Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Biomed Pharmacother ; 164: 114938, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37267635

ABSTRACT

In the current period of drug development, natural products have provided an unrivaled supply of anticancer medications. By modifying the cancer microenvironment and various signaling pathways, natural products and their derivatives and analogs play a significant role in cancer treatment. These substances are effective against several signaling pathways, particularly the cell death pathways (apoptosis and autophagy) and embryonic developmental pathways (Notch, Wnt, and Hedgehog pathways). Natural products have a long history, but more research is needed to understand their current function in the research and development of cancer treatments and the potential for natural products to serve as a significant source of therapeutic agents in the future. Several target-specific anticancer medications failed to treat cancer, necessitating research into natural compounds with multiple target properties. To help develop a better treatment plan for managing breast cancer, this review has outlined the anticancerous potential of several therapeutic approaches targeting the notch signaling system in breast tumors.


Subject(s)
Antineoplastic Agents , Biological Products , Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Hedgehog Proteins/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biological Products/pharmacology , Receptors, Notch/metabolism , Tumor Microenvironment
2.
Front Pharmacol ; 13: 901563, 2022.
Article in English | MEDLINE | ID: mdl-35873573

ABSTRACT

Immune checkpoint blockage targeting PD-L1 has led to breakthroughs in cancer treatment. Although anti-PD-L1-based immunotherapy has been approved as standard therapy in various cancer types, its therapeutic efficacy in most colorectal cancers (CRC) is still limited due to the low response to immunotherapy. Therefore, combining treatment with herbal medicines could be an alternative approach for treating CRC to overcome this limitation. Bojungikki-Tang (BJIKT), a herbal formula used in traditional Chinese medicine, clinically improves the quality of life for cancer patients and has been associated with antitumor and immune-modulating activities. However, the regulatory effect of BJIKT on the immune response in the tumor microenvironment remains largely uninvestigated. In this study, we verified the inhibitory effect of BJIKT on tumor growth and investigated the regulatory effect of combination therapy with BJIKT and anti-PD-L1 on antitumor immune responses in an MC38 CRC-bearing C57BL/6 mouse model. Immune profiling analysis by flow cytometry was used to characterize the exact cell types contributing to anticancer activities. Combination treatment with BJIKT and anti-PD-L1 therapy significantly suppressed tumor growth in MC38-bearing mice and increased the proportion of cytotoxic T lymphocytes and natural killer cells in tumor tissues. Furthermore, BJIKT suppressed the population of myeloid-derived suppressor cells, suggesting that this combination treatment effectively regulates the immunological function of T-cells by improving the tumor microenvironment. The herbal formula BJIKT can be a novel therapeutic option for improving anti-PD-L1-based immunotherapy in patients with CRC.

3.
Br J Cancer ; 123(12): 1720-1729, 2020 12.
Article in English | MEDLINE | ID: mdl-32963347

ABSTRACT

BACKGROUND: Recurrent and/or metastatic squamous cell carcinoma of head and neck (R/M SCCHN) is a common cancer with high recurrence and mortality. Current treatments have low response rates (RRs). METHODS: Fifty-three patients with R/M SCCHN received continuous oral buparlisib. In parallel, patient-derived xenografts (PDXs) were established in mice to evaluate resistance mechanisms and efficacy of buparlisib/cetuximab combination. Baseline and on-treatment tumour genomes and transcriptomes were sequenced. Based on the integrated clinical and PDX data, 11 patients with progression under buparlisib monotherapy were treated with a combination of buparlisib and cetuximab. RESULTS: For buparlisib monotherapy, disease control rate (DCR) was 49%, RR was 3% and median progression-free survival (PFS) and overall survival (OS) were 63 and 143 days, respectively. For combination therapy, DCR was 91%, RR was 18% and median PFS and OS were 111 and 206 days, respectively. Four PDX models were originated from patients enrolled in the current clinical trial. While buparlisib alone did not inhibit tumour growth, combination therapy achieved tumour inhibition in three of seven PDXs. Genes associated with apoptosis and cell-cycle arrest were expressed at higher levels with combination treatment than with buparlisib or cetuximab alone. CONCLUSIONS: The buparlisib/cetuximab combination has significant promise as a treatment strategy for R/M SCCHN. CLINICAL TRIAL REGISTRATION: NCT01527877.


Subject(s)
Aminopyridines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/therapeutic use , Head and Neck Neoplasms/drug therapy , Morpholines/therapeutic use , Adult , Aged , Aged, 80 and over , Aminopyridines/adverse effects , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cell Survival/drug effects , Cetuximab/adverse effects , DNA Copy Number Variations , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Humans , Male , Mice , Mice, Nude , Mice, SCID , Middle Aged , Morpholines/adverse effects , Mutation , Neoplasm Transplantation , Progression-Free Survival , Reproducibility of Results , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/mortality , Treatment Outcome , Up-Regulation/genetics , Whole Genome Sequencing/methods
4.
BMC Cancer ; 20(1): 316, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32293356

ABSTRACT

BACKGROUND: We investigated whether head and neck squamous cell carcinoma (HNSCC) patient-derived xenografts (PDXs) reaffirm patient responses to anti-cancer therapeutics. METHODS: Tumors from HNSCC patients were transplanted into immunodeficient mice and propagated via subsequent implantation. We evaluated established PDXs by histology, genomic profiling, and in vivo anti-cancer efficacy testing to confirm them as the authentic in vivo platform. RESULTS: From 62 HNSCCs, 15 (24%) PDXs were established. The primary cancer types were tongue (8), oropharynx (3), hypopharynx (1), ethmoid sinus cancer (1), supraglottic cancer (1), and parotid gland (1); six PDXs (40%) were established from biopsy specimens from advanced HNSCC. PDXs mostly retained donor characteristics and remained stable across passages. PIK3CA (H1047R), HRAS (G12D), and TP53 mutations (H193R, I195T, R248W, R273H, E298X) and EGFR, CCND1, MYC, and PIK3CA amplifications were identified. Using the acquisition method, biopsy showed a significantly higher engraftment rate when compared with that of surgical resection (100% [6/6] vs. 16.1% [9/56], P < 0.001). Specimens obtained from metastatic sites showed a significantly higher engraftment rate than did those from primary sites (100% [9/9] vs. 11.3% [6/53], P < 0.001). Three PDX models from HPV-positive tumors were established, as compared to 12 from HPV-negative (15.8% [3/19] and 27.9% [12/43] respectively, P = 0.311), suggesting that HPV positivity tends to show a low engraftment rate. Drug responses in PDX recapitulated the clinical responses of the matching patients with pan-HER inhibitors and pan-PI3K inhibitor. CONCLUSIONS: Genetically and clinically annotated HNSCC PDXs could be useful preclinical tools for evaluating biomarkers, therapeutic targets, and new drug discovery.


Subject(s)
Afatinib/administration & dosage , Aminopyridines/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Gene Regulatory Networks , Head and Neck Neoplasms/drug therapy , Methotrexate/administration & dosage , Morpholines/administration & dosage , Afatinib/pharmacology , Aminopyridines/pharmacology , Animals , Biopsy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Gene Amplification , Genetic Variation , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , High-Throughput Nucleotide Sequencing , Humans , Methotrexate/pharmacology , Mice , Morpholines/pharmacology , Papillomavirus Infections/drug therapy , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Patient-Specific Modeling , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 26(13): 3287-3295, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32269053

ABSTRACT

PURPOSE: Although first-line crizotinib treatment leads to clinical benefit in ROS1+ lung cancer, high prevalence of crizotinib-resistant ROS1-G2032R (ROS1G2032R) mutation and progression in the central nervous system (CNS) represents a therapeutic challenge. Here, we investigated the antitumor activity of repotrectinib, a novel next-generation ROS1/TRK/ALK-tyrosine kinase inhibitor (TKI) in ROS1+ patient-derived preclinical models. EXPERIMENTAL DESIGN: Antitumor activity of repotrectinib was evaluated in ROS1+ patient-derived preclinical models including treatment-naïve and ROS1G2032R models and was further demonstrated in patients enrolled in an on-going phase I/II clinical trial (NCT03093116). Intracranial antitumor activity of repotrectinib was evaluated in a brain-metastasis mouse model. RESULTS: Repotrectinib potently inhibited in vitro and in vivo tumor growth and ROS1 downstream signal in treatment-naïve YU1078 compared with clinically available crizotinib, ceritinib, and entrectinib. Despite comparable tumor regression between repotrectinib and lorlatinib in YU1078-derived xenograft model, repotrectinib markedly delayed the onset of tumor recurrence following drug withdrawal. Moreover, repotrectinib induced profound antitumor activity in the CNS with efficient blood-brain barrier penetrating properties. Notably, repotrectinib showed selective and potent in vitro and in vivo activity against ROS1G2032R. These findings were supported by systemic and intracranial activity of repotrectinib observed in patients enrolled in the on-going clinical trial. CONCLUSIONS: Repotrectinib is a novel next-generation ROS1-TKI with improved potency and selectivity against treatment-naïve and ROS1G2032R with efficient CNS penetration. Our findings suggest that repotrectinib can be effective both as first-line and after progression to prior ROS1-TKI.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Rearrangement , Lung Neoplasms/genetics , Macrocyclic Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Pyrazoles/pharmacology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Macrocyclic Compounds/therapeutic use , Mice , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Xenograft Model Antitumor Assays
6.
Cancer Res ; 80(11): 2257-2272, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32193288

ABSTRACT

Quiescent cancer cells are believed to cause cancer progression after chemotherapy through unknown mechanisms. We show here that human non-small cell lung cancer (NSCLC) cell line-derived, quiescent-like, slow-cycling cancer cells (SCC) and residual patient-derived xenograft (PDX) tumors after chemotherapy experience activating transcription factor 6 (ATF6)-mediated upregulation of various cytokines, which acts in a paracrine manner to recruit fibroblasts. Cancer-associated fibroblasts (CAF) underwent transcriptional upregulation of COX2 and type I collagen (Col-I), which subsequently triggered a slow-to-active cycling switch in SCC through prostaglandin E2 (PGE2)- and integrin/Src-mediated signaling pathways, leading to cancer progression. Both antagonism of ATF6 and cotargeting of Src/COX2 effectively suppressed cytokine production and slow-to-active cell cycling transition in SCC, withholding cancer progression. Expression of COX2 and Col-I and activation of Src were observed in patients with NSCLC who progressed while receiving chemotherapy. Public data analysis revealed significant association between COL1A1 and SRC expression and NSCLC relapse. Overall, these findings indicate that a proinflammatory niche created by the interplay between SCC and CAF triggers tumor progression. SIGNIFICANCE: Cotargeting COX2 and Src may be an effective strategy to prevent cancer progression after chemotherapy.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cytokines/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Activating Transcription Factor 6/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Celecoxib/administration & dosage , Cell Communication/drug effects , Cell Communication/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen Type I/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cytokines/biosynthesis , Dasatinib/administration & dosage , Disease Progression , Drug Resistance, Neoplasm , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Lung Neoplasms/metabolism , Mice , Mice, SCID , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , src-Family Kinases/antagonists & inhibitors
7.
EMBO Mol Med ; 11(12): e10581, 2019 12.
Article in English | MEDLINE | ID: mdl-31633304

ABSTRACT

Clinical benefit of ALK tyrosine kinase inhibitors (ALK-TKIs) in ALK-rearranged lung cancer has been limited by the inevitable development of acquired resistance, and bypass-molecular resistance mechanisms remain poorly understood. We investigated a novel therapeutic target through screening FDA-approved drugs in ALK-TKI-resistant models. Cerivastatin, the rate-limiting enzyme inhibitor of the mevalonate pathway, showed anti-cancer activity against ALK-TKI resistance in vitro/in vivo, accompanied by cytoplasmic retention and subsequent inactivation of transcriptional co-regulator YAP. The marked induction of YAP-targeted oncogenes (EGFR, AXL, CYR61, and TGFßR2) in resistant cells was abolished by cerivastatin. YAP silencing suppressed tumor growth in resistant cells, patient-derived xenografts, and EML4-ALK transgenic mice, whereas YAP overexpression decreased the responsiveness of parental cells to ALK inhibitor. In matched patient samples before/after ALK inhibitor treatment, nuclear accumulation of YAP was mainly detected in post-treatment samples. High expression of YAP in pretreatment samples was correlated with poor response to ALK-TKIs. Our findings highlight a crucial role of YAP in ALK-TKI resistance and provide a rationale for targeting YAP as a potential treatment option for ALK-rearranged patients with acquired resistance to ALK inhibitors.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle Proteins/metabolism , Lung Neoplasms/drug therapy , Transcription Factors/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lung Neoplasms/genetics , Mice , Mice, Nude , Transcription Factors/genetics
8.
Cancer Lett ; 466: 23-34, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31521695

ABSTRACT

The therapeutic targeting of oncogenic KRAS mutant-harboring (KRASMUT) non-small cell lung cancer (NSCLC) is an urgent unmet need in cancer therapy. Although NSCLC is often driven by epidermal growth factor receptor (EGFR) overexpression and/or mutations, no EGFR-targeted therapy is clinically available for KRASMUT NSCLC. In this study, we show that integrin ß3 expression is associated with the intrinsic resistance of KRASMUT NSCLCs to the anti-EGFR antibody cetuximab. Further analyses identified an integrin ß3-mediated ternary complex comprising NRP1-integrin ß3-KRASMUT and its downstream signaling of PI3K-Akt and RalB-TBK1 as a primary resistance mechanism of KRASMUT NSCLC to cetuximab treatment. Importantly, we demonstrate that the EGFR/NRP1 dual-targeting bispecific antibody, Ctx-TPP11, attenuates the downstream signaling driven by the ternary complex via the cellular co-internalization and degradation of the NRP1-coupled complex, resulting in the alleviation of cetuximab resistance in KRASMUT NSCLCs in vitro and in vivo, including patient-derived xenograft mouse models. Our study shows that the dual-targeting of EGFR and NRP1 with a bispecific antibody might be an effective therapeutic strategy for KRASMUT NSCLC.


Subject(s)
Antibodies, Bispecific/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Neuropilin-1/metabolism , Animals , Antibodies, Bispecific/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cetuximab , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Humans , Integrin beta3/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mutation , Neuropilin-1/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/drug effects , Ternary Complex Factors/drug effects , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 25(8): 2575-2587, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30670498

ABSTRACT

PURPOSE: Given that osimertinib is the only approved third-generation EGFR-TKI against EGFR activating and resistant T790M mutated non-small cell lung cancer (NSCLC), additional mutant-selective inhibitors with a higher efficacy, especially for brain metastases, with favorable toxicity profile are still needed. In this study, we investigated the antitumor efficacy of YH25448, an oral, mutant-selective, irreversible third-generation EGFR-TKI in preclinical models. EXPERIMENTAL DESIGN: Antitumor activity of YH25448 was investigated in vitro using mutant EGFR-expressing Ba/F3 cells and various lung cancer cell lines. In vivo antitumor efficacy, ability to penetrate the blood-brain barrier (BBB), and skin toxicity of YH25448 were examined and compared with those of osimertinib using cell lines and PDX model. RESULTS: Compared with osimertinib, YH25448 showed a higher selectivity and potency in kinase assay and mutant EGFR-expressing Ba/F3 cells. In various cell line models harboring EGFR activating and T790M mutation, YH25448 effectively inhibited EGFR downstream signaling pathways, leading to cellular apoptosis. When compared in vivo at equimolar concentrations, YH25448 produced significantly better tumor regression than osimertinib. Importantly, YH25448 induced profound tumor regression in brain metastasis model with excellent brain/plasma and tumor/brain area under the concentration-time curve value. YH25448 rarely suppressed the levels of p-EGFR in hair follicles, leading to less keratosis than osimertinib in animal model. The potent systemic and intracranial activity of YH25448 has been shown in an ongoing phase I/II clinical trial for advanced EGFR T790M mutated NSCLC (NCT03046992). CONCLUSIONS: Our findings suggest that YH25448 is a promising third-generation EGFR inhibitor, which may be more effective and better tolerated than the currently approved osimertinib.


Subject(s)
Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Acrylamides/chemistry , Acrylamides/pharmacology , Acrylamides/therapeutic use , Adult , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/genetics , Humans , Lung Neoplasms/diagnostic imaging , Male , Mice , Models, Molecular , Mutation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Structure-Activity Relationship , Treatment Outcome , Xenograft Model Antitumor Assays
10.
Lung Cancer ; 124: 168-178, 2018 10.
Article in English | MEDLINE | ID: mdl-30268457

ABSTRACT

BACKGROUND: Preclinical models that can better predict therapeutic activity in clinical trials are needed in this era of personalized cancer treatment. Herein, we established genomically and clinically annotated patient-derived xenografts (PDXs) from non-small-cell lung cancer (NSCLC) patients and investigated whether these PDXs would faithfully recapitulate patient responses to targeted therapy. METHODS: Patient-derived tumors were implanted in immunodeficient mice and subsequently expanded via re-implantation. Established PDXs were examined by light microscopy, genomic profiling, and in vivo drug testing, and the successful engraft rate was analyzed with the mutation profile, histology, or acquisition method. Finally, the drug responses of PDXs were compared with the clinical responses of the respective patients. RESULTS: Using samples from 122 patients, we established 41 NSCLC PDXs [30 adenocarcinoma (AD), 11 squamous cell carcinoma (SQ)], among which the following driver mutation were observed: 13 EGFR-mutant, 4 ALK-rearrangement, 1 ROS1-rearrangement, 1 PIK3CA-mutant, 1 FGFR1-amplification, and 2 KRAS-mutant. We rigorously characterized the relationship of clinical features to engraftment rate and latency rates. The engraft rates were comparable across histologic type. The AD engraft rate tended to be higher for surgically resected tissues relative to biopsies, whereas similar engraft rates was observed for SQ, irrespective of the acquisition method. Notably, EGFR-mutants demonstrated significantly longer latency time than EGFR-WT (86 vs. 37days, P = 0.007). The clinical responses were recapitulated by PDXs harboring driver gene alteration (EGFR, ALK, ROS1, or FGFR1) which regressed to their target inhibitors, suggesting that established PDXs comprise a clinically relevant platform. CONCLUSION: The establishment of genetically and clinically annotated NSCLC PDXs can yield a robust preclinical tool for biomarker, therapeutic target, and drug discovery.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Evaluation, Preclinical/methods , Lung Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Disease Models, Animal , Drug Discovery , ErbB Receptors/genetics , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Nude , Mice, SCID , Molecular Targeted Therapy , Mutation/genetics , Neoplasm Staging , Precision Medicine
11.
Nat Biomed Eng ; 2(4): 239-253, 2018 04.
Article in English | MEDLINE | ID: mdl-30936439

ABSTRACT

The accumulation of genetic and epigenetic alterations in cancer cells rewires cellular signalling pathways through changes in the patterns of protein-protein interactions (PPIs). Understanding these patterns may facilitate the design of tailored cancer therapies. Here, we show that single-molecule pull-down and co-immunoprecipitation techniques can be used to characterize signalling complexes of the human epidermal growth-factor receptor (HER) family in specific cancers. By analysing cancer-specific signalling phenotypes, including post-translational modifications and PPIs with downstream interactions, we found that activating mutations of the epidermal growth-factor receptor (EGFR) gene led to the formation of large protein complexes surrounding mutant EGFR proteins and to a reduction in the dependency of mutant EGFR signalling on phosphotyrosine residues, and that the strength of HER-family PPIs is correlated with the strength of the dependence of breast and lung adenocarcinoma cells on HER-family signalling pathways. Furthermore, using co-immunoprecipitation profiling to screen for EGFR-dependent cancers, we identified non-small-cell lung cancers that respond to an EGFR-targeted inhibitor. Our approach might help predict responses to targeted cancer therapies, particularly for cancers that lack actionable genomic mutations.


Subject(s)
ErbB Receptors/metabolism , Molecular Diagnostic Techniques/methods , Neoplasms/diagnosis , Protein Interaction Maps/physiology , Signal Transduction/physiology , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, SCID , Middle Aged , Neoplasms/metabolism
12.
J Thorac Oncol ; 12(3): 491-500, 2017 03.
Article in English | MEDLINE | ID: mdl-27836576

ABSTRACT

BACKGROUND: Anaplastic lymphoma receptor tyrosine kinase gene (ALK) fusion is a distinct molecular subclassification of NSCLC that is targeted by anaplastic lymphoma kinase (ALK) inhibitors. We established a transgenic mouse model that expresses tumors highly resembling human NSCLC harboring echinoderm microtubule associated protein like 4 gene (EML)-ALK fusion. We aimed to test an EML4-ALK transgenic mouse model as a platform for assessing the efficacy of ALK inhibitors and examining mechanisms of acquired resistance to ALK inhibitors. METHODS: Transgenic mouse lines harboring LoxP-STOP-LoxP-FLAGS-tagged human EML4-ALK (variant 1) transgene was established by using C57BL/6N mice. The transgenic mouse model with highly lung-specific, inducible expression of echinoderm microtubule associated protein like 4-ALK fusion protein was established by crossing the EML4-ALK transgenic mice with mice expressing Cre-estrogen receptor fusion protein under the control of surfactant protein C gene (SPC). Expression of EML4-ALK transgene was induced by intraperitoneally injecting mice with tamoxifen. When the lung tumor of the mice treated with the ALK inhibitor crizotinib for 2 weeks was measured, tumor shrinkage was observed. RESULTS: EML4-ALK tumor developed after 1 week of tamoxifen treatment. Echinoderm microtubule associated protein like 4-ALK was strongly expressed in the lung but not in other organs. ALK and FLAGS expressions were observed by immunohistochemistry. Treatment of EML4-ALK tumor-bearing mice with crizotinib for 2 weeks induced dramatic shrinkage of tumors with no signs of toxicity. Furthermore, prolonged treatment with crizotinib led to acquired resistance in tumors, resulting in regrowth and disease progression. The resistant tumor nodules revealed acquired ALK G1202R mutations. CONCLUSIONS: An EML4-ALK transgenic mouse model for study of drug resistance was successfully established with short duration of tumorigenesis. This model should be a strong preclinical model for testing efficacy of ALK TKIs, providing a useful tool for investigating the mechanisms of acquired resistance and pursuing novel treatment strategies in ALK-positive lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Disease Models, Animal , Lung Neoplasms/pathology , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/therapeutic use , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Male , Mice , Mice, Transgenic , Oncogene Proteins, Fusion/metabolism , Tumor Cells, Cultured
13.
Mol Cancer Ther ; 15(7): 1627-36, 2016 07.
Article in English | MEDLINE | ID: mdl-27196768

ABSTRACT

Although treatment of BRAF V600E-mutant non-small cell lung cancer (NSCLC(V600E)) with GSK2118436 has shown an encouraging efficacy, most patients develop resistance. To investigate the mechanisms of acquired resistance to GSK2118436 in NSCLC(V600E), we established GSK2118436-resistant (GSR) cells by exposing MV522 NSCLC(V600E) to increasing GSK2118436 concentrations. GSR cells displayed activated EGFR-RAS-CRAF signaling with upregulated EGFR ligands and sustained activation of ERK1/2, but not MEK1/2, in the presence of GSK2118436. Treatment of GSR cells with GSK2118436 enhanced EGFR-mediated RAS activity, leading to the formation of BRAF-CRAF dimers and transactivation of CRAF. Interestingly, sustained activation of ERK1/2 was partly dependent on receptor-interacting protein kinase-2 (RIP2) activity, but not on MEK1/2 activity. Combined BRAF and EGFR inhibition blocked reactivation of ERK signaling and improved efficacy in vitro and in vivo Our findings support the evaluation of combined BRAF and EGFR inhibition in NSCLC(V600E) with acquired resistance to BRAF inhibitors. Mol Cancer Ther; 15(7); 1627-36. ©2016 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , Imidazoles/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MAP Kinase Signaling System , Mutation , Oximes/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Amino Acid Substitution , Animals , Antineoplastic Agents/pharmacology , Autocrine Communication , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Codon , DNA Mutational Analysis , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Models, Biological , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Xenograft Model Antitumor Assays , ras Proteins/metabolism
14.
Lung Cancer ; 95: 57-64, 2016 May.
Article in English | MEDLINE | ID: mdl-27040853

ABSTRACT

OBJECTIVES: The epidermal growth factor receptor (EGFR) abnormalities including amplification, mutation, and overexpression are frequent in non-small cell lung cancer (NSCLC). We investigated in vitro and in vivo antitumor activity of ER2, a novel human anti-EGFR monoclonal antibody, in NSCLC. METHODS: A panel of NSCLC cell lines (A549, H460, H322, H358, H1299, HCC827, PC9, H1975, and PC9-GR) was used to evaluate in vitro antitumor activity of ER2 and cetuximab. The inhibitory effects of ER2 and cetuximab on downstream signaling were assessed by western blot. Secreted VEGF was measured by Human VEGF Quantikine ELISA kit. Antitumor effects of ER2 and cetuximab as single agents and in combination with cisplatin were evaluated in H322, HCC827 and A549 xenograft models. RESULTS: ER2 efficiently inhibits EGFR and its downstream signaling molecules including Akt and Erk1/2 in NSCLC cell lines with wild-type or mutant EGFR. ER2 inhibited cell viability of H322, HCC827 and A549 cells in a dose-dependent manner by inducing cell cycle arrest and apoptosis. Also, ER2 suppressed EGF-stimulated VEGF production as efficiently as cetuximab in H322, HCC827 and A549 cells. Moreover, ER2 alone and in combination with cisplatin showed a significant anti-tumor efficacy in xenograft mouse models. CONCLUSION: Taken together, ER2 has significant anti-tumor activity in in vitro and in vivo NSCLC models, suggesting a rationale for clinical development of ER2 in NSCLC.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Disease Models, Animal , Drug Synergism , ErbB Receptors/metabolism , Female , Humans , Lung Neoplasms/drug therapy , Mice , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
15.
Mol Cancer Ther ; 14(10): 2238-48, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26208525

ABSTRACT

RET rearrangement is a newly identified oncogenic mutation in lung adenocarcinoma (LADC). Activity of dovitinib (TKI258), a potent inhibitor of FGFR, VEGFR, and PDGFR, in RET-rearranged LADC has not been reported. The aims of the study are to explore antitumor effects and mechanisms of acquired resistance of dovitinib in RET-rearranged LADC. Using structural modeling and in vitro analysis, we demonstrated that dovitinib induced cell-cycle arrest at G0-G1 phase and apoptosis by selective inhibition of RET kinase activity and ERK1/2 signaling in RET-rearranged LC-2/ad cells. Strong antitumor effect of dovitinib was observed in an LC-2/ad tumor xenograft model. To identify the acquired resistance mechanisms to dovitinib, LC-2/ad cells were exposed to increasing concentrations of dovitinib to generate LC-2/ad DR cells. Gene-set enrichment analysis of gene expression and phosphor-kinase revealed that Src, a central gene in focal adhesion, was activated in LC-2/ad DR cells. Saracatinib, an src kinase inhibitor, suppressed ERK1/2 phosphorylation and growth of LC-2/ad DR cells. Taken together, these findings suggest that dovitinib can be a potential therapeutic option for RET-rearranged LADC, in which acquired resistance to dovitinib can be overcome by targeting Src.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Quinolones/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Antineoplastic Agents/chemistry , Apoptosis , Benzimidazoles/chemistry , Catalytic Domain , Cell Cycle Checkpoints , Cell Line, Tumor , Enzyme Activation , HEK293 Cells , Humans , Inhibitory Concentration 50 , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice, Nude , Models, Molecular , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-ret/chemistry , Quinolones/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
16.
Exp Cell Res ; 318(5): 539-49, 2012 Mar 10.
Article in English | MEDLINE | ID: mdl-22227409

ABSTRACT

p53, the major human tumor suppressor, appears to be related to sonic hedgehog (Shh)-Gli-mediated tumorigenesis. However, the role of p53 in tumor progression by the Shh-Gli signaling pathway is poorly understood. Herein we investigated the critical regulation of Gli3-p53 in tumorigenesis of colon cancer cells and the molecular mechanisms underlying these effects. RT-PCR analysis indicated that the mRNA level of Shh and Gli3 in colon tumor tissues was significantly higher than corresponding normal tissues (P<0.001). The inhibition of Gli3 by treatment with Gli3 siRNA resulted in a clear decrease in cell proliferation and enhanced the level of expression of p53 proteins compared to treatment with control siRNA. The half-life of p53 was dramatically increased by treatment with Gli3 siRNA. In addition, treatment with MG132 blocked MDM2-mediated p53 ubiquitination and degradation, and led to accumulation of p53 in Gli3 siRNA-overexpressing cells. Importantly, ectopic expression of p53 siRNA reduced the ability of Gli3 siRNA to suppress proliferation of those cells compared with the cells treated with Gli3 siRNA alone. Moreover, Gli3 siRNA sensitized colon cancer cells to treatment with anti-cancer agents (5-FU and bevacizumab). Taken together, our studies demonstrate that loss of Gli3 signaling leads to disruption of the MDM2-p53 interaction and strongly potentiate p53-dependent cell growth inhibition in colon cancer cells, indicating a basis for the rational use of Gli3 antagonists as a novel treatment option for colon cancer.


Subject(s)
Gene Expression , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Bevacizumab , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms , Fluorouracil/pharmacology , Gene Knockdown Techniques , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , Signal Transduction , Tumor Suppressor Protein p53/genetics , Ubiquitination , Zinc Finger Protein Gli3
18.
Exp Cell Res ; 317(12): 1746-62, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21570392

ABSTRACT

Bone morphogenetic proteins (BMPs) have been implicated in tumorigenesis and metastatic progression in various types of cancer cells, but the role and cellular mechanism in the invasive phenotype of gastric cancer cells is not known. Herein, we determined the roles of phosphoinositide 3-kinase (PI3K)/AKT, extracellular signal-regulated protein kinase (ERK), nuclear factor (NF)-κB, and matrix metalloproteinase (MMP) expression in BMP-2-mediated metastatic function in gastric cancer. We found that stimulation of BMP-2 in gastric cancer cells enhanced the phosphorylation of AKT and ERK. Accompanying activation of AKT and ERK kinase, BMP-2 also enhanced phosphorylation/degradation of IκBα and the nuclear translocation/activation of NF-κB. Interestingly, blockade of PI3K/AKT and ERK signaling using LY294002 and PD98059, respectively, significantly inhibited BMP-2-induced motility and invasiveness in association with the activation of NF-κB. Furthermore, BMP-2-induced MMP-9 expression and enzymatic activity was also significantly blocked by treatment with PI3K/AKT, ERK, or NF-κB inhibitors. Immunohistochemistry staining of 178 gastric tumor biopsies indicated that expression of BMP-2 and MMP-9 had a significant positive correlation with lymph node metastasis and a poor prognosis. These results indicate that the BMP-2 signaling pathway enhances tumor metastasis in gastric cancer by sequential activation of the PI3K/AKT or MAPK pathway followed by the induction of NF-κB and MMP-9 activity, indicating that BMP-2 has the potential to be a therapeutic molecular target to decrease metastasis.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intestinal Neoplasms/secondary , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/pathology , Apoptosis , Blotting, Western , Bone Morphogenetic Protein 2/genetics , Cell Adhesion , Cell Movement , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Humans , Immunoenzyme Techniques , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Luciferases/metabolism , Lymphatic Metastasis , Male , Matrix Metalloproteinase 9/genetics , Middle Aged , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Prognosis , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Rate , Tumor Cells, Cultured , Wound Healing
19.
Oncol Rep ; 22(3): 525-34, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19639199

ABSTRACT

Although dysregulation of bone morphogenetic protein (BMP) signaling has been linked to various types of cancers, the relationship between abnormal activation of these signaling pathways and tumorigenesis is not clear. The purpose of the current study was to clarify how BMP2 is involved in colon cancer aggressiveness. The data showed that SW480 and DLD-1 cells displayed different responses to short- and long-term exposure to BMP2. During the first 24 h of exposure to BMP2, these cells were growth-inhibited, whereas surviving cells became resistant to growth inhibition, showing epithelial-to-mesenchymal transformation (EMT) and enhanced motility and invasiveness. Interestingly, in highly metastatic mesenchymal colon carcinoma cells (CT26), blockade of BMP2 signaling by BMP2 siRNA prevented EMT, motility and invasiveness; rather, blockade of BMP2 signaling caused a mesenchymal-to-epithelial transition (MET). The levels of phosphorylated Akt were very different between the two cell types; the BMP2-sensitive SW480 and DLD-1 cells had much higher levels of expression than the BMP2-resistant SW480 and DLD-1 and CT26 cells. CT26 cells, following exposure to BMP2 and activation of Akt, escaped the EMT-induced cellular motility and invasiveness. Moreover, LY294002 treatment of BMP2-sensitive SW480 cells blocked cell growth and enhanced motility and invasiveness. Together, these results suggest that suppression of the PI3 kinase/Akt pathway is correlated with the development of BMP2 resistance and invasion in BMP2-induced EMT in colon cancer.


Subject(s)
Bone Morphogenetic Protein 2/physiology , Colonic Neoplasms/etiology , Epithelial Cells/pathology , Mesoderm/pathology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , Cell Line, Tumor , Cell Transformation, Neoplastic , Colonic Neoplasms/pathology , Humans , Neoplasm Invasiveness , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...