absence of stromal cell support. This pro-apoptotic effect was independent of CLL high-risk prognostic markers, was associated with production of reactive oxygen species and did not require caspase activation. Overall, these findings are evidence that Ulocuplumab (BMS-936564) has biological activity in CLL, highlight the relevance of the CXCR4-CXCL12 pathway as a therapeutic target in CLL, and provide biological rationale for ongoing clinical trials in CLL and other hematological malignancies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michelle R. Kuhne
- Bristol-Myers Squibb, Department of Cell Biology and Physiology, Redwood City, CA, USA
| | - Peter Sabbatini
- Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Lewis J. Cohen
- Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Suresh G. Shelat
- Department of Early Clinical and Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
| | | | - Thomas J. Kipps
- UCSD-Moores Cancer Center, La Jolla, CA, USA
- CLL Research Consortium, La Jolla, CA, USA
| | - Pina M. Cardarelli
- Bristol-Myers Squibb, Department of Cell Biology and Physiology, Redwood City, CA, USA
| | - Januario E. Castro
- UCSD-Moores Cancer Center, La Jolla, CA, USA
- CLL Research Consortium, La Jolla, CA, USA
| |
Collapse
|
220
|
Sun M, Tang H, Gao Y, Dai X, Yuan Y, Zhang C, Sun D. Constitutive expression and anticancer potency of a novel immunotoxin onconase-DV3. Oncol Rep 2016; 35:1987-94. [PMID: 26782924 DOI: 10.3892/or.2016.4570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/08/2015] [Indexed: 11/05/2022] Open
Abstract
Onconase is an RNase of the ribonuclease A superfamily that is purified from the Northern leopard frog (Rana pipiens). It targets several types of malignant tumors, digests cytoplasmic transfer RNA (tRNA), and causes tumor cell apoptosis. Onconase has been employed in clinical trials as an antitumor drug, and has revealed its valuable clinical activity in several types of tumors, particularly pleural mesothelioma. However, its inefficiency in targeting tumor cells and its non‑specific toxicity in normal tissues have diminished its clinical benefits. Furthermore, cyclization of the N-terminal glutamine residue (Gln), possesses more RNase activity than the structure of Met ahead of Glu in the N-terminal (99:1), which is more difficult for producing onconase by Pichia pastoris. Under the guidance of α-mating factor-pre (α-MF-pre) secretion signal, the secretion of the recombinant protein can reach a high level. In the present study, we constructed a constitutive expression vector for onconase-(DV3)2 (Onc-DV3) production in yeast Pichia pastoris with the GAP promoter, in which the Onc-DV3 gene is inserted downstream of the truncated Saccharomyces cerevisiae α-mating factor-pre (α-MF-pre) secretion signal. The immuno-RNase Onc-DV3 expressed a high level of production and bioactivity and possessed enhanced capability to deliver the Onc molecule to tumor cell monomeric counterparts. Notably, Onc-DV3 showed strong cytotoxicity to highly metastatic tumor cells, weak cytotoxicity to lowly metastatic tumor cells and no toxicity to normal cells. These results demonstrate that the specific toxicity to highly metastatic tumor cells has made Onc-DV3 a promising antitumor drug by using two copies of DV3 for the targeted delivery of onconase.
Collapse
Affiliation(s)
- Miaonan Sun
- Department of Biomedicine, Regeneration Medicine Institute, Jilin University, Changchun, Jilin 130061, P.R. China
| | - Huichun Tang
- Department of Biomedicine, Regeneration Medicine Institute, Jilin University, Changchun, Jilin 130061, P.R. China
| | - Yan Gao
- Jilin Zixin Pharmaceutical Co., Ltd., Changchun, Jilin 130041, P.R. China
| | - Xinxuan Dai
- Department of Biomedicine, Regeneration Medicine Institute, Jilin University, Changchun, Jilin 130061, P.R. China
| | - Yue Yuan
- Department of Biomedicine, Regeneration Medicine Institute, Jilin University, Changchun, Jilin 130061, P.R. China
| | - Chunmei Zhang
- Department of Biomedicine, Regeneration Medicine Institute, Jilin University, Changchun, Jilin 130061, P.R. China
| | - Dejun Sun
- Department of Biomedicine, Regeneration Medicine Institute, Jilin University, Changchun, Jilin 130061, P.R. China
| |
Collapse
|
221
|
Tulotta C, Stefanescu C, Beletkaia E, Bussmann J, Tarbashevich K, Schmidt T, Snaar-Jagalska BE. Inhibition of signaling between human CXCR4 and zebrafish ligands by the small molecule IT1t impairs the formation of triple-negative breast cancer early metastases in a zebrafish xenograft model. Dis Model Mech 2016; 9:141-53. [PMID: 26744352 PMCID: PMC4770151 DOI: 10.1242/dmm.023275] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/25/2015] [Indexed: 12/15/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and recurrent type of breast carcinoma that is associated with poor patient prognosis. Because of the limited efficacy of current treatments, new therapeutic strategies need to be developed. The CXCR4-CXCL12 chemokine signaling axis guides cell migration in physiological and pathological processes, including breast cancer metastasis. Although targeted therapies to inhibit the CXCR4-CXCL12 axis are under clinical experimentation, still no effective therapeutic approaches have been established to block CXCR4 in TNBC. To unravel the role of the CXCR4-CXCL12 axis in the formation of TNBC early metastases, we used the zebrafish xenograft model. Importantly, we demonstrate that cross-communication between the zebrafish and human ligands and receptors takes place and human tumor cells expressing CXCR4 initiate early metastatic events by sensing zebrafish cognate ligands at the metastatic site. Taking advantage of the conserved intercommunication between human tumor cells and the zebrafish host, we blocked TNBC early metastatic events by chemical and genetic inhibition of CXCR4 signaling. We used IT1t, a potent CXCR4 antagonist, and show for the first time its promising anti-tumor effects. In conclusion, we confirm the validity of the zebrafish as a xenotransplantation model and propose a pharmacological approach to target CXCR4 in TNBC. Summary: CXCR4-expressing human tumor cells respond to zebrafish cognate ligands and initiate metastatic events in a zebrafish xenograft model. The CXCR4 antagonist IT1t has promising tumor inhibitory effects.
Collapse
Affiliation(s)
- Claudia Tulotta
- Institute of Biology, Animal Sciences and Health, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Cristina Stefanescu
- Institute of Biology, Animal Sciences and Health, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elena Beletkaia
- Physics of Life Processes, Kamerligh Onnes-Huygens Laboratory, Leiden University, Niels Bohrweg 2, Leiden 2333 CA, The Netherlands
| | - Jeroen Bussmann
- Institute of Biology, Animal Sciences and Health, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | | | - Thomas Schmidt
- Physics of Life Processes, Kamerligh Onnes-Huygens Laboratory, Leiden University, Niels Bohrweg 2, Leiden 2333 CA, The Netherlands
| | - B Ewa Snaar-Jagalska
- Institute of Biology, Animal Sciences and Health, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
222
|
Cheng C, Yue W, Li L, Li S, Gao C, Si L, Tian H. Regulator of G-protein signaling 4: A novel tumor suppressor with prognostic significance in non-small cell lung cancer. Biochem Biophys Res Commun 2016; 469:384-91. [DOI: 10.1016/j.bbrc.2015.11.110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022]
|
223
|
Es-Haghi M, Soltanian S, Dehghani H. Perspective: Cooperation of Nanog, NF-κΒ, and CXCR4 in a regulatory network for directed migration of cancer stem cells. Tumour Biol 2015; 37:1559-65. [PMID: 26715265 DOI: 10.1007/s13277-015-4690-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/16/2015] [Indexed: 12/26/2022] Open
Abstract
Directed cell migration is a crucial mobility phase of cancer stem cells having stemness and tumorigenic characteristics. It is known that CXCR4 plays key roles in the perception of chemotactic gradients throughout the directed migration of CSCs. There are a number of complex signaling pathways and transcription factors that coordinate with CXCR4/CXCL12 axis during directed migration. In this review, we focus on some transcription factors such as Nanog, NF-κB, and Bmi-1 that cooperate with CXCR4/CXCL12 for the maintenance of stemness and induction of metastasis behavior in cancer stem cells.
Collapse
Affiliation(s)
- Masoumeh Es-Haghi
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sara Soltanian
- Department of Biology, Faculty of Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Hesam Dehghani
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran. .,Embryonic and Stem Cell Biology and Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
224
|
Lu J, Zhou WH, Ren L, Zhang YZ. CXCR4, CXCR7, and CXCL12 are associated with trophoblastic cells apoptosis and linked to pathophysiology of severe preeclampsia. Exp Mol Pathol 2015; 100:184-91. [PMID: 26721717 DOI: 10.1016/j.yexmp.2015.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/20/2015] [Accepted: 12/20/2015] [Indexed: 11/26/2022]
Abstract
Preeclampsia is a pregnancy disorder with sudden onset of maternal hypertension and proteinuria, which is characterized by defective cytotrophoblast invasion, increased apoptosis in cytotrophoblast, and diminished syncytial differentiation. In this study, samples from 11 mild preeclamptic patients, 18 severe preeclamptic patients, and 21 normal pregnant women were collected. The expression level of CXCL12 and its two receptors (CXCR4 and CXCR7) in these samples and their relationship with apoptosis were investigated. Morphological change of trophoblast cells that was observed by scanning electron microscope (SEM) indicated a significant tendency of apoptosis in the preeclamptic placenta. Immunohistochemical staining showed that expression level of three proteins was significantly lower in severe preeclamptic placentas compared with normal placentas (P<0.05), whereas no significant difference was found between mild preeclamptic and normal placentas (P>0.05). Real time quantitative PCR (RT-qPCR) and Western blot showed that both mRNA and protein expression level of CXCR4, CXCR7, and CXCL12 of trophoblasts were lower in the severe preeclampsia group than that in the normal group (P<0.05 for mRNA, P<0.01 for protein). In conclusion, our data revealed that the roles of CXCR4, CXCR7, and CXCL12 are associated with trophoblastic cells apoptosis and may be linked to the occurrence and development of severe preeclampsia.
Collapse
Affiliation(s)
- Jing Lu
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China
| | - Wen-Hui Zhou
- Medicine Center for Human Reproduction, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Liang Ren
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China
| | - Yuan-Zhen Zhang
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China.
| |
Collapse
|
225
|
Li YJ, Dai YL, Zhang WB, Li SJ, Tu CQ. Clinicopathological and prognostic significance of chemokine receptor CXCR4 in patients with bone and soft tissue sarcoma: a meta-analysis. Clin Exp Med 2015; 17:59-69. [PMID: 26678086 DOI: 10.1007/s10238-015-0405-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 12/06/2015] [Indexed: 02/05/2023]
Abstract
The prognostic significance of CXC chemokine receptor 4 (CXCR4) in patients with bone and soft tissue sarcomas remains controversial. To investigate the impact of its expression on survival and clinicopathological features, we performed a meta-analysis. Comprehensive literature searches were conducted in PubMed, Web of Science, Embase and Cochrane Library for relevant studies. In total, 12 studies with 997 sarcoma patients were included. CXCR4 expression was found to be significantly associated with poor overall survival (HR 2.37, 95 % CI 1.86-3.01; P < 0.001). Further, when the analysis was stratified by histological subtypes (bony sarcoma including osteosarcoma and Ewing sarcoma and soft tissue sarcoma including synovial sarcoma and rhabdomyosarcoma), statistical analysis method (multivariate analysis and univariate analysis) and CXCR4 measuring method (IHC or RT-PCR), the significant correlation to poor overall survival was also observed except for that in Ewing sarcoma and RT-PCR groups. As for clinicopathological features, CXCR4 expression was significantly associated with higher rate of metastasis (OR 6.97, 95 % CI 2.28-21.31; P = 0.001) and higher tumor stage (OR 7.55, 95 % CI 1.25-45.47; P = 0.027), but not associated with gender, age and tumor site. In conclusion, CXCR4 expression may be an effective predictive factor of poor prognosis and clinicopathological features for bone and soft tissue sarcomas. Further studies are needed to validate our findings.
Collapse
Affiliation(s)
- Yong-Jiang Li
- Department of Orthopedics, West China Hospital, Sichuan University, No. 37 Guoxuexiang, Chengdu, 610041, People's Republic of China
| | - Yi-Ling Dai
- College of Computer Science, Sichuan Normal University, Chengdu, 610068, People's Republic of China
| | - Wen-Biao Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, No. 37 Guoxuexiang, Chengdu, 610041, People's Republic of China
| | - Shuang-Jiang Li
- Department of Orthopedics, West China Hospital, Sichuan University, No. 37 Guoxuexiang, Chengdu, 610041, People's Republic of China
| | - Chong-Qi Tu
- Department of Orthopedics, West China Hospital, Sichuan University, No. 37 Guoxuexiang, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
226
|
Gou X, Wang R, Lam SSY, Hou J, Leung AYH, Sun D. Cell adhesion manipulation through single cell assembly for characterization of initial cell-to-cell interaction. Biomed Eng Online 2015; 14:114. [PMID: 26652601 PMCID: PMC4676142 DOI: 10.1186/s12938-015-0109-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/28/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell-to-cell interactions are complex processes that involve physical interactions, chemical binding, and biological signaling pathways. Identification of the functions of special signaling pathway in cell-to-cell interaction from the very first contact will help characterize the mechanism underlying the interaction and advance new drug discovery. METHODS This paper reported a case study of characterizing initial interaction between leukemia cancer cells and bone marrow stromal cells, through the use of an optical tweezers-based cell manipulation tool. Optical traps were used to assemble leukemia cells at different positions of the stromal cell layer and enable their interactions by applying a small trapping force to maintain the cell contact for a few minutes. Specific drug was used to inhibit the binding of molecules during receptor-ligand-mediated adhesion. RESULTS AND CONCLUSIONS Our results showed that the amount of adhesion molecule could affect cell adhesion during the first few minutes contact. We also found that leukemia cancer cells could migrate on the stromal cell layer, which was dependent on the adhesion state and activation triggered by specific chemokine. The reported approaches provided a new opportunity to investigate cell-to-cell interaction through single cell adhesion manipulation.
Collapse
Affiliation(s)
- Xue Gou
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
| | - Ran Wang
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
| | - Stephen S Y Lam
- Department of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Jundi Hou
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
| | - Anskar Y H Leung
- Department of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Dong Sun
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
227
|
Poty S, Désogère P, Goze C, Boschetti F, D'huys T, Schols D, Cawthorne C, Archibald SJ, Maëcke HR, Denat F. New AMD3100 derivatives for CXCR4 chemokine receptor targeted molecular imaging studies: synthesis, anti-HIV-1 evaluation and binding affinities. Dalton Trans 2015; 44:5004-16. [PMID: 25640878 DOI: 10.1039/c4dt02972k] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CXCR4 is a target of growing interest for the development of new therapeutic drugs and imaging agents as its role in multiple disease states has been demonstrated. AMD3100, a CXCR4 chemokine receptor antagonist that is in current clinical use as a haematopoietic stem cell mobilising drug, has been widely studied for its anti-HIV properties, potential to inhibit metastatic spread of certain cancers and, more recently, its ability to chelate radiometals for nuclear imaging. In this study, AMD3100 is functionalised on the phenyl moiety to investigate the influence of the structural modification on the anti-HIV-1 properties and receptor affinity in competition with anti-CXCR4 monoclonal antibodies and the natural ligand for CXCR4, CXCL12. The effect of complexation of nickel(II) in the cyclam cavities has been investigated. Two amino derivatives were obtained and are suitable intermediates for conjugation reactions to obtain CXCR4 molecular imaging agents. A fluorescent probe (BODIPY) and a precursor for (18)F (positron emitting isotope) radiolabelling were conjugated to validate this route to new CXCR4 imaging agents.
Collapse
Affiliation(s)
- Sophie Poty
- ICMUB (UMR CNRS 6302), 9 Av. Alain Savary, BP 47870 21000 Dijon, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Shi XY, Xiong LX, Xiao L, Meng C, Qi GY, Li WL. Downregulation of caveolin‑1 upregulates the expression of growth factors and regulators in co‑culture of fibroblasts with cancer cells. Mol Med Rep 2015; 13:744-52. [PMID: 26647977 PMCID: PMC4686091 DOI: 10.3892/mmr.2015.4610] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 10/22/2015] [Indexed: 12/18/2022] Open
Abstract
Reduced expression levels of caveolin-1 (Cav-1) in tumor stromal fibroblasts influences the occurrence and progression of tumors, particularly in breast cancer, but the relevant molecular mechanism is unclear. The present study aimed to clarify the potential mechanism underlying the promotion of tumor growth by reduced Cav-1 expression levels, by investigating Cav-1-targeted molecules in fibroblasts and breast cancer cells. The expression of growth factors in the ESF fibroblast cell line transfected with Cav-1 small interfering RNA (siRNA) was examined. The expression of apoptotic regulators in the BT474 breast cancer cell line that was co-cultured with the fibroblasts, was also investigated. The transfection of Cav-1-targeting siRNA in ESF cells resulted in efficient and specific inhibition of Cav-1 expression. The downregulation of Cav-1 increased the expression and secretion of stromal cell-derived factor-1 (SDF-1), epidermal growth factor (EGF) and fibroblast-specific protein-1 (FSP-1) in ESF cells. This resulted in the accelerated proliferation of the breast cancer cells. Tumor protein 53-induced glycolysis and apoptosis regulator (TIGAR) was upregulated in the BT474 cells under the condition of co-culture with Cav-1 siRNA fibroblasts, while levels of reactive oxygen species (ROS) were decreased, resulting in apoptosis inhibition in the breast cancer cells. These results demonstrated that the downregulation of Cav-1 promoted the growth of breast cancer cells through increasing SDF-1, EGF and FSP-1 in tumor stromal fibroblasts, and TIGAR levels in breast cancer cells. To the best of our knowledge, the present study supports the hypothesis that Cav-1 possesses tumor-suppressor properties, with the mechanism of Cav-1-dependent signaling involving the regulation of SDF-1, EGF, FSP-1 and TIGAR.
Collapse
Affiliation(s)
- Xiao-Yu Shi
- Key Laboratory of Medical Biology, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Xia Xiong
- Department of Pathophysiology, Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Liang Xiao
- Molecular Center Laboratory, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, P.R. China
| | - Chuang Meng
- Key Laboratory of Medical Biology, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Guan-Yun Qi
- Key Laboratory of Medical Biology, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wen-Lin Li
- Key Laboratory of Medical Biology, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
229
|
Marchese A. Monitoring Chemokine Receptor Trafficking by Confocal Immunofluorescence Microscopy. Methods Enzymol 2015; 570:281-92. [PMID: 26921951 DOI: 10.1016/bs.mie.2015.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Here, we describe a protocol to detect chemokine receptor CXCR4 by confocal immunofluorescence microscopy in HeLa cells treated with its chemokine ligand CXCL12. Typically, ligand-activated chemokine receptors undergo a multistep process of desensitization and/or internalization from the plasma membrane in order to terminate signaling. Once internalized to endosomes, chemokine receptors readily enter the recycling pathway and return to the cell surface, giving rise to resensitization of signaling. The chemokine receptor CXCR4, when activated by CXCL12 is also internalized to endosomes, but in contrast to many chemokine receptors it is mainly sorted to the degradative pathway, contributing to a loss in the cellular complement of CXCR4 and long-term downregulation of signaling. The trafficking of CXCR4 from early endosomes to lysosomes can be easily detected by confocal immunofluorescence microscopy by immunostaining fixed cells for the receptor and with markers of these vesicular compartments. This approach is advantageous because it can be used to identify factors that regulate the trafficking of CXCR4 from early endosomes to lysosomes. The protocol described here focuses on CXCR4, but it can be easily adapted to other chemokine receptors.
Collapse
Affiliation(s)
- Adriano Marchese
- Department of Pharmacology, Loyola University Chicago, Health Sciences Division, Maywood, Illinois, USA.
| |
Collapse
|
230
|
Kim JH, Graef AJ, Dickerson EB, Modiano JF. Pathobiology of Hemangiosarcoma in Dogs: Research Advances and Future Perspectives. Vet Sci 2015; 2:388-405. [PMID: 29061949 PMCID: PMC5644642 DOI: 10.3390/vetsci2040388] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/05/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022] Open
Abstract
Hemangiosarcoma (HSA) is an aggressive and common cancer in dogs. While cutaneous masses are often treatable by tumor excision, visceral tumors are almost always incurable. Treatment advances for this disease have been limited due to a poor understanding of the overall tumor biology. Based upon its histological appearance, HSA has been presumed to originate from transformed endothelial cells; however, accumulating data now suggest a pluripotent bone marrow progenitor as the cell of origin for this disease. More recently, the identification of a novel subclassification of HSAs has provided a foundation to further our understanding of the cellular characteristics of HSA tumor cells, along with those of the cells comprising the tumor microenvironment. These discoveries hold promise for the development of new approaches to improve treatments for canine HSA, as well as to establish the utility of this disease as a spontaneous model to understand the pathogenesis and develop new treatments for vascular tumors of humans. In this review, we will provide a brief historical perspective and pathobiology of canine HSA, along with a focus on the recent advances in the molecular and cellular understanding of these tumors. In addition, future directions that should continue to improve our understanding of HSA pathogenesis will be discussed.
Collapse
Affiliation(s)
- Jong-Hyuk Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Ashley J Graef
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Erin B Dickerson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jaime F Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
231
|
Liang AL, Qian HL, Zhang TT, Zhou N, Wang HJ, Men XT, Qi W, Zhang PP, Fu M, Liang X, Lin C, Liu YJ. Bifunctional fused polypeptide inhibits the growth and metastasis of breast cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5671-86. [PMID: 26527862 PMCID: PMC4621185 DOI: 10.2147/dddt.s90082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Breast cancer is the most common cancer and the leading cause of cancer-related death among women worldwide, with urgent need to develop new therapeutics. Targeted therapy is a promising strategy for breast cancer therapy. Stromal-derived factor-1/CXC chemokine receptor 4 (CXCR4) has been implicated in the metastasis of breast cancer, which renders it to be therapeutic target. This study aimed to evaluate the anticancer effect of fused TAT- DV1-BH3 polypeptide, an antagonist of CXCR4, and investigate the underlying mechanism for the cancer cell-killing effect in the treatment of breast cancer in vitro and in vivo. This results in a potent inhibitory effect of fused TAT-DV1-BH3 polypeptide on tumor growth and metastasis in nude mice bearing established MDA-MB-231 tumors. Fused TAT-DV1-BH3 polypeptide inhibited the proliferation of MDA-MB-231 and MCF-7 cells but did not affect that of HEK-293 cells. The fused TAT-DV1-BH3 polypeptide colocalized with mitochondria and exhibited a proapoptotic effect through the regulation of caspase-9 and -3. Furthermore, the fused TAT-DV1-BH3 polypeptide suppressed the migration and invasion of the highly metastatic breast cancer cell line MDA-MB-231 in a concentration-dependent manner. Notably, the DV1-mediated inhibition of the stromal-derived factor-1/CXCR4 pathway contributed to the antimetastasis effect, evident from the reduction in the level of phosphoinositide 3 kinase and matrix metalloproteinase 9 in MDA-MB-231 cells. Collectively, these results indicate that the apoptosis-inducing effect and migration- and invasion-suppressing effect explain the tumor regression and metastasis inhibition in vivo, with the involvement of caspase- and CXCR4-mediated signaling pathway. The data suggest that the fused TAT-DV1-BH3 polypeptide is a promising agent for the treatment of breast cancer, and more studies are warranted to fully elucidate the therapeutic targets and molecular mechanism.
Collapse
Affiliation(s)
- Ai-Ling Liang
- Medical Molecular Diagnostics Key Laboratory of Guangdong, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China ; Department of Biochemistry and Molecular Biology, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China ; Department of Clinical Biochemistry, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Hai-Li Qian
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Ting-Ting Zhang
- Medical Molecular Diagnostics Key Laboratory of Guangdong, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China ; Department of Biochemistry and Molecular Biology, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China ; Department of Clinical Biochemistry, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Ning Zhou
- Medical Molecular Diagnostics Key Laboratory of Guangdong, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China ; Department of Biochemistry and Molecular Biology, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China ; Department of Clinical Biochemistry, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Hai-Juan Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Xi-Ting Men
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Wei Qi
- Electroencephalogram Room, Guangdong Medical University Affiliated Hospital, Zhanjiang, Guangdong, People's Republic of China
| | - Ping-Ping Zhang
- Department of Orthopedics, Guangdong Medical University Affiliated Hospital, Zhanjiang, Guangdong, People's Republic of China
| | - Ming Fu
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Xiao Liang
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Chen Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yong-Jun Liu
- Medical Molecular Diagnostics Key Laboratory of Guangdong, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China ; Department of Biochemistry and Molecular Biology, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China ; Department of Clinical Biochemistry, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| |
Collapse
|
232
|
Chen Q, Zhong T. The association of CXCR4 expression with clinicopathological significance and potential drug target in prostate cancer: a meta-analysis and literature review. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5115-22. [PMID: 26379424 PMCID: PMC4567179 DOI: 10.2147/dddt.s82475] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
CXCR4/CXCL12 axis plays an important role in tumor growth, angiogenesis, metastasis, and therapeutic resistance. The aim of this study is to perform a meta-analysis and literature review to evaluate the association of CXCR4 expression with clinicopathological significance and prognosis in patients with prostate cancer (PCa). A detailed literature search was made in Medline, EMBASE, Web of Science, and Google Scholar for related research publications. The data were extracted and assessed independently. Analysis of pooled data was performed using Review Manager 5.2. Odds ratio (OR) with corresponding confidence intervals were calculated and summarized. The meta-analysis included a total of eleven studies and 630 patients. The rate of CXCR4 protein expression in PCa was significantly higher than in nonmalignant prostate tissues (OR =35.71, P<0.00001). The expression of CXCR4 protein was not significantly associated with Gleason score (P=0.73). However, the frequency of CXCR4 protein expression was significantly higher in T3–4 stage than in T1–2 stage of PCa (OR =2.35, P=0.001). The expression of CXCR4 protein was significantly associated with the presence of lymph node and bone metastasis of PCa: for lymph node metastasis positive versus negative, OR was 5.07 and P=0.0003, and for bone metastasis positive versus negative, OR was 7.03 and P=0.003. Cancer-specific survival of patients with PCa was significantly associated with CXCR4 protein expression, and the pooled Hazard ratio was 0.24 and P=0.002. In conclusion, the high expression of CXCR4 protein is a diagnostic biomarker of PCa, and it is significantly associated with T stages. The increased expression of CXCR4 protein is significantly associated with lymph nodes or bone metastasis, and CXCR4 is a poor prognosis predictor for patients with PCa. Taken together, our findings indicate that CXCR4 could be a target not only for the development of therapeutic intervention but also for the noninvasive monitoring of PCa progression.
Collapse
Affiliation(s)
- Qi Chen
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian, Shaanxi Province, People's Republic of China
| | - Tie Zhong
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian, Shaanxi Province, People's Republic of China
| |
Collapse
|
233
|
Soysal SD, Tzankov A, Muenst SE. Role of the Tumor Microenvironment in Breast Cancer. Pathobiology 2015; 82:142-52. [DOI: 10.1159/000430499] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
234
|
Cox BD, Mehta AK, DiRaddo JO, Liotta DC, Wilson LJ, Snyder JP. Structural analysis of CXCR4 - Antagonist interactions using saturation-transfer double-difference NMR. Biochem Biophys Res Commun 2015; 466:28-32. [PMID: 26301631 DOI: 10.1016/j.bbrc.2015.08.084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 08/19/2015] [Indexed: 11/15/2022]
Abstract
CXCR4 is a GPCR involved in leukocyte trafficking. Small molecule antagonists of the receptor may treat inflammatory disease, cancer and HIV. Here we probe the binding of a tetrahydroisoquinoline-based antagonist (TIQ-10) to CXCR4 using saturation transfer double-difference (STDD) NMR. STDD spectra were acquired using extracts from Chinese Hamster Ovary cells expressing membrane-embedded CXCR4. The experiments demonstrate competitive binding between TIQ-10 and established antagonists and provide the TIQ-10 - CXCR4 binding epitope. Molecular modeling of TIQ-10 into the binding pocket provides a pose consistent with STDD-derived interactions. This study paves the way for future investigations of GPCR-ligand interactions in a biological milieu for use in chemical biology, biochemistry, structural biology, and rational drug design.
Collapse
Affiliation(s)
- Bryan D Cox
- Emory University, Department of Chemistry, 1515 Dickey Dr., Atlanta, GA 30322, USA
| | - Anil K Mehta
- Emory University, Department of Chemistry, 1515 Dickey Dr., Atlanta, GA 30322, USA
| | - John O DiRaddo
- Emory University, Department of Chemistry, 1515 Dickey Dr., Atlanta, GA 30322, USA
| | - Dennis C Liotta
- Emory University, Department of Chemistry, 1515 Dickey Dr., Atlanta, GA 30322, USA
| | - Lawrence J Wilson
- Emory University, Department of Chemistry, 1515 Dickey Dr., Atlanta, GA 30322, USA
| | - James P Snyder
- Emory University, Department of Chemistry, 1515 Dickey Dr., Atlanta, GA 30322, USA.
| |
Collapse
|
235
|
Glycosaminoglycan silencing by engineered CXCL12 variants. FEBS Lett 2015; 589:2819-24. [DOI: 10.1016/j.febslet.2015.07.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 07/21/2015] [Accepted: 07/28/2015] [Indexed: 12/12/2022]
|
236
|
Thiele S, Mungalpara J, Steen A, Rosenkilde MM, Våbenø J. Determination of the binding mode for the cyclopentapeptide CXCR4 antagonist FC131 using a dual approach of ligand modifications and receptor mutagenesis. Br J Pharmacol 2015; 171:5313-29. [PMID: 25039237 DOI: 10.1111/bph.12842] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 06/25/2014] [Accepted: 07/08/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE The cyclopentapeptide FC131 (cyclo(-L-Arg(1) -L-Arg(2) -L-2-Nal(3) -Gly(4) -D-Tyr(5) -)) is an antagonist at the CXC chemokine receptor CXCR4, which plays a role in human immunodeficiency virus infection, cancer and stem cell recruitment. Binding modes for FC131 in CXCR4 have previously been suggested based on molecular docking guided by structure-activity relationship (SAR) data; however, none of these have been verified by in vitro experiments. EXPERIMENTAL APPROACH Heterologous (125) I-12G5-competition binding and functional assays (inhibition of CXCL12-mediated activation) of FC131 and three analogues were performed on wild-type CXCR4 and 25 receptor mutants. Computational modelling was used to rationalize the experimental data. KEY RESULTS The Arg(2) and 2-Nal(3) side chains of FC131 interact with residues in TM-3 (His(113) , Asp(171) ) and TM-5 (hydrophobic pocket) respectively. Arg(1) forms charge-charge interactions with Asp(187) in ECL-2, while D-Tyr(5) points to the extracellular side of CXCR4. Furthermore, the backbone of FC131 interacts with the chemokine receptor-conserved Glu(288) via two water molecules. Intriguingly, Tyr(116) and Glu(288) form a H-bond in CXCR4 crystal structures and mutation of either residue to Ala abolishes CXCR4 activity. CONCLUSIONS AND IMPLICATIONS Ligand modification, receptor mutagenesis and computational modelling approaches were used to identify the binding mode of FC131 in CXCR4, which was in agreement with binding modes suggested from previous SAR studies. Furthermore, insights into the mechanism for CXCR4 activation by CXCL12 were gained. The combined findings will facilitate future design of novel CXCR4 antagonists.
Collapse
Affiliation(s)
- S Thiele
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
237
|
Abstract
Deregulated inflammatory response plays a pivotal role in the initiation, development and progression of tumours. Potential molecular mechanism(s) that drive the establishment of an inflammatory-tumour microenvironment is not entirely understood owing to the complex cross-talk between pro-inflammatory and tumorigenic mediators such as cytokines, chemokines, oncogenes, enzymes, transcription factors and immune cells. These molecular mediators are critical linchpins between inflammation and cancer, and their activation and/or deactivation are influenced by both extrinsic (i.e. environmental and lifestyle) and intrinsic (i.e. hereditary) factors. At present, the research pertaining to inflammation-associated cancers is accumulating at an exponential rate. Interest stems from hope that new therapeutic strategies against molecular mediators can be identified to assist in cancer treatment and patient management. The present review outlines the various molecular and cellular inflammatory mediators responsible for tumour initiation, progression and development, and discusses the critical role of chronic inflammation in tumorigenesis.
Collapse
|
238
|
Progress toward rationally designed small-molecule peptide and peptidomimetic CXCR4 antagonists. Future Med Chem 2015; 7:1261-83. [DOI: 10.4155/fmc.15.64] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Over the last 5 years, X-ray structures of CXCR4 in complex with three different ligands (the small-molecule antagonist IT1t, the polypeptide antagonist CVX15 and the viral chemokine antagonist vMIP-II) have been released. In addition to the inherent scientific value of these specific X-ray structures, they provide a reliable structural foundation for studies of the molecular interactions between CXCR4 and its key peptide ligands (CXCL12 and HIV-1 gp120), and serve as valuable templates for further development of small-molecule CXCR4 antagonists with therapeutic potential. We here review recent computational studies of the molecular interactions between CXCR4 and its peptide ligands – based on the X-ray structures of CXCR4 – and the current status of small-molecule peptide and peptidomimetic CXCR4 antagonists.
Collapse
|
239
|
Nagaraju GP, Bramhachari PV, Raghu G, El-Rayes BF. Hypoxia inducible factor-1α: Its role in colorectal carcinogenesis and metastasis. Cancer Lett 2015; 366:11-8. [PMID: 26116902 DOI: 10.1016/j.canlet.2015.06.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 05/29/2015] [Accepted: 06/09/2015] [Indexed: 01/05/2023]
Abstract
Tumor growth creates a hypoxic microenvironment, which promotes angiogenesis and aggressive tumor growth and invasion. HIF1α is a central molecule involved in mediating these effects of hypoxia. In colorectal cancer (CRC), hypoxia stabilizes the transcription factor HIF1α, leading to the expression of genes that are involved in tumor vascularization, metastasis/migration, cell survival and chemo-resistance. Therefore, HIF1α is a rational target for the development of new therapeutics for CRC. This article reviews the central role of HIF1α in CRC angiogenesis, metastasis, and progression as well as the strategies to target HIF1α stabilization.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | | | - Godi Raghu
- Department of Biotechnology, Krishna University, Machilipatnam, AP-521001, India
| | - Bassel F El-Rayes
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
240
|
Krieg A, Riemer JC, Telan LA, Gabbert HE, Knoefel WT. CXCR4--A Prognostic and Clinicopathological Biomarker for Pancreatic Ductal Adenocarcinoma: A Meta-Analysis. PLoS One 2015; 10:e0130192. [PMID: 26091099 PMCID: PMC4474597 DOI: 10.1371/journal.pone.0130192] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/18/2015] [Indexed: 12/31/2022] Open
Abstract
Adenocarcinomas of the pancreatic duct (PDAC) are characteristically aggressive tumors that are extremely challenging to treat as curative surgical resection, the definitive treatment, is seldom possible. Regretably, most patients are diagnosed with metastatic disease at the time of initial presentation. In addition, current chemotherapeutic concepts that are used for advanced disease stages show frustrating results. Thus, there is an urgent need to identify novel therapeutic molecular targets that are associated with PDAC disease. Recently, the chemokine receptor CXCR4 has been demonstrated to be highly expressed in metastatic PDAC. However, the results of the published data on CXCR4 and its association with clinicopathological variables and prognosis in PDAC seem to be heterogeneous. Consequently, to clarify the relevance of CXCR4 as a biomarker in PDAC we performed a comprehensive literature search by using PubMed and Web of Science databases to identify articles that focused on the expression of CXCR4 in PDAC by using immunohistochemistry. Subsequently, data from nine relevant studies, encompassing 1183 patients were extracted, qualitatively assessed, and entered into a meta-analysis. By using a random effects model, the pooled hazard ratio of the seven studies that reported on patients overall survival revealed a correlation between expression of CXCR4 and poor prognosis (HR 1.49; 95% CI: 1.04-2.14; P = 0.03; I2 = 74%). Although heterogeneity became evident, subgroup analyses confirmed the prognostic value of CXCR4 in PDAC, especially in high-quality studies that performed multivariate analysis. In addition, meta-analysis revealed a strong association of CXCR4 expression with the UICC stage (OR: 3.40; 95% CI: 1.67-6.92; P = 0.0007; I2 = 0%) and metastatic disease (N-status: OR: 2.55; 95% CI: 1.56-4.15; P = 0.0002; I2 = 26%; recurrence to the liver: OR: 2.80; 95% CI: 1.48-5.29; P = 0.001; I2 = 0%). Taken together, our meta-analysis suggests that CXCR4 represents a useful prognostic biomarker in PDAC and might therefore be evaluated as a potential therapeutic target in the treatment of metastatic cancer disease of the pancreas.
Collapse
Affiliation(s)
- Andreas Krieg
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany
- * E-mail:
| | - Jasmin C. Riemer
- Institute of Pathology, Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany
| | - Leila A. Telan
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany
| | - Helmut E. Gabbert
- Institute of Pathology, Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany
| | - Wolfram T. Knoefel
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
241
|
|
242
|
Abstract
Chemokines mediate numerous physiological and pathological processes related primarily to cell homing and migration. The chemokine CXCL12, also known as stromal cell-derived factor-1, binds the G-protein-coupled receptor CXCR4, which, through multiple divergent pathways, leads to chemotaxis, enhanced intracellular calcium, cell adhesion, survival, proliferation, and gene transcription. CXCR4, initially discovered for its involvement in HIV entry and leukocytes trafficking, is overexpressed in more than 23 human cancers. Cancer cell CXCR4 overexpression contributes to tumor growth, invasion, angiogenesis, metastasis, relapse, and therapeutic resistance. CXCR4 antagonism has been shown to disrupt tumor-stromal interactions, sensitize cancer cells to cytotoxic drugs, and reduce tumor growth and metastatic burden. As such, CXCR4 is a target not only for therapeutic intervention but also for noninvasive monitoring of disease progression and therapeutic guidance. This review provides a comprehensive overview of the biological involvement of CXCR4 in human cancers, the current status of CXCR4-based therapeutic approaches, as well as recent advances in noninvasive imaging of CXCR4 expression.
Collapse
Affiliation(s)
- Samit Chatterjee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Babak Behnam Azad
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sridhar Nimmagadda
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
243
|
Handel TM. The Structure of a CXCR4:Chemokine Complex. Front Immunol 2015; 6:282. [PMID: 26097480 PMCID: PMC4457019 DOI: 10.3389/fimmu.2015.00282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/18/2015] [Indexed: 11/18/2022] Open
|
244
|
Totonchy JE, Clepper L, Phillips KG, McCarty OJT, Moses AV. CXCR7 expression disrupts endothelial cell homeostasis and causes ligand-dependent invasion. Cell Adh Migr 2015; 8:165-76. [PMID: 24710021 DOI: 10.4161/cam.28495] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The homeostatic function of endothelial cells (EC) is critical for a number of physiological processes including vascular integrity, immunity, and wound healing. Indeed, vascular abnormalities resulting from EC dysfunction contribute to the development and spread of malignancies. The alternative SDF-1/CXCL12 receptor CXCR7 is frequently and specifically highly expressed in tumor-associated vessels. In this study, we investigate whether CXCR7 contributes to vascular dysfunction by specifically examining the effect of CXCR7 expression on EC barrier function and motility. We demonstrate that CXCR7 expression in EC results in redistribution of CD31/PECAM-1 and loss of contact inhibition. Moreover, CXCR7+ EC are deficient in barrier formation. We show that CXCR7-mediated motility has no influence on angiogenesis but contributes to another motile process, the invasion of CXCR7+ EC into ligand-rich niches. These results identify CXCR7 as a novel manipulator of EC barrier function via alteration of PECAM-1 homophilic junctions. As such, aberrant expression of CXCR7 in the vasculature has the potential to disrupt vascular homeostasis and could contribute to vascular dysfunction in cancer systems.
Collapse
Affiliation(s)
- Jennifer E Totonchy
- Vaccine and Gene Therapy Institute; Oregon Health and Science University; Portland, OR USA
| | - Lisa Clepper
- Vaccine and Gene Therapy Institute; Oregon Health and Science University; Portland, OR USA
| | - Kevin G Phillips
- Department of Biomedical Engineering; Oregon Health and Science University; Portland, OR USA
| | - Owen J T McCarty
- Department of Biomedical Engineering; Oregon Health and Science University; Portland, OR USA
| | - Ashlee V Moses
- Vaccine and Gene Therapy Institute; Oregon Health and Science University; Portland, OR USA
| |
Collapse
|
245
|
Liao YX, Fu ZZ, Zhou CH, Shan LC, Wang ZY, Yin F, Zheng LP, Hua YQ, Cai ZD. AMD3100 reduces CXCR4-mediated survival and metastasis of osteosarcoma by inhibiting JNK and Akt, but not p38 or Erk1/2, pathways in in vitro and mouse experiments. Oncol Rep 2015; 34:33-42. [PMID: 25997540 PMCID: PMC4484610 DOI: 10.3892/or.2015.3992] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/29/2015] [Indexed: 12/27/2022] Open
Abstract
Osteosarcoma (OS) has an unfavorable prognosis and tends to metastasize to lung tissue. Although the CXCL12-CXCR4 axis appears to affect progression and metastasis in numerous tumors, its mechanism and downstream pathways in OS remain unclear. We used western blotting and flow cytometry to detect CXCR4 and CXCR7 expression in two OS cell lines (LM8 and Dunn). An MTT assay was used to evaluate the effects of CXCL12 and AMD3100, a specific CXCR4 antagonist, on cell viability. Flow cytometry was utilized to analyze changes in apoptosis induced by serum deprivation following treatment with CXCL12 and AMD3100. A Transwell assay was used to assess cell migration in response to CXCL12 and AMD3100. Western blotting was performed to identify the phosphorylation of signaling molecules (JNK, c-Jun, Akt, p38 and Erk1/2) and expression of caspase-3 and -8, and PARP. Mouse models were employed to evaluate AMD3100 inhibition of primary OS growth and lung metastasis in vivo. CXCR4 expression was detected in LM8 but not Dunn cells, and neither cell line expressed CXCR7. The addition of CXCL12 induced the survival and migration of serum-starved CXCR4+ LM8 cells activating JNK and Akt pathways, which were abrogated by adding AMD3100. However, similar results were not observed in CXCR4− Dunn cells. CXCL12 protected LM8, but not Dunn cells, from apoptosis induced by serum deprivation by suppressing PARP cleavage, which was partly reversed by AMD3100. In a mouse model, AMD3100 reduced primary tumor growth and lung metastasis compared with the controls. Thus, the CXCL12-CXCR4 axis regulated OS survival and metastasis through the JNK and Akt pathways, and blocking them with AMD3100 was found to be a potential OS treatment.
Collapse
Affiliation(s)
- Yu-Xin Liao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Ze-Ze Fu
- Department of Orthopaedics, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Cheng-Hao Zhou
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Lian-Cheng Shan
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Zhuo-Ying Wang
- Department of Orthopaedics, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Fei Yin
- Department of Orthopaedics, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Long-Po Zheng
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Ying-Qi Hua
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Zheng-Dong Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
246
|
Serum chemokine network correlates with chemotherapy in non-small cell lung cancer. Cancer Lett 2015; 365:57-67. [PMID: 25976768 DOI: 10.1016/j.canlet.2015.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 04/17/2015] [Accepted: 05/05/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Inflammation plays an important role in the microenvironment of lung cancer. The present study aimed to evaluate the association of inflammatory biomarker networks with chemotherapies for patients with non-small cell lung cancer (NSCLC). METHODS The sera of healthy non-smokers (n = 14) and patients with NSCLC (n = 50), 36 with adenocarcinoma and 14 with squamous cell carcinoma, were collected. Healthy patients were untreated, while those with NSCLC were either chemotherapy-naïve or had received one and two courses of chemotherapy. The cytokine concentrations were measured using multiplexed cytokine immunoassays. The clinical informatics was scored with a Digital Evaluation Score System (DESS) to assess the severity of the patients. All patients completed follow-up for up to 2 years. RESULTS Among the 40 mediators measured, 13 significantly differed between patients with lung cancer and healthy controls, while 18 differed between untreated patients and those with stage IV adenocarcinoma who had undergone the first and second chemotherapy courses. The protein network of cytokines in NSCLC after multiple courses of chemotherapy was similar to that of normal persons. MIP-3α is the most crucial biomarker for predicting survival rates in NSCLC patients. CONCLUSIONS Our data identify an NSCLC-specific profile of inflammatory mediators that may be useful for cancer sub-classification, as well as the evaluation of therapeutic effects and overall survival.
Collapse
|
247
|
Mühlethaler-Mottet A, Liberman J, Ascenção K, Flahaut M, Balmas Bourloud K, Yan P, Jauquier N, Gross N, Joseph JM. The CXCR4/CXCR7/CXCL12 Axis Is Involved in a Secondary but Complex Control of Neuroblastoma Metastatic Cell Homing. PLoS One 2015; 10:e0125616. [PMID: 25955316 PMCID: PMC4425663 DOI: 10.1371/journal.pone.0125616] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/24/2015] [Indexed: 12/16/2022] Open
Abstract
Neuroblastoma (NB) is one of the most deadly solid tumors of the young child, for which new efficient and targeted therapies are strongly needed. The CXCR4/CXCR7/CXCL12 chemokine axis has been involved in the progression and organ-specific dissemination of various cancers. In NB, CXCR4 expression was shown to be associated to highly aggressive undifferentiated tumors, while CXCR7 expression was detected in more differentiated and mature neuroblastic tumors. As investigated in vivo, using an orthotopic model of tumor cell implantation of chemokine receptor-overexpressing NB cells (IGR-NB8), the CXCR4/CXCR7/CXCL12 axis was shown to regulate NB primary and secondary growth, although without any apparent influence on organ selective metastasis. In the present study, we addressed the selective role of CXCR4 and CXCR7 receptors in the homing phase of metastatic dissemination using an intravenous model of tumor cell implantation. Tail vein injection into NOD-scid-gamma mice of transduced IGR-NB8 cells overexpressing CXCR4, CXCR7, or both receptors revealed that all transduced cell variants preferentially invaded the adrenal gland and typical NB metastatic target organs, such as the liver and the bone marrow. However, CXCR4 expression favored NB cell dissemination to the liver and the lungs, while CXCR7 was able to strongly promote NB cell homing to the adrenal gland and the liver. Finally, coexpression of CXCR4 and CXCR7 receptors significantly and selectively increased NB dissemination toward the bone marrow. In conclusion, CXCR4 and CXCR7 receptors may be involved in a complex and organ-dependent control of NB growth and selective homing, making these receptors and their inhibitors potential new therapeutic targets.
Collapse
Affiliation(s)
- Annick Mühlethaler-Mottet
- Department of Pediatrics, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| | - Julie Liberman
- Department of Pediatrics, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
| | - Kelly Ascenção
- Department of Pediatrics, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
| | - Marjorie Flahaut
- Department of Pediatrics, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
| | - Katia Balmas Bourloud
- Department of Pediatrics, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
| | - Pu Yan
- Department of Pathology, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Jauquier
- Department of Pediatrics, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
| | - Nicole Gross
- Department of Pediatrics, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
| | - Jean-Marc Joseph
- Department of Pediatrics, University Hospital CHUV, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
248
|
Chen Q, Zhou Z, Shan L, Zeng H, Hua Y, Cai Z. The importance of Src signaling in sarcoma. Oncol Lett 2015; 10:17-22. [PMID: 26170970 DOI: 10.3892/ol.2015.3184] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 01/22/2015] [Indexed: 12/23/2022] Open
Abstract
Src is a tyrosine kinase that is of significance in tumor biology. The present review focuses on Src, its molecular structure, and role in cancer, in addition to its expression and function in sarcoma. In addition, the feasibility of Src as a potential drug target for the treatment of sarcoma is also discussed. Previous studies have suggested that Src has essential functions in cell proliferation, apoptosis, invasion, metastasis and the tumor microenvironment. Thus, it may be a potential target for cancer therapy. Src has been found to enhance proliferation, reduce apoptosis and promote metastasis in certain subtypes of sarcoma, including osteosarcoma, chondrosarcoma and Ewing's sarcoma. Furthermore, a number of novel effective therapeutic agents, such as SI-83, which target Src have been investigated in vitro and in vivo. Bosutinib and dasatinib, which inhibit Src, have been approved by the U.S. Food and Drug Administration for the treatment of chronic myelogenous leukemia. In addition, vandetanib is approved for the treatment of medullary thyroid cancer. Furthermore, the Src inhibitor, saracatinib, is currently in clinical trials for the treatment of a variety of solid tumors, including breast and lung cancers. Thus, Src is considered to be an important factor in sarcoma progression and may present a novel clinical therapeutic target. This review demonstrates the importance and clinical relevance of Src in sarcoma, and discusses a number of small molecular inhibitors of src kinase, such as dasatinib and sarcatinib, which are currently in clinical trials for the treatment of sarcoma patients.
Collapse
Affiliation(s)
- Quanchi Chen
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Zifei Zhou
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Liancheng Shan
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Hui Zeng
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Zhengdong Cai
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
249
|
Construction of a CXCL12-KDEL fusion gene to inhibit head and neck squamous cell carcinoma metastasis by intracellular sequestration of CXCR4. BIOMED RESEARCH INTERNATIONAL 2015; 2015:195828. [PMID: 25866764 PMCID: PMC4383411 DOI: 10.1155/2015/195828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/18/2014] [Accepted: 08/19/2014] [Indexed: 12/30/2022]
Abstract
The CXCL12-CXCR4 biological axis consisting of the chemotactic factor CXCL12 and its specific receptor CXCR4 plays an important role in oral cancer metastasis. High expression of CXCR4 may help oral squamous cancer cells invade local tissues and metastasize to lymph nodes. No obvious association was observed between CXCL12 expression and lymph node metastasis, suggesting that CXCL12 chemotaxis may only be related to CXCR4 expression on the tumor cell membrane. KDEL can be retained by receptors on the surface of the intracellular endoplasmic reticulum (ER) and also be called an ER retention signal sequence. So we adopted the KDEL sequence in this study to generate a CXCL12-KDEL fusion protein in combination with a traceable E-tag label. As such, CXCL12 was retained in the ER. Specific receptor CXCR4 binds to the CXCL12-KDEL, was also retained in the ER, and was thus prevented from reaching the oral squamous cancer cell surface. We reduced the cell surface level of CXCR4 and called the technique “intracellular sequestration.” By this way, we have finished blocking of CXCL12-CXCR4 biological axis and inhibiting lymph node metastasis of oral carcinoma.
Collapse
|
250
|
Panneerselvam J, Jin J, Shanker M, Lauderdale J, Bates J, Wang Q, Zhao YD, Archibald SJ, Hubin TJ, Ramesh R. IL-24 inhibits lung cancer cell migration and invasion by disrupting the SDF-1/CXCR4 signaling axis. PLoS One 2015; 10:e0122439. [PMID: 25775124 PMCID: PMC4361489 DOI: 10.1371/journal.pone.0122439] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/11/2015] [Indexed: 01/17/2023] Open
Abstract
Background The stromal cell derived factor (SDF)-1/chemokine receptor (CXCR)-4 signaling pathway plays a key role in lung cancer metastasis and is molecular target for therapy. In the present study we investigated whether interleukin (IL)-24 can inhibit the SDF-1/CXCR4 axis and suppress lung cancer cell migration and invasion in vitro. Further, the efficacy of IL-24 in combination with CXCR4 antagonists was investigated. Methods Human H1299, A549, H460 and HCC827 lung cancer cell lines were used in the present study. The H1299 lung cancer cell line was stably transfected with doxycycline-inducible plasmid expression vector carrying the human IL-24 cDNA and used in the present study to determine the inhibitory effects of IL-24 on SDF-1/CXCR4 axis. H1299 and A549 cell lines were used in transient transfection studies. The inhibitory effects of IL-24 on SDF1/CXCR4 and its downstream targets were analyzed by quantitative RT-PCR, western blot, luciferase reporter assay, flow cytometry and immunocytochemistry. Functional studies included cell migration and invasion assays. Principal Findings Endogenous CXCR4 protein expression levels varied among the four human lung cancer cell lines. Doxycycline-induced IL-24 expression in the H1299-IL24 cell line resulted in reduced CXCR4 mRNA and protein expression. IL-24 post-transcriptionally regulated CXCR4 mRNA expression by decreasing the half-life of CXCR4 mRNA (>40%). Functional studies showed IL-24 inhibited tumor cell migration and invasion concomitant with reduction in CXCR4 and its downstream targets (pAKTS473, pmTORS2448, pPRAS40T246 and HIF-1α). Additionally, IL-24 inhibited tumor cell migration both in the presence and absence of the CXCR4 agonist, SDF-1. Finally, IL-24 when combined with CXCR4 inhibitors (AMD3100, SJA5) or with CXCR4 siRNA demonstrated enhanced inhibitory activity on tumor cell migration. Conclusions IL-24 disrupts the SDF-1/CXCR4 signaling pathway and inhibits lung tumor cell migration and invasion. Additionally, IL-24, when combined with CXCR4 inhibitors exhibited enhanced anti-metastatic activity and is an attractive therapeutic strategy for lung metastasis.
Collapse
Affiliation(s)
- Janani Panneerselvam
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jiankang Jin
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Manish Shanker
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jason Lauderdale
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jonathan Bates
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Qi Wang
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Yan D. Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | | | - Timothy J. Hubin
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Chemistry, Southwestern Oklahoma State University, Weatherford, Oklahoma, United States of America
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Graduate Program in Biomedical Sciences, Oklahoma City, Oklahoma, United States of America
- * E-mail: (RR)
| |
Collapse
|