Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
Biomedicines ; 11(2)2023 Jan 26.
Article in English | MEDLINE | ID: mdl-36830899

ABSTRACT

Given that, even after multimodal therapy, early-stage lung cancer (LC) often recurs, novel prognostic markers to help guide therapy are highly desired. The mRNA levels of cell division cycle 25C (CDC25C), a phosphatase that regulates G2/M cell cycle transition in malignant cells, correlate with poor clinical outcomes in lung adenocarcinoma (LUAD). However, whether CDC25C protein detected by immunohistochemistry can serve as a prognostic marker in LUAD is yet unknown. We stained an LC tissue array and a cohort of 61 LUAD tissue sections for CDC25C and searched for correlations between CDC25C staining score and the pathological characteristics of the tumors and the patients' clinical outcomes. Clinical data were retrieved from our prospectively maintained departmental database. We found that high expression of CDC25C was predominant among poorly differentiated LUAD (p < 0.001) and in LUAD > 1cm (p < 0.05). Further, high expression of CDC25C was associated with reduced disease-free survival (p = 0.03, median follow-up of 39 months) and with a trend for reduced overall survival (p = 0.08). Therefore, high expression of CDC25C protein in LUAD is associated with aggressive histological features and with poor outcomes. Larger studies are required to further validate CDC25C as a prognostic marker in LUAD.

2.
Front Immunol ; 13: 1026185, 2022.
Article in English | MEDLINE | ID: mdl-36685577

ABSTRACT

Given the need to improve the efficacy of standard-of-care immunotherapy (anti-CTLA-4 + anti-PD-1) in human malignant pleural mesothelioma (hMPM), we thoroughly characterized the immunobiology of the AB12 murine mesothelioma (MM) model, aiming to increase its accuracy in predicting the response of hMPM to immunotherapy and in designing novel anti-hMPM treatments. Specifically, we used immunologic, transcriptomic and survival analyses, to synchronize the MM tumor growth phases and immune evolution with the histo-molecular and immunological characteristics of hMPM while also determining the anti-MM efficacy of standard-of-care anti-hMPM immunotherapy as a benchmark that novel therapeutics should meet. We report that early-, intermediate- and advanced- AB12 tumors are characterized by a bell-shaped anti-tumor response that peaks in intermediate tumors and decays in advanced tumors. We further show that intermediate- and advanced- tumors match with immune active ("hot") and immune inactive ("cold") hMPM respectively, and that they respond to immunotherapy in a manner that corresponds well with its performance in real-life settings. Finally, we show that in advanced tumors, addition of cisplatin to anti CTLA-4 + anti PD-1 can extend mice survival and invigorate the decaying anti-tumor response. Therefore, we highlight this triple combination as a worthy candidate to improve clinical outcomes in hMPM.


Subject(s)
Mesothelioma, Malignant , Mesothelioma , Humans , Animals , Mice , Mesothelioma, Malignant/drug therapy , Mesothelioma/drug therapy , Mesothelioma/pathology , Cisplatin/therapeutic use , Immunotherapy , Immunity
3.
J Thorac Cardiovasc Surg ; 155(5): 2164-2175.e1, 2018 05.
Article in English | MEDLINE | ID: mdl-29429629

ABSTRACT

OBJECTIVE: Past studies are inconsistent with regard to the role of matrix metalloproteinase 12 in lung tumorigenesis. This is due, in part, to differential tumorigenesis based on tumor-derived versus immune-derived matrix metalloproteinase 12 expression. Our study aims to thoroughly dissect the role of matrix metalloproteinase 12 in lung tumorigenesis. METHODS: We tested matrix metalloproteinase 12 expression and the association with prognosis using a tissue array and a published non-small cell lung cancer gene expression database. In addition, we characterized the contribution of matrix metalloproteinase 12 to tumor propagation in the lung using a series of in vitro and in vivo studies. RESULTS: Tumor cells of a diverse set of human lung cancers stained positive for matrix metalloproteinase 12, and high matrix metalloproteinase 12 mRNA levels in the tumor were associated with reduced survival. The lung microenvironment stimulated endogenous production of matrix metalloproteinase 12 in lung cancer cells (human 460 lung cancer cell line, Lewis lung carcinoma). In vitro, matrix metalloproteinase 12 knockout Lewis lung carcinoma and Lewis lung carcinoma cells had the same proliferation rate, but Lewis lung carcinoma showed increased invasiveness. In vivo, deficiency of matrix metalloproteinase 12 in Lewis lung carcinoma cells, but not in the host, reduced tumor growth and invasiveness. CONCLUSIONS: We suggest that tumor cell-derived matrix metalloproteinase 12 promotes tumor propagation in the lung and that in the context of pulmonary malignancies matrix metalloproteinase 12 should further be tested as a potential novel therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Lewis Lung/enzymology , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Movement , Lung Neoplasms/enzymology , Matrix Metalloproteinase 12/metabolism , Animals , Biomarkers, Tumor/genetics , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Matrix Metalloproteinase 12/genetics , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Signal Transduction
4.
Cancer Res ; 78(6): 1471-1483, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29259008

ABSTRACT

CXCR4 expression in neuroblastoma tumors correlates with disease severity. In this study, we describe mechanisms by which CXCR4 signaling controls neuroblastoma tumor growth and response to therapy. We found that overexpression of CXCR4 or stimulation with CXCL12 supports neuroblastoma tumorigenesis. Moreover, CXCR4 inhibition with the high-affinity CXCR4 antagonist BL-8040 prevented tumor growth and reduced survival of tumor cells. These effects were mediated by the upregulation of miR-15a/16-1, which resulted in downregulation of their target genes BCL-2 and cyclin D1, as well as inhibition of ERK. Overexpression of miR-15a/16-1 in cells increased cell death, whereas antagomirs to miR-15a/16-1 abolished the proapoptotic effects of BL-8040. CXCR4 overexpression also increased miR-15a/16-1, shifting their oncogenic dependency from the BCL-2 to the ERK signaling pathway. Overall, our results demonstrate the therapeutic potential of CXCR4 inhibition in neuroblastoma treatment and provide a rationale to test combination therapies employing CXCR4 and BCL-2 inhibitors to increase the efficacy of these agents.Significance: These results provide a mechanistic rationale for combination therapy of CXCR4 and BCL-2 inhibitors to treat a common and commonly aggressive pediatric cancer.Cancer Res; 78(6); 1471-83. ©2017 AACR.


Subject(s)
Brain Neoplasms/pathology , MicroRNAs/metabolism , Neuroblastoma/pathology , Receptors, CXCR4/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , MicroRNAs/genetics , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Peptides/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Xenograft Model Antitumor Assays
5.
Front Immunol ; 8: 1432, 2017.
Article in English | MEDLINE | ID: mdl-29218043

ABSTRACT

Chemokines and their receptors play critical roles in the progression of autoimmunity and inflammation. Typically, multiple chemokines are involved in the development of these pathologies. Indeed, targeting single chemokines or chemokine receptors has failed to achieve significant clinical benefits in treating autoimmunity and inflammation. Moreover, the binding of host atypical chemokine receptors to multiple chemokines as well as the binding of chemokine-binding proteins secreted by various pathogens can serve as a strategy for controlling inflammation. In this work, promiscuous chemokine-binding peptides that could bind and inhibit multiple inflammatory chemokines, such as CCL2, CCL5, and CXCL9/10/11, were selected from phage display libraries. These peptides were cloned into human mutated immunoglobulin Fc-protein fusions (peptibodies). The peptibodies BKT120Fc and BKT130Fc inhibited the ability of inflammatory chemokines to induce the adhesion and migration of immune cells. Furthermore, BKT120Fc and BKT130Fc also showed a significant inhibition of disease progression in a variety of animal models for autoimmunity and inflammation. Developing a novel class of antagonists that can control the courses of diseases by selectively blocking multiple chemokines could be a novel way of generating effective therapeutics.

6.
Clin Cancer Res ; 23(7): 1733-1747, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-27697999

ABSTRACT

Purpose: To explore the functional consequences of possible cross-talk between the CXCR4/CXCL12 and the sphingosine-1-phosphate (S1P) pathways in multiple myeloma (MM) cells and to evaluate the effect of S1P targeting with the FTY720 modulator as a potential anti-MM therapeutic strategy.Experimental Design and Results: S1P targeting with FTY720 induces MM cell apoptosis. The combination of FTY720 with the SPHK1 inhibitor SKI-II results in synergistic inhibition of MM growth. CXCR4/CXCL12-enhanced expression correlates with reduced MM cell sensitivity to both FTY720 and SKI-II inhibitors, and with SPHK1 coexpression in both cell lines and primary MM bone marrow (BM) samples, suggesting regulative cross-talk between the CXCR4/CXCL12 and SPHK1 pathways in MM cells. FTY720 was found to directly target CXCR4. FTY720 profoundly reduces CXCR4 cell-surface levels and abrogates the CXCR4-mediated functions of migration toward CXCL12 and signaling pathway activation. Moreover, FTY720 cooperates with bortezomib, inducing its cytotoxic activity and abrogating the bortezomib-mediated increase in CXCR4 expression. FTY720 effectively targets bortezomib-resistant cells and increases their sensitivity to bortezomib, promoting DNA damage. Finally, in a recently developed novel xenograft model of CXCR4-dependent systemic MM with BM involvement, FTY720 treatment effectively reduces tumor burden in the BM of MM-bearing mice. FTY720 in combination with bortezomib demonstrates superior tumor growth inhibition and abrogates bortezomib-induced CXCR4 increase on MM cells.Conclusions: Altogether, our work identifies a cross-talk between the S1P and CXCR4 pathways in MM cells and provides a preclinical rationale for the therapeutic application of FTY720 in combination with bortezomib in patients with MM. Clin Cancer Res; 23(7); 1733-47. ©2016 AACR.


Subject(s)
Chemokine CXCL12/genetics , Multiple Myeloma/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptors, CXCR4/genetics , Animals , Apoptosis/drug effects , Bortezomib/administration & dosage , DNA Damage/drug effects , Drug Synergism , Fingolimod Hydrochloride/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysophospholipids/genetics , Lysophospholipids/metabolism , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/genetics , Sphingosine/metabolism , Xenograft Model Antitumor Assays
7.
J Thorac Oncol ; 10(1): 46-58, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25654727

ABSTRACT

BACKGROUND: The existing shortage of animal models that properly mimic the progression of early-stage human lung cancer from a solitary confined tumor to an invasive metastatic disease hinders accurate characterization of key interactions between lung cancer cells and their stroma. We herein describe a novel orthotopic animal model that addresses these concerns and consequently serves as an attractive platform to study tumor-stromal cell interactions under conditions that reflect early-stage lung cancer. METHODS: Unlike previous methodologies, we directly injected small numbers of human or murine lung cancer cells into murine's left lung and longitudinally monitored disease progression. Next, we used green fluorescent protein-tagged tumor cells and immuno-fluorescent staining to determine the tumor's microanatomic distribution and to look for tumor-infiltrating immune cells and stromal cells. Finally, we compared chemokine gene expression patterns in the tumor and lung microenvironment. RESULTS: We successfully generated a solitary pulmonary nodule surrounded by normal lung parenchyma that grew locally and spread distally over time. Notably, we found that both fibroblasts and leukocytes are recruited to the tumor's margins and that distinct myeloid cell attracting and CCR2-binding chemokines are specifically induced in the tumor microenvironment. CONCLUSION: Our orthotopic lung cancer model closely mimics the pathologic sequence of events that characterizes early-stage human lung cancer propagation. It further introduces new means to monitor tumor-stromal cell interactions and offers unique opportunities to test therapeutic targets under conditions that reflect early-stage lung cancer. We argue that for such purposes our model is superior to lung cancer models that are based either on genetic induction of epithelial transformation or on ectopic transplantation of malignant cells.


Subject(s)
Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Disease Models, Animal , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Transplantation, Heterologous , Tumor Microenvironment , Xenograft Model Antitumor Assays
8.
Oncotarget ; 5(22): 11283-96, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25526031

ABSTRACT

Multiple myeloma (MM) cells specifically attract peripheral-blood monocytes, while interaction of MM with bone marrow stromal cells (BMSCs) significantly increased monocyte recruitment (p<0.01). The CXCL12 chemokine, produced by both the MM and BMSCs, was found to be a critical regulator of monocyte migration. CXCL12 production was up-regulated under MM-BMSCs co-culture conditions, whereas blockage with anti-CXCR4 antibodies significantly abrogated monocyte recruitment toward a MM-derived conditioned medium (p<0.01). Furthermore, elevated levels of CXCL12 were detected in MM, but not in normal BM samples, whereas malignant MM cells often represented the source of increased CXCL12 in the BM. Blood-derived macrophages effectively supported MM cells proliferation and protected them from chemotherapy-induced apoptosis. Importantly, MM cells affected macrophage polarization, elevating the expression of M2-related scavenger receptor CD206 in macrophages and blocking LPS-induced TNFα secretion (a hallmark of M1 response). Of note, MM-educated macrophages suppressed T-cell proliferation and IFNγ production in response to activation. Finally, increased numbers of CXCR4-expressing CD163+CD206+ macrophages were detected in the BM of MM patients (n=25) in comparison to MGUS (n=11) and normal specimens (n=8). Taken together, these results identify macrophages as important players in MM tumorogenicity, and recognize the CXCR4/CXCL12 axis as a critical regulator of MM-stroma interactions and microenvironment formation.


Subject(s)
Chemokine CXCL12/metabolism , Macrophages/pathology , Multiple Myeloma/pathology , Receptors, CXCR4/metabolism , Adult , Aged , Apoptosis/physiology , Cell Line, Tumor , Cell Polarity/physiology , Cell Proliferation/physiology , Female , Humans , Macrophages/metabolism , Male , Middle Aged , Multiple Myeloma/metabolism , Phenotype , Signal Transduction
9.
Mol Cancer Ther ; 13(5): 1155-69, 2014 May.
Article in English | MEDLINE | ID: mdl-24502926

ABSTRACT

Functional role of CXCR4 in chronic myelogenous leukemia (CML) progression was evaluated. Elevated CXCR4 significantly increased the in vitro survival and proliferation in response to CXCL12. CXCR4 stimulation resulted in activation of extracellular signal-regulated kinase (Erk)-1/2, Akt, S6K, STAT3, and STAT5 prosurvival signaling pathways. In accordance, we found that in vitro treatment with CXCR4 antagonist BKT140 directly inhibited the cell growth and induced cell death of CML cells. Combination of BKT140 with suboptimal concentrations of imatinib significantly increased the anti-CML effect. BKT140 induced apoptotic cell death, decreasing the levels of HSP70 and HSP90 chaperones and antiapoptotic proteins BCL-2 and BCL-XL, subsequently promoting the release of mitochondrial factors cytochrome c and SMAC/Diablo. Bone marrow (BM) stromal cells (BMSC) markedly increased the proliferation of CML cells and protected them from imatinib-induced apoptosis. Furthermore, BMSCs elevated proto-oncogene BCL6 expression in the CML cells in response to imatinib treatment, suggesting the possible role of BCL6 in stroma-mediated TKI resistance. BKT140 reversed the protective effect of the stroma, effectively promoted apoptosis, and decreased BCL6 levels in CML cells cocultured with BMSCs. BKT140 administration in vivo effectively reduced the growth of subcutaneous K562-produced xenografts. Moreover, the combination of BKT140 with low-dose imatinib markedly inhibited tumor growth, achieving 95% suppression. Taken together, our data indicate the importance of CXCR4/CXCL12 axis in CML growth and CML-BM stroma interaction. CXCR4 inhibition with BKT140 antagonist efficiently cooperated with imatinib in vitro and in vivo. These results provide the rational basis for CXCR4-targeted therapy in combination with TKI to override drug resistance and suppress residual disease.


Subject(s)
Benzamides/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Oligopeptides/pharmacology , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Stromal Cells/drug effects , Stromal Cells/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Chemokine CXCL12/metabolism , Chemokine CXCL12/pharmacology , Disease Models, Animal , Drug Synergism , Gene Expression , Humans , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Proto-Oncogene Mas , Receptors, CXCR4/genetics , Signal Transduction/drug effects , Tumor Burden/drug effects , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 20(2): 469-79, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24246358

ABSTRACT

PURPOSE: CXCR4 plays an important role in the retention of stem cells within the bone marrow. BKT140 (4F-benzoyl-TN14003) is a 14-residue bio stable synthetic peptide, which binds CXCR4 with a greater affinity compared with plerixafor (4 vs. 84 nmol/L). Studies in mice demonstrated the efficient and superior mobilization and transplantation of stem cells collected with GCSF-BKT140, compared with those obtained when using stem cells obtained with each one of these mobilizing agent alone. These results have served as a platform for the present clinical phase I study. EXPERIMENTAL DESIGN: Eighteen patients with multiple myeloma who were preparing for their first autologous stem cell transplantation were included. Patients received a standard multiple myeloma mobilization regimen, consisting of 3 to 4 g/m(2) cyclophosphamide (day 0), followed by granulocyte colony-stimulating factor (G-CSF) at 5 µg/kg/d starting on day 5 and administered between 8 and 10 pm until the end of stem cell collection. A single injection of BKT140 (0.006, 0.03, 0.1, 0.3, and 0.9 mg/kg) was administered subcutaneously on day 10 in the early morning, followed by G-CSF 12 hours later. RESULTS: BKT140 was well tolerated at all concentrations, and none of the patients developed grade 3 and 4 toxicity. A single administration of BKT140 at the highest dose, 0.9 mg/kg, resulted in a robust mobilization and collection of CD34(+) cells (20.6 ± 6.9 × 10(6)/kg), which were obtained through a single apheresis. All transplanted patients received ∼5.3 × 10(6) CD34(+) cells/kg, which rapidly engrafted (n = 17). The median time to neutrophil and platelet recovery was 12 and 14 days, respectively, at the highest dose (0.9 mg/kg). CONCLUSIONS: When combined with G-CSF, BKT140 is a safe and efficient stem cell mobilizer that enabled the collection of a high number of CD34(+) cells in 1 and 2 aphaeresis procedures, resulting in successful engraftment.


Subject(s)
Antineoplastic Agents/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Oligopeptides/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Adult , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Blood Cell Count , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Transplantation , Humans , Male , Mice , Middle Aged , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Protein Binding , Receptors, CXCR4/metabolism , Treatment Outcome
11.
Br J Haematol ; 163(2): 248-59, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23906028

ABSTRACT

Platelets are the terminal differentiation product of megakaryocytes (MKs). Cytokines, such as thrombopoietin (TPO), are known to influence different steps in MK development; however, the complex differentiation and platelet localization processes are not fully understood. MKs express the receptor CXCR4 and have been shown to migrate in response to CXCL12 and to increase their platelet production. In this study, we studied the role of CXCR4 in platelet production with the high affinity CXCR4 antagonist, BKT140. Single and sequential administration of BKT140 significantly increased the number of MKs and haematopoietic progenitors (HPCs) within the bone marrow (BM). Increased megakaryopoiesis was associated with increased platelet production. Single and sequential administration of BKT140 also increased the number of HPCs in the blood. In a model of 5-fluorouracil-induced thrombocytopenia, BKT140 significantly reduced the severity and duration of thrombocytopenia and cytopenia when administered before and after chemotherapy. Our results demonstrated that the CXCR4 antagonist, BKT140, mediated unique beneficial effects by stimulating megakaryopoiesis and platelet production. These results provide evidence for the possible therapeutic use of BKT140 for modulating platelet numbers in thrombocytopenic conditions.


Subject(s)
Blood Platelets/cytology , Blood Platelets/drug effects , Oligopeptides/administration & dosage , Receptors, CXCR4/antagonists & inhibitors , Thrombopoiesis/drug effects , Thrombopoiesis/physiology , Animals , Antineoplastic Agents/adverse effects , Blood Platelets/metabolism , Bone Marrow/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Leukocyte Count , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/drug effects , Mice , Platelet Count , Thrombocytopenia/chemically induced , Thrombocytopenia/drug therapy , Thrombopoietin/pharmacology
12.
Clin Cancer Res ; 19(13): 3495-507, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23637121

ABSTRACT

PURPOSE: Chemokine axis CXCR4/CXCL12 is critically involved in the survival and trafficking of normal and malignant B lymphocytes. Here, we investigated the effect of high-affinity CXCR4 antagonist BKT140 on lymphoma cell growth and rituximab-induced cytotoxicity in vitro and in vivo. EXPERIMENTAL DESIGN: In vitro efficacy of BKT140 alone or in combination with rituximab was determined in non-Hodgkin lymphoma (NHL) cell lines and primary samples from bone marrow aspirates of patients with NHL. In vivo efficacy was evaluated in xenograft models of localized and disseminated NHL with bone marrow involvement. RESULTS: Antagonizing CXCR4 with BKT140 resulted in significant inhibition of CD20+ lymphoma cell growth and in the induction of cell death, respectively. Combination of BKT140 with rituximab significantly enhanced the apoptosis against the lymphoma cells in a dose-dependent manner. Moreover, rituximab induced CXCR4 expression in lymphoma cell lines and primary lymphoma cells, suggesting the possible interaction between CD20 and CXCR4 pathways in NHL. Primary bone marrow stromal cells (BMSC) further increased CXCR4 expression and protected NHL cells from rituximab-induced apoptosis, whereas BKT140 abrogated this protective effect. Furthermore, BKT140 showed efficient antilymphoma activity in vivo in the xenograft model of disseminated NHL with bone marrow involvement. BKT140 treatment inhibited the local tumor progression and significantly reduced the number of NHL cells in the bone marrow. Combined treatment of BKT140 with rituximab further decreased the number of viable lymphoma cells in the bone marrow, achieving 93% reduction. CONCLUSIONS: These findings suggest the possible role of CXCR4 in NHL progression and response to rituximab and provide the scientific basis for the development of novel CXCR4-targeted therapies for refractory NHL.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD20/metabolism , Antineoplastic Agents/pharmacology , Lymphoma, Non-Hodgkin/metabolism , Oligopeptides/pharmacology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Bone Marrow/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Microenvironment/drug effects , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/pathology , Mice , Rituximab , Xenograft Model Antitumor Assays
13.
Hepatology ; 58(3): 1021-30, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23526353

ABSTRACT

UNLABELLED: Human hepatocellular carcinoma (HCC) is an inflammation-induced cancer, which is the third-leading cause of cancer mortality worldwide. We investigated the role of the chemokine receptors, CCR5 and CCR1, in regulating inflammation and tumorigenesis in an inflammation-induced HCC model in mice. Multidrug resistance 2 gene (Mdr2)-knockout (Mdr2-KO) mice spontaneously develop chronic cholestatic hepatitis and fibrosis that is eventually followed by HCC. We generated two new strains from the Mdr2-KO mouse, the Mdr2:CCR5 and the Mdr2:CCR1 double knockouts (DKOs), and set out to compare inflammation and tumorigenesis among these strains. We found that in Mdr2-KO mice lacking the chemokine receptor, CCR5 (Mdr2:CCR5 DKO mice), but not CCR1 (Mdr2:CCR1 DKO), macrophage recruitment and trafficking to the liver was significantly reduced. Furthermore, in the absence of CCR5, reduced inflammation was also associated with reduced periductal accumulation of CD24(+) oval cells and abrogation of fibrosis. DKO mice for Mdr2 and CCR5 exhibited a significant decrease in tumor incidence and size. CONCLUSIONS: Our results indicate that CCR5 has a critical role in both the development and progression of liver cancer. Therefore, we propose that a CCR5 antagonist can serve for HCC cancer prevention and treatment.


Subject(s)
Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/physiopathology , Hepatitis, Chronic/complications , Liver Cirrhosis/complications , Liver Neoplasms/etiology , Liver Neoplasms/physiopathology , Receptors, CCR5/physiology , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/physiology , Animals , Carcinoma, Hepatocellular/epidemiology , Chemokine CCL5/physiology , Disease Models, Animal , Disease Progression , Hepatitis, Chronic/genetics , Incidence , Liver/pathology , Liver/physiopathology , Liver Cirrhosis/genetics , Liver Neoplasms/epidemiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR1/deficiency , Receptors, CCR1/genetics , Receptors, CCR1/physiology , Receptors, CCR5/deficiency , Receptors, CCR5/genetics , ATP-Binding Cassette Sub-Family B Member 4
14.
PLoS One ; 6(9): e24856, 2011.
Article in English | MEDLINE | ID: mdl-21949768

ABSTRACT

OBJECTIVES: Autocrine and paracrine chemokine/chemokine receptor-based interactions promote non-small-cell-lung-cancer (NSCLC) carcinogenesis. CCL20/CCR6 interactions are involved in prostatic and colonic malignancy pathogenesis. The expression and function of CCL20/CCR6 and its related Th-17 type immune response in NSCLC is not yet defined. We sought to characterize the role of the CCL20/CCR6/IL-17 axis in NSCLC tumor growth. METHODS: A specialized histopathologist blindly assessed CCL20/CCR6 expression levels in 49 tissue samples of NSCLC patients operated in our department. Results were correlated to disease progression. Colony assays, ERK signaling and chemokine production were measured to assess cancer cell responsiveness to CCL20 and IL-17 stimulation. RESULTS: CCL20 was highly expressed in the majority (38/49, 77.5%) of tumor samples. Only a minority of samples (8/49, 16.5%) showed high CCR6 expression. High CCR6 expression was associated with a shorter disease-free survival (P = 0.008) and conferred a disease stage-independent 4.87-fold increased risk for disease recurrence (P = 0.0076, CI 95% 1.52-15.563). Cancerous cell colony-forming capacity was increased by CCL20 stimulation; this effect was dependent in part on ERK phosphorylation and signaling. IL-17 expression was detected in NSCLC; IL-17 potentiated the production of CCL20 by cancerous cells. CONCLUSION: Our findings suggest that the CCL20/CCR6 axis promotes NSCLC disease progression. CCR6 is identified as a potential new prognostic marker and the CCL20/CCR6/IL-17 axis as a potential new therapeutic target. Larger scale studies are required to consolidate these observations.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Chemokine CCL20/metabolism , Disease Progression , Interleukin-17/metabolism , Lung Neoplasms/pathology , Receptors, CCR6/metabolism , Aged , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Lung Neoplasms/enzymology , Male , Phosphorylation , Signal Transduction
15.
Cytokine ; 54(3): 249-57, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21376626

ABSTRACT

Chemokines were shown to govern the trafficking of immune cells and may also play important roles in the survival and activation of these cells. We report here that under physiological conditions, the bone marrow (BM), spleen, blood and liver of Ccr5, but not of Ccr1-deficient mice, contain reduced numbers of NK cells. NK cells in the BM of Ccr5-deficient mice proliferate to a lesser extent compared to WT mice. Furthermore, spleen NK cells derived from Ccr5-deficient mice that were transplanted into irradiated recipients failed to proliferate in the host. Ccr5, but not Ccr1-deficient NK cells, failed to migrate in vitro in response to RANTES and MIP-1ß but not MIP-1ß or SDF-1 and had reduced activation, lower expression levels of NK cell markers and a slightly reduced capacity to adhere to target cells and stimulate their killing. Using the polyI:C mouse model for NK trafficking, we found that in the absence of Ccr5, but not Ccr1, NK cells failed to accumulate in the liver. In contrast, using the influenza viral infection as a model to evaluate NK cell proliferation, we found that Ccr5-deficient NK cells in the BM had a higher proliferation rate than WT NK cells. These results suggest a role for Ccr5 in NK cell proliferation and circulation under physiological conditions and a complex role for Ccr5 in determining the fate of NK cells under pathological conditions.


Subject(s)
Gene Expression Regulation , Killer Cells, Natural/metabolism , Receptors, CCR5/deficiency , Receptors, CCR5/physiology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Female , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Liver/metabolism , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Phenotype , Poly I-C/metabolism , Spleen/metabolism , Virus Diseases/metabolism
16.
Exp Hematol ; 39(3): 282-92, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21138752

ABSTRACT

OBJECTIVE: The chemokine receptor CXCR4 and its ligand CXCL12 are involved in the progression and dissemination of a diverse number of solid and hematological malignancies. Binding CXCL12 to CXCR4 activates a variety of intracellular signal transduction pathways that regulate cell chemotaxis, adhesion, survival, proliferation, and apoptosis. MATERIALS AND METHODS: Here, we demonstrate that the CXCR4 antagonist, 4F-benzoyl-TN14003 (BKT140), but not AMD3100, exhibits a CXCR4-dependent preferential cytotoxicity toward malignant cells of hematopoietic origin. BKT140 significantly and preferentially stimulated multiple myeloma apoptotic cell death. BKT140 treatment induced morphological changes, phosphatidylserine externalization, decreased mitochondrial membrane potential, caspase-3 activation, sub-G1 arrest, and DNA double-stranded breaks. RESULTS: In vivo, subcutaneous injections of BKT140 significantly reduced, in a dose-dependent manner, the growth of human acute myeloid leukemia and multiple myeloma xenografts. Tumors from animals treated with BKT140 were smaller in size and weights, had larger necrotic areas and high apoptotic scores. CONCLUSIONS: Taken together, these results suggest a potential therapeutic use for BKT140 in multiple myeloma and leukemia patients.


Subject(s)
Apoptosis/drug effects , Leukemia, Myeloid, Acute/drug therapy , Multiple Myeloma/drug therapy , Peptides/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Animals , Anti-HIV Agents/pharmacology , Antineoplastic Agents , Benzylamines , Chemokine CXCL12/metabolism , Cyclams , HL-60 Cells , Heterocyclic Compounds/pharmacology , Humans , Jurkat Cells , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, SCID , Multiple Myeloma/metabolism , Neoplasm Transplantation , Receptors, CXCR4/metabolism
17.
J Interferon Cytokine Res ; 30(6): 439-49, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20235626

ABSTRACT

Influenza pandemics are imminent and represent a major world health concern. Since vaccinations are expected to be less efficient in the coming years due to newly emerging influenza virus strains, novel antiviral therapies are urgently needed. Here, we show that influenza-infected mice, capable of clearing the virus in the early stages of infection, failed to control inflammation and death. Sequential administration of Interferon-gamma (IFN-gamma) at early stage of the infection protected infected mice from death in a NK cell-dependent manner. IFN-gamma treatment stimulated NK cell proliferation and function and increased their number in the bone marrow, blood, spleen, and infected lungs, keeping viral clearance intact. In parallel, IFN-gamma treatment significantly reduced the number of T cells and NKT cells in the lungs at the inflammatory phase following infection. Thus, rapidly clearing the virus and reducing inflammation by shaping the cellular and cytokine profiles in the early stages of infection may favorably change the fate of influenza pathogenesis.


Subject(s)
Influenza A virus/immunology , Interferon-gamma/administration & dosage , Killer Cells, Natural/drug effects , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Animals , Cell Count , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Hemagglutination Tests , Immunophenotyping , Influenza A virus/pathogenicity , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Lung/pathology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Spleen/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Load/drug effects
18.
Liver Int ; 29(8): 1273-84, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19538448

ABSTRACT

INTRODUCTION: Progressive liver failure may develop following removal of a large part of the liver or transplantation of a small for size liver graft. The pathophysiology of this clinical syndrome is only partially understood. METHODS: We assessed liver damage and hepatocyte 5-bromo-2'-deoxyuridine (BrdU) incorporation following partial hepatectomy (PH) in C57BL/6, BALB/C and immune-deficient mice. Hepatic lymphocyte subpopulations were characterized. Lipopolysaccharide (LPS) treatment and bowel decontamination determined the role of gut antigens. RESULTS: Discrete, round necrotic lesions were observed as early as 2 h following 70%, but not 30% PH. In immune competent mice the extent of hepatocyte necrosis inversely correlated with BrdU incorporation. T, natural killer and natural killer T cells were recruited to the liver early after PH; however, only T-cell depletion abrogated hepatic necrosis. Hepatic injury was significantly reduced in non-obese diabetic/severe combined immunodeficient mice undergoing PH, while BrdU incorporation was not affected. Liver injury was augmented by LPS injection and reduced by gut decontamination. CONCLUSIONS: A distinct pattern of early focal hepatic necrosis is observed following extensive PH in mice. T cells infiltrating the liver immediately after PH and gut-derived antigens are indispensable for the observed liver necrosis and may thus provide therapeutic targets to ameliorate liver damage following PH.


Subject(s)
Gastric Mucosa/immunology , Hepatectomy/adverse effects , Liver Cirrhosis/etiology , Liver Regeneration/physiology , Liver/pathology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Blocking/pharmacology , Antigen Presentation , Antigens, Bacterial/immunology , Bone Marrow Transplantation , Bromodeoxyuridine/metabolism , Cell Proliferation , Disease Models, Animal , Intestines/drug effects , Intestines/immunology , Intestines/microbiology , Lipopolysaccharides/pharmacology , Liver/immunology , Liver/metabolism , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Specific Pathogen-Free Organisms
19.
PLoS One ; 4(4): e5125, 2009.
Article in English | MEDLINE | ID: mdl-19340288

ABSTRACT

The chemokine receptor CXCR4 and its ligand CXCL12 is overexpressed in the majority of tumors and is critically involved in the development and metastasis of these tumors. CXCR4 is expressed in malignant tumor cells whereas its ligand SDF-1 (CXCL12) is expressed mainly by cancer associated fibroblasts (CAF). Similarly to CXCR4, the chemokine CCL20 is overexpressed in variety of tumors; however its role and regulation in tumors is not fully clear. Here, we show that the chemokine receptor CXCR4 stimulates the production of the chemokine CCL20 and that CCL20 stimulates the proliferation and adhesion to collagen of various tumor cells. Furthermore, overexpression of CCL20 in tumor cells promotes growth and adhesion in vitro and increased tumor growth and invasiveness in vivo. Moreover, neutralizing antibodies to CCL20 inhibit the in vivo growth of tumors that either overexpress CXCR4 or CCL20 or naturally express CCL20. These results reveal a role for CCL20 in CXCR4-dependent and -independent tumor growth and suggest a therapeutic potential for CCL20 and CCR6 antagonists in the treatment of CXCR4- and CCL20-dependent malignancies.


Subject(s)
Cell Division/physiology , Chemokine CCL20/physiology , Neoplasms/pathology , Receptors, CXCR4/physiology , Cell Adhesion , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Neoplasm Invasiveness , Neoplasms/blood supply , Neovascularization, Pathologic , Polymerase Chain Reaction
20.
Stem Cells ; 25(9): 2158-66, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17525235

ABSTRACT

An increase in the number of stem cells in blood following mobilization is required to enhance engraftment after high-dose chemotherapy and improve transplantation outcome. Therefore, an approach that improves stem cell mobilization is essential. The interaction between CXCL12 and its receptor, CXCR4, is involved in the retention of stem cells in the bone marrow. Therefore, blocking CXCR4 may result in mobilization of hematopoietic progenitor and stem cells. We have found that the CXCR4 antagonist known as 4F-benzoyl-TN14003 (T-140) can induce mobilization of hematopoietic stem cells and progenitors within a few hours post-treatment in a dose-dependent manner. Furthermore, although T-140 can also increase the number of white blood cells (WBC) in blood, including monocytes, B cells, and T cells, it had no effect on mobilizing natural killer cells. T-140 was found to efficiently synergize with granulocyte colony-stimulating factor (G-CSF) in its ability to mobilize WBC and progenitors, as well as to induce a 660-fold increase in the number of erythroblasts in peripheral blood. Comparison between the CXCR4 antagonists T-140 and AMD3100 showed that T-140 with or without G-CSF was significantly more potent in its ability to mobilize hematopoietic stem cells and progenitors into blood. These results demonstrate that different CXCR4 antagonists may have different therapeutic potentials.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Peptides/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Drug Synergism , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Transplantation , Leukocytes/cytology , Leukocytes/drug effects , Mice , Mice, Inbred C57BL , Peptides/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL