Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
JCI Insight ; 8(4)2023 02 22.
Article in English | MEDLINE | ID: mdl-36810257

ABSTRACT

Inhibitors of the DNA damage signaling kinase ATR increase tumor cell killing by chemotherapies that target DNA replication forks but also kill rapidly proliferating immune cells including activated T cells. Nevertheless, ATR inhibitor (ATRi) and radiotherapy (RT) can be combined to generate CD8+ T cell-dependent antitumor responses in mouse models. To determine the optimal schedule of ATRi and RT, we determined the impact of short-course versus prolonged daily treatment with AZD6738 (ATRi) on responses to RT (days 1-2). Short-course ATRi (days 1-3) plus RT caused expansion of tumor antigen-specific, effector CD8+ T cells in the tumor-draining lymph node (DLN) at 1 week after RT. This was preceded by acute decreases in proliferating tumor-infiltrating and peripheral T cells and a rapid proliferative rebound after ATRi cessation, increased inflammatory signaling (IFN-ß, chemokines, particularly CXCL10) in tumors, and an accumulation of inflammatory cells in the DLN. In contrast, prolonged ATRi (days 1-9) prevented the expansion of tumor antigen-specific, effector CD8+ T cells in the DLN, and entirely abolished the therapeutic benefit of short-course ATRi with RT and anti-PD-L1. Our data argue that ATRi cessation is essential to allow CD8+ T cell responses to both RT and immune checkpoint inhibitors.


Subject(s)
Neoplasms , Animals , Mice , Neoplasms/pathology , Sulfonamides , Immunity , Antigens, Neoplasm
2.
Cell Rep ; 40(12): 111371, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130512

ABSTRACT

ATR kinase is a central regulator of the DNA damage response (DDR) and cell cycle checkpoints. ATR kinase inhibitors (ATRi's) combine with radiation to generate CD8+ T cell-dependent responses in mouse models of cancer. We show that ATRi's induce cyclin-dependent kinase 1 (CDK1)-dependent origin firing across active replicons in CD8+ T cells activated ex vivo while simultaneously decreasing the activity of rate-limiting enzymes for nucleotide biosynthesis. These pleiotropic effects of ATRi induce deoxyuridine (dU) contamination in genomic DNA, R loops, RNA-DNA polymerase collisions, and interferon-α/ß (IFN-α/ß). Remarkably, thymidine rescues ATRi-induced dU contamination and partially rescues death and IFN-α/ß expression in proliferating CD8+ T cells. Thymidine also partially rescues ATRi-induced cancer cell death. We propose that ATRi-induced dU contamination contributes to dose-limiting leukocytopenia and inflammation in the clinic and CD8+ T cell-dependent anti-tumor responses in mouse models. We conclude that ATR is essential to limit dU contamination in genomic DNA and IFN-α/ß expression.


Subject(s)
CD8-Positive T-Lymphocytes , CDC2 Protein Kinase , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , CD8-Positive T-Lymphocytes/metabolism , CDC2 Protein Kinase/metabolism , Cell Death , Cell Line, Tumor , DNA , DNA Damage , DNA-Directed DNA Polymerase/metabolism , Deoxyuridine , Genomics , Interferon-alpha/metabolism , Interferon-beta , Mice , Nucleotides/metabolism , Protein Kinase Inhibitors/pharmacology , RNA , Thymidine/pharmacology
3.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33351788

ABSTRACT

Human lung adenocarcinoma (LUAD) in current or former smokers exhibits a high tumor mutational burden (TMB) and distinct mutational signatures. Syngeneic mouse models of clinically relevant smoking-related LUAD are lacking. We established and characterized a tobacco-associated, transplantable murine LUAD cell line, designated FVBW-17, from a LUAD induced by the tobacco carcinogen 4-(methylnitrosoamino)-1-(3-pyridyl)-1-butanone in the FVB/N mouse strain. Whole-exome sequencing of FVBW-17 cells identified tobacco-associated KrasG12D and Trp53 mutations and a similar mutation profile to that of classic alkylating agents with a TMB greater than 500. FVBW-17 cells transplanted subcutaneously, via tail vein, and orthotopically generated tumors that were histologically similar to human LUAD in FVB/N mice. FVBW-17 tumors expressed programmed death ligand 1 (PD-L1), were infiltrated with CD8+ T cells, and were responsive to anti-PD-L1 therapy. FVBW-17 cells were also engineered to express green fluorescent protein and luciferase to facilitate detection and quantification of tumor growth. Distant metastases to lung, spleen, liver, and kidney were observed from subcutaneously transplanted tumors. This potentially novel cell line is a robust representation of human smoking-related LUAD biology and provides a much needed preclinical model in which to test promising new agents and combinations, including immune-based therapies.


Subject(s)
Adenocarcinoma of Lung/chemically induced , B7-H1 Antigen/metabolism , Carcinogens/toxicity , Lung Neoplasms/chemically induced , Nitrosamines/toxicity , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Mice , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Smoke/adverse effects , Nicotiana/toxicity , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
4.
Cancer Immunol Res ; 7(8): 1258-1266, 2019 08.
Article in English | MEDLINE | ID: mdl-31239318

ABSTRACT

Multiple studies have associated the transcription factor IRF1 with tumor-suppressive activities. Here, we report an opposite tumor cell-intrinsic function of IRF1 in promoting tumor growth. IRF1-deficient tumor cells showed reduced tumor growth in MC38 and CT26 colon carcinoma and B16 melanoma mouse models. This reduction in tumor growth was dependent on host CD8+ T cells. Detailed profiling of tumor-infiltrating leukocytes did not show changes in the various T-cell and myeloid cell populations. However, CD8+ T cells that had infiltrated IRF1-deficieint tumors in vivo exhibited enhanced cytotoxicity. IRF1-deficient tumor cells lost the ability to upregulate PD-L1 expression in vitro and in vivo and were more susceptible to T-cell-mediated killing. Induced expression of PD-L1 in IRF1-deficient tumor cells restored tumor growth. These results indicate differential activity of IRF1 in tumor escape.


Subject(s)
B7-H1 Antigen/genetics , Gene Expression Regulation, Neoplastic , Immunomodulation , Interferon Regulatory Factor-1/metabolism , Animals , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Humans , Immunologic Memory , Immunomodulation/genetics , Interferon Regulatory Factor-1/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental , Mice , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
J Clin Invest ; 128(9): 3926-3940, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29952768

ABSTRACT

DNA-damaging chemotherapy and radiation therapy are integrated into the treatment paradigm of the majority of cancer patients. Recently, immunotherapy that targets the immunosuppressive interaction between programmed death 1 (PD-1) and its ligand PD-L1 has been approved for malignancies including non-small cell lung cancer, melanoma, and head and neck squamous cell carcinoma. ATR is a DNA damage-signaling kinase activated at damaged replication forks, and ATR kinase inhibitors potentiate the cytotoxicity of DNA-damaging chemotherapies. We show here that the ATR kinase inhibitor AZD6738 combines with conformal radiation therapy to attenuate radiation-induced CD8+ T cell exhaustion and potentiate CD8+ T cell activity in mouse models of Kras-mutant cancer. Mechanistically, AZD6738 blocks radiation-induced PD-L1 upregulation on tumor cells and dramatically decreases the number of tumor-infiltrating Tregs. Remarkably, AZD6738 combines with conformal radiation therapy to generate immunologic memory in complete responder mice. Our work raises the possibility that a single pharmacologic agent may enhance the cytotoxic effects of radiation while concurrently potentiating radiation-induced antitumor immune responses.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/radiation effects , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/radiotherapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Sulfoxides/pharmacology , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/radiotherapy , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chemoradiotherapy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/radiotherapy , Humans , Indoles , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/radiation effects , Mice , Mice, Inbred BALB C , Morpholines , Neoplasms, Experimental/immunology , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/pharmacokinetics , Radiotherapy, Conformal , Sulfonamides , Sulfoxides/pharmacokinetics
6.
Sci Rep ; 7: 41892, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28145510

ABSTRACT

We show that ATM kinase inhibition using AZ31 prior to 9 or 9.25 Gy total body irradiation (TBI) reduced median time to moribund in mice to 8 days. ATR kinase inhibition using AZD6738 prior to TBI did not reduce median time to moribund. The striking finding associated with ATM inhibition prior to TBI was increased crypt loss within the intestine epithelium. ATM inhibition reduced upregulation of p21, an inhibitor of cyclin-dependent kinases, and blocked G1 arrest after TBI thereby increasing the number of S phase cells in crypts in wild-type but not Cdkn1a(p21CIP/WAF1)-/- mice. In contrast, ATR inhibition increased upregulation of p21 after TBI. Thus, ATM activity is essential for p21-dependent arrest while ATR inhibition may potentiate arrest in crypt cells after TBI. Nevertheless, ATM inhibition reduced median time to moribund in Cdkn1a(p21CIP/WAF1)-/- mice after TBI. ATM inhibition also increased cell death in crypts at 4 h in Cdkn1a(p21CIP/WAF1)-/-, earlier than at 24 h in wild-type mice after TBI. In contrast, ATR inhibition decreased cell death in crypts in Cdkn1a(p21CIP/WAF1)-/- mice at 4 h after TBI. We conclude that ATM activity is essential for p21-dependent and p21-independent mechanisms that radioprotect intestinal crypts and that ATM inhibition promotes GI syndrome after TBI.


Subject(s)
Acute Radiation Syndrome/drug therapy , G1 Phase/drug effects , Gastrointestinal Diseases/drug therapy , Intestinal Mucosa/drug effects , Protein Kinase Inhibitors/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gamma Rays/adverse effects , Indoles , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Mice , Mice, Inbred C57BL , Morpholines , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinolines/pharmacokinetics , Quinolines/pharmacology , Quinolines/therapeutic use , Radiation-Protective Agents/pharmacokinetics , Radiation-Protective Agents/therapeutic use , Sulfonamides , Sulfoxides/pharmacokinetics , Sulfoxides/pharmacology , Sulfoxides/therapeutic use
7.
J Pharm Biomed Anal ; 138: 158-165, 2017 May 10.
Article in English | MEDLINE | ID: mdl-28213176

ABSTRACT

The ATM kinase inhibitor AZ31 and ATR kinase inhibitor AZD6738 are in various phases of preclinical and clinical evaluation for their ability to potentiate chemoradiation. To support the preclinical evaluation of their pharmacokinetics, we developed and validated an LC-MS/MS assay for the simultaneous quantification of AZ31 and AZD6738 in mouse plasma. A "dilute and shoot" method was used to precipitate proteins from a sample volume of 50µL. Chromatographic separation was achieved using a Phenomenex Polar-RP column and a gradient mobile phase consisting of methanol-water with 0.1% formic acid. Detection was accomplished using a Waters Quattro Micro mass spectrometer in positive ionization mode. The assay utilizing 50µL sample was linear from 10 to 5000ng/mL and determined to be both accurate (-8.2 to 8.6%) and precise (<5.4% CV) and achieved the criteria for U.S. FDA guidance for bioanalytical method validation. Quantification was achieved in mouse tissue homogenate using a separate 200µL sample preparation. This LC-MS/MS assay will be essential for determining the tissue distribution and pharmacokinetics in future mouse studies.


Subject(s)
Alloys/chemistry , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Chromatography, Liquid/methods , Plasma/chemistry , Pyrimidines/chemistry , Sulfoxides/chemistry , Tandem Mass Spectrometry/methods , Animals , Biological Assay/methods , Drug Stability , Formates/chemistry , Indoles , Methanol/chemistry , Mice , Morpholines , Pyrimidines/blood , Reproducibility of Results , Sulfonamides , Sulfoxides/blood
8.
Oncotarget ; 6(42): 44289-305, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26517239

ABSTRACT

ATR and ATM are DNA damage signaling kinases that phosphorylate several thousand substrates. ATR kinase activity is increased at damaged replication forks and resected DNA double-strand breaks (DSBs). ATM kinase activity is increased at DSBs. ATM has been widely studied since ataxia telangiectasia individuals who express no ATM protein are the most radiosensitive patients identified. Since ATM is not an essential protein, it is widely believed that ATM kinase inhibitors will be well-tolerated in the clinic. ATR has been widely studied, but advances have been complicated by the finding that ATR is an essential protein and it is widely believed that ATR kinase inhibitors will be toxic in the clinic. We describe AZD6738, an orally active and bioavailable ATR kinase inhibitor. AZD6738 induces cell death and senescence in non-small cell lung cancer (NSCLC) cell lines. AZD6738 potentiates the cytotoxicity of cisplatin and gemcitabine in NSCLC cell lines with intact ATM kinase signaling, and potently synergizes with cisplatin in ATM-deficient NSCLC cells. In contrast to expectations, daily administration of AZD6738 and ATR kinase inhibition for 14 consecutive days is tolerated in mice and enhances the therapeutic efficacy of cisplatin in xenograft models. Remarkably, the combination of cisplatin and AZD6738 resolves ATM-deficient lung cancer xenografts.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ataxia Telangiectasia Mutated Proteins/deficiency , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/administration & dosage , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Sulfoxides/administration & dosage , Administration, Oral , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Biological Availability , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Death/drug effects , Cell Line, Tumor , Cellular Senescence/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Drug Synergism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Indoles , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Morpholines , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , RNA Interference , Sulfonamides , Sulfoxides/pharmacokinetics , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
9.
Oncotarget ; 6(1): 56-70, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25474141

ABSTRACT

Recent clinical data in lung cancer suggests that epigenetically targeted therapy may selectively enhance chemotherapeutic sensitivity. There have been few if any studies rigorously evaluating this hypothesized priming effect. Here we describe a series of investigations testing whether epigenetic priming with azacitidine and entinostat increases sensitivity of NSCLC to cytotoxic agents. We noted no differences in chemosensitivity following treatment with epigenetic therapy in in vitro assays of viability and colony growth. Using cell line and patient derived xenograft (PDX) models, we also observed no change in responsiveness to cisplatin in vivo. In select models, we noted differential responses to irinotecan treatment in vivo. In vitro epigenetic therapy prior to tumor implantation abrogated response of H460 xenografts to irinotecan. Conversely, in vitro epigenetic therapy appeared to sensitize A549 xenografts (tumor growth inhibition 51%, vs. 22% in mock-pretreated control). In vivo epigenetic therapy enhanced the response of adenocarcinoma PDX to irinotecan. Taken together, these data do not support broadly applicable epigenetic priming in NSCLC. Priming effects may be context-specific, dependent on both tumor and host factors. Further preclinical study is necessary to determine whether, and in which contexts, priming with epigenetic therapy has potential to enhance chemotherapeutic efficacy in NSCLC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Azacitidine/pharmacology , Benzamides/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Pyridines/pharmacology , Animals , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Line, Tumor , Cell Survival , DNA Methylation , Drug Evaluation, Preclinical , Epigenesis, Genetic , Female , Histones/chemistry , Humans , Irinotecan , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation
10.
Expert Opin Biol Ther ; 13(9): 1273-85, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23859704

ABSTRACT

INTRODUCTION: Epigenetics refers to heritable modifications of DNA and associated chromatin components that influence gene expression without altering DNA coding sequence. Epigenetic dysregulation is a central contributor to oncogenesis and is increasingly a focus of interest in cancer therapeutic research. Two key levels of aberrant epigenetic control are DNA methylation and histone acetylation. Primary regulators of these epigenetic changes include DNA methyltransferases (DNMTs) and histone deacetylases (HDACs). AREAS COVERED: This review focuses on epigenetic changes in non-small-cell lung cancer and recent preclinical and clinical studies targeting these changes. DNMT inhibitors were previously explored at or near maximally tolerated doses, levels at which these agents are cytotoxic but have suboptimal effects on DNA methylation. Use of these inhibitors at substantially lower doses, in combination with HDAC inhibitors, can promote re-expression of silenced tumor suppressor genes, can result in major clinical responses and may alter tumor responsiveness to subsequent cytotoxic therapies. EXPERT OPINION: Combinatorial epigenetic therapy has demonstrated encouraging clinical activity, but many relevant questions remain. Global strategies influencing the epigenome may have both positive and potential negative long-term effects on cancer progression. Further clinical investigation of this approach, including exploratory studies to define predictive biomarkers, is warranted.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , DNA Modification Methylases/antagonists & inhibitors , Epigenesis, Genetic , Histone Deacetylase Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Drug Resistance, Neoplasm , Humans , Lung Neoplasms/enzymology
11.
Cancer Discov ; 1(7): 598-607, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22586682

ABSTRACT

UNLABELLED: Epigenetic alterations are strongly associated with the development of cancer. We conducted a phase I/II trial of combined epigenetic therapy with azacitidine and entinostat, inhibitors of DNA methylation and histone deacetylation, respectively, in extensively pretreated patients with recurrent metastatic non-small cell lung cancer. This therapy is well tolerated, and objective responses were observed, including a complete response and a partial response in a patient who remains alive and without disease progression approximately 2 years after completing protocol therapy. Median survival in the entire cohort was 6.4 months (95% CI 3.8-9.2), comparing favorably with existing therapeutic options. Demethylation of a set of 4 epigenetically silenced genes known to be associated with lung cancer was detectable in serial blood samples in these patients and was associated with improved progression-free (P = 0.034) and overall survival (P = 0.035). Four of 19 patients had major objective responses to subsequent anticancer therapies given immediately after epigenetic therapy. SIGNIFICANCE: This study demonstrates that combined epigenetic therapy with low-dose azacitidine and entinostat results in objective, durable responses in patients with solid tumors and defines a blood-based biomarker that correlates with clinical benefit.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Azacitidine/administration & dosage , Azacitidine/adverse effects , Benzamides/administration & dosage , Benzamides/adverse effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cohort Studies , Combined Modality Therapy , DNA Methylation/drug effects , Disease Progression , Disease-Free Survival , Epigenesis, Genetic , Female , Genetic Therapy , Histones/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Middle Aged , Pyridines/administration & dosage , Pyridines/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...