Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 114(38): 10220-10225, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874539

ABSTRACT

Contrast-enhanced MRI is typically used to follow treatment response and progression in patients with glioblastoma (GBM). However, differentiating tumor progression from pseudoprogression remains a clinical dilemma largely unmitigated by current advances in imaging techniques. Noninvasive imaging techniques capable of distinguishing these two conditions could play an important role in the clinical management of patients with GBM and other brain malignancies. We hypothesized that PET probes for deoxycytidine kinase (dCK) could be used to differentiate immune inflammatory responses from other sources of contrast-enhancement on MRI. Orthotopic malignant gliomas were established in syngeneic immunocompetent mice and then treated with dendritic cell (DC) vaccination and/or PD-1 mAb blockade. Mice were then imaged with [18F]-FAC PET/CT and MRI with i.v. contrast. The ratio of contrast enhancement on MRI to normalized PET probe uptake, which we term the immunotherapeutic response index, delineated specific regions of immune inflammatory activity. On postmortem examination, FACS-based enumeration of intracranial tumor-infiltrating lymphocytes directly correlated with quantitative [18F]-FAC PET probe uptake. Three patients with GBM undergoing treatment with tumor lysate-pulsed DC vaccination and PD-1 mAb blockade were also imaged before and after therapy using MRI and a clinical PET probe for dCK. Unlike in mice, [18F]-FAC is rapidly catabolized in humans; thus, we used another dCK PET probe, [18F]-clofarabine ([18F]-CFA), that may be more clinically relevant. Enhanced [18F]-CFA PET probe accumulation was identified in tumor and secondary lymphoid organs after immunotherapy. Our findings identify a noninvasive modality capable of imaging the host antitumor immune response against intracranial tumors.


Subject(s)
Glioblastoma/diagnostic imaging , Animals , Cell Line , Female , Glioblastoma/therapy , Humans , Immunotherapy , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Positron-Emission Tomography
2.
Sci Rep ; 6: 29032, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27358011

ABSTRACT

Metastatic renal cell carcinoma (mRCC) is nearly incurable and accounts for most of the mortality associated with RCC. Von Hippel Lindau (VHL) is a tumour suppressor that is lost in the majority of clear cell RCC (ccRCC) cases. Its role in regulating hypoxia-inducible factors-1α (HIF-1α) and -2α (HIF-2α) is well-studied. Recent work has demonstrated that VHL knock down induces an epithelial-mesenchymal transition (EMT) phenotype. In this study we showed that a CRISPR/Cas9-mediated knock out of VHL in the RENCA model leads to morphologic and molecular changes indicative of EMT, which in turn drives increased metastasis to the lungs. RENCA cells deficient in HIF-1α failed to undergo EMT changes upon VHL knockout. RNA-seq revealed several HIF-1α-regulated genes that are upregulated in our VHL knockout cells and whose overexpression signifies an aggressive form of ccRCC in the cancer genome atlas (TCGA) database. Independent validation in a new clinical dataset confirms the upregulation of these genes in ccRCC samples compared to adjacent normal tissue. Our findings indicate that loss of VHL could be driving tumour cell dissemination through stabilization of HIF-1α in RCC. A better understanding of the mechanisms involved in this phenomenon can guide the search for more effective treatments to combat mRCC.


Subject(s)
Carcinoma, Renal Cell/secondary , Disease Models, Animal , Kidney Neoplasms/pathology , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Animals , Bacterial Proteins , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Movement , Datasets as Topic , Endonucleases , Epithelial-Mesenchymal Transition , Female , Gene Editing , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Lung Neoplasms/secondary , Mice , Mice, Knockout , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , RNA, Guide, Kinetoplastida , Von Hippel-Lindau Tumor Suppressor Protein/genetics
3.
Cancer Res ; 75(22): 4742-52, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26471360

ABSTRACT

Malignant ascites is a common complication in the late stages of epithelial ovarian cancer (EOC) that greatly diminishes the quality of life of patients. Malignant ascites is a known consequence of vascular dysfunction, but current approved treatments are not effective in preventing fluid accumulation. In this study, we investigated an alternative strategy of targeting macrophage functions to reverse the vascular pathology of malignant ascites using fluid from human patients and an immunocompetent murine model (ID8) of EOC that mirrors human disease by developing progressive vascular disorganization and leakiness culminating in massive ascites. We demonstrate that the macrophage content in ascites fluid from human patients and the ID8 model directly correlates with vascular permeability. To further substantiate macrophages' role in the pathogenesis of malignant ascites, we blocked macrophage function in ID8 mice using a colony-stimulating factor 1 receptor kinase inhibitor (GW2580). Administration of GW2580 in the late stages of disease resulted in reduced infiltration of protumorigenic (M2) macrophages and dramatically decreased ascites volume. Moreover, the disorganized peritoneal vasculature became normalized and sera from GW2580-treated ascites protected against endothelial permeability. Therefore, our findings suggest that macrophage-targeted treatment may be a promising strategy toward a safe and effective means to control malignant ascites of EOC.


Subject(s)
Anisoles/pharmacology , Ascites/prevention & control , Capillary Permeability/drug effects , Macrophages/drug effects , Neoplasms, Glandular and Epithelial/complications , Ovarian Neoplasms/complications , Pyrimidines/pharmacology , Animals , Ascites/etiology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunohistochemistry , Mice , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
4.
PLoS One ; 8(9): e73650, 2013.
Article in English | MEDLINE | ID: mdl-24023896

ABSTRACT

Tumor-specific adenoviral vectors comprise a fruitful gene-based diagnostic imaging and therapy research area for advanced stage of cancer, including metastatic disease. However, clinical translation of viral vectors has encountered considerable obstacles, largely due to host immune responses against the virus. Here, we explored the utilization of an immunosuppressant, rapamycin, to circumvent the anti-adenovirus immunity in immunocompetent murine prostate cancer models. Rapamycin diminished adenoviral-induced acute immune response by inhibiting NF-κB activation; it also reduced the scale and delayed the onset of inflammatory cytokine secretion. Further, we found that rapamycin abrogated anti-adenovirus antibody production and retarded the function of myeloid cells and lymphocytes that were activated upon viral administration in pre-immunized hosts. Thus, the co-administration of rapamycin prolonged and enhanced adenovirus-delivered transgene expression in vivo, and thereby augmented the imaging capability of adenoviral vectors in both bioluminescent and positron emission tomography modalities. Furthermore, we showed that despite an excellent response of cancer cells to a cytotoxic gene therapeutic vector in vitro, only minimal therapeutic effects were observed in vivo in pre-immunized mice. However, when we combined gene therapy with transient immunosuppression, complete tumor growth arrest was achieved. Overall, transient immunosuppression by rapamycin was able to boost the diagnostic utility and therapeutic potentials of adenoviral vectors.


Subject(s)
Adenoviridae/genetics , Immunosuppression Therapy/methods , Immunosuppressive Agents/pharmacology , Molecular Imaging , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Sirolimus/pharmacology , Adaptive Immunity/drug effects , Animals , Cell Line, Tumor , Ganciclovir/pharmacology , Genetic Therapy/adverse effects , Genetic Vectors/genetics , Immunity, Innate/drug effects , Immunization , Male , Mice , Molecular Imaging/adverse effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Safety , Thymidine Kinase/genetics , Transgenes/genetics
5.
Blood ; 115(7): 1461-71, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-20008303

ABSTRACT

Tumor-infiltrating myeloid cells (TIMs) support tumor growth by promoting angiogenesis and suppressing antitumor immune responses. CSF-1 receptor (CSF1R) signaling is important for the recruitment of CD11b(+)F4/80(+) tumor-associated macrophages (TAMs) and contributes to myeloid cell-mediated angiogenesis. However, the impact of the CSF1R signaling pathway on other TIM subsets, including CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSCs), is unknown. Tumor-infiltrating MDSCs have also been shown to contribute to tumor angiogenesis and have recently been implicated in tumor resistance to antiangiogenic therapy, yet their precise involvement in these processes is not well understood. Here, we use the selective pharmacologic inhibitor of CSF1R signaling, GW2580, to demonstrate that CSF-1 regulates the tumor recruitment of CD11b(+)Gr-1(lo)Ly6C(hi) mononuclear MDSCs. Targeting these TIM subsets inhibits tumor angiogenesis associated with reduced expression of proangiogenic and immunosuppressive genes. Combination therapy using GW2580 with an anti-VEGFR-2 antibody synergistically suppresses tumor growth and severely impairs tumor angiogenesis along with reverting at least one TIM-mediated antiangiogenic compensatory mechanism involving MMP-9. These data highlight the importance of CSF1R signaling in the recruitment and function of distinct TIM subsets, including MDSCs, and validate the benefits of targeting CSF1R signaling in combination with antiangiogenic drugs for the treatment of solid cancers.


Subject(s)
Anisoles/pharmacology , Carcinoma, Lewis Lung/drug therapy , Cell Movement/drug effects , Lung Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Pyrimidines/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Lung Neoplasms/pathology , Macrophages/cytology , Male , Matrix Metalloproteinase 9/metabolism , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/pathology , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rats , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...