Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Exp Med ; 216(9): 2150-2169, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31239386

ABSTRACT

We have identified a precursor that differentiates into granulocytes in vitro and in vivo yet belongs to the monocytic lineage. We have termed these cells monocyte-like precursors of granulocytes (MLPGs). Under steady state conditions, MLPGs were absent in the spleen and barely detectable in the bone marrow (BM). In contrast, these cells significantly expanded in tumor-bearing mice and differentiated to polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Selective depletion of monocytic cells had no effect on the number of granulocytes in naive mice but decreased the population of PMN-MDSCs in tumor-bearing mice by 50%. The expansion of MLPGs was found to be controlled by the down-regulation of Rb1, but not IRF8, which is known to regulate the expansion of PMN-MDSCs from classic granulocyte precursors. In cancer patients, putative MLPGs were found within the population of CXCR1+CD15-CD14+HLA-DR-/lo monocytic cells. These findings describe a mechanism of abnormal myelopoiesis in cancer and suggest potential new approaches for selective targeting of MDSCs.


Subject(s)
Monocytes/pathology , Myeloid-Derived Suppressor Cells/pathology , Neoplasms/pathology , Neutrophils/pathology , Adult , Aged , Animals , Cell Differentiation , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Retinoblastoma Binding Proteins/metabolism
2.
Clin Cancer Res ; 25(9): 2783-2794, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30765391

ABSTRACT

PURPOSE: BRAF and MEK inhibitors (BRAFi and MEKi) are actively used for the treatment of metastatic melanoma in patients with BRAFV600E mutation in their tumors. However, the development of resistance to BRAFi and MEKi remains a difficult clinical challenge with limited therapeutic options available to these patients. In this study, we investigated the mechanism and potential therapeutic utility of combination BRAFi and adoptive T-cell therapy (ACT) in melanoma resistant to BRAFi. EXPERIMENTAL DESIGN: Investigations were performed in vitro and in vivo with various human melanoma cell lines sensitive and resistant to BRAFi as well as patient-derived xenografts (PDX) derived from patients. In addition, samples were evaluated from patients on a clinical trial of BRAFi in combination with ACT. RESULTS: Herein we report that in human melanoma cell lines, senstitive and resistant to BRAFi and in PDX from patients who progressed on BRAFi and MEKi therapy, BRAFi caused transient upregulation of mannose-6-phosphate receptor (M6PR). This sensitized tumor cells to CTLs via uptake of granzyme B, a main component of the cytotoxic activity of CTLs. Treatment of mice bearing resistant tumors with BRAFi enhanced the antitumor effect of patients' TILs. A pilot clinical trial of 16 patients with metastatic melanoma who were treated with the BRAFi vemurafenib followed by therapy with TILs demonstrated a significant increase of M6PR expression on tumors during vemurafenib treatment. CONCLUSIONS: BRAF-targeted therapy sensitized resistant melanoma cells to CTLs, which opens new therapeutic opportunities for the treatment of patients with BRAF-resistant disease.See related commentary by Goff and Rosenberg, p. 2682.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Protein Kinase Inhibitors , T-Lymphocytes
3.
Cancer Cell ; 32(5): 654-668.e5, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29136508

ABSTRACT

Tumor-associated macrophages (TAM) contribute to all aspects of tumor progression. Use of CSF1R inhibitors to target TAM is therapeutically appealing, but has had very limited anti-tumor effects. Here, we have identified the mechanism that limited the effect of CSF1R targeted therapy. We demonstrated that carcinoma-associated fibroblasts (CAF) are major sources of chemokines that recruit granulocytes to tumors. CSF1 produced by tumor cells caused HDAC2-mediated downregulation of granulocyte-specific chemokine expression in CAF, which limited migration of these cells to tumors. Treatment with CSF1R inhibitors disrupted this crosstalk and triggered a profound increase in granulocyte recruitment to tumors. Combining CSF1R inhibitor with a CXCR2 antagonist blocked granulocyte infiltration of tumors and showed strong anti-tumor effects.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Monocytes/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms, Experimental/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Cancer-Associated Fibroblasts/drug effects , Cell Line, Tumor , Granulocytes/metabolism , Histone Deacetylase 2 , Humans , Imidazoles/pharmacology , Macrophages/metabolism , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/metabolism , Tumor Burden/drug effects
4.
Sarcoma ; 2015: 614736, 2015.
Article in English | MEDLINE | ID: mdl-26880867

ABSTRACT

Purpose. Patients with large >5 cm, high-grade resectable soft tissue sarcomas (STS) have the highest risk of distant metastases. Previously we have shown that dendritic cell (DC) based vaccines show consistent immune responses. Methods. This was a Phase I single institution study of neoadjuvant radiation with DC injections on 18 newly diagnosed high-risk STS patients. Neoadjuvant treatment consisted of 50 Gy of external beam radiation (EBRT), given in 25 fractions delivered five days/week, combined with four intratumoral injections of DCs followed by complete resection. The primary endpoint was to establish the immunological response to neoadjuvant therapy and obtain data on its clinical safety and outcomes. Results. There were no unexpected toxicities or serious adverse events. Twelve out of 18 (67%) patients were alive, of which an encouraging 11/18 (61%) were alive with no systemic recurrence over a period of 2-8 years. Favorable immunological responses correlated with clinical responses in some cases. Conclusions. This study provides clinical support to using dendritic cell injections along with radiation in sarcomas, which when used optimally in combination can help clinical outcomes in soft tissue sarcoma. Study registration number is NCT00365872.

5.
Cancer Immunol Immunother ; 63(10): 1009-21, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24943275

ABSTRACT

There is a significant body of evidence demonstrating that radiation therapy (XRT) enhances the effect of immune therapy. However, the precise mechanisms by which XRT potentiates the immunotherapy of cancer remain elusive. Here, we report that XRT potentiates the effect of immune therapy via induction of autophagy and resultant trafficking of mannose-6-phopsphate receptor (MPR) to the cell surface. Irradiation of different tumor cells caused substantial up-regulation of MPR on the cell surface in vitro and in vivo. Down-regulation of MPR in tumor cells with shRNA completely abrogated the combined effect of XRT and immunotherapy (CTLA4 antibody) in B16F10-bearing mice without changes in the tumor-specific responses of T cells. Radiation-induced MPR up-regulation was the result of redistribution of the receptor to the cell surface. This effect was caused by autophagy with redirection of MPR to autophagosomes in a clathrin-dependent manner. In autophagosomes, MPR lost its natural ligands, which resulted in subsequent trafficking of empty receptor(s) back to the surface. Together, our data demonstrated a novel mechanism by which XRT can enhance the effect of immunotherapy and the molecular mechanism of this process.


Subject(s)
Immunotherapy/methods , Neoplasms, Experimental/therapy , Receptor, IGF Type 2/metabolism , Animals , Autophagy/immunology , Autophagy/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/radiotherapy , Random Allocation , Up-Regulation
6.
PLoS One ; 8(1): e54506, 2013.
Article in English | MEDLINE | ID: mdl-23349911

ABSTRACT

MicroRNAs (miRNAs) are important and ubiquitous regulators of gene expression that can suppress their target genes by translational inhibition as well as mRNA destruction. Cell type-specific miRNA expression patterns have been successfully exploited for targeting the expression of experimental and therapeutic gene constructs, for example to reduce pathogenic effects of cancer virotherapy in normal tissues. In order to avoid liver damage associated with systemic or intrahepatic delivery of oncolytic adenoviruses we have introduced the concept of suppressing adenovirus replication in hepatic cells by inserting target elements for the liver-specific miR122 into the viral genome. Here we show using ex vivo cultured tissue specimens that six perfectly complementary miR122 target sites in the 3' untranslated region of the viral E1A gene are sufficient in the absence of any other genetic modifications to prevent productive replication of serotype 5 adenovirus (Ad5) in normal human liver. This modification did not compromise the replicative capacity of the modified virus in cancer tissue derived from a colon carcinoma liver metastasis or its oncolytic potency in a human lung cancer xenograft mouse model. Unlike wild-type Ad5, the modified virus did not result in increased serum levels of liver enzymes in infected mice. These results provide a strong preclinical proof of concept for the use of miR122 target sites for reducing the risk of liver damage caused by oncolytic adenoviruses, and suggest that ectopic miR122 target elements should be considered as an additional safety measure included in any therapeutic virus or viral vector posing potential hazard to the liver.


Subject(s)
Adenoviridae/genetics , Colonic Neoplasms/genetics , Genetic Therapy , MicroRNAs/genetics , Oncolytic Viruses/genetics , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Colonic Neoplasms/drug therapy , Colonic Neoplasms/virology , Gene Expression Regulation , Genome, Viral , Humans , Liver/metabolism , Liver/virology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/virology , Mice , MicroRNAs/metabolism , Neoplasm Metastasis , Organ Specificity , Transplantation, Heterologous , Virus Replication/genetics
7.
PLoS One ; 6(11): e26810, 2011.
Article in English | MEDLINE | ID: mdl-22073198

ABSTRACT

Development of new cancer treatments focuses increasingly on the relation of cancer tissue with its microenvironment. A major obstacle for the development of new anti-cancer therapies has been the lack of relevant animal models that would reproduce all the events involved in disease progression from the early-stage primary tumor until the development of mature metastatic tissue. To this end, we have developed a readily imageable mouse model of colorectal cancer featuring highly reproducible formation of spontaneous liver metastases derived from intrasplenic primary tumors. We optimized several experimental variables, and found that the correct choice of cell line and the genetic background, as well as the age of the recipient mice, were critical for establishing a useful model system. Among a panel of colorectal cancer cell lines tested, the epithelial carcinoma HT29 line was found to be the most suitable in terms of producing homogeneous tumor growth and metastases. In our hands, SCID mice at the age of 125 days or older were the most suitable in supporting consistent HT29 tumor growth after splenic implantation followed by reproducible metastasis to the liver. A magnetic resonance imaging (MRI) protocol was optimized for use with this mouse model, and demonstrated to be a powerful method for analyzing the antitumor effects of an experimental therapy. Specifically, we used this system to with success to verify by MRI monitoring the efficacy of an intrasplenically administered oncolytic adenovirus therapy in reducing visceral tumor load and development of liver metastases. In summary, we have developed a highly optimized mouse model for liver metastasis of colorectal cancer, which allows detection of the tumor load at the whole body level and enables an accurate timing of therapeutic interventions to target different stages of cancer progression and metastatic development.


Subject(s)
Disease Models, Animal , Neoplasm Metastasis/pathology , Neoplasms, Experimental/therapy , Animals , Cell Line, Tumor , Humans , Magnetic Resonance Imaging , Mice , Mice, SCID
8.
J Transl Med ; 8: 80, 2010 Aug 23.
Article in English | MEDLINE | ID: mdl-20727221

ABSTRACT

BACKGROUND: Colorectal cancer is often a deadly disease and cannot be cured at metastatic stage. Oncolytic adenoviruses have been considered as a new therapeutic option for treatment of refractory disseminated cancers, including colorectal cancer. The safety data has been excellent but tumor transduction and antitumor efficacy especially in systemic administration needs to be improved. METHODS: Here, the utility of αvß integrin targeting moiety Arg-Gly-Asp (RGD) in the Lys-Lys-Thr-Lys (KKTK) domain of the fiber shaft or in the HI-loop of adenovirus serotype 5 for increased tumor targeting and antitumor efficacy was evaluated. To this end, novel spleen-to-liver metastatic colorectal cancer mouse model was used and the antitumor efficacy was evaluated with magnetic resonance imaging (MRI). RESULTS: Both modifications (RGD in the HI-loop or in the fiber shaft) increased gene transfer efficacy in colorectal cancer cell lines and improved tumor-to-normal ratio in systemic administration of the vector. CONCLUSIONS: Antitumor potency was not compromised with RGD modified viruses suggesting increased safety profile and tumor specificity.


Subject(s)
Adenoviridae/chemistry , Adenoviridae/genetics , Colorectal Neoplasms/therapy , Magnetic Resonance Imaging , Oligopeptides/metabolism , Receptors, Vitronectin/metabolism , Adenoviridae/physiology , Animals , Capsid/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cytotoxicity, Immunologic , Gene Transfer Techniques , Genetic Vectors/pharmacokinetics , Humans , Liver Neoplasms/secondary , Mice , Neoplasm Metastasis , Organ Specificity , Splenic Neoplasms/secondary , Tissue Distribution , Virus Replication , Xenograft Model Antitumor Assays
9.
Hum Gene Ther ; 20(10): 1214-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19537946

ABSTRACT

Tumor targeting on systemic adenovirus administration is key to improve the prospects of adenovirus-mediated gene therapy and virus therapy of cancer. Despite many genetic and ligand conjugation approaches this objective remains elusive. Ablation of human adenovirus type 5 (Ad5) binding to its natural receptors in airway epithelial cells, that is, the coxsackievirus and adenovirus receptor (CAR) and integrins, does not impact on the preeminent liver tropism of adenovirus in the bloodstream. This is explained by a distinct entry pathway mediated by blood factors and heparan sulfates. Mutation of the KKTK heparin sulfate-binding domain of the fiber shaft to GATK results in liver transduction detargeting, but it is not compatible with otherwise useful HI-loop tumor-targeting ligand insertions such as the insertion of Arg-Gly-Asp (RGD). To circumvent this problem we have mutated the KKTK domain to RGDK, and analyzed the liver-detargeting and tumor-targeting transduction properties of this replacement mutant. Similar to RGD at the HI-loop, RGD at this new shaft location efficiently enhances the infectivity of adenovirus and improves the tumor-to-liver transduction ratio in vivo.


Subject(s)
Adenoviridae/genetics , Heparan Sulfate Proteoglycans/chemistry , Neoplasms/therapy , Neoplasms/virology , Oligopeptides/chemistry , Animals , Cell Line, Tumor , Genetic Vectors/genetics , Humans , Liver/enzymology , Liver/virology , Luciferases/metabolism , Mice , Protein Structure, Tertiary , Recombination, Genetic/genetics , Tissue Distribution , Transduction, Genetic
10.
PLoS One ; 3(8): e2917, 2008 Aug 13.
Article in English | MEDLINE | ID: mdl-18698374

ABSTRACT

Clinical trials have confirmed the safety of selectively oncolytic adenoviruses for treatment of advanced cancers. However, increasingly effective viruses could result in more toxicity and therefore it would be useful if replication could be abrogated if necessary. We analyzed viruses containing the cyclooxygenase-2 (Cox-2) or vascular endothelial growth factor (VEGF) promoter for controlling replication. Anti-inflammatory agents can lower Cox-2 protein levels and therefore we hypothesized that also the promoter might be affected. As Cox-2 modulates expression of VEGF, also the VEGF promoter might be controllable. First, we evaluated the effect of anti-inflammatory agents on promoter activity or adenovirus infectivity in vitro. Further, we analyzed the oncolytic potency of the viruses in vitro and in vivo with and without the reagents. Moreover, the effect of on virus replication was analyzed. We found that RGD-4C or Ad5/3 modified fibers improved the oncolytic potency of the viruses in vitro and in vivo. We found that both promoters could be downregulated with dexamethasone, sodium salicylate, or salicylic acid. Oncolytic efficacy correlated with the promoter activity and in vitro virus production could be abrogated with the substances. In vivo, we saw good therapeutic efficacy of the viruses in a model of intravenous therapy of metastatic cervical cancer, but the inhibitory effect of dexamethasone was not strong enough to provide significant differences in a complex in vivo environment. Our results suggest that anti-inflammatory drugs may affect the replication of adenovirus, which might be relevant in case of replication associated side effects.


Subject(s)
Adenoviridae , Oncolytic Virotherapy/methods , Uterine Cervical Neoplasms/rehabilitation , Uterine Cervical Neoplasms/virology , Cell Division , Cell Line, Tumor , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , HeLa Cells , Humans , Oncolytic Viruses/genetics , Promoter Regions, Genetic , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...