Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 181(2): 442-459.e29, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302573

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is a powerful tool for defining cellular diversity in tumors, but its application toward dissecting mechanisms underlying immune-modulating therapies is scarce. We performed scRNA-seq analyses on immune and stromal populations from colorectal cancer patients, identifying specific macrophage and conventional dendritic cell (cDC) subsets as key mediators of cellular cross-talk in the tumor microenvironment. Defining comparable myeloid populations in mouse tumors enabled characterization of their response to myeloid-targeted immunotherapy. Treatment with anti-CSF1R preferentially depleted macrophages with an inflammatory signature but spared macrophage populations that in mouse and human expresses pro-angiogenic/tumorigenic genes. Treatment with a CD40 agonist antibody preferentially activated a cDC population and increased Bhlhe40+ Th1-like cells and CD8+ memory T cells. Our comprehensive analysis of key myeloid subsets in human and mouse identifies critical cellular interactions regulating tumor immunity and defines mechanisms underlying myeloid-targeted immunotherapies currently undergoing clinical testing.


Subject(s)
Colonic Neoplasms/pathology , Myeloid Cells/metabolism , Single-Cell Analysis/methods , Adult , Aged , Aged, 80 and over , Animals , Base Sequence/genetics , CD8-Positive T-Lymphocytes/immunology , China , Colonic Neoplasms/therapy , Colorectal Neoplasms/pathology , Dendritic Cells/immunology , Female , Humans , Immunotherapy , Macrophages/immunology , Male , Mice , Middle Aged , Sequence Analysis, RNA/methods , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Cancer Immunol Immunother ; 72(5): 1327-1335, 2023 May.
Article in English | MEDLINE | ID: mdl-36394642

ABSTRACT

Type I interferon-mediated activation of immune cells can facilitate the generation of productive tumor antigen-specific T cell responses in solid tumors. The cGAS/STING DNA sensing pathway is a critical upstream mediator of type I interferon production and is an important regulator of anti-tumor immunity. Numerous STING pathway agonists are now being tested in clinical trials, but the effectiveness of this approach is not yet clear and a better understanding of the relative importance of this pathway in various tumor settings is needed. We have evaluated syngeneic tumor models with different baseline inflammatory states to determine the contributions of STING activity in both tumor and non-tumor cellular compartments to anti-tumor immune responses. We find that productive anti-tumor immune responses in the poorly immunogenic B16F10 model show a strong dependence on STING expression in non-tumor cells. In the immunogenic MC38 model, constitutive STING activation in tumor cells can partially bypass the requirement for STING-dependent activity from immune cells. Our findings reveal multiple, context-dependent roles for STING activity in the regulation of anti-tumor immunity and the response to immunotherapy. In preclinical models where STING is basally active, checkpoint inhibition is more likely to have a therapeutic effect and removal of STING signaling from either the tumor or the non-tumor compartment has a minimal effect. Removal of STING signaling in both, however, diminishes the efficacy derived from checkpoint therapy. Further work is needed to understand the heterogeneity of STING signaling in patients, both in tumor cells and the tumor microenvironment, and the best means of harnessing this pathway to generate anti-tumor immunity and improve therapeutic outcomes.


Subject(s)
Interferon Type I , Neoplasms , Humans , DNA , Immunity, Innate , Immunotherapy , Signal Transduction , Tumor Microenvironment
3.
Cancer Immunol Immunother ; 70(8): 2401-2410, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33511454

ABSTRACT

Tumor-associated macrophages (TAMs) are abundant in solid tumors where they exhibit immunosuppressive and pro-tumorigenic functions. Inhibition of TAM proliferation and survival through CSF1R blockade has been widely explored as a cancer immunotherapy. To further define mechanisms regulating CSF1R-targeted therapies, we systematically evaluated the effect of anti-CSF1R treatment on tumor growth and tumor microenvironment (TME) inflammation across multiple murine models. Despite substantial macrophage depletion, anti-CSF1R had minimal effects on the anti-tumor immune response in mice bearing established tumors. In contrast, anti-CSF1R treatment concurrent with tumor implantation resulted in more robust tumor growth inhibition and evidence of enhanced anti-tumor immunity. Our findings suggest only minor contributions of CSF1R-dependent TAMs to the inflammatory state of the TME in established tumors, that immune landscape heterogeneity across different tumor models can influence anti-CSF1R activity, and that alternative treatment schedules and/or TAM depletion strategies may be needed to maximize the clinical benefit of this approach.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/immunology , Neoplasms/therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Tumor-Associated Macrophages/drug effects , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Female , Immunotherapy/methods , Inflammation/drug therapy , Inflammation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology
4.
Bioorg Med Chem Lett ; 23(23): 6396-400, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24120542

ABSTRACT

A series of aminooxadiazoles was optimized for inhibition of Cdc7. Early lead isoquinoline 1 suffered from modest cell potency (cellular IC50=0.71 µM measuring pMCM2), low selectivity against structurally related kinases, and high IV clearance in rats (CL=18 L/h/kg). Extensive optimization resulted in azaindole 26 (Cdc7 IC50=1.1 nM, pMCM2 IC50=32 nM) that demonstrated robust lowering of pMCM2 in a mouse pharmacodynamic (PD) model when dosed orally. Modifications to improve the pharmacokinetic profile of this series were guided by trapping experiments with glutathione in rat hepatocytes.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Oxadiazoles/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Disease Models, Animal , Female , Mice , Mice, Nude , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
5.
Bioorg Med Chem Lett ; 21(18): 5206-9, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21840217

ABSTRACT

A bis-amide antagonist of Smoothened, a seven-transmembrane receptor in the Hedgehog signaling pathway, was discovered via high throughput screening. In vitro and in vivo experiments demonstrated that the bis-amide was susceptible to N-acyl transferase mediated amide scission. Several bioisosteric replacements of the labile amide that maintained in vitro potency were identified and shown to be metabolically stable in vitro and in vivo.


Subject(s)
Acyltransferases/antagonists & inhibitors , Amides/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Acyltransferases/metabolism , Amides/chemistry , Amides/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , High-Throughput Screening Assays , Molecular Structure , Stereoisomerism , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 20(12): 3618-22, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20493695

ABSTRACT

The Hedgehog (Hh) signaling pathway regulates cell proliferation and differentiation in developing tissues, and abnormal activation of the Hh pathway has been linked to several tumor subsets. As a transducer of Hh signaling, the GPCR-like protein Smoothened (Smo) is a promising target for disruption of unregulated Hh signaling. A series of 1-amino-4-arylphthalazines was developed as potent and orally bioavailable inhibitors of Smo. A representative compound from this class demonstrated significant tumor volume reduction in a mouse medulloblastoma model.


Subject(s)
Phthalazines/chemistry , Phthalazines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Cell Line, Tumor , Cytochrome P-450 Enzyme System/drug effects , Drug Design , Hedgehog Proteins , Humans , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Mice , Phthalazines/chemical synthesis , Signal Transduction , Smoothened Receptor
7.
Bioorg Med Chem Lett ; 20(15): 4607-10, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20594845

ABSTRACT

Pyridopyridazine antagonists of the hedgehog signaling pathway are described. Designed to optimize our previously described phthalazine smoothened antagonists, a representative compound eliminates a PXR liability while retaining potency and in vitro metabolic stability. Moreover, the compound has improved efficacy in a hedgehog/smoothened signaling mouse pharmacodynamic model.


Subject(s)
Hedgehog Proteins/antagonists & inhibitors , Phthalazines/chemistry , Piperazines/chemistry , Pyridazines/chemistry , Receptors, Steroid/chemistry , Animals , Hedgehog Proteins/metabolism , Humans , Mice , Microsomes, Liver/metabolism , Phthalazines/chemical synthesis , Phthalazines/pharmacokinetics , Piperazines/chemical synthesis , Piperazines/pharmacokinetics , Pregnane X Receptor , Pyridazines/chemical synthesis , Pyridazines/pharmacokinetics , Rats , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Receptors, Steroid/metabolism , Signal Transduction , Smoothened Receptor , Structure-Activity Relationship , Tylosin/analogs & derivatives
8.
J Biol Methods ; 7(1): e125, 2020.
Article in English | MEDLINE | ID: mdl-32206674

ABSTRACT

Bone marrow chimeras represent a key tool employed to understand biological contributions stemming from the hematopoietic versus the stromal compartment. In most institutions, cesium irradiators are used to lethally irradiate recipient animals prior to the injection of donor bone marrow. Cesium irradiators, however, have significant liabilities-including concerns around domestic security. Recently, X-ray irradiators have been implemented as a potential alternative to cesium sources. Only a small number of publications in the literature have attempted to compare these two modalities and, in most cases, the emphasis was on irradiation of human blood productions. We were able to find only a single study that directly compared X-ray and cesium technologies in the generation of murine bone marrow chimeras, a standard laboratory practice. This study focused on chimerism in the blood of recipient animals. In the present study, we begin by comparing cesium and X-ray based sources for irradiation, then transition to using X-ray-based systems for immunology models with an emphasis on immunotherapy of cancer in immunocompetent mouse models-specifically evaluating chimerism in the blood, spleen, and tumor microenvironment. While our data demonstrate that the two platforms are functionally comparable and suggest that X-ray based technology is a suitable alternative to cesium sources. We also highlight a difference in chimerism between the peripheral (blood, spleen) and tumor compartments that is observed using both technologies. While the overall degree of chimerism in the peripheral tissues is very high, the degree of chimerism in the tumor is cell type specific with T and NK cells showing lower chimerism than other cell types.

9.
Mol Cancer Ther ; 13(4): 880-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24526162

ABSTRACT

Acute myeloid leukemia (AML) remains a serious unmet medical need. Despite high remission rates with chemotherapy standard-of-care treatment, the disease eventually relapses in a major proportion of patients. Activating Fms-like tyrosine kinase 3 (FLT3) mutations are found in approximately 30% of patients with AML. Targeting FLT3 receptor tyrosine kinase has shown encouraging results in treating FLT3-mutated AML. Responses, however, are not sustained and acquired resistance has been a clinical challenge. Treatment options to overcome resistance are currently the focus of research. We report here the preclinical evaluation of AMG 925, a potent, selective, and bioavailable FLT3/cyclin-dependent kinase 4 (CDK4) dual kinase inhibitor. AMG 925 inhibited AML xenograft tumor growth by 96% to 99% without significant body weight loss. The antitumor activity of AMG 925 correlated with the inhibition of STAT5 and RB phosphorylation, the pharmacodynamic markers for inhibition of FLT3 and CDK4, respectively. In addition, AMG 925 was also found to inhibit FLT3 mutants (e.g., D835Y) that are resistant to the current FLT3 inhibitors (e.g., AC220 and sorafenib). CDK4 is a cyclin D-dependent kinase that plays an essential central role in regulating cell proliferation in response to external growth signals. A critical role of the CDK4-RB pathway in cancer development has been well established. CDK4-specific inhibitors are being developed for treating RB-positive cancer. AMG 925, which combines inhibition of two kinases essential for proliferation and survival of FLT3-mutated AML cells, may improve and prolong clinical responses.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Naphthyridines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Nude , Naphthyridines/pharmacokinetics , Naphthyridines/therapeutic use , Neoplasms, Experimental , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/pharmacology , Signal Transduction/drug effects , Sorafenib , U937 Cells , Xenograft Model Antitumor Assays
10.
Mol Cancer Ther ; 10(12): 2298-308, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21926191

ABSTRACT

The signaling pathway of the receptor tyrosine kinase MET and its ligand hepatocyte growth factor (HGF) is important for cell growth, survival, and motility and is functionally linked to the signaling pathway of VEGF, which is widely recognized as a key effector in angiogenesis and cancer progression. Dysregulation of the MET/VEGF axis is found in a number of human malignancies and has been associated with tumorigenesis. Cabozantinib (XL184) is a small-molecule kinase inhibitor with potent activity toward MET and VEGF receptor 2 (VEGFR2), as well as a number of other receptor tyrosine kinases that have also been implicated in tumor pathobiology, including RET, KIT, AXL, and FLT3. Treatment with cabozantinib inhibited MET and VEGFR2 phosphorylation in vitro and in tumor models in vivo and led to significant reductions in cell invasion in vitro. In mouse models, cabozantinib dramatically altered tumor pathology, resulting in decreased tumor and endothelial cell proliferation coupled with increased apoptosis and dose-dependent inhibition of tumor growth in breast, lung, and glioma tumor models. Importantly, treatment with cabozantinib did not increase lung tumor burden in an experimental model of metastasis, which has been observed with inhibitors of VEGF signaling that do not target MET. Collectively, these data suggest that cabozantinib is a promising agent for inhibiting tumor angiogenesis and metastasis in cancers with dysregulated MET and VEGFR signaling.


Subject(s)
Anilides/therapeutic use , Cell Growth Processes/drug effects , Neoplasm Metastasis/prevention & control , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Pyridines/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Anilides/pharmacology , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Nude , Models, Biological , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
11.
Proc Natl Acad Sci U S A ; 102(44): 15901-6, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16247015

ABSTRACT

Metastasis of primary tumors leads to a very poor prognosis for patients suffering from cancer. Although it is well established that not every tumor will eventually metastasize, it is less clear whether primary tumors acquire genetic alterations in a stochastic process at a late stage, which make them invasive, or whether genetic alterations acquired early in the process of tumor development drive primary tumor growth and determine whether this tumor is going to be metastatic. To address this issue, we tested genes identified in a large-scale comparative genomic hybridization analysis of primary tumor for their ability to confer metastatic properties on a cancer cell. We identified amplification of the ACK1 gene in primary tumors, which correlates with poor prognosis. We further show that overexpression of Ack1 in cancer cell lines can increase the invasive phenotype of these cells both in vitro and in vivo and leads to increased mortality in a mouse model of metastasis. Biochemical studies show that Ack1 is involved in extracellular matrix-induced integrin signaling, ultimately activating signaling processes like the activation of the small GTPase Rac. Taken together, this study supports a theory from Bernards and Weinberg [Bernards, R. & Weinberg, R. A. (2002) Nature 418, 823], which postulates that the tendency to metastasize is largely predetermined.


Subject(s)
Gene Amplification , Neoplasm Metastasis/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Animals , Cell Line, Tumor , Crk-Associated Substrate Protein/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha3beta1/metabolism , Lung Neoplasms/secondary , Mice , Neoplasm Transplantation , Prognosis , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Transplantation, Heterologous , Tumor Cells, Cultured , rac GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL