Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 38(31): 5933-5941, 2019 08.
Article in English | MEDLINE | ID: mdl-31285551

ABSTRACT

Two out of 41 non-small cell lung cancer patients enrolled in a clinical study were found with a somatic CRAF mutation in their tumor, namely CRAFP261A and CRAFP207S. To our knowledge, both mutations are novel in lung cancer and CRAFP261A has not been previously reported in cancer. Expression of CRAFP261A in HEK293T cells and BEAS-2B lung epithelial cells led to increased ERK pathway activation in a dimer-dependent manner, accompanied with loss of CRAF phosphorylation at the negative regulatory S259 residue. Moreover, stable expression of CRAFP261A in mouse embryonic fibroblasts and BEAS-2B cells led to anchorage-independent growth. Consistent with a previous report, we could not observe a gain-of-function with CRAFP207S. Type II but not type I RAF inhibitors suppressed the CRAFP261A-induced ERK pathway activity in BEAS-2B cells, and combinatorial treatment with type II RAF inhibitors and a MEK inhibitor led to a stronger ERK pathway inhibition and growth arrest. Our findings suggest that the acquisition of a CRAFP261A mutation can provide oncogenic properties to cells, and that such cells are sensitive to combined MEK and type II RAF inhibitors. CRAF mutations should be diagnostically and therapeutically explored in lung and perhaps other cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mutation , Proto-Oncogene Proteins c-raf/genetics , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , HEK293 Cells , Humans , Lung Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-raf/antagonists & inhibitors
2.
BMC Res Notes ; 11(1): 751, 2018 Oct 22.
Article in English | MEDLINE | ID: mdl-30348204

ABSTRACT

OBJECTIVE: Our aim was to produce a mono-biotinylated single domain antibody ('nanobody') specific for the epidermal growth factor receptor (EGFR), which is overexpressed in many cancer cells. The binding of the nanobody and its function are tested in cancer cells. The construct could be used to carry variable therapeutic or diagnostic load using biotin-streptavidin bridging. RESULTS: The EGFR-specific 7D12 nanobody was genetically fused to an IgA hinge linker and to a C-terminal biotin ligase acceptor sequence, allowing mono-biotinylation in E. coli. Expression was in strain BL21-DE3 from a T7 RNA polymerase driven pET22b vector. The biotinylated nanobody, isolated from the periplasm, was purified using streptavidin-mutein affinity chromatography. Final yields were up to 5 mg/l of cell culture. We showed that the construct could bind to EGFR expressing A431 epidermoid carcinoma cells, and to transiently transformed EGFR overexpressing HEK293T cells and not to EGFR negative control cells. The specificity for the EGFR was further demonstrated by immunoprecipitation. To test the functionality, PC9 non-small cell lung cancer cells were treated with mono-biotinylated nanobody or with streptavidin-coupled tetravalent nanobodies. Both were able to block mutant EGFR phosphorylation and slow down growth of PC9 cells. Tetravalent nanobodies were able to downregulate AKT phosphorylation.


Subject(s)
ErbB Receptors/immunology , Escherichia coli , Periplasm , Single-Domain Antibodies , Streptavidin , Cell Line, Tumor , HEK293 Cells , Humans
3.
Oncotarget ; 9(47): 28731-28744, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29983892

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can selectively induce apoptosis in cancer cells without causing damage to normal cells. However, some tumors are resistant to TRAIL monotherapy and clinical studies assessing targeted agents towards the TRAIL receptor have failed to show robust therapeutic activity. Evidence has shown that standard anti-mitotic drugs can induce synergistic apoptosis upon combination with TRAIL via cell cycle arrest. Polo like kinase-1 (PLK1) plays a critical role in different stages of cell cycle progression and mitosis. A number of investigations have demonstrated that PLK1 inhibition causes cell cycle arrest and mitotic catastrophe in non-small cell lung cancer (NSCLC), and we thus postulated that PLK1 inhibition could enhance TRAIL-induced apoptosis. We demonstrate that the combination of a TRAIL receptor agonist and a PLK1 inhibitor synergistically reduces cell viability, and strongly increases apoptosis in NSCLC cellular models. Consistent with our in vitro observations, this drug combination also significantly reduces tumor growth in vivo. Our data additionally reveal that G2/M cell cycle arrest and downregulation of Mcl-1 and signal transducer and activator of transcription 3 (STAT3) activity following PLK1 inhibition may contribute to the sensitization of TRAIL-induced apoptosis in NSCLC. Together, these data support the further exploration of combined TRAIL and PLK1 inhibition in the treatment of NSCLC.

4.
Lung Cancer ; 90(2): 167-74, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26342549

ABSTRACT

INTRODUCTION: Lung cancer remains the leading cause of cancer-related mortality worldwide, with metastatic disease frequently a prominent feature at the time of diagnosis. The role of NSCLC-derived EGFR mutations in cancer cell proliferation and survival has been widely reported, but little is known about the function of these mutations in invasive growth and metastasis. In this study, we sought to evaluate the intrinsic invasive properties of NSCLC cells with differing EGFR status and examine possible therapeutic targets that can abrogate invasive growth. MATERIALS AND METHODS: Collagen-based assays and 3D cell cultures were used to assess morphological features, actin cytoskeleton dynamics and the invasive capacity of NSCLC cell lines with differing EGFR status. The role of the RhoA/ROCK/MYPT1 and EGFR/HER pathways in NSCLC-related invasion was investigated by pharmacological inhibition and RNA interference techniques. RESULTS: We demonstrate a positive correlation between EGFR mutational/amplification status and invasive capacity. Knockdown of wild-type and mutant EGFR leads to depletion of active and total MYPT1 levels. Combined pharmacological inhibition or genetic ablation of ROCK/EGFR suppresses the hallmarks of cancer cells and abrogates the invasive phenotype in EGFR-dependent NSCLC cells. CONCLUSIONS: These observations suggest that combined targeting of the ROCK and EGFR/HER pathways may be a potential therapeutic approach in limiting invasive growth in NSCLC.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Neoplasm Invasiveness/prevention & control , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lung Neoplasms/metabolism , Mutation/drug effects , Phenotype , Signal Transduction/drug effects
5.
Oncotarget ; 6(24): 20132-44, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-25992771

ABSTRACT

KRAS is a frequently mutated oncogene in lung cancer and among the most refractory to EGFR targeted therapy. Recently, preclinical evidence in pancreatic cancer has demonstrated that mutant KRAS can be regulated by EGFR. However, the distinct correlation between the EGFR/HER family members and mutant KRAS has not been investigated. Here, we show that non-small cell lung cancer cell lines harboring differing isoforms of mutant KRAS, can be broadly divided into EGFR/HER dependent and EGFR/HER independent groups. Combined therapeutic targeting of EGFR, HER2 and HER3 in isoforms regulated by extracellular growth signals promotes in vitro and in vivo efficacy. We also provide evidence that depletion of EGFR via RNA interference specifically abolishes the EGFR/KRAS interaction in the dependent subset. Taken together, these findings suggest that upstream inhibition of the EGFR/HER receptors may be effective in treating a subset of KRAS mutant lung cancers.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Erlotinib Hydrochloride/administration & dosage , Female , Genes, ras , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Molecular Targeted Therapy , Mutation , Random Allocation , Signal Transduction , Xenograft Model Antitumor Assays
6.
PLoS One ; 8(3): e59708, 2013.
Article in English | MEDLINE | ID: mdl-23527257

ABSTRACT

Epidermal growth factor receptor (EGFR) and c-MET receptors are expressed on many non-small cell lung cancer (NSCLC) cells. Current single agent therapeutic targeting of a mutant EGFR has a high efficacy in the clinic, but is not curative. Here, we investigated the combination of targeting EGFR and c-MET pathways in NSCLC cells resistant to receptor tyrosine kinase inhibitors (TKIs), using RNA interference and inhibition by TKIs. Different NSCLC cell lines with various genomic characteristics (H358, H1650 and H1975) were transfected with EGFR-specific-siRNA, T790M-specific-siRNA, c-MET siRNA or the combination. Subsequently EGFR TKIs (gefitinib, erlotinib or afatinib) or monoclonal antibody cetuximab were combined respectively with the c-MET-specific TKI su11274 in NSCLC cell lines. The cell proliferation, viability, caspase-3/7 activity and apoptotic morphology were monitored by spectrophotometry, fluorimetry and fluorescence microscopy. The combined effect of EGFR TKIs, or cetuximab and su11274, was evaluated using a combination index. The results showed that the cell lines that were relatively resistant to EGFR TKIs, especially the H1975 cell line containing the resistance T790M mutation, were found to be more sensitive to EGFR-specific-siRNA. The combination of EGFR siRNA plus c-MET siRNA enhanced cell growth inhibition, apoptosis induction and inhibition of downstream signaling in EGFR TKI resistant H358, H1650 and H1975 cells, despite the absence of activity of the c-MET siRNA alone. EGFR TKIs or cetuximab plus su11274 were also consistently superior to either agent alone. The strongest biological effect was observed when afatinib, an irreversible pan-HER blocker was combined with su11274, which achieved a synergistic effect in the T790M mutant H1975 cells. In a conclusion, our findings offer preclinical proof of principle for combined inhibition as a promising treatment strategy for NSCLC, especially for patients in whom current EGFR-targeted treatments fail due to the presence of the T790M-EGFR-mutation or high c-MET expression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Indoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Afatinib , Analysis of Variance , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Fluorometry , Gefitinib , Humans , Microscopy, Fluorescence , Proto-Oncogene Proteins c-met/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spectrophotometry
7.
Acta Med Indones ; 43(3): 178-84, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21979283

ABSTRACT

AIM: to validate isolation and cultivation methods of bone marrow mesenchymal stem cells (BM-MSCs) from iliac crest, and to compare biological characteristics of BM-MSCs from different age groups for preparation of autologous stem cell therapy in cartilage defect. METHODS: patients undergoing spinal surgery were selected and grouped according to age. Iliac crest bone marrow from the patients was aspirated. BM-MSCs were isolated from the bone marrow and then cultivated. Their biological characteristics including morphological appearances and surface biomarkers were evaluated. Growth curves were observed. Sterility and Mycoplasma tests were also performed for quality assessment of BM-MSCs culture procedure. RESULTS: in average, cultivated-BM-MSCs reached the number of 7.56-22.95 x 106 in 4-7 weeks period. BM-MSCs of all age groups showed the same quality of morphology, shape and surface biomarkers (CD105+, CD73+, CD34-, CD45-, CD14-, CD19-, HLA-DR-). CONCLUSION: our procedures in isolating and cultivating of BM-MSCs have reached required amount for implantation into the cartilage lesion. In addition, the cultivated-BM-MSCs' biological characteristics were also in accordance with International Society of Cell Therapy (ISCT) MSCs criteria.


Subject(s)
Antigens, CD/analysis , Bone Marrow Cells , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Adolescent , Adult , Age Factors , Cartilage/injuries , Cartilage/surgery , Cell Proliferation , Culture Media , Female , HLA-DR Antigens/analysis , Humans , Ilium , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/microbiology , Middle Aged , Mycoplasma/isolation & purification , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...