Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 84(10): 1964-1979.e6, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759628

ABSTRACT

The role of the mitochondrial electron transport chain (ETC) in regulating ferroptosis is not fully elucidated. Here, we reveal that pharmacological inhibition of the ETC complex I reduces ubiquinol levels while decreasing ATP levels and activating AMP-activated protein kinase (AMPK), the two effects known for their roles in promoting and suppressing ferroptosis, respectively. Consequently, the impact of complex I inhibitors on ferroptosis induced by glutathione peroxidase 4 (GPX4) inhibition is limited. The pharmacological inhibition of complex I in LKB1-AMPK-inactivated cells, or genetic ablation of complex I (which does not trigger apparent AMPK activation), abrogates the AMPK-mediated ferroptosis-suppressive effect and sensitizes cancer cells to GPX4-inactivation-induced ferroptosis. Furthermore, complex I inhibition synergizes with radiotherapy (RT) to selectively suppress the growth of LKB1-deficient tumors by inducing ferroptosis in mouse models. Our data demonstrate a multifaceted role of complex I in regulating ferroptosis and propose a ferroptosis-inducing therapeutic strategy for LKB1-deficient cancers.


Subject(s)
AMP-Activated Protein Kinases , Electron Transport Complex I , Ferroptosis , Animals , Female , Humans , Mice , AMP-Activated Protein Kinase Kinases/genetics , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Cell Line, Tumor , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Ferroptosis/genetics , Ferroptosis/drug effects , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/drug effects , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/drug therapy , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Xenograft Model Antitumor Assays
2.
Mol Cell ; 81(8): 1698-1714.e6, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33626321

ABSTRACT

The DREAM complex orchestrates cell quiescence and the cell cycle. However, how the DREAM complex is deregulated in cancer remains elusive. Here, we report that PAF (PCLAF/KIAA0101) drives cell quiescence exit to promote lung tumorigenesis by remodeling the DREAM complex. PAF is highly expressed in lung adenocarcinoma (LUAD) and is associated with poor prognosis. Importantly, Paf knockout markedly suppressed LUAD development in mouse models. PAF depletion induced LUAD cell quiescence and growth arrest. PAF is required for the global expression of cell-cycle genes controlled by the repressive DREAM complex. Mechanistically, PAF inhibits DREAM complex formation by binding to RBBP4, a core DREAM subunit, leading to transactivation of DREAM target genes. Furthermore, pharmacological mimicking of PAF-depleted transcriptomes inhibited LUAD tumor growth. Our results unveil how the PAF-remodeled DREAM complex bypasses cell quiescence to promote lung tumorigenesis and suggest that the PAF-DREAM axis may be a therapeutic vulnerability in lung cancer.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/genetics , Kv Channel-Interacting Proteins/genetics , Lung Neoplasms/genetics , Lung/pathology , Repressor Proteins/genetics , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Carcinogenesis/pathology , Cell Division/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , NIH 3T3 Cells , Transcriptional Activation/genetics , Transcriptome/genetics
3.
Nature ; 593(7860): 586-590, 2021 05.
Article in English | MEDLINE | ID: mdl-33981038

ABSTRACT

Ferroptosis, a form of regulated cell death that is induced by excessive lipid peroxidation, is a key tumour suppression mechanism1-4. Glutathione peroxidase 4 (GPX4)5,6 and ferroptosis suppressor protein 1 (FSP1)7,8 constitute two major ferroptosis defence systems. Here we show that treatment of cancer cells with GPX4 inhibitors results in acute depletion of N-carbamoyl-L-aspartate, a pyrimidine biosynthesis intermediate, with concomitant accumulation of uridine. Supplementation with dihydroorotate or orotate-the substrate and product of dihydroorotate dehydrogenase (DHODH)-attenuates or potentiates ferroptosis induced by inhibition of GPX4, respectively, and these effects are particularly pronounced in cancer cells with low expression of GPX4 (GPX4low). Inactivation of DHODH induces extensive mitochondrial lipid peroxidation and ferroptosis in GPX4low cancer cells, and synergizes with ferroptosis inducers to induce these effects in GPX4high cancer cells. Mechanistically, DHODH operates in parallel to mitochondrial GPX4 (but independently of cytosolic GPX4 or FSP1) to inhibit ferroptosis in the mitochondrial inner membrane by reducing ubiquinone to ubiquinol (a radical-trapping antioxidant with anti-ferroptosis activity). The DHODH inhibitor brequinar selectively suppresses GPX4low tumour growth by inducing ferroptosis, whereas combined treatment with brequinar and sulfasalazine, an FDA-approved drug with ferroptosis-inducing activity, synergistically induces ferroptosis and suppresses GPX4high tumour growth. Our results identify a DHODH-mediated ferroptosis defence mechanism in mitochondria and suggest a therapeutic strategy of targeting ferroptosis in cancer treatment.


Subject(s)
Dihydroorotate Dehydrogenase/metabolism , Ferroptosis , Mitochondria/metabolism , Neoplasms/enzymology , Animals , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Dihydroorotate Dehydrogenase/genetics , Female , Gene Deletion , Humans , Lipid Peroxidation , Metabolomics , Mice, Nude , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Xenograft Model Antitumor Assays
4.
Nature ; 590(7846): 504-508, 2021 02.
Article in English | MEDLINE | ID: mdl-33536620

ABSTRACT

Amplification of chromosomal region 8p11-12 is a common genetic alteration that has been implicated in the aetiology of lung squamous cell carcinoma (LUSC)1-3. The FGFR1 gene is the main candidate driver of tumorigenesis within this region4. However, clinical trials evaluating FGFR1 inhibition as a targeted therapy have been unsuccessful5. Here we identify the histone H3 lysine 36 (H3K36) methyltransferase NSD3, the gene for which is located in the 8p11-12 amplicon, as a key regulator of LUSC tumorigenesis. In contrast to other 8p11-12 candidate LUSC drivers, increased expression of NSD3 correlated strongly with its gene amplification. Ablation of NSD3, but not of FGFR1, attenuated tumour growth and extended survival in a mouse model of LUSC. We identify an LUSC-associated variant NSD3(T1232A) that shows increased catalytic activity for dimethylation of H3K36 (H3K36me2) in vitro and in vivo. Structural dynamic analyses revealed that the T1232A substitution elicited localized mobility changes throughout the catalytic domain of NSD3 to relieve auto-inhibition and to increase accessibility of the H3 substrate. Expression of NSD3(T1232A) in vivo accelerated tumorigenesis and decreased overall survival in mouse models of LUSC. Pathological generation of H3K36me2 by NSD3(T1232A) reprograms the chromatin landscape to promote oncogenic gene expression signatures. Furthermore, NSD3, in a manner dependent on its catalytic activity, promoted transformation in human tracheobronchial cells and growth of xenografted human LUSC cell lines with amplification of 8p11-12. Depletion of NSD3 in patient-derived xenografts from primary LUSCs containing NSD3 amplification or the NSD3(T1232A)-encoding variant attenuated neoplastic growth in mice. Finally, NSD3-regulated LUSC-derived xenografts were hypersensitive to bromodomain inhibition. Thus, our work identifies NSD3 as a principal 8p11-12 amplicon-associated oncogenic driver in LUSC, and suggests that NSD3-dependency renders LUSC therapeutically vulnerable to bromodomain inhibition.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Histones/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Nuclear Proteins/metabolism , Animals , Biocatalysis , Carcinogenesis/genetics , Carcinoma, Squamous Cell/genetics , Female , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Lung Neoplasms/genetics , Male , Methylation , Mice , Models, Molecular , Mutation , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Xenograft Model Antitumor Assays
5.
Nature ; 597(7878): 732-737, 2021 09.
Article in English | MEDLINE | ID: mdl-34526717

ABSTRACT

Epidermal growth factor receptor (EGFR) mutations typically occur in exons 18-21 and are established driver mutations in non-small cell lung cancer (NSCLC)1-3. Targeted therapies are approved for patients with 'classical' mutations and a small number of other mutations4-6. However, effective therapies have not been identified for additional EGFR mutations. Furthermore, the frequency and effects of atypical EGFR mutations on drug sensitivity are unknown1,3,7-10. Here we characterize the mutational landscape in 16,715 patients with EGFR-mutant NSCLC, and establish the structure-function relationship of EGFR mutations on drug sensitivity. We found that EGFR mutations can be separated into four distinct subgroups on the basis of sensitivity and structural changes that retrospectively predict patient outcomes following treatment with EGFR inhibitors better than traditional exon-based groups. Together, these data delineate a structure-based approach for defining functional groups of EGFR mutations that can effectively guide treatment and clinical trial choices for patients with EGFR-mutant NSCLC and suggest that a structure-function-based approach may improve the prediction of drug sensitivity to targeted therapies in oncogenes with diverse mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Afatinib/therapeutic use , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Drug Repositioning , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Exons , Female , Humans , Lung Neoplasms/genetics , Mice , Molecular Docking Simulation , Mutation , Structure-Activity Relationship
6.
Bioorg Med Chem ; 58: 116673, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35189561

ABSTRACT

To identify potential new reagents and biomarkers for early lung cancer detection we combined the use of a novel preclinical isogenic model of human lung epithelial cells comparing non-malignant cells with those transformed to full malignancy using defined oncogenic changes and our on-bead two color (red and green stained cells) (OBTC) peptoid combinatorial screening methodology. The preclinical model used normal parent lung epithelial cells (HBEC3-KT, labeled with green dye) and isogenic fully malignant transformed derivatives (labeled with a red dye) via the sequential introduction of key genetic alterations of p53 knockdown, oncogenic KRAS and overexpression of cMYC (HBEC3p53, KRAS, cMYC). Using the unbiased OBTC screening approach, we tested 100,000 different peptoids and identified only one (named JM3A) that bound to the surface of the HBEC3p53, KRAS, cMYC cells (red cells) but not HBEC3-KT cells (green cells). Using the JM3A peptoid and proteomics, we identified the protein bound as vimentin using multiple validation approaches. These all confirmed the cell surface expression of vimentin (CSV) on transformed (HBEC3p53, KRAS, cMYC) but not on untransformed (HBEC3-KT) cells. JM3A coupled with fluorophores was able to detect and stain cell surface vimentin on very early stage lung cancers but not normal lung epithelial cells in a fashion comparable to that using anti-vimentin antibodies. We conclude: using a combined isogenic preclinical model of lung cancer and two color screening of a large peptoid library, we have identified differential expression of cell surface vimentin (CSV) after malignant transformation of lung epithelial cells, and developed a new peptoid reagent (JM3A) for detection of CSV which works well in staining of early stage NSCLCs. This new, highly specific, easy to prepare, CSV detecting JM3A peptoid provides an important new reagent for identifying cancer cells in early stage tumors as well as a resource for detection and isolating of CSV expressing circulating tumor cells.


Subject(s)
Epithelial Cells/metabolism , Lung Neoplasms/metabolism , Peptoids/metabolism , Vimentin/genetics , Cell Line , Humans , Lung Neoplasms/pathology , Molecular Structure , Peptoids/chemistry , Vimentin/metabolism
8.
Crit Rev Clin Lab Sci ; 57(6): 400-414, 2020 09.
Article in English | MEDLINE | ID: mdl-32662342

ABSTRACT

Since Coronavirus Disease 2019 (COVID-19) first emerged in December 2019, the disease has rapidly evolved into a pandemic that threatens societies around the world. As soon as the causative pathogen, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was identified and its genome sequence determined, a laboratory diagnosis based on nucleic acid amplification technologies was quickly established and has played essential roles in the confirmation of a clinical diagnosis. Serological testing for antibodies against SARS-CoV-2 is becoming available for complementary diagnosis, identification of convalescent plasma, and epidemiologic studies. Additional laboratory biochemical tests, including monitoring the change in blood cells, blood gas, coagulation, liver function, cardiac markers, and inflammatory responses such as cytokine levels in plasma, are also critical in combating COVID-19. Nevertheless, with overwhelming numbers of patients and potentially large numbers of asymptomatic cases, clinical laboratories encounter enormous challenges in diagnostic approaches that can rapidly and accurately identify infected persons. Strategies that can effectively detect disease progression in order to stratify patients for appropriate care, and that can thereby prevent exacerbation of the disease, are urgently needed. This review discusses the laboratory's role and challenges in combating COVID-19.


Subject(s)
Betacoronavirus/physiology , Clinical Laboratory Services , Coronavirus Infections/therapy , Pneumonia, Viral/therapy , Amino Acid Sequence , COVID-19 , Coronavirus Infections/blood , Coronavirus Infections/diagnosis , Coronavirus Infections/epidemiology , Humans , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/diagnosis , Pneumonia, Viral/epidemiology , SARS-CoV-2 , Viral Proteins/chemistry
9.
Cancer ; 125(21): 3738-3748, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31287557

ABSTRACT

BACKGROUND: Patient-derived xenograft (PDX) models increasingly are used in translational research. However, the engraftment rates of patient tumor samples in immunodeficient mice to PDX models vary greatly. METHODS: Tumor tissue samples from 308 patients with non-small cell lung cancer were implanted in immunodeficient mice. The patients were followed for 1.5 to approximately 6Ā years. The authors performed histological analysis of PDXs and some residual tumor tissues in mice with failed PDX growth at 1Ā year after implantation. Quantitative polymerase chain reaction and enzyme-linked immunoadsorbent assay were performed to measure the levels of Epstein-Barr virus genes and human immunoglobulin G in PDX samples. Patient characteristics were compared for PDX growth and overall survival as outcomes using Cox regression analyses. Disease staging was based on the 7th TNM staging system. RESULTS: The overall engraftment rate for PDXs from patients with non-small cell lung cancer was 34%. Squamous cell carcinomas had a higher engraftment rate (53%) compared with adenocarcinomas. Tumor samples from patients with stage II and stage III disease and from larger tumors were found to have relatively high engraftment rates. Patients whose tumors successfully engrafted had worse overall survival, particularly those individuals with adenocarcinoma, stage III or stage IV disease, and moderately differentiated tumors. Lymphoma formation was one of the factors associated with engraftment failure. Human CD8-positive and CD20-positive cells were detected in residual samples of tumor tissue that failed to generate a PDX at 1Ā year after implantation. Human immunoglobulin G was detected in the plasma of mice that did not have PDX growth at 14Ā months after implantation. CONCLUSIONS: The results of the current study indicate that the characteristics of cancer cells and the tumor immune microenvironment in primary tumors both can affect engraftment of a primary tumor sample.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Disease Models, Animal , Lung Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Antigens, CD20/immunology , Antigens, CD20/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Heterografts , Humans , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Staging , Xenograft Model Antitumor Assays/methods
10.
Cancer ; 124(5): 1061-1069, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29178133

ABSTRACT

BACKGROUND: Next-generation sequencing of cell-free DNA (cfDNA) has been shown to be a useful noninvasive test for detecting mutations in solid tumors. METHODS: Targeted gene sequencing was performed with a panel of 263 cancer-related genes for cfDNA and genomic DNA of peripheral blood mononuclear cells (PBMCs) obtained from presurgical specimens of 6 lung cancer patients, and mutation calls in these samples were compared with those of primary tumors and corresponding patient-derived xenografts (PDXs). RESULTS: Approximately 67% of the mutations detected in the tumor samples (primary tumors and/or PDXs) were also detected in genomic DNA from PBMCs as background mutations. These background mutations consisted of germline polymorphisms and a group of mutations with low allele frequencies, mostly <10%. These variants with a low allele frequency were repeatedly detected in all types of samples from the same patients and at similarly low allele frequency levels in PBMCs from different patients; this indicated that their detection might be derived from common causes, such as homologous sequences in the human genome. Allele frequencies of mutations detected in both primary tumors and cfDNA showed 2 patterns: 1) low allele frequencies (approximately 1%-10%) in cfDNA but high allele frequencies (usually >10% or >3-fold increase) in primary tumors and further enrichment in PDXs and 2) similar allele frequencies across samples. CONCLUSIONS: Because only a small fraction of total cfDNA might be derived from tumor cells, only mutations with the first allele frequency pattern may be regarded as tumor-specific mutations in cfDNA. Effective filtering of background mutations will be required to improve the accuracy of mutation calls in cfDNA. Cancer 2018;124:1061-9. Ā© 2017 American Cancer Society.


Subject(s)
Circulating Tumor DNA/genetics , DNA, Neoplasm/genetics , High-Throughput Nucleotide Sequencing/methods , Leukocytes, Mononuclear/metabolism , Lung Neoplasms/genetics , Mutation , Female , Gene Frequency , Genomics/methods , Humans , Lung Neoplasms/pathology , Male , Neoplasm Staging
11.
Oncologist ; 23(9): 1008-1015, 2018 09.
Article in English | MEDLINE | ID: mdl-29700208

ABSTRACT

BACKGROUND: Occurrence at a younger age has been demonstrated to be associated with a distinct biology in non-small cell lung cancer. However, genomics and clinical characteristics among younger patients with lung adenocarcinoma remain to be determined. Here we studied the potentially targetable genetic alterations by next-generation sequencing (NGS) assay in young Chinese patients with lung adenocarcinoma. MATERIALS AND METHODS: Seventy-one surgically resected lung adenocarcinoma tissue samples from patients aged less than 45 years were collected with informed consent from all patients. Targeted NGS assays were used to identify actionable genetic alterations in the cancer tissues. Additionally, the genomic and clinicopathologic characteristics of 106 patients with lung adenocarcinoma who received NGS testing over the same period were analyzed retrospectively. RESULTS: The frequencies of targetable genetic alterations in 177 patients with lung adenocarcinoma were analyzed by defined age categories, which unveiled a distinctive molecular profile in the younger group, aged less than 45 years. Notably, higher frequency of ALK and HER2 genetic alterations were associated with young age. However, a reverse trend was observed for KRAS, STK11 and EGFR exon 20 mutations, which were more frequently identified in the older group, aged more than 46 years. Furthermore, concurrent EGFR/TP53 mutations were much more prevalent in the younger patients (81.6% vs. 46.8%), which might have a poor response to treatment with epidermal growth factor receptor tyrosine kinase inhibitor. CONCLUSION: In this study, NGS assay revealed a distinctive genetic profile in younger patients with adenocarcinoma. High frequency of concurrent EGFR/TP53 mutations was found in the younger patients, which especially warranted personalized treatment in this population. IMPLICATIONS FOR PRACTICE: Further investigation is needed to understand the genomics and clinical characteristics of young patients with lung adenocarcinoma. In the present study, hybrid capture-based next-generation sequencing assays were used to identify targeted genetic alterations in young lung adenocarcinoma patients. Young patients with lung adenocarcinoma, aged less than 45 years, harbored a higher frequency of ALK and HER2 genetic alterations compared with patients aged more than 46 years. Dramatically, concurrent EGFR/TP53 mutations were much more prevalent in younger patients, which had a poor response to treatment with epidermal growth factor receptor kinase inhibitor. These results reveal a distinctive genetic profile in younger patients with adenocarcinoma, which might improve the treatment of this subpopulation.


Subject(s)
Adenocarcinoma of Lung/genetics , Genetic Profile , Lung Neoplasms/genetics , Adenocarcinoma of Lung/pathology , China , Humans , Lung Neoplasms/pathology , Middle Aged
12.
J Transl Med ; 16(1): 328, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30477533

ABSTRACT

BACKGROUND: Because patient-derived xenografts (PDXs) are grown in immunodeficient mouse strains, PDXs are regarded as lacking an immune microenvironment. However, whether patients' immune cells co-exist in PDXs remains uncharacterized. METHODS: We cultured small pieces of lung PDX tissue in media containing human interleukin-2 and characterized the proliferated lymphocytes by flow cytometric assays with antibodies specific for human immune cell surface markers. Presence of immune cells in PDXs was also determined by immunohistochemical staining. RESULTS: Human tumor-infiltrating lymphocytes (TILs) were cultured from nine of 25 PDX samples (36%). The mean time of PDX growth in immunodeficient mice before obtaining TILs in culture was 113Ā days (range 63-292Ā days). The TILs detected in PDXs were predominantly human CD8+ T cells, CD4+ T cells, or CD19+ B cells, depending on cases. DNA fingerprint analysis showed that the TILs originated from the same patients as the PDXs. Further analysis of two PDX-derived CD8+ T cells showed that they were PD-1-, CD45RO+, and either CD62L+ or CD62L-, suggesting they were likely memory T cells. Immunohistochemical staining showed that human T cells (CD8+ or CD4+), B cells (CD19+), and macrophages (CD68+) were present in stroma or intraepithelial cancer structures and that human PD-L1 was expressed in stromal cells. Moreover, the patient-derived immune cells in PDX can be passaged to the F2 generation and may migrate to spleens of PDX-bearing mice. CONCLUSIONS: Patient-derived immune cells co-exist in early passages of PDXs in some lung cancer PDX models. The CD8+ cells from PDXs were likely memory T cells. These results suggest that PDXs can be used for evaluating the functionality of immune components in tumor microenvironments.


Subject(s)
Lung Neoplasms/immunology , Lung Neoplasms/pathology , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Humans , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/immunology , Stromal Cells/pathology
13.
BMC Cancer ; 18(1): 1082, 2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30409126

ABSTRACT

Recent breakthroughs in targeted therapy and immunotherapy have revolutionized the treatment of lung cancer, the leading cause of cancer-related deaths in the United States and worldwide. Here we provide an overview of recent progress in immune checkpoint blockade therapy for treatment of non-small cell lung cancer (NSCLC), and discuss biomarkers associated with the treatment responses, mechanisms underlying resistance and strategies to overcome resistance. The success of immune checkpoint blockade therapies is driven by immunogenicity of tumor cells, which is associated with mutation burden and neoantigen burden in cancers. Lymphocyte infiltration in cancer tissues and interferon-ƎĀ³-induced PD-L1 expression in tumor microenvironments may serve as surrogate biomarkers for adaptive immune resistance and likelihood of responses to immune checkpoint blockade therapy. In contrast, weak immunogenicity of, and/or impaired antigen presentation in, tumor cells are primary causes of resistance to these therapies. Thus, approaches that increase immunogenicity of cancer cells and/or enhance immune cell recruitment to cancer sites will likely overcome resistance to immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Clinical Trials as Topic , Humans , Immunotherapy/methods , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Treatment Outcome
14.
Blood ; 125(19): 2968-73, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25712988

ABSTRACT

Tyrosine kinase inhibitors (TKIs) are used as a frontline therapy for BCR-ABL(+) acute lymphoblastic leukemia (ALL). However, resistance to TKI therapy arises rapidly, and its underlying molecular mechanisms are poorly understood. In this study, we identified a novel cascade of events initiated by TKIs and traversing through mesenchymal stem cells (MSCs) to leukemic cells, leading to resistance. MSCs exposed to TKIs acquired a new functional status with the expression of genes encoding for chemo-attractants, adhesion molecules, and prosurvival growth factors, and this priming enabled leukemic cells to form clusters underneath the MSCs. This cluster formation was associated with the protection of ALL cells from therapy as leukemic cells switched from BCR-ABL signaling to IL-7R/Janus kinase signaling to survive in the MSC milieu. Our findings illustrate a novel perspective in the evolution of TKI resistance and provide insights for advancing the treatment of BCR-ABL(+) ALL.


Subject(s)
Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/metabolism , Mesenchymal Stem Cells/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/drug effects , Fusion Proteins, bcr-abl/genetics , Gene Expression Profiling , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured
15.
Mol Cell ; 36(4): 547-59, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19941816

ABSTRACT

Increased transcriptional activity of beta-catenin resulting from Wnt/Wingless-dependent or -independent signaling has been detected in many types of human cancer, but the underlying mechanism of Wnt-independent regulation remains unclear. We demonstrate here that EGFR activation results in disruption of the complex of beta-catenin and alpha-catenin, thereby abrogating the inhibitory effect of alpha-catenin on beta-catenin transactivation via CK2alpha-dependent phosphorylation of alpha-catenin at S641. ERK2, which is activated by EGFR signaling, directly binds to CK2alpha via the ERK2 docking groove and phosphorylates CK2alpha primarily at T360/S362, subsequently enhancing CK2alpha activity toward alpha-catenin phosphorylation. In addition, levels of alpha-catenin S641 phosphorylation correlate with levels of ERK1/2 activity in human glioblastoma specimens and with grades of glioma malignancy. This EGFR-ERK-CK2-mediated phosphorylation of alpha-catenin promotes beta-catenin transactivation and tumor cell invasion. These findings highlight the importance of the crosstalk between EGFR and Wnt pathways in tumor development.


Subject(s)
Casein Kinase II/metabolism , Epidermal Growth Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Transcriptional Activation/drug effects , alpha Catenin/metabolism , beta Catenin/genetics , Amino Acid Sequence , Binding Sites , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Molecular Sequence Data , Neoplasm Invasiveness , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , alpha Catenin/chemistry , beta Catenin/metabolism
16.
Clin Lab ; 63(7): 1251-1259, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28792692

ABSTRACT

BACKGROUND: Identification of biomarkers of pancreatic neuroendocrine tumors (PNETs) is important for stratification of the prognosis. Ki-67 index was the significant prognostic factor for PNETs. Recently, MMP-9, DJ-1, and α-1-B glycoprotein (A1BG) were shown to be the prognostic markers in some malignant tumors except for PNETs. The aim of this study was to compare these tumor markers for predicting disease-free survival in surgically resected PNETs. METHODS: A retrospective review of patients pathologically diagnosed with PNETs at our institution from January 2012 to January 2014 was conducted. Tumor specimens were stained by immunochemistry for MMP-9, DJ-1, A1BG, and Ki-67. Clinicopathologic findings and these 4 tumor markers of patients with PNETs were analyzed. Prognostic factors were determined by univariate and multivariate analyses. RESULTS: A total of 40 patients were selected. Of 40 tumors, 19 (47.5%) were positive for MMP-9, 21 (52.5%) for DJ-1, 18 (45%) for A1BG, and 24 (60%) for Ki-67 > 2%. Four (10%) tumors expressed none of the markers. MMP-9 and DJ-1 expression were more frequently observed in distant metastasis, higher WHO grade, and lymph node metastasis. Similarly, those with Ki-67 > 2% had larger tumor, higher WHO grade, lymph node invasion and larger surgical procedures. A1BG had no substantial effect on distant metastasis. Univariate analyses revealed that grade 2/3 histology, distant metastasis, lymph nodes metastasis, high expression of MMP-9, high expression of DJ-1, and Ki-67 > 2% were predictive of inferior disease-free survival for PNETs (all p < 0.05). In multivariable analysis, the significant poor factors associated with disease-free survival were grade 2/3 histology, distant metastasis, lymph node metastasis, and Ki-67 > 2% (all p < 0.05). CONCLUSIONS: Of the 4 markers studied, only Ki-67 was the independent prognostic factor. Expression of MMP-9 and DJ-1 correlated with invasion, metastasis, disease progression, and poor prognosis in PNETs, while A1BG had no prognostic value in PNETs. It is anticipated that these findings can serve as useful clinical survival predictors, especially in the setting of resected disease.


Subject(s)
Biomarkers, Tumor/analysis , Neuroendocrine Tumors/surgery , Pancreatic Neoplasms/surgery , Disease-Free Survival , Humans , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Prognosis , Retrospective Studies
17.
N Engl J Med ; 369(6): 507-16, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23782157

ABSTRACT

BACKGROUND: Bruton's tyrosine kinase (BTK) is a mediator of the B-cell-receptor signaling pathway implicated in the pathogenesis of B-cell cancers. In a phase 1 study, ibrutinib, a BTK inhibitor, showed antitumor activity in several types of non-Hodgkin's lymphoma, including mantle-cell lymphoma. METHODS: In this phase 2 study, we investigated oral ibrutinib, at a daily dose of 560 mg, in 111 patients with relapsed or refractory mantle-cell lymphoma. Patients were enrolled into two groups: those who had previously received at least 2 cycles of bortezomib therapy and those who had received less than 2 complete cycles of bortezomib or had received no prior bortezomib therapy. The primary end point was the overall response rate. Secondary end points were duration of response, progression-free survival, overall survival, and safety. RESULTS: The median age was 68 years, and 86% of patients had intermediate-risk or high-risk mantle-cell lymphoma according to clinical prognostic factors. Patients had received a median of three prior therapies. The most common treatment-related adverse events were mild or moderate diarrhea, fatigue, and nausea. Grade 3 or higher hematologic events were infrequent and included neutropenia (in 16% of patients), thrombocytopenia (in 11%), and anemia (in 10%). A response rate of 68% (75 patients) was observed, with a complete response rate of 21% and a partial response rate of 47%; prior treatment with bortezomib had no effect on the response rate. With an estimated median follow-up of 15.3 months, the estimated median response duration was 17.5 months (95% confidence interval [CI], 15.8 to not reached), the estimated median progression-free survival was 13.9 months (95% CI, 7.0 to not reached), and the median overall survival was not reached. The estimated rate of overall survival was 58% at 18 months. CONCLUSIONS: Ibrutinib shows durable single-agent efficacy in relapsed or refractory mantle-cell lymphoma. (Funded by Pharmacyclics and others; ClinicalTrials.gov number, NCT01236391.)


Subject(s)
Lymphoma, Mantle-Cell/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Adenine/analogs & derivatives , Administration, Oral , Adult , Agammaglobulinaemia Tyrosine Kinase , Aged , Aged, 80 and over , Drug Resistance, Neoplasm , Female , Humans , Lymphocyte Count , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Piperidines , Protein Kinase Inhibitors/adverse effects , Pyrazoles/adverse effects , Pyrimidines/adverse effects , Recurrence , Survival Analysis
18.
Acta Biochim Biophys Sin (Shanghai) ; 48(1): 27-38, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26350096

ABSTRACT

Activating mutations of oncogenic RAS genes are frequently detected in human cancers. The studies in genetically engineered mouse models (GEMMs) reveal that Kras-activating mutations predispose mice to early onset tumors in the lung, pancreas, and gastrointestinal tract. Nevertheless, most of these tumors do not have metastatic phenotypes. Metastasis occurs when tumors acquire additional genetic changes in other cancer driver genes. Studies on clinical specimens also demonstrated that KRAS mutations are present in premalignant tissues and that most of KRAS mutant human cancers have co-mutations in other cancer driver genes, including TP53, STK11, CDKN2A, and KMT2C in lung cancer; APC, TP53, and PIK3CA in colon cancer; and TP53, CDKN2A, SMAD4, and MED12 in pancreatic cancer. Extensive efforts have been devoted to develop therapeutic agents that target enzymes involved in RAS posttranslational modifications, that inhibit downstream effectors of RAS signaling pathways, and that kill RAS mutant cancer cells through synthetic lethality. Recent clinical studies have revealed that sorafenib, a pan-RAF and VEGFR inhibitor, has impressive benefits for KRAS mutant lung cancer patients. Combination therapy of MEK inhibitors with either docetaxel, AKT inhibitors, or PI3K inhibitors also led to improved clinical responses in some KRAS mutant cancer patients. This review discusses knowledge gained from GEMMs, human cancer cells, and patient-related studies on RAS-mediated tumorigenesis and anti-RAS therapy. Emerging evidence demonstrates that RAS mutant cancers are heterogeneous because of the presence of different mutant alleles and/or co-mutations in other cancer driver genes. Effective subclassifications of RAS mutant cancers may be necessary to improve patients' outcomes through personalized precision medicine.


Subject(s)
Genetic Engineering/methods , Neoplasms/genetics , Neoplasms/metabolism , ras Proteins/metabolism , Adenocarcinoma/genetics , Alleles , Animals , Cell Line, Tumor , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic , Humans , Mice , Mutation , Neoplasm Metastasis , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Precision Medicine , Signal Transduction
19.
Chin J Cancer ; 34(7): 295-309, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26134262

ABSTRACT

The molecular characterization of various cancers has shown that cancers with the same origins, histopathologic diagnoses, and clinical stages can be highly heterogeneous in their genetic and epigenetic alterations that cause tumorigenesis. A number of cancer driver genes with functional abnormalities that trigger malignant transformation and that are required for the survival of cancer cells have been identified. Therapeutic agents targeting some of these cancer drivers have been successfully developed, resulting in substantial improvements in clinical symptom amelioration and outcomes in a subset of cancer patients. However, because such therapeutic drugs often benefit only a limited number of patients, the successes of clinical development and applications rely on the ability to identify those patients who are sensitive to the targeted therapies. Thus, biomarkers that can predict treatment responses are critical for the success of precision therapy for cancer patients and of anticancer drug development. This review discusses the molecular heterogeneity of lung cancer pathogenesis; predictive biomarkers for precision medicine in lung cancer therapy with drugs targeting epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 receptor tyrosine kinase (ROS1), and immune checkpoints; biomarkers associated with resistance to these therapeutics; and approaches to identify predictive biomarkers in anticancer drug development. The identification of predictive biomarkers during anticancer drug development is expected to greatly facilitate such development because it will increase the chance of success or reduce the attrition rate. Additionally, such identification will accelerate the drug approval process by providing effective patient stratification strategies in clinical trials to reduce the sample size required to demonstrate clinical benefits.


Subject(s)
Biomarkers, Tumor , Lung Neoplasms , Pharmacogenetics , Precision Medicine , Anaplastic Lymphoma Kinase , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm , ErbB Receptors , Genes, erbB-1 , Humans , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases
20.
Toxicol Appl Pharmacol ; 281(3): 303-9, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25448049

ABSTRACT

NSC-743380 (1-[(3-chlorophenyl)-methyl]-1H-indole-3-carbinol) is in early stages of development as an anticancer agent. Two metabolites reflect sequential conversion of the carbinol functionality to a carboxaldehyde and the major metabolite, 1-[(3-chlorophenyl)-methyl]-1H-indole-3-carboxylic acid. In an exploratory toxicity study in rats, NSC-743380 induced elevations in liver-associated serum enzymes and biliary hyperplasia. Biliary hyperplasia was observed 2 days after dosing orally for 2 consecutive days at 100mg/kg/day. Notably, hepatotoxicity and biliary hyperplasia were observed after oral administration of the parent compound, but not when major metabolites were administered. The toxicities of a structurally similar but pharmacologically inactive molecule and a structurally diverse molecule with a similar efficacy profile in killing cancer cells in vitro were compared to NSC-743380 to explore scaffold versus target-mediated toxicity. Following two oral doses of 100mg/kg/day given once daily on two consecutive days, the structurally unrelated active compound produced hepatic toxicity similar to NSC-743380. The structurally similar inactive compound did not, but, lower exposures were achieved. The weight of evidence implies that the hepatotoxicity associated with NSC-743380 is related to the anticancer activity of the parent molecule. Furthermore, because biliary hyperplasia represents an unmanageable and non-monitorable adverse effect in clinical settings, this model may provide an opportunity for investigators to use a short-duration study design to explore biomarkers of biliary hyperplasia.


Subject(s)
Acute Disease , Biliary Tract Diseases/chemically induced , Biliary Tract/drug effects , Indoles/adverse effects , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Biliary Tract/metabolism , Biliary Tract/pathology , Biliary Tract Diseases/blood , Biliary Tract Diseases/metabolism , Biliary Tract Diseases/pathology , Biomarkers/blood , Biotransformation , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacokinetics , Hyperplasia , Indoles/administration & dosage , Indoles/blood , Indoles/metabolism , Indoles/pharmacokinetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver/physiopathology , Male , Random Allocation , Rats, Inbred F344 , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL