Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nature ; 628(8009): 872-877, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570682

ABSTRACT

Propionic acidaemia is a rare disorder caused by defects in the propionyl-coenzyme A carboxylase α or ß (PCCA or PCCB) subunits that leads to an accumulation of toxic metabolites and to recurrent, life-threatening metabolic decompensation events. Here we report interim analyses of a first-in-human, phase 1/2, open-label, dose-optimization study and an extension study evaluating the safety and efficacy of mRNA-3927, a dual mRNA therapy encoding PCCA and PCCB. As of 31 May 2023, 16 participants were enrolled across 5 dose cohorts. Twelve of the 16 participants completed the dose-optimization study and enrolled in the extension study. A total of 346 intravenous doses of mRNA-3927 were administered over a total of 15.69 person-years of treatment. No dose-limiting toxicities occurred. Treatment-emergent adverse events were reported in 15 out of the 16 (93.8%) participants. Preliminary analysis suggests an increase in the exposure to mRNA-3927 with dose escalation, and a 70% reduction in the risk of metabolic decompensation events among 8 participants who reported them in the 12-month pretreatment period.


Subject(s)
Propionic Acidemia , Propionyl-Coenzyme A Carboxylase , RNA, Messenger , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Male , Young Adult , Administration, Intravenous , Dose-Response Relationship, Drug , Propionic Acidemia/genetics , Propionic Acidemia/therapy , Propionyl-Coenzyme A Carboxylase/genetics , Propionyl-Coenzyme A Carboxylase/metabolism , RNA, Messenger/administration & dosage , RNA, Messenger/adverse effects , RNA, Messenger/genetics , RNA, Messenger/therapeutic use
3.
J Biol Chem ; 297(5): 101163, 2021 11.
Article in English | MEDLINE | ID: mdl-34481841

ABSTRACT

Inactivation of p53 is present in almost every tumor, and hence, p53-reactivation strategies are an important aspect of cancer therapy. Common mechanisms for p53 loss in cancer include expression of p53-negative regulators such as MDM2, which mediate the degradation of wildtype p53 (p53α), and inactivating mutations in the TP53 gene. Currently, approaches to overcome p53 deficiency in these cancers are limited. Here, using non-small cell lung cancer and glioblastoma multiforme cell line models, we show that two alternatively spliced, functional truncated isoforms of p53 (p53ß and p53γ, comprising exons 1 to 9ß or 9γ, respectively) and that lack the C-terminal MDM2-binding domain have markedly reduced susceptibility to MDM2-mediated degradation but are highly susceptible to nonsense-mediated decay (NMD), a regulator of aberrant mRNA stability. In cancer cells harboring MDM2 overexpression or TP53 mutations downstream of exon 9, NMD inhibition markedly upregulates p53ß and p53γ and restores activation of the p53 pathway. Consistent with p53 pathway activation, NMD inhibition induces tumor suppressive activities such as apoptosis, reduced cell viability, and enhanced tumor radiosensitivity, in a relatively p53-dependent manner. In addition, NMD inhibition also inhibits tumor growth in a MDM2-overexpressing xenograft tumor model. These results identify NMD inhibition as a novel therapeutic strategy for restoration of p53 function in p53-deficient tumors bearing MDM2 overexpression or p53 mutations downstream of exon 9, subgroups that comprise approximately 6% of all cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , Mutation , Nonsense Mediated mRNA Decay , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53 , A549 Cells , Animals , Humans , Mice , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
4.
J Thorac Oncol ; 16(4): 601-609, 2021 04.
Article in English | MEDLINE | ID: mdl-33388476

ABSTRACT

INTRODUCTION: Plasma-based circulating tumor DNA (ctDNA) is an established biomarker for molecular profiling with emerging applications in disease monitoring in multiple tumor types, including, NSCLC. However, determinants of ctDNA shedding and correlation with tumor burden are incompletely understood, particularly in advanced-stage disease. METHODS: We retrospectively analyzed ctDNA-based and tissue-based genomic data and imaging from 144 patients with NSCLC. Tumor burden was quantified with computed tomography (CT) and brain magnetic resonance imaging for the overall cohort and 18F-fludeoxyglucose positron emission tomography-CT in a subset of patients. RESULTS: There was a moderate but statistically significant correlation between ctDNA variant allele frequency and multiple imaging measures of tumor burden such as CT volume (rho = 0.34, p ≤ 0.0001) and metabolic tumor volume (rho = 0.36, p = 0.003). This correlation was strongest in KRAS-mutant tumors (rho = 0.56, p ≤ 0.001), followed by TP53 mutants (rho = 0.43, p ≤ 0.0001), and weakest in EGFR-mutated (EGFR+) tumors (rho = 0.24, p = 0.077). EGFR+ tumors with EGFR copy number gain had significantly higher variant allele frequency than EGFR+ without copy number gain (p ≤ 0.00001). In multivariable analysis, TP53 and EGFR mutations, visceral metastasis, and tumor burden were independent predictors of increased ctDNA shedding. CONCLUSIONS: Levels of detectable ctDNA were affected not only by tumor burden but also by tumor genotype. The genotype-specific differences observed may be due to variations in DNA shedding and cellular turnover. These findings have implications for the emerging use of ctDNA in NSCLC disease monitoring and early detection.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Circulating Tumor DNA , Lung Neoplasms , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Circulating Tumor DNA/genetics , Genotype , Humans , Lung Neoplasms/genetics , Mutation , Retrospective Studies
5.
Mod Pathol ; 32(11): 1698-1707, 2019 11.
Article in English | MEDLINE | ID: mdl-31231128

ABSTRACT

Hybrid oncocytic/chromophobe tumor (HOCT) of the kidney represents a poorly understood clinicopathologic entity with pathologic features that overlap between benign renal oncocytoma (RO) and malignant chromophobe renal cell carcinoma (ChRCC). Consequently, characterization of HOCT and its separation from the foregoing entities are clinically important. The aim of this study was to describe the pathologic and molecular features of HOCT and to compare them with those of RO and ChRCC. We retrospectively identified a cohort of 73 cases with renal oncocytic tumors (19 RO, 27 HOCT, and 27 ChRCC) for whom clinical follow-up data were available by 2 tertiary care hospitals. All cases were sporadic except for 2 HOCTs that were associated with Birt-Hogg-Dubé syndrome. Lesional tissues were retrieved for molecular analysis. We performed targeted gene sequencing of all exons of 261 cancer related genes on a subset of HOCT samples (n = 16). Gene expression profiling of a customized codeset was conducted on 19 RO, 24 HOCT, and 27 ChRCC samples. Clinicopathologic characteristics as well as DNA copy number alterations, mutational and transcriptional features of HOCT derived from sequencing and expression profiling data are described and compared to those in RO and ChRCC. HOCTs were more frequently multifocal and did not exhibit mutations in genes that are recurrently mutated in RO or ChRCC but showed copy number alterations primarily involving losses in chromosomes 1 and X/Y. The mRNA transcript data show that HOCT can be separated from RO and ChRCC. Hence, HOCT appears to represent a distinct renal tumor entity with genomic features that are intermediate between those of RO and ChRCC.


Subject(s)
Adenoma, Oxyphilic/genetics , Adenoma, Oxyphilic/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Aged , Biomarkers, Tumor/analysis , Female , Humans , Male , Middle Aged , Retrospective Studies , Transcriptome
6.
Cancer Discov ; 9(5): 646-661, 2019 05.
Article in English | MEDLINE | ID: mdl-30777870

ABSTRACT

Despite recent advances in the use of immunotherapy, only a minority of patients with small cell lung cancer (SCLC) respond to immune checkpoint blockade (ICB). Here, we show that targeting the DNA damage response (DDR) proteins PARP and checkpoint kinase 1 (CHK1) significantly increased protein and surface expression of PD-L1. PARP or CHK1 inhibition remarkably potentiated the antitumor effect of PD-L1 blockade and augmented cytotoxic T-cell infiltration in multiple immunocompetent SCLC in vivo models. CD8+ T-cell depletion reversed the antitumor effect, demonstrating the role of CD8+ T cells in combined DDR-PD-L1 blockade in SCLC. We further demonstrate that DDR inhibition activated the STING/TBK1/IRF3 innate immune pathway, leading to increased levels of chemokines such as CXCL10 and CCL5 that induced activation and function of cytotoxic T lymphocytes. Knockdown of cGAS and STING successfully reversed the antitumor effect of combined inhibition of DDR and PD-L1. Our results define previously unrecognized innate immune pathway-mediated immunomodulatory functions of DDR proteins and provide a rationale for combining PARP/CHK1 inhibitors and immunotherapies in SCLC. SIGNIFICANCE: Our results define previously unrecognized immunomodulatory functions of DDR inhibitors and suggest that adding PARP or CHK1 inhibitors to ICB may enhance treatment efficacy in patients with SCLC. Furthermore, our study supports a role of innate immune STING pathway in DDR-mediated antitumor immunity in SCLC.See related commentary by Hiatt and MacPherson, p. 584.This article is highlighted in the In This Issue feature, p. 565.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA Damage , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Membrane Proteins/immunology , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/immunology , Animals , Apoptosis/drug effects , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Female , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymphocyte Activation/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyrazines/pharmacology , Pyrazoles/pharmacology , Random Allocation , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
JAMA Oncol ; 5(1): 67-73, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30267032

ABSTRACT

Importance: In recurrent human papilloma virus (HPV)-driven cancer, immune checkpoint blockade with anti-programmed cell death 1 (PD-1) antibodies produces tumor regression in only a minority of patients. Therapeutic HPV vaccines have produced strong immune responses to HPV-16, but vaccination alone has been ineffective for invasive cancer. Objective: To determine whether the efficacy of nivolumab, an anti-PD-1 immune checkpoint antibody, is amplified through treatment with ISA 101, a synthetic long-peptide HPV-16 vaccine inducing HPV-specific T cells, in patients with incurable HPV-16-positive cancer. Design, Setting, and Participants: In this single-arm, single-center phase 2 clinical trial, 24 patients with incurable HPV-16-positive cancer were enrolled from December 23, 2015, to December 12, 2016. Duration of follow-up for censored patients was 12.2 months through August 31, 2017. Interventions: The vaccine ISA101, 100 µg/peptide, was given subcutaneously on days 1, 22, and 50. Nivolumab, 3 mg/kg, was given intravenously every 2 weeks beginning day 8 for up to 1 year. Main Outcomes and Measures: Assessment of efficacy reflected in the overall response rate (per Response Evaluation Criteria in Solid Tumors, version 1.1). Results: Of the 24 patients (4 women and 20 men; 22 with oropharyngeal cancer; median age, 60 years [range, 36-73 years]), the overall response rate was 33% (8 patients; 90% CI, 19%-50%). Median duration of response was 10.3 months (95% CI, 10.3 months to inestimable). Five of 8 patients remain in response. Median progression-free survival was 2.7 months (95% CI, 2.5-9.4 months). Median overall survival was 17.5 months (95% CI, 17.5 months to inestimable). Grades 3 to 4 toxicity occurred in 2 patients (asymptomatic grade 3 transaminase level elevation in 1 patient and grade 4 lipase elevation in 1 patient), requiring discontinuation of nivolumab therapy. Conclusions and Relevance: The overall response rate of 33% and median overall survival of 17.5 months is promising compared with PD-1 inhibition alone in similar patients. A randomized clinical trial to confirm the contribution of HPV-16 vaccination to tumoricidal effects of PD-1 inhibition is warranted for further study. Trial Registration: ClinicalTrials.gov identifier: NCT02426892.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Human papillomavirus 16/drug effects , Neoplasms/drug therapy , Nivolumab/therapeutic use , Papillomavirus Infections/drug therapy , Papillomavirus Vaccines/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Antineoplastic Agents, Immunological/adverse effects , Disease Progression , Female , Human papillomavirus 16/immunology , Humans , Male , Middle Aged , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/virology , Nivolumab/adverse effects , Papillomavirus Infections/immunology , Papillomavirus Infections/mortality , Papillomavirus Infections/virology , Papillomavirus Vaccines/adverse effects , Programmed Cell Death 1 Receptor/immunology , Progression-Free Survival , Time Factors
8.
Cell Metab ; 27(5): 977-987.e4, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29628419

ABSTRACT

Adoptive T cell therapy (ACT) produces durable responses in some cancer patients; however, most tumors are refractory to ACT and the molecular mechanisms underlying resistance are unclear. Using two independent approaches, we identified tumor glycolysis as a pathway associated with immune resistance in melanoma. Glycolysis-related genes were upregulated in melanoma and lung cancer patient samples poorly infiltrated by T cells. Overexpression of glycolysis-related molecules impaired T cell killing of tumor cells, whereas inhibition of glycolysis enhanced T cell-mediated antitumor immunity in vitro and in vivo. Moreover, glycolysis-related gene expression was higher in melanoma tissues from ACT-refractory patients, and tumor cells derived from these patients exhibited higher glycolytic activity. We identified reduced levels of IRF1 and CXCL10 immunostimulatory molecules in highly glycolytic melanoma cells. Our findings demonstrate that tumor glycolysis is associated with the efficacy of ACT and identify the glycolysis pathway as a candidate target for combinatorial therapeutic intervention.


Subject(s)
Glycolysis , Immunotherapy, Adoptive , Lung Neoplasms/therapy , Melanoma/therapy , T-Lymphocytes/transplantation , Animals , Cell Line, Tumor , Chemokine CXCL10/metabolism , Female , Humans , Interferon Regulatory Factor-1/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Male , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred C57BL
9.
Oncotarget ; 9(18): 14268-14284, 2018 Mar 06.
Article in English | MEDLINE | ID: mdl-29581842

ABSTRACT

While several molecular targets have been identified for adenocarcinoma (ACA) of the lung, similar drivers with squamous cell carcinoma (SCC) are sparse. We compared signaling pathways and potential therapeutic targets in lung SCC and ACA tumors using reverse phase proteomic arrays (RPPA) from two independent cohorts of resected early stage NSCLC patients: a testing set using an MDACC cohort (N=140) and a validation set using the Cancer Genome Atlas (TCGA) cohorts. We identified multiple potentially targetable proteins upregulated in SCC, including NRF2, Keap1, PARP, TrkB, and Chk2. Of these potential targets, we found that TrkB also had significant increases in gene expression in SCC as compared to adenocarcinoma. Thus, we next validated the upregulation of TrkB both in vitro and in vivo and found that it was constitutively expressed at high levels in a subset of SCC cell lines. Furthermore, we found that TrkB inhibition suppressed tumor growth, invasiveness and sensitized SCC cells to tyrosine kinase EGFR inhibition in a cell-specific manner.

10.
Cancer Immunol Res ; 6(2): 163-177, 2018 02.
Article in English | MEDLINE | ID: mdl-29339375

ABSTRACT

Expression of the multikinase inhibitor encoded by the tumor suppressor gene TUSC2 (also known as FUS1) is lost or decreased in non-small cell lung carcinoma (NSCLC). TUSC2 delivered systemically by nanovesicles has mediated tumor regression in clinical trials. Because of the role of TUSC2 in regulating immune cells, we assessed TUSC2 efficacy on antitumor immune responses alone and in combination with anti-PD-1 in two Kras-mutant syngeneic mouse lung cancer models. TUSC2 alone significantly reduced tumor growth and prolonged survival compared with anti-PD-1. When combined, this effect was significantly enhanced, and correlated with a pronounced increases in circulating and splenic natural killer (NK) cells and CD8+ T cells, and a decrease in regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and T-cell checkpoint receptors PD-1, CTLA-4, and TIM-3. TUSC2 combined with anti-PD-1 induced tumor infiltrating more than NK and CD8+ T cells and fewer MDSCs and Tregs than each agent alone, both in subcutaneous tumor and in lung metastases. NK-cell depletion abrogated the antitumor effect and Th1-mediated immune response of this combination, indicating that NK cells mediate TUSC2/anti-PD-1 synergy. Release of IL15 and IL18 cytokines and expression of the IL15Rα chain and IL18R1 were associated with NK-cell activation by TUSC2. Immune response-related gene expression in the tumor microenvironment was altered by combination treatment. These data provide a rationale for immunogene therapy combined with immune checkpoint blockade in the treatment of NSCLC. Cancer Immunol Res; 6(2); 163-77. ©2018 AACR.


Subject(s)
Immunogenetics/methods , Killer Cells, Natural/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Suppressor Proteins/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Tumor Suppressor Proteins/genetics
11.
Oncotarget ; 8(43): 73419-73432, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-29088717

ABSTRACT

Small cell lung cancer (SCLC) is a recalcitrant cancer for which no new treatments have been approved in over 30 years. While molecular subtyping now guides treatment selection for patients with non-small cell lung cancer and other cancers, SCLC is still treated as a single disease entity. Using model-based clustering, we found two major proteomic subtypes of SCLC characterized by either high thyroid transcription factor-1 (TTF1)/low cMYC protein expression or high cMYC/low TTF1. Applying "drug target constellation" (DTECT) mapping, we further show that protein levels of TTF1 and cMYC predict response to targeted therapies including aurora kinase, Bcl2, and HSP90 inhibitors. Levels of TTF1 and DLL3 were also highly correlated in preclinical models and patient tumors. TTF1 (used in the diagnosis lung cancer) could therefore be used as a surrogate of DLL3 expression to identify patients who may respond to the DLL3 antibody-drug conjugate rovalpituzumab tesirine. These findings suggest that TTF1, cMYC or other protein markers identified here could be used to identify subgroups of SCLC patients who may respond preferentially to several emerging targeted therapies.

12.
Oncotarget ; 8(49): 86369-86383, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29156801

ABSTRACT

Human cancer cell lines are the most frequently used preclinical models in the study of cancer biology and the development of therapeutics. Although anatomically diverse, human papillomavirus (HPV)-driven cancers have a common etiology and similar mutations that overlap with but are distinct from those found in HPV-negative cancers. Building on prior studies that have characterized subsets of head and neck squamous cell carcinoma (HNSCC) and cervical squamous cell carcinoma (CESC) cell lines separately, we performed genomic, viral gene expression, and viral integration analyses on 74 cell lines that include all readily-available HPV-positive (9 HNSCC, 8 CESC) and CESC (8 HPV-positive, 2 HPV-negative) cell lines and 55 HPV-negative HNSCC cell lines. We used over 700 human tumors for comparison. Mutation patterns in the cell lines were similar to those of human tumors. We confirmed HPV viral protein and mRNA expression in the HPV-positive cell lines. We found HPV types in three CESC cell lines that are distinct from those previously reported. We found that cell lines and tumors had similar patterns of viral gene expression; there were few sites of recurrent HPV integration. As seen in tumors, HPV integration did appear to alter host gene expression in cell lines. The HPV-positive cell lines had higher levels of p16 and lower levels of Rb protein expression than did the HPV-negative lines. Although the number of HPV-positive cell lines is limited, our results suggest that these cell lines represent suitable models for studying HNSCC and CESC, both of which are common and lethal.

13.
Sci Transl Med ; 9(415)2017 Nov 08.
Article in English | MEDLINE | ID: mdl-29118262

ABSTRACT

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) resistance mediated by T790M-independent mechanisms remains a major challenge in the treatment of non-small cell lung cancer (NSCLC). We identified a targetable mechanism of EGFR inhibitor resistance whereby stress hormones activate ß2-adrenergic receptors (ß2-ARs) on NSCLC cells, which cooperatively signal with mutant EGFR, resulting in the inactivation of the tumor suppressor, liver kinase B1 (LKB1), and subsequently induce interleukin-6 (IL-6) expression. We show that stress and ß2-AR activation promote tumor growth and EGFR inhibitor resistance, which can be abrogated with ß-blockers or IL-6 inhibition. IL-6 was associated with a worse outcome in EGFR TKI-treated NSCLC patients, and ß-blocker use was associated with lower IL-6 concentrations and improved benefit from EGFR inhibitors. These findings provide evidence that chronic stress hormones promote EGFR TKI resistance via ß2-AR signaling by an LKB1/CREB (cyclic adenosine 3',5'-monophosphate response element-binding protein)/IL-6-dependent mechanism and suggest that combinations of ß-blockers with EGFR TKIs merit further investigation as a strategy to abrogate resistance.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Epinephrine/pharmacology , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Norepinephrine/pharmacology , Protein Kinase Inhibitors/therapeutic use , AMP-Activated Protein Kinase Kinases , Adrenergic beta-Antagonists/pharmacology , Afatinib , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , ErbB Receptors/metabolism , Humans , Interleukin-6/metabolism , Lung Neoplasms/pathology , Mutation/genetics , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
14.
Clin Cancer Res ; 23(20): 6239-6253, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28698200

ABSTRACT

Purpose: Drugs targeting DNA repair and cell-cycle checkpoints have emerged as promising therapies for small-cell lung cancer (SCLC). Among these, the WEE1 inhibitor AZD1775 has shown clinical activity in a subset of SCLC patients, but resistance is common. Understanding primary and acquired resistance mechanisms will be critical for developing effective WEE1 inhibitor combinations.Experimental Design: AZD1775 sensitivity in SCLC cell lines was correlated with baseline expression level of 200 total or phosphorylated proteins measured by reverse-phase protein array (RPPA) to identify predictive markers of primary resistance. We further established AZD1775 acquired resistance models to identify mechanism of acquired resistance. Combination regimens were tested to overcome primary and acquired resistance to AZD1775 in in vitro and in vivo SCLC models.Results: High-throughput proteomic profiling demonstrate that SCLC models with primary resistance to AZD1775 express high levels of AXL and phosphorylated S6 and that WEE1/AXL or WEE1/mTOR inhibitor combinations overcome resistance in vitro and in vivo Furthermore, AXL, independently and via mTOR, activates the ERK pathway, leading to recruitment and activation of another G2-checkpoint protein, CHK1. AZD1775 acquired resistance models demonstrated upregulation of AXL, pS6, and MET, and resistance was overcome with the addition of AXL (TP0903), dual-AXL/MET (cabozantinib), or mTOR (RAD001) inhibitors.Conclusions: AXL promotes resistance to WEE1 inhibition via downstream mTOR signaling and resulting activation of a parallel DNA damage repair pathway, CHK1. These findings suggest rational combinations to enhance the clinical efficacy of AZD1775, which is currently in clinical trials for SCLC and other malignancies. Clin Cancer Res; 23(20); 6239-53. ©2017 AACR.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm , Lung Neoplasms/metabolism , Nuclear Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Small Cell Lung Carcinoma/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
15.
J Cell Sci ; 130(15): 2551-2563, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28600325

ABSTRACT

Nucleosome assembly proceeds through DNA replication-coupled or replication-independent mechanisms. For skeletal myocytes, whose nuclei have permanently exited the cell cycle, replication-independent assembly is the only mode available for chromatin remodeling. For this reason, any nucleosome composition alterations accompanying transcriptional responses to physiological signals must occur through a DNA replication-independent pathway. HIRA is the histone chaperone primarily responsible for replication-independent incorporation of histone variant H3.3 across gene bodies and regulatory regions. Thus, HIRA would be expected to play an important role in epigenetically regulating myocyte gene expression. The objective of this study was to determine the consequence of eliminating HIRA from mouse skeletal myocytes. At 6 weeks of age, myofibers lacking HIRA showed no pathological abnormalities; however, genes involved in transcriptional regulation were downregulated. By 6 months of age, myofibers lacking HIRA exhibited hypertrophy, sarcolemmal perforation and oxidative damage. Genes involved in muscle growth and development were upregulated, but those associated with responses to cellular stresses were downregulated. These data suggest that elimination of HIRA produces a hypertrophic response in skeletal muscle and leaves myofibers susceptible to stress-induced degeneration.


Subject(s)
Cell Cycle Proteins/deficiency , Histone Chaperones/deficiency , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Oxidative Stress , Transcription Factors/deficiency , Animals , Hypertrophy , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Muscular Diseases/pathology
16.
Cancer Res ; 77(14): 3870-3884, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28490518

ABSTRACT

Effective targeted therapies for small-cell lung cancer (SCLC), the most aggressive form of lung cancer, remain urgently needed. Here we report evidence of preclinical efficacy evoked by targeting the overexpressed cell-cycle checkpoint kinase CHK1 in SCLC. Our studies employed RNAi-mediated attenuation or pharmacologic blockade with the novel second-generation CHK1 inhibitor prexasertib (LY2606368), currently in clinical trials. In SCLC models in vitro and in vivo, LY2606368 exhibited strong single-agent efficacy, augmented the effects of cisplatin or the PARP inhibitor olaparib, and improved the response of platinum-resistant models. Proteomic analysis identified CHK1 and MYC as top predictive biomarkers of LY2606368 sensitivity, suggesting that CHK1 inhibition may be especially effective in SCLC with MYC amplification or MYC protein overexpression. Our findings provide a preclinical proof of concept supporting the initiation of a clinical efficacy trial in patients with platinum-sensitive or platinum-resistant relapsed SCLC. Cancer Res; 77(14); 3870-84. ©2017 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Cisplatin/pharmacology , Lung Neoplasms/drug therapy , Phthalazines/pharmacology , Piperazines/pharmacology , Small Cell Lung Carcinoma/drug therapy , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Cisplatin/administration & dosage , Drug Synergism , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Phthalazines/administration & dosage , Piperazines/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Pyrazines/administration & dosage , Pyrazines/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Small Cell Lung Carcinoma/enzymology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology
17.
Oncotarget ; 8(17): 28575-28587, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28212573

ABSTRACT

Small cell lung cancer (SCLC) is one of the most aggressive forms of cancer, with a 5-year survival <7%. A major barrier to progress is the absence of predictive biomarkers for chemotherapy and novel targeted agents such as PARP inhibitors. Using a high-throughput, integrated proteomic, transcriptomic, and genomic analysis of SCLC patient-derived xenografts (PDXs) and profiled cell lines, we identified biomarkers of drug sensitivity and determined their prevalence in patient tumors. In contrast to breast and ovarian cancer, PARP inhibitor response was not associated with mutations in homologous recombination (HR) genes (e.g., BRCA1/2) or HRD scores. Instead, we found several proteomic markers that predicted PDX response, including high levels of SLFN11 and E-cadherin and low ATM. SLFN11 and E-cadherin were also significantly associated with in vitro sensitivity to cisplatin and topoisomerase1/2 inhibitors (all commonly used in SCLC). Treatment with cisplatin or PARP inhibitors downregulated SLFN11 and E-cadherin, possibly explaining the rapid development of therapeutic resistance in SCLC. Supporting their functional role, silencing SLFN11 reduced in vitro sensitivity and drug-induced DNA damage; whereas ATM knockdown or pharmacologic inhibition enhanced sensitivity. Notably, SCLC with mesenchymal phenotypes (i.e., loss of E-cadherin and high epithelial-to-mesenchymal transition (EMT) signature scores) displayed striking alterations in expression of miR200 family and key SCLC genes (e.g., NEUROD1, ASCL1, ALDH1A1, MYCL1). Thus, SLFN11, EMT, and ATM mediate therapeutic response in SCLC and warrant further clinical investigation as predictive biomarkers.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Nuclear Proteins/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Ataxia Telangiectasia Mutated Proteins/metabolism , Biomarkers , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , Drug Resistance, Neoplasm/genetics , Gene Expression , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism
18.
Cancer Lett ; 392: 71-82, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28126323

ABSTRACT

The genomic alterations identified in head and neck squamous cell carcinoma (HNSCC) tumors have not resulted in any changes in clinical care, making the development of biomarker-driven targeted therapy for HNSCC a major translational gap in knowledge. To fill this gap, we used 59 molecularly characterized HNSCC cell lines and found that mutations of AJUBA, SMAD4 and RAS predicted sensitivity and resistance to treatment with inhibitors of polo-like kinase 1 (PLK1), checkpoint kinases 1 and 2, and WEE1. Inhibition or knockdown of PLK1 led to cell-cycle arrest at the G2/M transition and apoptosis in sensitive cell lines and decreased tumor growth in an orthotopic AJUBA-mutant HNSCC mouse model. AJUBA protein expression was undetectable in most AJUBA-mutant HNSCC cell lines, and total PLK1 and Bora protein expression were decreased. Exogenous expression of wild-type AJUBA in an AJUBA-mutant cell line partially rescued the phenotype of PLK1 inhibitor-induced apoptosis and decreased PLK1 substrate inhibition, suggesting a threshold effect in which higher drug doses are required to affect PLK1 substrate inhibition. PLK1 inhibition was an effective therapy for HNSCC in vitro and in vivo. However, biomarkers to guide such therapy are lacking. We identified AJUBA, SMAD4 and RAS mutations as potential candidate biomarkers of response of HNSCC to treatment with these mitotic inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Head and Neck Neoplasms/drug therapy , LIM Domain Proteins/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/antagonists & inhibitors , Checkpoint Kinase 2/metabolism , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Genotype , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Mice, Nude , Molecular Targeted Therapy , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phenotype , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , RNA Interference , Signal Transduction/drug effects , Smad4 Protein/genetics , Squamous Cell Carcinoma of Head and Neck , Thiophenes/pharmacology , Time Factors , Transfection , Tumor Burden/drug effects , Urea/analogs & derivatives , Urea/pharmacology , Xenograft Model Antitumor Assays , ras Proteins/genetics , Polo-Like Kinase 1
19.
Oncotarget ; 7(30): 47998-48010, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27384992

ABSTRACT

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are effective against non-small cell lung cancer (NSCLC) with activating EGFR mutations, but resistance is inevitable. Mechanisms of acquired resistance include T790M mutations and epithelial-mesenchymal transition (EMT). One potential strategy for overcoming this resistance is the inhibition of polo-like kinase 1 (PLK1) based on our previous studies showing that mesenchymal NSCLC cell lines are more sensitive to PLK1 inhibition than epithelial cell lines. To determine the extent to which PLK1 inhibition overcomes EGFR TKI resistance we measured the effects of the PLK1 inhibitor volasertib alone and in combination with the EGFR inhibitor erlotinib in vitro and in vivo in EGFR mutant NSCLC cell lines with acquired resistance to erlotinib. Two erlotinib-resistant cell lines that underwent EMT had higher sensitivity to volasertib, which caused G2/M arrest and apoptosis, than their parental cells. In all NSCLC cell lines with T790M mutations, volasertib markedly reduced erlotinib resistance. All erlotinib-resistant NSCLC cell lines with T790M mutations had higher sensitivity to erlotinib plus volasertib than to erlotinib alone, and the combination treatment caused G2/M arrest and apoptosis. Compared with either agent alone, the combination treatment also caused significantly more DNA damage and greater reductions in tumor size. Our results suggest that PLK1 inhibition is clinically effective against NSCLC that becomes resistant to EGFR inhibition through EMT or the acquisition of a T790M mutation. These results uncover new functions of PLK1 inhibition in the treatment of NSCLC with acquired resistance to EGFR TKIs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease-Free Survival , Drug Resistance, Neoplasm , Drug Synergism , Erlotinib Hydrochloride/administration & dosage , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Nude , Mutation , Protein Kinase Inhibitors/administration & dosage , Pteridines/administration & dosage , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
20.
Cancer Inform ; 15: 81-9, 2016.
Article in English | MEDLINE | ID: mdl-27199546

ABSTRACT

With increasing use of publicly available gene expression data sets, the quality of the expression data is a critical issue for downstream analysis, gene signature development, and cross-validation of data sets. Thus, identifying reliable expression measurements by leveraging multiple mRNA expression platforms is an important analytical task. In this study, we propose a statistical framework for selecting reliable measurements between platforms by modeling the correlations of mRNA expression levels using a beta-mixture model. The model-based selection provides an effective and objective way to separate good probes from probes with low quality, thereby improving the efficiency and accuracy of the analysis. The proposed method can be used to compare two microarray technologies or microarray and RNA sequencing measurements. We tested the approach in two matched profiling data sets, using microarray gene expression measurements from the same samples profiled on both Affymetrix and Illumina platforms. We also applied the algorithm to mRNA expression data to compare Affymetrix microarray data with RNA sequencing measurements. The algorithm successfully identified probes/genes with reliable measurements. Removing the unreliable measurements resulted in significant improvements for gene signature development and functional annotations.

SELECTION OF CITATIONS
SEARCH DETAIL
...