Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Am Chem Soc ; 144(17): 7634-7645, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35438987

ABSTRACT

Artificial antigen-presenting cells (aAPCs) constructed by integrating T cell activation ligands on biocompatible materials hold great potential in tumor immunotherapy. However, it remains challenging to develop aAPCs, which could mimic the characteristics of natural APCs, thereby realizing antigen-specific T cells activation in vivo. Here, we report the first effort to construct natural lymphocyte-based homologous targeting aAPCs (LC-aAPCs) with lipid-DNA-mediated noninvasive live cell surface engineering. Through a predesigned bottom-up self-assembly path, we achieved natural-APC-mimicking distribution of T cell activation ligands on LC-aAPCs, which would enable the optimized T cell activation. Moreover, the lipid-DNA-mediated self-assembly occurring on lipid bilayers would not affect the functions of homing receptors expressed on lymphocyte. Therefore, such LC-aAPCs could actively migrate to peripheral lymphatic organs and then effectively activate antigen-specific T cells. Combined with an immune checkpoint inhibitor, such LC-aAPCs could effectively inhibit the growth of different tumor models. Thus, our work provides a new design of aAPCs for in vivo applications in tumor immunotherapy, and the lipid-DNA-mediated noninvasive live cell surface engineering would be a powerful tool for designing cell-based therapeutics.


Subject(s)
Artificial Cells , Neoplasms , Antigen-Presenting Cells , DNA/metabolism , Humans , Immunotherapy , Ligands , Lipids , Neoplasms/metabolism , Neoplasms/therapy , T-Lymphocytes
2.
Gastric Cancer ; 19(1): 116-27, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25618371

ABSTRACT

BACKGROUND: Gastric cancer (GC) is an aggressive malignancy whose mechanisms of development and progression are poorly understood. The identification of prognosis-related genomic loci and genes may suffer from the relatively small case numbers and a lack of systematic validation in previous studies. METHODS: Array-based comparative genomic hybridization (aCGH) coupled with patient clinical information was applied to identify prognosis-related loci and genes with high-frequency recurrent gains in 129 GC patients. The candidate loci and genes were then validated using an independent cohort of 384 patients through branched DNA signal amplification analysis (QuantiGene assays). RESULTS: In the 129 patients, a copy number gain of three chromosome regions-namely, 8q22 (including ESRP1 and CCNE2), 8q24 (including MYC and TNFRSF11B), and 20q11-q13 (including SRC, MMP9, and CSE1L)--conferred poor survival for patients. In addition, the correlation between the branched DNA signal amplification analysis results and the aCGH results was analyzed in 73 of these 129 patients, and MYC, TNFRSF11B, ESRP1, CSE1L, and MMP9 were found to be well correlated. Further validation using an independent cohort (n = 384) verified that only MYC and TNFRSF11B within 8q24 are related to survival. Patients with gains in both MYC and TNFRSF11B had poorer survival than those with no gains, particularly those with noncardia GC. Gains in both of these genes were also a significant independent prognostic indicator. CONCLUSIONS: Our results revealed that copy number gains in MYC and TNFRSF11B located at 8q24 are associated with survival in GC, particularly noncardia GC.


Subject(s)
Chromosomes, Human, Pair 8 , Genes, myc , Osteoprotegerin/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Adult , Aged , Aged, 80 and over , Cohort Studies , Comparative Genomic Hybridization/methods , Female , Gene Amplification , Gene Dosage , Humans , Male , Middle Aged , Prognosis , Stomach Neoplasms/pathology , Survival Analysis
3.
Genes Chromosomes Cancer ; 54(3): 168-76, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25407564

ABSTRACT

To recommend a reliable and clinically realistic RET/PTC rearrangement detection assay for papillary thyroid carcinoma (PTC), we compared multiplex quantitative polymerase chain reaction (qPCR), fluorescence in situ hybridization (FISH), and immunohistochemistry (IHC). RET/PTC rearrangement was detected using either RET break-apart FISH followed by multicolor FISH to confirm CCDC6/RET or NCOA4/RET fusions, or by multiplex qPCR to detect 14 RET/PTC subtypes with simultaneous RET mRNA expression. RET protein expression was detected by IHC. The specificity and sensitivity of multiplex qPCR and IHC were calculated using break-apart FISH as a reference. Among 73 PTC patients with sufficient tissue available for FISH and multiplex qPCR, 10 cases were defined as RET/PTC positive by both assays, including eight CCDC6/RET and two NCOA4/RET fusions with relatively high RET mRNA. In addition, multiplex qPCR identified another two CCDC6/RET fusion positive cases, but with low RET mRNA expression. IHC staining identified 11 RET positive cases among 39 patients with available samples. In comparison to FISH, multiplex qPCR displayed 100% sensitivity and 97% specificity to detect RET/PTC fusions, while IHC was neither sensitive nor specific. Our data reveal that both multiplex qPCR and FISH assays are equally applicable for detection of RET/PTC rearrangements. Break-apart FISH methodology is highly recommended for the wider screening of RET rearrangements (regardless of partner genes), while multiplex qPCR is preferred to identify all known fusion types using one assay, provided mRNA expression is also measured. IHC analysis could potentially provide an additional method of fusion detection dependent on further optimization of assay conditions and scoring cutoffs.


Subject(s)
Carcinoma/genetics , In Situ Hybridization, Fluorescence , Proto-Oncogene Proteins c-ret/genetics , Real-Time Polymerase Chain Reaction , Thyroid Neoplasms/genetics , Translocation, Genetic , Carcinoma, Papillary , Humans , Immunohistochemistry , Thyroid Cancer, Papillary
4.
J Transl Med ; 13: 116, 2015 Apr 11.
Article in English | MEDLINE | ID: mdl-25889497

ABSTRACT

BACKGROUND: Genetic amplification of HER2 drives tumorigenesis and cancer progression in a subset of patients with gastric cancer (GC), and treatment with trastuzumab, a humanized HER2-neutralizing antibody, improves the overall survival rate of HER2-positive patients. However, a considerable portion of the patients does not respond to trastuzumab and the molecular mechanisms underlying the intrinsic resistance to anti-HER2 therapy in GC is not fully understood. METHODS: We performed whole-transcriptome sequencing on 21 HER2-positive tumor specimens from Chinese GC patients. Whole genome sequencing was performed on the three samples with HER2 fusion to discover the DNA integration structure. A multicolor FISH assay for HER2 split screening was conducted to confirm HER2 fusion and IHC (HercepTest™) was used to detect the membranous expression of HER2. Fusion cDNA were transfected into NIH/3T3 cells and generate stable cell line by lentivirus. The expression of exogenous HER2 fusion proteins and pHER2 were examined by western blot analysis. In vitro efficacy studies were also conducted by PD assay and softagar assay in cell line expression wild type and fusion HER2. T-DM1 was used to assess its binding to NIH/3T3 cells ectopically expressing wild-type and fusion HER2. Finally, the anti-tumor efficacy of trastuzumab was tested in NIH/3 T3 xenografts expressing the HER2 fusion variants. RESULTS: We identified three new HER2 fusions with ZNF207, MDK, or NOS2 in 21 HER2-amplified GC samples (14%; 3/21). Two of the fusions, ZNF207-HER2, and MDK-HER2, which are oncogenic, lead to aberrant activation of HER2 kinase. Treatment with trastuzumab inhibited tumor growth significantly in xenografts expressing MDK-HER2 fusion. In contrast, trastuzumab had no effect on the growth of xenografts expressing ZNF207-HER2 fusion, due to its inability to bind to trastuzumab. CONCLUSIONS: Our results provide the molecular basis of a novel resistance mechanism to trastuzumab-based anti-HER2 therapy, supporting additional molecule stratification within HER2-positive GC patients for more effective therapy options.


Subject(s)
Genes, erbB-2 , Oncogenes , Stomach Neoplasms/genetics , Animals , Base Sequence , DNA Primers , Gene Fusion , Humans , In Situ Hybridization, Fluorescence , Mice , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction
5.
BMC Cancer ; 15: 454, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26040563

ABSTRACT

BACKGROUND: MAPK7/ERK5 (extracellular-signal-regulated kinase 5) functions within a canonical three-tiered MAPK (mitogen activated protein kinase) signaling cascade comprising MEK (MAPK/ERK kinase) 5, MEKK(MEK kinase) 2/3 and ERK5 itself. Despite being the least well studied of the MAPK-modules, evidence supports a role for MAPK7-signaling in the pathology of several cancer types. METHODS AND RESULTS: Fluorescence in situ hybridization (FISH) analysis identified MAPK7 gene amplification in 4% (3/74) of non-small cell lung cancers (NSCLC) (enriched to 6% (3/49) in squamous cell carcinoma) and 2% (2/95) of squamous esophageal cancers (sqEC). Immunohistochemical (IHC) analysis revealed a good correlation between MAPK7 gene amplification and protein expression. MAPK7 was validated as a proliferative oncogenic driver by performing in vitro siRNA knockdown of MAPK7 in tumor cell lines. Finally, a novel MEK5/MAPK7 co-transfected HEK293 cell line was developed and used for routine cell-based pharmacodynamic screening. Phosphorylation antibody microarray analysis also identified novel downstream pharmacodynamic (PD) biomarkers of MAPK7 kinase inhibition in tumor cells (pMEF2A and pMEF2D). CONCLUSIONS: Together, these data highlight a broader role for dysregulated MAPK7 in driving tumorigenesis within niche populations of highly prevalent tumor types, and describe current efforts in establishing a robust drug discovery screening cascade.


Subject(s)
Carcinoma, Squamous Cell/genetics , Drug Screening Assays, Antitumor , Esophageal Neoplasms/genetics , Lung Neoplasms/genetics , Mitogen-Activated Protein Kinase 7/genetics , Protein Kinase Inhibitors/pharmacology , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/chemistry , Cell Proliferation/genetics , Esophageal Neoplasms/chemistry , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Lung Neoplasms/chemistry , MEF2 Transcription Factors/metabolism , Mitogen-Activated Protein Kinase 7/analysis , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Phosphorylation , Signal Transduction
6.
BMC Cancer ; 15: 171, 2015 Mar 23.
Article in English | MEDLINE | ID: mdl-25881079

ABSTRACT

BACKGROUND: To determine the prevalence of RET rearrangement genes, RET copy number gains and expression in tumor samples from four Phase III non-small-cell lung cancer (NSCLC) trials of vandetanib, a selective inhibitor of VEGFR, RET and EGFR signaling, and to determine any association with outcome to vandetanib treatment. METHODS: Archival tumor samples from the ZODIAC ( NCT00312377 , vandetanib ± docetaxel), ZEAL ( NCT00418886 , vandetanib ± pemetrexed), ZEPHYR ( NCT00404924 , vandetanib vs placebo) and ZEST ( NCT00364351 , vandetanib vs erlotinib) studies were evaluated by fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) in 944 and 1102 patients. RESULTS: The prevalence of RET rearrangements by FISH was 0.7% (95% CI 0.3-1.5%) among patients with a known result. Seven tumor samples were positive for RET rearrangements (vandetanib, n = 3; comparator, n = 4). 2.8% (n = 26) of samples had RET amplification (innumerable RET clusters, or ≥7 copies in > 10% of tumor cells), 8.1% (n = 76) had low RET gene copy number gain (4-6 copies in ≥40% of tumor cells) and 8.3% (n = 92) were RET expression positive (signal intensity ++ or +++ in >10% of tumor cells). Of RET-rearrangement-positive patients, none had an objective response in the vandetanib arm and one patient responded in the comparator arm. Radiologic evidence of tumor shrinkage was observed in two patients treated with vandetanib and one treated with comparator drug. The objective response rate was similar in the vandetanib and comparator arms for patients positive for RET copy number gains or RET protein expression. CONCLUSIONS: We have identified prevalence for three RET biomarkers in a population predominated by non-Asians and smokers. RET rearrangement prevalence was lower than previously reported. We found no evidence of a differential benefit for efficacy by IHC and RET gene copy number gains. The low prevalence of RET rearrangements (0.7%) prevents firm conclusions regarding association of vandetanib treatment with efficacy in the RET rearrangement NSCLC subpopulation. TRIAL REGISTRATION: Randomized Phase III clinical trials ( NCT00312377 , ZODIAC; NCT00418886 , ZEAL; NCT00364351 , ZEST; NCT00404924 , ZEPHYR).


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Piperidines/therapeutic use , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Quinazolines/therapeutic use , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Female , Gene Amplification , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Prevalence , Retrospective Studies , Translocation, Genetic , Treatment Outcome
7.
Genes Chromosomes Cancer ; 53(11): 883-94, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24935174

ABSTRACT

Gastric cancer is the second leading cause of death from cancer worldwide, with an approximately 20% 5-year survival rate. To identify molecular subtypes associated with the clinical prognosis, in addition to genetic aberrations for potential targeted therapeutics, we conducted a comprehensive whole-genome analysis of 131 Chinese gastric cancer tissue specimens using whole-genome array comparative genomic hybridization. The analyses revealed gene focal amplifications, including CTSB, PRKCI, PAK1, STARD13, KRAS, and ABCC4, in addition to ERBB2, FGFR2, and MET. The growth of PAK1-amplified gastric cancer cells in vitro and in vivo was inhibited when the corresponding mRNA was knocked down. Furthermore, both KRAS amplification and KRAS mutation were identified in the gastric cancer specimens. KRAS amplification was associated with worse clinical outcomes, and the KRAS gene mutation predicted sensitivity to the MEK1/2 inhibitor AZD6244 in gastric cancer cell lines. In summary, amplified PAK1, as well as KRAS amplification/mutation, may represent unique opportunities for developing targeted therapeutics for the treatment of gastric cancer.


Subject(s)
Gene Dosage , Genome, Human , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics , p21-Activated Kinases/genetics , ras Proteins/genetics , Benzimidazoles/pharmacology , Chromosomal Instability , Cohort Studies , Female , Gene Amplification , Gene Expression Profiling , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Male , Middle Aged , Mutation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Rate , p21-Activated Kinases/metabolism , ras Proteins/metabolism
8.
Mol Cancer ; 13: 43, 2014 Mar 02.
Article in English | MEDLINE | ID: mdl-24580730

ABSTRACT

BACKGROUND: Tumor-associated macrophages (TAMs) are the major component of tumor-infiltrating leukocytes. TAMs are heterogeneous, with distinct phenotypes influenced by the microenvironment surrounding tumor tissues, but relatively little is known about the key molecular in these cells that contribute to malignant phenotypes. Autophagic activity is a critical factor in tumor development that contributes to enhancing cellular fitness and survival in the hostile tumor microenvironment. However, the molecular basis and relations between autophagy and TAMs polarization remain unclear. METHODS: Cathepsin S (Cat S) expression was analyzed in human colon carcinoma and normal colon tissues. In vivo effects were evaluated using PancO2 subcutaneous tumor model and SL4 hepatic metastasis model. Immunofluorescence staining, flow cytometry and real-time PCR were done to examine TAMs polarization. Western blotting assay, transmission electron microscopy, mCherry-GFP-LC3 transfection and DQ-BSA degradation assays were carried out to determine its role in regulating autophagy. RESULTS: In the present study, we showed that the enhanced expression of Cat S correlated with the severity of histologic grade as well as clinical stage, metastasis, and recurrence, which are known indicators of a relatively poor prognosis of human colon carcinoma. Cat S knockout led to decreased tumor growth and metastasis. Moreover, Cat S knockout inhibited M2 macrophage polarization during tumor development. We further demonstrated that Cat S was required for not only autophagic flux but also the fusion processes of autophagosomes and lysosomes in TAMs. Importantly, we found that Cat S contributed to tumor development by regulating the M2 phenotype of TAMs through the activation of autophagy. CONCLUSIONS: These results indicated that Cat S-mediated autophagic flux is an important mechanism for inducing M2-type polarization of TAMs, which leads to tumor development. These data provide strong evidence for a tumor-promoting role of autophagy in TAMs and suggest Cat S could be a potential target for cancer therapy.


Subject(s)
Autophagy , Cathepsins/metabolism , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Macrophages/immunology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Blotting, Western , Cell Polarity , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Real-Time Polymerase Chain Reaction , Tumor Microenvironment/immunology
9.
Tumour Biol ; 35(3): 2437-44, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24197981

ABSTRACT

Brain metastasis (BM) is a leading cause of death in patients with non-small cell lung cancer (NSCLC). EGFR mutations in primary NSCLC lesions have been associated with sensitivity to EGFR tyrosine kinase inhibitor (TKI). Therefore, it has become important to understand EGFR mutation status in BM lesions of NSCLC, and its clinical implications. BM samples of 136 NSCLC patients from South China, in which 15 had paired primary lung tumors, were retrospectively analyzed for EGFR mutation by amplification mutation refractory system (ARMS). Effect of BM EGFR mutations on progression-free survival (PFS) and overall survival (OS) was evaluated by Kaplan-Meier curves and log-rank test. EGFR mutations were detected in 52.9% (72 of 136) of the BM lesions, with preference in female and never-smokers. A concordance rate of 93.3% (14 of 15) was found between the primary NSCLC and corresponding BM. Positive prediction value of testing primary NSCLCs for BM EGFR mutation is 100.0 %, and negative prediction value is 87.5%. Median PFS of BM surgery was 12 and 10 months (P = 0.594) in the wild-type and mutant group, respectively. Median OS of BM surgery was 24.5 and 15 months (P = 0.248) in the wild-type and mutant group, respectively. In conclusion, EGFR mutation status is highly concordant between the primary NSCLC and corresponding BM. The primary NSCLC could be used as surrogate samples to predict EGFR mutation status in BM lesions or vice versa. Moreover, EGFR mutations showed no significant effect on PFS or OS of NSCLCs with BM.


Subject(s)
Asian People/genetics , Brain Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Adult , Aged , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , DNA Mutational Analysis , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction
10.
J Transl Med ; 11: 241, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24088382

ABSTRACT

INTRODUCTION: Activation of the PI3K/AKT pathway is a common phenomenon in cancer due to multiple mechanisms, including mutation of PI3KCA, loss or mutation of PTEN, or over-expression of receptor tyrosine kinases. We recently developed a novel AKT kinase inhibitor, AZD5363, and demonstrated that HGC27, a cell line harboring both PI3KCA mutation and PTEN loss, displayed the greatest sensitivity to this AKT inhibitor in vitro and in vivo. CASE PREPARATION: To further elucidate the correlation between AZD5363 response and genetic alterations in gastric cancer (GC) and identify GC patients with both PI3KCA mutations and PTEN loss, we investigated the effects of pharmacological inhibition of AKT on a panel of 20 GC cell lines and genetic aberrations in tumor samples from a cohort of Chinese GC patients. We demonstrated that GC cells with PI3KCA mutations were selectively sensitive to AZD5363. Disease linkage studies showed that PI3KCA activating mutations or PTEN loss were found in 2.7% (4/150) and 23% (14/61) of Chinese GC patients respectively. To further dissect the role of PI3KCA mutation and PTEN loss in response to AKT inhibition, we tested the antitumor activity of AZD5363 in two patient-derived GC xenograft (PDGCX) models harboring either PI3KCA mutation or PTEN loss. Our data indicated that AZD5363 monotherapy treatment led to a moderate response in the PI3KCA mutant PDGCX model. Whilst monotherapy AZD5363 or Taxotere were ineffective in the PTEN negative PDGCX model, significant anti-tumor activity was observed when AZD5363 was combined with Taxotere. CONCLUSION: Our results indicated that PI3KCA mutation is an important determinant of response to AKT inhibition in GC and combination with AZD5363 can overcome innate resistance to Taxotere in a PTEN loss PDGCX model. It is suggested that AKT inhibitor is an attractive option for treatment of a new segment of GC patients with aberrant PI3K/AKT signaling.


Subject(s)
Nuclear Proteins/genetics , PTEN Phosphohydrolase/deficiency , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Stomach Neoplasms/drug therapy , Taxoids/therapeutic use , Transcription Factors/genetics , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asian People , Cell Line, Tumor , Cell Proliferation/drug effects , China , Disease Models, Animal , Docetaxel , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Mutation/genetics , PTEN Phosphohydrolase/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Taxoids/pharmacology , Treatment Outcome
11.
J Transl Med ; 11: 168, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23842453

ABSTRACT

BACKGROUND: Patient-derived tumor xenograft models have been established and increasingly used for preclinical studies of targeted therapies in recent years. However, patient-derived non-small cell lung cancer (NSCLC) xenograft mouse models are relatively few in number and are limited in their degree of genetic characterization and validation. In this study, we aimed to establish a variety of patient-derived NSCLC models and characterize these for common genetic aberrations to provide more informative models for preclinical drug efficacy testing. METHODS: NSCLC tissues from thirty-one patients were collected and implanted into immunodeficient mice. Established xenograft models were characterized for common genetic aberrations, including detection of gene mutations within EGFR and KRAS, and genetic amplification of FGFR1 and cMET. Finally, gefitinib anti-tumor efficacy was tested in these patient-derived NSCLC xenograft models. RESULTS: Ten passable patient-derived NSCLC xenograft models were established by implantation of NSCLC specimens of thirty-one patients into immunodeficient mice. Genetic aberrations were detected in six of the models, including one model with an EGFR activating mutation (Exon19 Del), one model with KRAS mutation, one model with both KRAS mutation and cMET gene amplification, and three models with FGFR1 amplification. Anti-tumor efficacy studies using gefitinib demonstrated that the EGFR activating mutation model had superior sensitivity and that the KRAS mutation models were resistant to gefitinib. The range of gefitinib responses in the patient-derived NSCLC xenograft models were consistent with the results reported from clinical trials. Furthermore, we observed that patient-derived NSCLC models with FGFR1 gene amplification were insensitive to gefitinib treatment. CONCLUSIONS: Ten patient-derived NSCLC xenograft models were established containing a variety of genetic aberrations including EGFR activating mutation, KRAS mutation, and FGFR1 and cMET amplification. Gefitinib anti-tumor efficacy in these patient-derived NSCLC xenografts containing EGFR and KRAS mutation was consistent with the reported results from previous clinical trials. Thus, data from our panel of patient-derived NSCLC xenograft models confirms the utility of these models in furthering our understanding of this disease and aiding the development of personalized therapies for NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/genetics , Lung Neoplasms/metabolism , Mutation , Receptor, Fibroblast Growth Factor, Type 1/genetics , ras Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Mutational Analysis , Disease Models, Animal , Female , Gefitinib , Genes, ras , Genetic Variation , Humans , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Proto-Oncogene Proteins c-met/genetics , Quinazolines/pharmacology , Xenograft Model Antitumor Assays
12.
Bioorg Med Chem Lett ; 23(4): 979-84, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23317569

ABSTRACT

This Letter describes the medicinal chemistry effort towards a series of novel imidazo[1,5-a]pyrazine derived inhibitors of ACK1. Virtual screening led to the discovery of the initial hit, and subsequent exploration of structure-activity relationships and optimization of drug metabolism and pharmacokinetic properties led to the identification of potent, selective and orally bioavailable ACK1 inhibitors.


Subject(s)
Imidazoles/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazines/chemistry , Administration, Oral , Animals , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Mice , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Structure-Activity Relationship
13.
Biomater Sci ; 11(16): 5605-5617, 2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37404020

ABSTRACT

Many patients, especially those with chronic diseases, would benefit from personalized drugs that could modulate the treatment regimen. Tailored drug delivery via microneedle patches (MNPs) has emerged as a promising technology to address this problem. However, it is still difficult to modulate the treatment regimen in one MNP. Here, multiple treatment regimens were achieved by the same MNP functionalized with modifiable nanocontainers (NCs). The MNPs were biphasic in design, resulting in approximately a twice as high drug loading capacity than that of traditional dissolving MNPs. The drug-loaded NCs could have a zero-order release rate for at least 20 d in vitro. Furthermore, three model MNPs, Type-A (100% drug), Type-B (50% drug and 50% NCs) and Type-C (100% NCs) were generated to simulate various personalized dosing needs. In vivo application of these models could achieve effective therapeutic drug concentrations in the first 12 h and adjusted the duration of effective drug action from 24 h to 96 h and 144 h, respectively, with outstanding biocompatibility. These findings indicate that this device holds significant promise for personalized drug delivery.


Subject(s)
Drug Delivery Systems , Needles , Humans , Drug Delivery Systems/methods , Pharmaceutical Preparations
14.
EMBO J ; 27(14): 1985-94, 2008 Jul 23.
Article in English | MEDLINE | ID: mdl-18566589

ABSTRACT

The insulin-like growth factor-1 receptor (IGF1R) is a receptor tyrosine kinase (RTK) that has a critical role in mitogenic signalling during embryogenesis and an antiapoptotic role in the survival and progression of many human tumours. Here, we present the crystal structure of the tyrosine kinase domain of IGF1R (IGF1RK), in its unphosphorylated state, in complex with a novel compound, cis-3-[3-(4-methyl-piperazin-l-yl)-cyclobutyl]-1-(2-phenyl-quinolin-7-yl)-imidazo[1,5-a]pyrazin-8-ylamine (PQIP), which we show is a potent inhibitor of both the unphosphorylated (basal) and phosphorylated (activated) states of the kinase. PQIP interacts with residues in the ATP-binding pocket and in the activation loop, which confers specificity for IGF1RK and the highly related insulin receptor (IR) kinase. In this crystal structure, the IGF1RK active site is occupied by Tyr1135 from the activation loop of an symmetry (two-fold)-related molecule. This dimeric arrangement affords, for the first time, a visualization of the initial trans-phosphorylation event in the activation loop of an RTK, and provides a molecular rationale for a naturally occurring mutation in the activation loop of the IR that causes type II diabetes mellitus.


Subject(s)
Receptor, IGF Type 1/chemistry , Binding Sites , Crystallography, X-Ray , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Humans , Imidazoles/pharmacology , Mutation , Phosphorylation , Pyrazines/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism
15.
J Transl Med ; 10: 180, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22935382

ABSTRACT

BACKGROUND: Trastuzumab is currently approved for the clinical treatment of breast and gastric cancer patients with HER-2 positive tumors, but not yet for the treatment of esophageal carcinoma patients, whose tumors typically show 5 ~ 35% HER-2 gene amplification and 0 ~ 56% HER-2 protein expression. This study aimed to investigate the therapeutic efficacy of Trastuzumab in patient-derived esophageal squamous cell carcinoma xenograft (PDECX) mouse models. METHODS: PDECX models were established by implanting patient esophageal squamous cell carcinoma (ESCC) tissues into immunodeficient (SCID/nude) mice. HER-2 gene copy number (GCN) and protein expression were determined in xenograft tissues and corresponding patient EC samples by FISH and IHC analysis. Trastuzumab anti-tumor efficacy was evaluated within these PDECX models (n = 8 animals/group). Furthermore, hotspot mutations of EGFR, K-ras, B-raf and PIK3CA genes were screened for in the PDECX models and their corresponding patient's ESCC tissues. Similarity between the PDECX models and their corresponding patient's ESCC tissue was confirmed by histology, morphology, HER-2 GCN and mutation. RESULTS: None of the PDECX models (or their corresponding patient's ESCC tissues) harbored HER-2 gene amplification. IHC staining showed HER-2 positivity (IHC 2+) in 2 PDECX models and negativity in 3 PDECX models. Significant tumor regression was observed in the Trastuzumab-treated EC044 HER-2 positive model (IHC 2+). A second HER-2 positive (IHC 2+) model, EC039, harbored a known PIK3CA mutation and showed strong activation of the AKT signaling pathway and was insensitive to Trastuzumab treatment, but could be resensitised using a combination of Trastuzumab and AKT inhibitor AZD5363. In summary, we established 5 PDECX mouse models and demonstrated tumor regression in response to Trastuzumab treatment in a HER-2 IHC 2+ model, but resistance in a HER-2 IHC 2+/PIK3CA mutated model. CONCLUSIONS: This study demonstrates Trastuzumab-induced tumor regressions in HER-2 positive tumors, and highlights PIK3CA mutation as a potential resistance mechanism to Trastuzumab treatment in pre-clinical patient-derived EC xenograft models.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Transplantation, Heterologous , Animals , Base Sequence , DNA Primers , Disease Models, Animal , Female , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Nude , Mice, SCID , Polymerase Chain Reaction , Trastuzumab
16.
Bioorg Med Chem Lett ; 21(4): 1176-80, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21251824

ABSTRACT

Preclinical and emerging clinical evidence suggests that inhibiting insulin-like growth factor 1 receptor (IGF-1R) signaling may offer a promising therapeutic strategy for the treatment of several types of cancer. This Letter describes the medicinal chemistry effort towards a series of 8-amino-imidazo[1,5-a]pyrazine derived inhibitors of IGF-1R which features a substituted quinoline moiety at the C1 position and a cyclohexyl linking moiety at the C3 position. Lead optimization efforts which included the optimization of structure-activity relationships and drug metabolism and pharmacokinetic properties led to the identification of compound 9m, a potent, selective and orally bioavailable inhibitor of IGF-1R with in vivo efficacy in an IGF-driven mouse xenograft model.


Subject(s)
Antineoplastic Agents/chemistry , Benzimidazoles/chemistry , Imidazoles/chemistry , Protein Kinase Inhibitors/chemistry , Pyrazines/chemistry , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Benzimidazoles/pharmacokinetics , Benzimidazoles/therapeutic use , Mice , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyrazines/pharmacokinetics , Pyrazines/therapeutic use , Receptor, IGF Type 1/metabolism , Structure-Activity Relationship , Transplantation, Heterologous
17.
Cell Res ; 30(1): 34-49, 2020 01.
Article in English | MEDLINE | ID: mdl-31811277

ABSTRACT

Metastasis, the development of secondary malignant growths at a distance from a primary tumor, is the cause of death for 90% of cancer patients, but little is known about how metastatic cancer cells adapt to and colonize new tissue environments. Here, using clinical samples, patient-derived xenograft (PDX) samples, PDX cells, and primary/metastatic cell lines, we discovered that liver metastatic colorectal cancer (CRC) cells lose their colon-specific gene transcription program yet gain a liver-specific gene transcription program. We showed that this transcription reprogramming is driven by a reshaped epigenetic landscape of both typical enhancers and super-enhancers. Further, we identified that the liver-specific transcription factors FOXA2 and HNF1A can bind to the gained enhancers and activate the liver-specific gene transcription, thereby driving CRC liver metastasis. Importantly, similar transcription reprogramming can be observed in multiple cancer types. Our data suggest that reprogrammed tissue-specific transcription promotes metastasis and should be targeted therapeutically.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Transcriptional Activation , Animals , Cell Line, Tumor , Cellular Reprogramming , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Enhancer Elements, Genetic , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/physiology , Humans , Liver/metabolism , Liver Neoplasms/metabolism , Mice, Inbred BALB C , Mice, Nude , Organ Specificity , Transcriptome
18.
Bioorg Med Chem ; 16(3): 1359-75, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17983756

ABSTRACT

A series of novel, potent quinolinyl-derived imidazo[1,5-a]pyrazine IGF-IR (IGF-1R) inhibitors--most notably, cis-3-(3-azetidin-1-ylmethylcyclobutyl)-1-(2-phenylquinolin-7-yl)imidazo[1,5-a]pyrazin-8-ylamine (AQIP)--is described. Synthetic details, structure-activity relationships, and in vitro biological activity are reported for the series. Key in vitro and in vivo biological results for AQIP are reported, including: inhibition of ligand-stimulated autophosphorylation of IGF-IR and downstream pathways in 3T3/huIGFIR cells; inhibition of proliferation and induction of DNA fragmentation in human tumor cell lines; a pharmacokinetic profile suitable for once-per-day oral dosing; antitumor activity in a 3T3/huIGFIR xenograft model; and effects on insulin and glucose levels.


Subject(s)
Imidazoles/chemical synthesis , Imidazoles/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyrazines/chemical synthesis , Pyrazines/pharmacology , Quinolines/chemistry , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Blood Glucose/metabolism , Cell Line , Dogs , Female , Humans , Imidazoles/chemistry , Insulin/blood , Ligands , Mice , Molecular Structure , Protein Kinase Inhibitors/chemistry , Pyrazines/chemistry , Rats , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Xenograft Model Antitumor Assays
19.
Mol Cancer Ther ; 6(8): 2158-67, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17671083

ABSTRACT

Insulin-like growth factor-I receptor (IGF-IR) and its ligands, IGF-I and IGF-II, are up-regulated in a variety of human cancers. In tumors, such as colorectal, non-small cell lung, ovarian, and pediatric cancers, which may drive their own growth and survival through autocrine IGF-II expression, the role of IGF-IR is especially critical. Here, we present a novel small-molecule IGF-IR kinase inhibitor, cis-3-[3-(4-methyl-piperazin-l-yl)-cyclobutyl]-1-(2-phenyl-quinolin-7-yl)-imidazo[1,5-a]pyrazin-8-ylamine (PQIP), which displayed a cellular IC(50) of 19 nmol/L for inhibition of ligand-dependent autophosphorylation of human IGF-IR with 14-fold cellular selectivity relative to the human insulin receptor. PQIP showed minimal activity against a panel of 32 other protein kinases. It also abolished the ligand-induced activation of downstream phosphorylated AKT and phosphorylated extracellular signal-regulated kinase 1/2 in both IGF-IR transfectant cells and a GEO human colorectal cancer cell line. Analysis of GEO cells revealed a significant level of both phosphorylated IGF-IR and IGF-II expression. Furthermore, inactivation of IGF-II in conditioned GEO culture medium by a neutralizing antibody diminished IGF-IR activation, indicating the presence of a functional IGF-II/IGF-IR autocrine loop in GEO cells. Once daily oral dosing of PQIP induced robust antitumor efficacy in GEO xenografts. The antitumor efficacy correlated with the degree and duration of inhibition of tumor IGF-IR phosphorylation in vivo by this compound. Moreover, when mice were treated for 3 days with a dose of PQIP that maximally inhibited tumor growth, only minor changes in blood glucose were observed. Thus, PQIP represents a potent and selective IGF-IR kinase inhibitor that is especially efficacious in an IGF-II-driven human tumor model.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/pathology , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Autocrine Communication/drug effects , Blood Glucose/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , Humans , Imidazoles/administration & dosage , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Insulin-Like Growth Factor II/metabolism , Mice , Mice, Nude , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrazines/administration & dosage , Pyrazines/chemistry , Pyrazines/pharmacokinetics , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
20.
Sci Signal ; 11(547)2018 09 11.
Article in English | MEDLINE | ID: mdl-30206136

ABSTRACT

The Hippo signaling pathway regulates organ size and plays critical roles in maintaining tissue growth, homeostasis, and regeneration. Dysregulated in a wide spectrum of cancers, in mammals, this pathway is regulated by two key effectors, YAP and TAZ, that may functionally overlap. We found that TAZ promoted liver inflammation and tumor development. The expression of TAZ, but not YAP, in human liver tumors positively correlated with the expression of proinflammatory cytokines. Hyperactivated TAZ induced substantial myeloid cell infiltration into the liver and the secretion of proinflammatory cytokines through a TEAD-dependent mechanism. Furthermore, tumors with hyperactivated YAP and TAZ had distinct transcriptional signatures, which included the increased expression of inflammatory cytokines in TAZ-driven tumors. Our study elucidated a previously uncharacterized link between TAZ activity and inflammatory responses that influence tumor development in the liver.


Subject(s)
DNA-Binding Proteins/genetics , Inflammation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Liver/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Animals , Cell Cycle Proteins , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling/methods , Hippo Signaling Pathway , Humans , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Inbred C57BL , Mutation , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , TEA Domain Transcription Factors , Trans-Activators , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL