Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
2.
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
3.
Nat Commun ; 8(1): 1954, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29203865

ABSTRACT

Subunit vaccines have been investigated in over 1000 clinical trials of cancer immunotherapy, but have shown limited efficacy. Nanovaccines may improve efficacy but have rarely been clinically translated. By conjugating molecular vaccines with Evans blue (EB) into albumin-binding vaccines (AlbiVax), here we develop clinically promising albumin/AlbiVax nanocomplexes that self-assemble in vivo from AlbiVax and endogenous albumin for efficient vaccine delivery and potent cancer immunotherapy. PET pharmacoimaging, super-resolution microscopies, and flow cytometry reveal almost 100-fold more efficient co-delivery of CpG and antigens (Ags) to lymph nodes (LNs) by albumin/AlbiVax than benchmark incomplete Freund's adjuvant (IFA). Albumin/AlbiVax elicits ~10 times more frequent peripheral antigen-specific CD8+ cytotoxic T lymphocytes with immune memory than IFA-emulsifying vaccines. Albumin/AlbiVax specifically inhibits progression of established primary or metastatic EG7.OVA, B16F10, and MC38 tumors; combination with anti-PD-1 and/or Abraxane further potentiates immunotherapy and eradicates most MC38 tumors. Albumin/AlbiVax nanocomplexes are thus a robust platform for combination cancer immunotherapy.


Subject(s)
Cancer Vaccines/pharmacology , Lymph Nodes/drug effects , Melanoma, Experimental/pathology , T-Lymphocytes, Cytotoxic/drug effects , Adjuvants, Immunologic/pharmacology , Albumin-Bound Paclitaxel/pharmacology , Albumins , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Drug Delivery Systems , Freund's Adjuvant/pharmacology , Immunotherapy , Lipids/pharmacology , Lymph Nodes/pathology , Mice , Molecular Docking Simulation , Nanostructures , Positron-Emission Tomography
4.
Oncotarget ; 8(43): 73387-73406, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-29088715

ABSTRACT

Expression of the chemokine receptor CXCR4 by many cancers correlates with aggressive clinical behavior. As part of the initial studies in a project whose goal was to quantify CXCR4 expression on cancers non-invasively, we examined CXCR4 expression in cancer samples by immunohistochemistry using a validated anti-CXCR4 antibody. Among solid tumors, we found expression of CXCR4 on significant percentages of major types of kidney, lung, and pancreatic adenocarcinomas, and, notably, on metastases of clear cell renal cell carcinoma and squamous cell carcinoma of the lung. We found particularly high expression of CXCR4 on adrenocortical cancer (ACC) metastases. Microarrays of ACC metastases revealed correlations between expression of CXCR4 and other chemokine system genes, particularly CXCR7/ACKR3, which encodes an atypical chemokine receptor that shares a ligand, CXCL12, with CXCR4. A first-in-human study using 64Cu-plerixafor for PET in an ACC patient prior to resection of metastases showed heterogeneity among metastatic nodules and good correlations among PET SUVs, CXCR4 staining, and CXCR4 mRNA. Additionally, we were able to show that CXCR4 expression correlated with the rates of growth of the pulmonary lesions in this patient. Further studies are needed to understand better the role of CXCR4 in ACC and whether targeting it may be beneficial. In this regard, non-invasive methods for assessing CXCR4 expression, such as PET using 64Cu-plerixafor, should be important investigative tools.

5.
Clin Immunol ; 183: 316-324, 2017 10.
Article in English | MEDLINE | ID: mdl-28939272

ABSTRACT

Th-17 type immune response that occurs in multiple sclerosis (MS) is linked to CCR6-CCL20 interaction. We confirmed the dependency on CCR6 in EAE development. Vaccination of mice with hCCL20, but not mCCL20, produced anti-murine CCL20 and ameliorated EAE. The EAE clinical score negatively correlated with anti CCL20 levels. A beneficial effect was transferred by sera from hCCL20-immunized mice. Immunized mice with cyclic peptide that include a bacterial outer membrane protein A (ompA), that share homology sequence with hCCL20 produced anti CCL20, anti ompA and anti-cyclic peptide. Immunization of mice with ompA or the cyclic peptide ameliorated EAE. The cyclic peptide inhibited CCL20 activity in an adhesion assay. A significantly higher level of anti CCL20 were found in healthy individuals compared to RR-MS patients. There was no similar difference for anti-CXCL10. Natural or induced immunization against CCL20 confer protection against EAE and may be beneficial in MS.


Subject(s)
Chemokine CCL20/immunology , Encephalitis/prevention & control , Hashimoto Disease/prevention & control , Multiple Sclerosis/prevention & control , Animals , Cell Adhesion , Chemokine CCL20/administration & dosage , Computational Biology , Female , Immunization , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, CCR6/genetics , Receptors, CCR6/metabolism , T-Lymphocytes
6.
J Nucl Med ; 58(4): 590-597, 2017 04.
Article in English | MEDLINE | ID: mdl-27879373

ABSTRACT

One of the major design considerations for a drug is its pharmacokinetics in the blood. A drug with a short half-life in the blood is less available at a target organ. Such a limitation dictates treatment with either high doses or more frequent doses, both of which may increase the likelihood of undesirable side effects. To address the need for additional methods to improve the blood half-life of drugs and molecular imaging agents, we developed an "add-on" molecule that contains 3 groups: a truncated Evans blue dye molecule that binds to albumin with a low micromolar affinity and provides a prolonged half-life in the blood; a metal chelate that allows radiolabeling for imaging and radiotherapy; and maleimide for easy conjugation to drug molecules. Methods: The truncated Evans blue molecule was conjugated with the chelator NOTA or DOTA, and the resulting conjugate was denoted as NMEB or DMEB, respectively. As a proof of concept, we coupled NMEB and DMEB to c(RGDfK), which is a small cyclic arginine-glycine-aspartic acid (RGD) peptide, for targeting integrin αvß3 NMEB and DMEB were radiolabeled with 64Cu and 90Y, respectively, and tested in xenograft models. Results: The resulting radiolabeled conjugates showed a prolonged circulation half-life and enhanced tumor accumulation in integrin αvß3-expressing tumors. Tumor uptake was markedly improved over that with NOTA- or DOTA-conjugated c(RGDfK). Tumor radiotherapy experiments in mice with 90Y-DMEB-RGD showed promising results; existing tumors were eliminated. Conclusion: Conjugation of our novel add-on molecule, NMEB or DMEB, to potential tracers or therapeutic agents improved blood half-life and tumor uptake and could transform such agents into theranostic entities.


Subject(s)
Evans Blue/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Animals , Cell Line, Tumor , Copper Radioisotopes , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds/chemistry , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Integrin alphaVbeta3/metabolism , Mice , Oligopeptides/chemistry , Positron-Emission Tomography , Radiochemistry , Radiopharmaceuticals/chemistry , Tissue Distribution , Yttrium Radioisotopes/therapeutic use
7.
Angew Chem Int Ed Engl ; 54(43): 12777-81, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26308650

ABSTRACT

Azido (18) F-arenes are important and versatile building blocks for the radiolabeling of biomolecules via Huisgen cycloaddition ("click chemistry") for positron emission tomography (PET). However, routine access to such clickable agents is challenged by inefficient and/or poorly defined multistep radiochemical approaches. A high-yielding direct radiofluorination for azido (18) F-arenes was achieved through the development of an ortho-oxygen-stabilized iodonium derivative (OID). This OID strategy addresses an unmet need for a reliable azido (18) F-arene clickable agent for bioconjugation reactions. A ssDNA aptamer was radiolabeled with this agent and visualized in a xenograft mouse model of human colon cancer by PET, which demonstrates that this OID approach is a convenient and highly efficient way of labeling and tracking biomolecules.


Subject(s)
Aptamers, Nucleotide/chemistry , Cell Adhesion Molecules/analysis , Colon/pathology , Colonic Neoplasms/diagnosis , DNA, Single-Stranded/chemistry , Positron-Emission Tomography/methods , Receptor Protein-Tyrosine Kinases/analysis , Animals , Azides/chemistry , Click Chemistry , Female , Fluorine Radioisotopes/chemistry , HCT116 Cells , Humans , Mice , Radiopharmaceuticals/chemistry
8.
J Nucl Med ; 56(11): 1780-1785, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26315836

ABSTRACT

UNLABELLED: Protein tyrosine kinase-7 (PTK7), a member of receptor tyrosine kinase superfamily initially identified as colon carcinoma kinase-4, is highly expressed in various human malignancies. Its expression was found to correlate with aggressive biologic behaviors such as increased cell proliferation, invasiveness, and migration. Despite the importance and unmet need of imaging PTK7 in vivo, there is currently no clinically relevant method to visualize tumoral PTK7 expression noninvasively such as PET or SPECT. This study aimed to develop a specific, selective, and high-affinity PET radioligand based on single-stranded DNA aptamer to address this challenge. METHODS: Sgc8, a 41-oligonucleotide that targets to PTK7, was labeled with (18)F using a 2-step radiochemical synthesis, which featured a direct 1-step radiofluorination on the distinctive spirocyclic hypervalent iodine(III) precursor to give (18)F-fluorobenzyl azide followed by copper-mediated click conjugation with Sgc8-alkyne. (18)F-Sgc8 was evaluated in vitro and in vivo in 2 cell lines, HCT116 and U87MG, which express high and low amounts of PTK7, respectively. RESULTS: Sgc8 was labeled efficiently with (18)F in an isolated radiochemical yield of 62% ± 2%, non-decay-corrected based on (18)F-fluorobenzyl azide. (18)F-Tr-Sgc8 was found to possess high-affinity binding to both cell lines, with binding affinity values of 2.7 ± 0.6 nM for HCT116 and 16.9 ± 2.1 nM for U87MG. In vivo PET imaging clearly visualized PTK7 expression in HCT116 xenografted mice, with tumor uptake of 0.76 ± 0.09 percentage injected dose per gram (%ID/g) at 30 min after injection for the subcutaneous tumor model and greater than 1.5 %ID/g for the liver metastasis model. U87MG xenograft tumors had much lower tracer accumulation (0.13 ± 0.06 %ID/g at 30 min after injection), which was consistent with the lower expression of PTK7 in this tumor model. The labeled aptamer was rapidly cleared from the blood through the kidneys and bladder to give high tumor-to-blood and tumor-to-muscle ratios of 7.29 ± 1.51 and 10.25 ± 2.08, respectively. CONCLUSION: The (18)F-radiolabeling methodology shown here is a robust procedure for labeling aptamers and similar chemical moieties and can be applied to many different targets. Quantification of PTK7 using (18)F-Tr-Sgc8 may be suitable for clinical translation and might help in the future to select and monitor appropriate therapies.


Subject(s)
Aptamers, Nucleotide/chemistry , Cell Adhesion Molecules/biosynthesis , DNA, Single-Stranded/genetics , Radiopharmaceuticals/chemistry , Receptor Protein-Tyrosine Kinases/biosynthesis , Animals , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Fluorine Radioisotopes , Gene Expression Regulation, Enzymologic/genetics , Humans , Isotope Labeling , Mice , Neoplasms, Experimental/diagnostic imaging , Positron-Emission Tomography , Receptor Protein-Tyrosine Kinases/genetics , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Xenograft Model Antitumor Assays
9.
Transpl Immunol ; 33(2): 125-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26209354

ABSTRACT

Hematopoietic chimerism established by allogeneic bone marrow transplantation is known to promote donor-specific organ allograft tolerance; however, clinical application is limited by the need for toxic host conditioning and "megadoses" of donor bone marrow cells. A potential solution to this problem has been suggested by the observation that recipient bone marrow mobilization by the CXCR4 antagonist AMD3100 promotes chimerism in congenic bone marrow transplantation experiments in mice. Here we report that a single subcutaneous dose of 10 mg/kg AMD3100 in recipient C57BL/6 mice was able to enhance hematopoietic chimerism when complete MHC-mismatched BALB/c donor bone marrow cells were transplanted 1h after drug dosing. However, levels of chimerism measured 30 days post-transplantation were not sustained when mice were reexamined on day 90 post-transplantation. Moreover, transient chimerism induced by this protocol did not support robust donor-specific skin allograft tolerance. Using the same transient immunosuppression protocol, we confirmed that "megadoses" of donor bone marrow cells could induce durable chimerism associated with donor-specific skin allograft tolerance without AMD3100 pre-treatment. We conclude that in this protocol AMD3100 pretreatment may empty bone marrow niches that become reoccupied by allogeneic donor hematopoietic progenitor cells but not by true long-lived donor hematopoietic stem cells, resulting in short-lived chimerism and failure to support durable donor-specific allograft tolerance.


Subject(s)
Bone Marrow Transplantation , Hematopoietic Stem Cells/drug effects , Heterocyclic Compounds/administration & dosage , Receptors, CXCR4/antagonists & inhibitors , Skin Transplantation , Animals , Benzylamines , Chimerism , Cyclams , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/physiology , Heterocyclic Compounds/adverse effects , Histocompatibility Antigens/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Stem Cell Niche/drug effects , Transplantation Conditioning , Transplantation Tolerance/drug effects
10.
J Nucl Med ; 56(4): 616-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25698784

ABSTRACT

UNLABELLED: Tenascin-C is an extracellular matrix glycoprotein that is expressed by injured tissues and by various cancers. Recent publications showed that tenascin-C expression by cancer lesions predicts tumor growth, metastasis, and angiogenesis, suggesting tenascin-C as a potential therapeutic target. Currently there is no noninvasive method to determine tumoral tenascin-C expression in vivo. To address the need for an agent to image and quantify tenascin-C, we report the development of a radioactive PET tracer based on a tenascin-C-specific single-stranded DNA aptamer (tenascin-C aptamer). METHODS: Tenascin-C aptamer was radiolabeled with (18)F and (64)Cu. PET imaging studies for the evaluation of tumor uptake and pharmacokinetics of tenascin-C aptamer were performed in comparison to a nonspecific scrambled aptamer (Sc aptamer). RESULTS: The labeled tenascin-C aptamer provided clear visualization of tenascin-C-positive but not tenascin-C-negative tumors. The uptake of tenascin-C aptamer was significantly higher than that of Sc aptamer in tenascin-C-positive tumors. The labeled tenascin-C aptamer had fast clearance from the blood and other nonspecific organs through the kidneys, resulting in high tumor contrast. CONCLUSION: Our data suggest that suitably labeled tenascin-C aptamer can be used as a PET tracer to image tumor expression of tenascin-C with a high tumor-to-background ratio and might provide insightful and personalized medical data that will help determine appropriate treatment and monitoring.


Subject(s)
Aptamers, Nucleotide/chemistry , Copper Radioisotopes/chemistry , Fluorine Radioisotopes/chemistry , Positron-Emission Tomography/methods , Tenascin/chemistry , Animals , Cell Line, Tumor , DNA, Single-Stranded/chemistry , Diagnostic Imaging , Female , Humans , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasm Transplantation , Nucleic Acid Conformation , Nucleotides/chemistry , Radioactive Tracers , Radiopharmaceuticals
11.
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
12.
J Immunol ; 192(4): 1778-86, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442439

ABSTRACT

It was reported that host defense against pulmonary Klebsiella pneumoniae infection requires IL-22, which was proposed to be of T cell origin. Supporting a role for IL-22, we found that Il22(-/-) mice had decreased survival compared with wild-type mice after intratracheal infection with K. pneumoniae. Surprisingly, however, Rag2(-/-) mice did not differ from wild-type mice in survival or levels of IL-22 in the lungs postinfection with K. pneumoniae. In contrast, K. pneumoniae-infected Rag2(-/-)Il2rg(-/-) mice failed to produce IL-22. These data suggested a possible role for NK cells or other innate lymphoid cells in host defense and production of IL-22. Unlike NK cell-like innate lymphoid cells that produce IL-22 and display a surface phenotype of NK1.1(-)NKp46(+)CCR6(+), lung NK cells showed the conventional phenotype, NK1.1(+)NKp46(+)CCR6(-). Mice depleted of NK cells using anti-asialo GM1 showed decreased survival and higher lung bacterial counts, as well as increased dissemination of K. pneumoniae to blood and liver, compared with control-treated mice. NK cell depletion also led to decreased production of IL-22 in the lung. Within 1 d postinfection, although there was no increase in the number of lung NK cells, a subset of lung NK cells became competent to produce IL-22, and such cells were found in both wild-type and Rag2(-/-) mice. Our data suggest that, during pulmonary infection of mice with K. pneumoniae, conventional NK cells are required for optimal host defense, which includes the production of IL-22.


Subject(s)
Interleukins/metabolism , Killer Cells, Natural/immunology , Klebsiella Infections/immunology , Pneumonia/immunology , Animals , Antigens, Ly/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukins/biosynthesis , Interleukins/genetics , Klebsiella Infections/mortality , Klebsiella pneumoniae/immunology , Lung/cytology , Lung/immunology , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Pneumonia/mortality , Receptors, CCR6/metabolism , Survival , Interleukin-22
13.
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
14.
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
16.
Theranostics ; 3(1): 76-84, 2013.
Article in English | MEDLINE | ID: mdl-23382787

ABSTRACT

CXCR4 was found to be expressed by many different types of human cancers and its expression has been correlated with tumor aggressiveness, poor prognosis and resistance to chemotherapy. CXCR4 was also shown to contribute to metastatic seeding of organs that express its ligand CXCL12 and support the survival of these cells. These findings suggest that CXCR4 is a potentially attractive therapeutic target, and several antagonists and antibodies for this receptor were developed and are under clinical evaluation. Quantifying CXCR4 expression non-invasively might aid in prognostication as a mean for personalized therapy and post treatment monitoring. Multiple attempts were done over the recent years to develop imaging agents for CXCR4 using different technologies including PET, SPECT, fluorescent and bioluminescence, and will be reviewed in this paper.


Subject(s)
Molecular Imaging/methods , Receptors, CXCR4/metabolism , Animals , Fluorescence , Humans , Luminescent Measurements , Positron-Emission Tomography , Tomography, Emission-Computed, Single-Photon
17.
Hepatology ; 58(1): 192-204, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23423643

ABSTRACT

UNLABELLED: Chronic inflammation is strongly associated with an increased risk for hepatocellular carcinoma (HCC) development. The multidrug resistance 2 (Mdr2)-knockout (KO) mouse (adenosine triphosphate-binding cassette b4(-/-) ), a model of inflammation-mediated HCC, develops chronic cholestatic hepatitis at an early age and HCC at an adult age. To delineate factors contributing to hepatocarcinogenesis, we compared the severity of early chronic hepatitis and late HCC development in two Mdr2-KO strains: Friend virus B-type/N (FVB) and C57 black 6 (B6). We demonstrated that hepatocarcinogenesis was significantly less efficient in the Mdr2-KO/B6 mice versus the Mdr2-KO/FVB mice; this difference was more prominent in males. Chronic hepatitis in the Mdr2-KO/B6 males was more severe at 1 month of age but was less severe at 3 months of age in comparison with age-matched Mdr2-KO/FVB males. A comparative genome-scale gene expression analysis of male livers of both strains at 3 months of age revealed both common and strain-specific aberrantly expressed genes, including genes associated with the regulation of inflammation, the response to oxidative stress, and lipid metabolism. One of these regulators, galectin-1 (Gal-1), possesses both anti-inflammatory and protumorigenic activities. To study its regulatory role in the liver, we transferred the Gal-1-KO mutation (lectin galactoside-binding soluble 1(-/-) ) from the B6 strain to the FVB strain, and we demonstrated that endogenous Gal-1 protected the liver against concanavalin A-induced hepatitis with the B6 genetic background but not the FVB genetic background. CONCLUSION: Decreased chronic hepatitis in Mdr2-KO/B6 mice at the age of 3 months correlated with a significant retardation of liver tumor development in this strain versus the Mdr2-KO/FVB strain. We found candidate factors that may determine strain-specific differences in the course of chronic hepatitis and HCC development in the Mdr2-KO model, including inefficient anti-inflammatory activity of the endogenous lectin Gal-1 in the FVB strain.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Galectin 1/physiology , Hepatitis, Chronic/pathology , Liver Neoplasms/pathology , Mice, Inbred Strains/genetics , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Carcinoma, Hepatocellular/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Concanavalin A , Hepatitis, Chronic/complications , Hepatitis, Chronic/etiology , Liver/metabolism , Liver Neoplasms/etiology , Male , Methionine Adenosyltransferase/biosynthesis , Mice , Mice, Knockout , ATP-Binding Cassette Sub-Family B Member 4
18.
J Invest Dermatol ; 133(1): 164-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22895364

ABSTRACT

A subset of CC chemokine receptor-6(+) (CCR6(+)), γδ-low (GDL) T cells that express Th17 cytokines in mouse skin participates in IL-23-induced psoriasiform dermatitis. We use CCR6-deficient (knockout, KO) and wild-type (WT) mice to analyze skin trafficking patterns of GDL T cells and function-blocking mAbs to determine the role of CCR6 in IL-23-mediated dermatitis. Herein, CCL20 was highly upregulated in IL-23-injected WT mouse ear skin as early as 24 hours after initial treatment, and large numbers of CCR6(+) cells were observed in the epidermis of IL-23-injected WT mice. Anti-CCL20 mAbs reduced psoriasiform dermatitis and blocked recruitment of GDL T cells to the epidermis. In CCR6 KO mice, GDL T cells failed to accumulate in the epidermis after IL-23 treatment, but the total numbers of GDL T cells in the dermis of WT and CCR6 KO mice were equivalent. There was an ∼70% reduction in the proportion of IL-22(+) GDL T cells in the dermis of CCR6 KO mice (vs WT mice), suggesting that effector function and epidermal recruitment of GDL T cells are impaired in CCR6-deficient mice. Thus, these data show that CCR6 regulates epidermal trafficking of γδ-T-cell subsets in the skin and suggest the potential of CCR6 as a therapeutic target for psoriasis.


Subject(s)
Dermatitis/immunology , Epidermis/immunology , Interleukin-23/adverse effects , Psoriasis/immunology , Receptors, CCR6/immunology , T-Lymphocyte Subsets/immunology , Animals , Antibodies, Blocking/pharmacology , Cell Movement/drug effects , Cell Movement/immunology , Chemokine CCL20/immunology , Dermatitis/pathology , Epidermis/drug effects , Epidermis/pathology , Interleukins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Psoriasis/chemically induced , Psoriasis/pathology , Receptors, Antigen, T-Cell, gamma-delta/drug effects , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, CCR6/genetics , T-Lymphocyte Subsets/drug effects , Interleukin-22
19.
J Thorac Cardiovasc Surg ; 144(5): 1167-1175.e1, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22925564

ABSTRACT

OBJECTIVES: CXCR4/CXCL12 interactions promote non-small cell lung cancer (NSCLC) growth and dissemination. Furthermore, this axis might promote NSCLC resistance to chemotherapy and/or radiotherapy. Therefore, the CXCR4/CXCL12 axis constitutes an attractive therapeutic target for the treatment of NSCLC. We aimed to characterize the therapeutic efficacy of the novel CXCR4 antagonist BKT140 against human NSCLC. METHODS: We determined the CXCR4 expression in 5 NSCLC cell lines (H358, A549, H460, H1299, and L4). We then tested the colony-forming capacity and proliferation of these cells in the presence of CXCL12 and BKT140. Next, we measured the in vivo growth of A549 and H460 xenografts with or without BKT140 treatment. Finally, we examined, in vitro, the potential antiproliferative effect of BKT140 combined with cisplatin or paclitaxel and after irradiation of NSCLC cells. RESULTS: All tested cell lines expressed CXCR4 and showed increased colony formation in response to CXCL12 stimulation. BKT140 reduced the colony-forming capacity of NSCLC cells. Proliferation assays demonstrated both cytotoxic and cytostatic properties for this peptide. H460 cells were the most sensitive to BKT140 and A549 cells the least. Subcutaneous administration of BKT140 significantly delayed the development of H460 xenografts and showed a similar trend for A549 xenografts. Finally, the antiproliferative effects of BKT140 appears to be additive to those of chemotherapeutic drugs and radiotherapy. CONCLUSIONS: Targeting the CXCL12/CXCR4 axis with BKT140 attenuated NSCLC cells tumor growth and augmented the effects of chemotherapy and radiotherapy. Future research will benefit from delineating the downstream mechanism of BKT140 action and defining BKT140 susceptibility markers.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Oligopeptides/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL2/metabolism , Chemoradiotherapy , Cisplatin/administration & dosage , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Mice , Mice, Nude , Oligopeptides/administration & dosage , Paclitaxel/administration & dosage , Receptors, CXCR4/metabolism , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
Mol Imaging Biol ; 14(1): 106-14, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21347799

ABSTRACT

PURPOSE: Expression of CXCR4 in cancers has been correlated with poor prognosis and increased metastasis. Quantifying CXCR4 expression non-invasively might aid in prognostication and monitoring therapy. We evaluated a radiolabeled antagonist of CXCR4, 64Cu-AMD3100, as a positron-emitting imaging agent. PROCEDURES: CXCR4-transfected or non-transfected cell lines were injected into mice to form xenografts. Accumulation of 64Cu-AMD3100 in tumors was analyzed by small-animal PET and biodistribution assays. RESULTS: 64Cu-AMD3100 accumulated in CXCR4-expressing, but not CXCR4-negative, tumors. For CXCR4-expressing tumors, tumor-to-blood and tumor-to-muscle ratios were 23-41 and 50-59, respectively, depending on tumor type. Excess of unlabeled Cu-AMD3100 or AMD3100 significantly reduced 64Cu-AMD3100 accumulation in CXCR4-expressing tumors. Human-absorbed dose calculations predicted a dose limit of 444 MBq. CONCLUSIONS: CXCR4 can be imaged in tumors using 64Cu-AMD3100. Dosimetry studies suggest that imaging in humans is feasible. We conclude that 64Cu-AMD3100 should be investigated as a potential agent for imaging and quantifying CXCR4 in tumors.


Subject(s)
Copper Radioisotopes , Heterocyclic Compounds , Neoplasms, Experimental/diagnostic imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals , Receptors, CXCR4/biosynthesis , Animals , Benzylamines , CHO Cells , Carcinoma, Lewis Lung/diagnostic imaging , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor , Copper Radioisotopes/chemistry , Copper Radioisotopes/pharmacokinetics , Cricetinae , Cricetulus , Cyclams , Female , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacokinetics , Humans , Liver Neoplasms, Experimental/diagnostic imaging , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Imaging/methods , Neoplasms, Experimental/metabolism , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/metabolism , Radiation Dosage , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Receptors, CXCR4/genetics , Tissue Distribution , Transfection , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL