45 results on '"Jingxuan Pan"'
Search Results
2. Correction to: Ponatinib efficiently kills imatinib-resistant chronic eosinophilic leukemia cells harboring gatekeeper mutant T674I FIP1L1-PDGFRα: roles of Mcl-1 and β-catenin
- Author
-
Yanli Jin, Ke Ding, Honglin Li, Mengzhu Xue, Xiaoke Shi, Chengyan Wang, and Jingxuan Pan
- Subjects
Male ,Cancer Research ,Receptor, Platelet-Derived Growth Factor alpha ,Oncogene Proteins, Fusion ,Fluorescent Antibody Technique ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Electrophoretic Mobility Shift Assay ,Transfection ,Mice ,Microscopy, Electron, Transmission ,Hypereosinophilic Syndrome ,Animals ,Humans ,RC254-282 ,beta Catenin ,Cell Proliferation ,mRNA Cleavage and Polyadenylation Factors ,Mice, Inbred BALB C ,Imidazoles ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Correction ,Xenograft Model Antitumor Assays ,Molecular Docking Simulation ,Pyridazines ,Oncology ,Drug Resistance, Neoplasm ,Molecular Medicine ,Myeloid Cell Leukemia Sequence 1 Protein - Abstract
T674I FIP1L1-PDGFRα in a subset of chronic eosinophilic leukemia (CEL) is a gatekeeper mutation that is resistant to many tyrosine kinase inhibitors (TKIs) (e.g., imatinib, nilotinib and dasatinib), similar to T315I Bcr-Abl. Therefore, novel TKIs effective against T674I FIP1L1-PDGFRα are needed. Ponatinib (AP24534) is a novel orally bioavailable TKI against T315I Bcr-Abl, but it is not clear whether ponatinib is effective against T674I FIP1L1-PDGFRα. The purpose of this study was to examine the effect of ponatinib on T674I FIP1L1-PDGFRα.Molecular docking analysis in silico was performed. The effects of ponatinib on PDGFRα signaling pathways, apoptosis and cell cycling were examined in EOL-1, BaF3 cells expressing either wild type (WT) or T674I FIP1L1-PDGFRα. The in vivo antitumor activity of ponatinib was evaluated with xenografted BaF3-T674I FIP1L1-PDGFRα cells in nude mice models.Molecular docking analysis revealed that ponatinib could bind to the DFG (Asp-Phe-Gly)-out state of T674I PDGFRα. Ponatinib potently inhibited the phosphorylation of WT and T674I FIP1L1-PDGFRα and their downstream signaling molecules (e.g., Stat3, Stat5). Ponatinib strikingly inhibited the growth of both WT and T674I FIP1L1-PDGFRα-carrying CEL cells (IC50: 0.004-2.5 nM). It induced apoptosis in CEL cells with caspase-3-dependent cleavage of Mcl-1, and inhibited tyrosine phosphorylation of β-catenin to decrease its stability and pro-survival functions. In vivo, ponatinib abrogated the growth of xenografted BaF3-T674I FIP1L1-PDGFRα cells in nude mice.Ponatinib is a pan-FIP1L1-PDGFRα inhibitor, and clinical trials are warranted to investigate its efficacy in imatinib-resistant CEL.
- Published
- 2021
3. Transcriptional inhibition by CDK7/9 inhibitor SNS-032 abrogates oncogene addiction and reduces liver metastasis in uveal melanoma
- Author
-
Shenglan Liu, Jingxuan Pan, Jing Zhang, and Qianyun Ye
- Subjects
Male ,Uveal Neoplasms ,0301 basic medicine ,Cancer Research ,RHOA ,Cell ,Cell Cycle Proteins ,Apoptosis ,Cell motility ,MMP9 ,Metastasis ,Mice ,0302 clinical medicine ,Uveal melanoma ,Oncogene Addiction ,Liver metastasis ,Melanoma ,Oxazoles ,biology ,Kinase ,Liver Neoplasms ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,KLF4 ,Cyclin-Dependent Kinases ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Self-renewal ,Transcription ,Signal Transduction ,SNS-032 ,lcsh:RC254-282 ,Kruppel-Like Factor 4 ,03 medical and health sciences ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Transcription factor ,Research ,Correction ,RhoA ,Cancer stem-like cells ,medicine.disease ,Cyclin-Dependent Kinase 9 ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Thiazoles ,030104 developmental biology ,biology.protein ,Cancer research ,Cyclin-Dependent Kinase-Activating Kinase ,Transcription Factors - Abstract
Background Life of patients with uveal melanoma (UM) is largely threatened by liver metastasis. Little is known about the drivers of liver organotropic metastasis in UM. The elevated activity of transcription of oncogenes is presumably to drive aspects of tumors. We hypothesized that inhibition of transcription by cyclin-dependent kinase 7/9 (CDK7/9) inhibitor SNS-032 diminished liver metastasis by abrogating the putative oncogenes in charge of colonization, stemness, cell motility of UM cells in host liver microenvironment. Methods The effects of SNS-032 on the expression of the relevant oncogenes were examined by qRT-PCR and Western blotting analysis. Proliferative activity, frequency of CSCs and liver metastasis were evaluated by using NOD-SCID mouse xenograft model and NOG mouse model, respectively. Results The results showed that CDK7/9 were highly expressed in UM cells, and SNS-032 significantly suppressed the cellular proliferation, induced apoptosis, and inhibited the outgrowth of xenografted UM cells and PDX tumors in NOD-SCID mice, repressed the cancer stem-like cell (CSC) properties through transcriptional inhibition of stemness-related protein Krüppel-like factor 4 (KLF4), inhibited the invasive phonotypes of UM cells through matrix metalloproteinase 9 (MMP9). Mechanistically, SNS-032 repressed the c-Myc-dependent transcription of RhoA gene, and thereby lowered the RhoA GTPase activity and actin polymerization, and subsequently inhibited cell motility and liver metastasis. Conclusions In conclusion, we validate a set of transcription factors which confer metastatic traits (e.g., KLF4 for CSCs, c-Myc for cell motility) in UM cells. Our results identify SNS-032 as a promising therapeutic agent, and warrant a clinical trial in patients with metastatic UM.
- Published
- 2019
- Full Text
- View/download PDF
4. Correction to: Salinomycin effectively eliminates cancer stem-like cells and obviates hepatic metastasis in uveal melanoma
- Author
-
Jingfeng Zhou, Hailin Zou, Wei Dai, Shubo Wang, Jing Zhang, Shenglan Liu, Jingxuan Pan, and Yun Wang
- Subjects
Cancer Research ,business.industry ,Melanoma ,Cancer ,Correction ,Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Hepatic metastasis ,chemistry.chemical_compound ,Text mining ,Oncology ,chemistry ,medicine ,Cancer research ,Molecular Medicine ,business ,Salinomycin - Abstract
An amendment to this paper has been published and can be accessed via the original article.
- Published
- 2021
5. CHEK1 and circCHEK1_246aa evoke chromosomal instability and induce bone lesion formation in multiple myeloma
- Author
-
Chunyan Gu, Wang Wang, Fenghuang Zhan, Jingxuan Pan, Tingting Xu, Ye Yang, Dirk Hose, Yajun Wang, Xiaozhu Tang, Siegfried Janz, Artur Jurczyszyn, Mengjie Guo, Rongfang Wei, Anja Seckinger, Yanxin Zhang, Meral Beksac, Hematology, and Basic (bio-) Medical Sciences
- Subjects
0301 basic medicine ,Cancer Research ,Proliferation ,Osteoclasts ,Multiple Myeloma/genetics ,Biology ,Bone and Bones ,Chromosomal Instability/genetics ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Osteoclast ,In vivo ,Chromosomal Instability ,medicine ,Animals ,Humans ,Bone and Bones/pathology ,CHEK1 ,Multiple myeloma ,RC254-282 ,Checkpoint Kinase 1/genetics ,Cell growth ,Kinase ,hematology ,Research ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Transfection ,RNA, Circular ,medicine.disease ,Molecular biology ,In vitro ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,RNA, Circular/genetics ,Drug resistance ,Osteoclasts/metabolism ,Checkpoint Kinase 1 ,Molecular Medicine ,Heterografts ,Bone marrow ,circCHEK1_246aa ,Multiple Myeloma - Abstract
Background Multiple myeloma (MM) is still incurable and characterized by clonal expansion of plasma cells in the bone marrow (BM). Therefore, effective therapeutic interventions must target both myeloma cells and the BM niche. Methods Cell proliferation, drug resistance, and chromosomal instability (CIN) induced by CHEK1 were confirmed by Giemsa staining, exon sequencing, immunofluorescence and xenograft model in vivo. Bone lesion was evaluated by Tartrate-resistant acid phosphatase (TRAP) staining. The existence of circCHEK1_246aa was evaluated by qPCR, Sanger sequencing and Mass Spectrometer. Results We demonstrated that CHEK1 expression was significantly increased in human MM samples relative to normal plasma cells, and that in MM patients, high CHEK1 expression was associated with poor outcomes. Increased CHEK1 expression induced MM cellular proliferation and evoked drug-resistance in vitro and in vivo. CHEK1-mediated increases in cell proliferation and drug resistance were due in part to CHEK1-induced CIN. CHEK1 activated CIN, partly by phosphorylating CEP170. Interestingly, CHEK1 promoted osteoclast differentiation by upregulating NFATc1 expression. Intriguingly, we discovered that MM cells expressed circCHEK1_246aa, a circular CHEK1 RNA, which encoded and was translated to the CHEK1 kinase catalytic center. Transfection of circCHEK1_246aa increased MM CIN and osteoclast differentiation similarly to CHEK1 overexpression, suggesting that MM cells could secrete circCHEK1_246aa in the BM niche to increase the invasive potential of MM cells and promote osteoclast differentiation. Conclusions Our findings suggest that targeting the enzymatic catalytic center encoded by CHEK1 mRNA and circCHEK1_246aa is a promising therapeutic modality to target both MM cells and BM niche.
- Published
- 2021
6. Verification of EZH2 as a druggable target in metastatic uveal melanoma
- Author
-
Huasheng Yang, Jingxuan Pan, Yun Wang, Ping Zhang, Jing Zhang, Huijing Ye, Hailin Zou, and Bei Jin
- Subjects
Uveal Neoplasms ,0301 basic medicine ,Cancer Research ,Indoles ,Pyridones ,Apoptosis ,Mice, SCID ,macromolecular substances ,Biology ,lcsh:RC254-282 ,Metastasis ,Small hairpin RNA ,Mice ,03 medical and health sciences ,Uveal melanoma ,0302 clinical medicine ,Circulating tumor cell ,Mice, Inbred NOD ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,EZH2 ,Melanoma ,Cell Proliferation ,GNA11 ,Research ,Tumor Suppressor Proteins ,Cancer ,Motility ,Cancer stem-like cells ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Hepatic metastasis ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Molecular Medicine ,Ubiquitin Thiolesterase ,GNAQ - Abstract
Background Hepatic metastasis develops in ~ 50% of uveal melanoma (UM) patients with no effective treatments. Although GNAQ/GNA11 mutations are believed to confer pathogenesis of UM, the underlying mechanism of liver metastasis remains poorly understood. Given that profound epigenetic evolution may occur in the long journey of circulating tumor cells (CTCs) to distant organs, we hypothesized that EZH2 endowed tumor cells with enhanced malignant features (e.g., stemness and motility) during hepatic metastasis in UM. We aimed to test this hypothesis and explore whether EZH2 was a therapeutic target for hepatic metastatic UM patients. Methods Expression of EZH2 in UM was detected by qRT-PCR, Western blotting and immunohistochemistry staining. Proliferation, apoptosis, cancer stem-like cells (CSCs) properties, migration and invasion were evaluated under circumstances of treatment with either EZH2 shRNA or EZH2 inhibitor GSK126. Antitumor activity and frequency of CSCs were determined by xenografted and PDX models with NOD/SCID mice. Hepatic metastasis was evaluated with NOG mice. Results We found that EZH2 overexpressed in UM promoted the growth of UM; EZH2 increased the percentage and self-renewal of CSCs by miR-29c-DVL2-β-catenin signaling; EZH2 facilitates migration and invasion of UM cells via RhoGDIγ-Rac1 axis. Targeting EZH2 either by genetics or small molecule inhibitor GSK126 decreased CSCs and motility and abrogated the liver metastasis of UM. Conclusions These findings validate EZH2 as a druggable target in metastatic UM patients, and may shed light on the understanding and interfering the complicated metastatic process.
- Published
- 2020
- Full Text
- View/download PDF
7. Antitumor Effects of Blocking Protein Neddylation in T315I-BCR-ABL Leukemia Cells and Leukemia Stem Cells
- Author
-
Chang Liu, Xin Du, Yangqiu Li, Juan Li, Jingxuan Pan, Jingfeng Zhou, Danian Nie, Yanli Jin, and Yuhong Lu
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Fusion Proteins, bcr-abl ,Antigens, CD34 ,Antineoplastic Agents ,Apoptosis ,Cyclopentanes ,Ubiquitin-Activating Enzymes ,Biology ,03 medical and health sciences ,Protein neddylation ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Point Mutation ,Gene silencing ,Molecular Targeted Therapy ,neoplasms ,Leukemia, Experimental ,Myeloid leukemia ,Imatinib ,medicine.disease ,Fusion protein ,Mice, Inbred C57BL ,Leukemia ,Pyrimidines ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Imatinib Mesylate ,Neoplastic Stem Cells ,Cancer research ,Stem cell ,medicine.drug ,Chronic myelogenous leukemia - Abstract
Imatinib revolutionized the treatment of chronic myeloid leukemia (CML), but drug resistance and disease recurrence remain a challenge. In this study, we suggest a novel strategy based on blocking protein neddylation to address BCR-ABL point mutations and leukemia stem cells (LSC) that lie at the root of imatinib-resistant recurrences. On the basis of the finding that the NEDD8-activating enzyme subunit NAE1 is overexpressed in CML cells, we hypothesized that the function of certain neddylation-dependent protein substrates might be targeted to therapeutic ends in imatinib-resistant CML cells and LSCs. In support of this hypothesis, we demonstrated that the NAE1 inhibitor MLN4924 induced G2–M-phase arrest and apoptosis in bulk CML cells with wild-type p53, regardless of their T315I mutation status in BCR-ABL. Moreover, MLN4924 inhibited the survival and self-renewal of primary human CML CD34+ cells and LSCs in CML-bearing mice via accumulation of p27kip1 in the nucleus. Notably, p27kip1 silencing attenuated the suppressive effect of MLN4924 on the maintenance of LSCs in CML-bearing mice. Taken together, our findings offer a preclinical proof of concept for targeting protein neddylation as a novel therapeutic strategy to override mutational and LSC-derived imatinib resistance in CML. Significance: These findings highlight a mediator of protein neddylation, a type of protein turnover mechanism, as a viable therapeutic target against imatinib-resistant forms of chronic myelogenous leukemia. Cancer Res; 78(6); 1522–36. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
8. Pristimerin effectively inhibits the malignant phenotypes of uveal melanoma cells by targeting NF-κB pathway
- Author
-
Biao Zhang, Jingxuan Pan, and Jing Zhang
- Subjects
Uveal Neoplasms ,0301 basic medicine ,Cancer Research ,Cell Survival ,Cell ,Population ,Apoptosis ,Biology ,Vinblastine ,03 medical and health sciences ,0302 clinical medicine ,NF-KappaB Inhibitor alpha ,Cell Movement ,Cell Line, Tumor ,medicine ,Humans ,Molecular Targeted Therapy ,Viability assay ,Clonogenic assay ,education ,Melanoma ,Cell Proliferation ,education.field_of_study ,Oncogene ,Tumor Necrosis Factor-alpha ,NF-kappa B ,Transcription Factor RelA ,Drug Synergism ,Cell cycle ,Triterpenes ,Histone Deacetylase Inhibitors ,IκBα ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Pentacyclic Triterpenes ,Signal Transduction - Abstract
Uveal melanoma (UM) is a highly aggressive intraocular malignancy that lacks any effective targeted-therapy. Neither survival nor prognosis has been improved for the past decades in patients with metastatic UM. NF‑κB pathway is reported to be abnormally activated in UM. However, the role of NF‑κB pathway as a potential therapeutical target in UM remains unclear. Here, the effect of pristimerin, a potent inhibitor of NF‑κB pathway, on UM cells in terms of growth, apoptosis, motility, invasion and cancer stem-like cells (CSCs) was evaluated in vitro. We showed that pristimerin suppressed tumor necrosis factor α (TNFα)-induced IκBα phosphorylation, translocation of p65, and expression of NF‑κB-dependent genes. Moreover, pristimerin decreased cell viability and clonogenic ability of UM cells. A synergistic effect was observed in the treatment of pristimerin combined with vinblastine, a frontline therapeutic agent, in UM. Pristimerin led to a significant increase in the Annexin V+ cell population as measured by flow cytometry. We also observed that pristimerin impaired the abilities of migration and invasion in UM cells. Furthermore, pristimerin eliminated the ALDH+ cells and weakened serial re-plating ability of melanosphere. Collectively, pristimerin shows remarkable anticancer activities in UM cells through inactivating NF‑κB pathway, revealing that pristimerin may be a promising therapeutic agent in UM.
- Published
- 2017
- Full Text
- View/download PDF
9. Preclinical development of a novel BCR-ABL T315I inhibitor against chronic myeloid leukemia
- Author
-
Anna Maria Barbuti, Dong-Hua Yang, Xin Zhang, Pranav Gupta, Jingxuan Pan, Ke Ding, Jingfeng Zhou, Guan-Nan Zhang, Sabesan Yoganathan, Nishant Karadkhelkar, and Zhe-Sheng Chen
- Subjects
0301 basic medicine ,Cancer Research ,Mutant ,Fusion Proteins, bcr-abl ,Apoptosis ,Fusion gene ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Animals ,Humans ,neoplasms ,Protein Kinase Inhibitors ,Myeloproliferative neoplasm ,Cell Proliferation ,Chemistry ,Kinase ,Myeloid leukemia ,Imatinib ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Pyrimidines ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Benzamides ,Mutation ,Cancer research ,Imatinib Mesylate ,medicine.drug ,Signal Transduction - Abstract
Chronic Myeloid Leukemia (CML) is a myeloproliferative neoplasm primarily due to the presence of the BCR-ABL fusion gene that produces the constitutively active protein, BCR-ABL. Imatinib, a BCR-ABL-targeted drug, is a first-line drug for the treatment of CML. Resistance to imatinib occurs as a result of mutations in the BCR-ABL kinase domains. In this study, we evaluated S116836, a novel BCR-ABL inhibitor, for its anti-cancer efficacy in the wild-type (WT) and T315I mutant BCR-ABL. S116836 was efficacious in BaF3 cells with WT or T315I mutated BCR-ABL genotypes. S116836 inhibits the phosphorylation of BCR-ABL and its downstream signaling in BaF3/WT and BaF3/T315I cells. Mechanistically, S116836 arrests the cells in the G0/G1 phase of cell cycle, induces apoptosis, increases ROS production, and decreases GSH production in BaF3/WT and BaF3/T315I cells. Moreover, in mouse tumor xenografts, S116836 significantly inhibits the growth and volume of tumors expressing the WT or T315I mutant BCR-ABL without causing significant cardiotoxicity. Overall, our results indicate that S116836 significantly inhibits the imatinib-resistant T315I BCR-ABL mutation and could be a novel drug candidate for treating imatinib-resistant CML patients.
- Published
- 2019
10. Salinomycin effectively eliminates cancer stem-like cells and obviates hepatic metastasis in uveal melanoma
- Author
-
Wei Dai, Yun Wang, Shubo Wang, Jingxuan Pan, Jingfeng Zhou, Hailin Zou, Shenglan Liu, and Jing Zhang
- Subjects
0301 basic medicine ,Uveal Neoplasms ,Cancer Research ,Apoptosis ,Mice, SCID ,Metastasis ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Uveal melanoma ,Mice, Inbred NOD ,Melanoma ,Salinomycin ,Membrane Potential, Mitochondrial ,education.field_of_study ,Molecular Structure ,Liver Neoplasms ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Molecular Medicine ,Twist1 ,Population ,Antineoplastic Agents ,Biology ,lcsh:RC254-282 ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,education ,Cell Proliferation ,Pyrans ,Dose-Response Relationship, Drug ,Cell growth ,Research ,Cancer ,Cancer stem-like cells ,medicine.disease ,Xenograft Model Antitumor Assays ,Transplantation ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Cancer research - Abstract
Background Uveal melanoma (UM) is the most common primary intraocular tumor. Hepatic metastasis is the major and direct death-related reason in UM patients. Given that cancer stem-like cells (CSCs) are roots of metastasis, targeting CSCs may be a promising strategy to overcome hepatic metastasis in UM. Salinomycin, which has been identified as a selective inhibitor of CSCs in multiple types of cancer, may be an attractive agent against CSCs thereby restrain hepatic metastasis in UM. The objective of the study is to explore the antitumor activity of salinomycin against UM and clarify its underlying mechanism. Methods UM cells were treated with salinomycin, and its effects on cell proliferation, apoptosis, migration, invasion, CSCs population, and the related signal transduction pathways were determined. The in vivo antitumor activity of salinomycin was evaluated in the NOD/SCID UM xenograft model and intrasplenic transplantation liver metastasis mouse model. Results We found that salinomycin remarkably obviated growth and survival in UM cell lines and in a UM xenograft mouse model. Meanwhile, salinomycin significantly eliminated CSCs and efficiently hampered hepatic metastasis in UM liver metastasis mouse model. Mechanistically, Twist1 was fundamental for the salinomycin-enabled CSCs elimination and migration/invasion blockage in UM cells. Conclusions Our findings suggest that targeting UM CSCs by salinomycin is a promising therapeutic strategy to hamper hepatic metastasis in UM. These results provide the first pre-clinical evidence for further testing of salinomycin for its antitumor efficacy in UM patients with hepatic metastasis.
- Published
- 2019
11. Anthelmintic Niclosamide Disrupts the Interplay of p65 and FOXM1/β-catenin and Eradicates Leukemia Stem Cells in Chronic Myelogenous Leukemia
- Author
-
Bei Jin, Jingxuan Pan, Ke Ding, Juan Li, Chengyan Wang, and Xin Du
- Subjects
0301 basic medicine ,Cancer Research ,Fusion Proteins, bcr-abl ,Apoptosis ,Mice, SCID ,0302 clinical medicine ,Mice, Inbred NOD ,hemic and lymphatic diseases ,Protein Interaction Mapping ,Cell Self Renewal ,RNA, Small Interfering ,Promoter Regions, Genetic ,beta Catenin ,Niclosamide ,Anthelmintics ,Feedback, Physiological ,Mice, Knockout ,Gene Expression Regulation, Leukemic ,Graft Survival ,Neoplasm Proteins ,Leukemia ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Heterografts ,RNA Interference ,Stem cell ,Tyrosine kinase ,Interleukin Receptor Common gamma Subunit ,medicine.drug ,Chromatin Immunoprecipitation ,Antineoplastic Agents ,Biology ,03 medical and health sciences ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Forkhead Box Protein M1 ,Transcription Factor RelA ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Cancer research ,FOXM1 ,K562 Cells ,Chromatin immunoprecipitation ,K562 cells ,Chronic myelogenous leukemia - Abstract
Purpose: Leukemia stem cells (LSC), which are insensitive to tyrosine kinase inhibitors (TKI), are an important source of TKI resistance and disease relapse in chronic myelogenous leukemia (CML). Obstacles to eradicating LSCs include limited understanding of the regulation network of LSCs. The current study aimed to examine the interplay between NF-κB and FOXM1/β-catenin, and the effect of its chemical intervention on CML LSCs. Experimental Design: The interplay between NF-κB and FOXM1/β-catenin was analyzed by reciprocal coimmunoprecipitation (co-IP) and chromatin immunoprecipitation (ChIP) assay in CML cells. The effect of disturbing NF-κB and FOXM1/β-catenin by niclosamide on the self-renewal capacity and survival of LSCs was evaluated in vitro in human primary CML CD34+ cells and in vivo in CML mice. Results: Reciprocal co-IP experiments showed physical interaction of p65 and FOXM1. p65 promoted transcription of FOXM1 gene. ChIP assay revealed recruitment of p65 on the promoter of FOXM1 gene. Conversely, FOXM1 and β-catenin positively regulated the nuclear translocation and transcriptional activity of NF-κB in CML cells. Niclosamide disrupted the positive feedback loop between NF-κB and FOXM1/β-catenin, thereby impairing the self-renewal capacity and survival of CML LSCs. Niclosamide decreased the long-term engraftment of human CML LSCs in NOD-SCID IL2Rγ chain-deficient (NOG) mice, and prolonged the survival of CML mice. Conclusions: Interaction of p65 with FOXM1/β-catenin is critical in CML and its disruption by niclosamide eradicates LSCs. These findings may improve the understanding of a self-renewal regulatory mechanism of LSCs and offer a rationale-based approach to eliminate LSCs in CML. Clin Cancer Res; 23(3); 789–803. ©2016 AACR.
- Published
- 2017
- Full Text
- View/download PDF
12. [Corrigendum] Anthelmintic pyrvinium pamoate blocks Wnt/β-catenin and induces apoptosis in multiple myeloma cells
- Author
-
Fang Xu, Yingjie Zhu, Yuhong Lu, Zhi Yu, Jun Zhong, Yangqiu Li, and Jingxuan Pan
- Subjects
Cancer Research ,Oncology - Published
- 2020
- Full Text
- View/download PDF
13. Anthelmintic pyrvinium pamoate blocks Wnt/β-catenin and induces apoptosis in multiple myeloma cells
- Author
-
Jingxuan Pan, Yingjie Zhu, Yangqiu Li, Jun Zhong, Yuhong Lu, Fang Xu, and Zhi Yu
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Oncogene ,Chemistry ,Cell growth ,Cellular differentiation ,Myeloid leukemia ,Articles ,Cell cycle ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Bone marrow ,Corrigendum - Abstract
Multiple myeloma (MM) is a malignancy of the bone marrow. The median survival time of patients with MM is only 5 years, with patients frequently experiencing relapse. Currently, there is no effective therapy for recurrent MM. The results of the present study indicated that pyrvinium pamoate (PP), a US Food and Drug Administration-approved oral anthelmintic drug, exhibited potent antitumor activity in MM cells in vitro. It is demonstrated that PP inhibited MM cell proliferation and mediated apoptosis. Notably, PP markedly promoted the degradation of β-catenin and abrogated its phosphorylation. PP triggered apoptosis in MM cells by inducing the release of cytochrome c and downregulating the expression of myeloid leukemia cell differentiation protein. In addition, PP effectively induced cell death in primary MM cells. In conclusion, PP may be a promising agent for the clinical treatment of MM.
- Published
- 2018
- Full Text
- View/download PDF
14. PTEN Is Fundamental for Elimination of Leukemia Stem Cells Mediated by GSK126 Targeting EZH2 in Chronic Myelogenous Leukemia
- Author
-
Chang Liu, Wei Dai, Yun Wang, Jingxuan Pan, Yangqiu Li, Xin Du, Jingfeng Zhou, Juan Li, Yanli Jin, Yuhong Lu, and Danian Nie
- Subjects
0301 basic medicine ,Cancer Research ,Indoles ,medicine.drug_class ,Pyridones ,CD34 ,Fusion Proteins, bcr-abl ,macromolecular substances ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,Myelogenous ,hemic and lymphatic diseases ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,PTEN ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,neoplasms ,Cell Proliferation ,ABL ,biology ,Dose-Response Relationship, Drug ,business.industry ,TOR Serine-Threonine Kinases ,PTEN Phosphohydrolase ,medicine.disease ,Xenograft Model Antitumor Assays ,Leukemia ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Gene Knockdown Techniques ,Cancer research ,biology.protein ,Neoplastic Stem Cells ,Stem cell ,business ,Proto-Oncogene Proteins c-akt ,Chronic myelogenous leukemia - Abstract
Purpose: Leukemia stem cells (LSCs) are an important source of tyrosine kinase inhibitor resistance and disease relapse in patients with chronic myelogenous leukemia (CML). Targeting LSCs may be an attractive strategy to override this thorny problem. Given that EZH2 was overexpressed in primary CML CD34+ cells, our purpose in this study was to evaluate the effects of targeting EZH2 on CML LSCs and clarify its underlying mechanism. Experimental Design: Human primary CML CD34+ cells and retrovirally BCR–ABL-driven CML mouse models were employed to evaluate the effects of suppression of EZH2 by GSK126- or EZH2-specific shRNA in vitro and in vivo. Recruitment of EZH2 and H3K27me3 on the promoter of tumor-suppressor gene PTEN in CML cells was measured by chromatin immunoprecipitation assay. Results: Our results showed that pharmacologic inhibition of EZH2 by GSK126 not only elicited apoptosis and restricted cell growth in CML bulk leukemia cells, but also decreased LSCs in CML CD34+ cells while sparing those from normal bone marrow CD34+ cells. Suppression of EZH2 by GSK126 or specific shRNA prolonged survival of CML mice and reduced the number of LSCs in mice. EZH2 knockdown resulted in elevation of PTEN and led to impaired recruitment of EZH2 and H3K27me3 on the promoter of PTEN gene. The effect of EZH2 knockdown in the CML mice was at least partially reversed by PTEN knockdown. Conclusions: These findings improve the understanding of the epigenetic regulation of stemness in CML LSCs and warrant clinical trial of GSK126 in refractory patients with CML. Clin Cancer Res; 24(1); 145–57. ©2017 AACR.
- Published
- 2017
15. The Anti-malarial Drug Artesunate Blocks Wnt/β-catenin Pathway and Inhibits Growth, Migration and Invasion of Uveal Melanoma Cells
- Author
-
Jingxuan Pan and Lei Zheng
- Subjects
0301 basic medicine ,Adult ,Uveal Neoplasms ,Cancer Research ,Cell Survival ,Artesunate ,Antineoplastic Agents ,Apoptosis ,03 medical and health sciences ,chemistry.chemical_compound ,Antimalarials ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Drug Discovery ,Medicine ,Humans ,Neoplasm Invasiveness ,Viability assay ,Phosphorylation ,Melanoma ,Wnt Signaling Pathway ,beta Catenin ,Cell Proliferation ,Pharmacology ,Analysis of Variance ,Glycogen Synthase Kinase 3 beta ,business.industry ,Cell growth ,Wnt signaling pathway ,Cell migration ,medicine.disease ,030104 developmental biology ,Oncology ,chemistry ,Vincristine ,030220 oncology & carcinogenesis ,Catenin ,Cancer research ,Drug Therapy, Combination ,business ,Signal Transduction - Abstract
Background: Uveal melanoma is the most common primary intraocular malignancy in adults. So far, there have been no effective targeted therapeutic agents in patients with uveal melanoma. Artesunate is a semi-synthetic derivative of artemisinin extracted from traditional Chinese medicine Artemisia annua L for treatment of severe and multidrug-resistant malaria. Besides its antimalarial activity, artesunate is identified as an anti-cancer drug due to the inhibition of Wnt/β- catenin pathway in multiple types of cancer. However, the effect of artesunate on uveal melanoma remains unknown. Objective: We evaluated the anti-tumor effects of artesunate on uveal melanoma cells, and analyzed in terms of Wnt/β-catenin pathway, cell growth, cell death, cell migration, invasion and cancer stemlike cells (CSCs) properties. Methods: Primary (92.1, Mel270) and metastatic (Omm1 and Omm2.3) uveal melanoma cells were used. Immunofluorescence staining, dual luciferase reporter assay, Western blotting, MTS, soft agar cloning technique, Annexin V/PI analyses, wound healing scratch assay, in vitro transwell migration and invasion assays, aldehyde dehydrogenase (ALDH) analyses and melanosphere formation assay et al. were carried out. Results: Artesunate suppressed the phosphorylation of GSK3β at S9, and lowered the protein level of β-catenin and its downstream targets (c-Myc, cyclin D1). Artesunate potently inhibited cell viability and colony formation ability. Treatment with artesunate significantly induced apoptosis. In addition, artesunate significantly reduced the migration and invasion of uveal melanoma cells, impaired the traits of CSCs in vitro. Conclusion: Artesunate may be a potential interest for the therapy of uveal melanoma.
- Published
- 2017
16. Novel thiazole amine class tyrosine kinase inhibitors induce apoptosis in human mast cells expressing D816V KIT mutation
- Author
-
Mengjie Shen, Jingxuan Pan, Ke Ding, Yanli Jin, and Deping Wang
- Subjects
Male ,Cancer Research ,Time Factors ,Genotype ,medicine.drug_class ,Mice, Nude ,Antineoplastic Agents ,Apoptosis ,Transfection ,Receptor tyrosine kinase ,Tyrosine-kinase inhibitor ,Mice ,chemistry.chemical_compound ,Cell Line, Tumor ,Survivin ,medicine ,Animals ,Humans ,Mast Cells ,Molecular Targeted Therapy ,Amines ,Phosphorylation ,Systemic mastocytosis ,Protein Kinase Inhibitors ,Mice, Inbred BALB C ,Dose-Response Relationship, Drug ,biology ,Drug Synergism ,Tyrosine phosphorylation ,Mastocytoma ,medicine.disease ,Xenograft Model Antitumor Assays ,Molecular biology ,Proto-Oncogene Proteins c-kit ,Thiazoles ,Phenotype ,Imatinib mesylate ,Oncology ,chemistry ,Mutation ,biology.protein ,Cancer research ,RNA Interference ,Tyrosine kinase ,Signal Transduction - Abstract
Gain-of-function mutations of receptor tyrosine kinase KIT play a critical role in the pathogenesis of systemic mastocytosis (SM) and gastrointestinal stromal tumors. D816V KIT mutation, found in ∼80% of SM, is resistant to the currently available tyrosine kinase inhibitors (TKIs) (e.g. imatinib mesylate). Therefore, development of promising TKIs for the treatment of D816V KIT mutation is still urgently needed. We synthesized thiazole amine compounds and chose one representative designated 126332 to investigate its effect on human mast cells expressing KIT mutations. We found 126332 inhibited the phosphorylation of KIT and its downstream signaling molecules Stat3 and Stat5. 126332 inhibited the proliferation of D816V KIT expressing cells. 126332 induced apoptosis and downregulated levels of Mcl-1 and survivin. Furthermore, 126332 inhibited the tyrosine phosphorylation of β-catenin, inhibited β-catenin-mediated transcription and DNA binding of TCF. Moreover, 126332 also exhibited in vivo antineoplastic activity against cells harboring D816V mutation. Our findings suggest thiazole amine compounds may be promising agents for the treatment of diseases caused by KIT mutation.
- Published
- 2014
- Full Text
- View/download PDF
17. SAHA and S116836, a novel tyrosine kinase inhibitor, synergistically induce apoptosis in imatinib-resistant chronic myelogenous leukemia cells
- Author
-
Xin Du, Waiyi Zou, Lijing Cui, Juan Li, Jingxuan Pan, Ke Ding, and Qiangui Bu
- Subjects
Cancer Research ,Cell Survival ,medicine.drug_class ,Antineoplastic Agents ,Apoptosis ,Pharmacology ,Biology ,Hydroxamic Acids ,Piperazines ,Tyrosine-kinase inhibitor ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Humans ,Viability assay ,Protein Kinase Inhibitors ,neoplasms ,Cell Proliferation ,Vorinostat ,Cell growth ,Drug Synergism ,Imatinib ,medicine.disease ,XIAP ,Dasatinib ,Pyrimidines ,Imatinib mesylate ,Oncology ,Drug Resistance, Neoplasm ,Benzamides ,Imatinib Mesylate ,Cancer research ,Tyrosine ,Molecular Medicine ,Research Paper ,Chronic myelogenous leukemia ,medicine.drug - Abstract
Limited treatment options are available for chronic myelogenous leukemia (CML) patients who develop imatinib mesylate (IM) resistance. Here we proposed a novel combination regimen, a co-administration of S116836, a novel small molecule multi-targeted tyrosine kinase inhibitor that was synthesized by rational design, and histone deacetylases inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA), to overcome IM resistance in CML. S116836 at low concentrations used in the present study mildly downregulates auto-tyrosine phosphorylation of Bcr-Abl. SAHA, an FDA-approved HDACi drug, at 1 μM has modest anti-tumor activity in treating CML. However, we found a synergistic interaction between SAHA and S116836 in Bcr-Abl-positive CML cells that were sensitive or resistant to IM. Exposure of KBM5 and KBM5-T315I cells to minimal or non-toxic concentrations of SAHA and S116836 synergistically reduced cell viability and induced cell death. Co-treatment with SAHA and S116838 repressed the expressions of anti-apoptosis proteins, such as Mcl-1 and XIAP, but promoted Bim expression and mitochondrial damage. Of importance, treatment with both drugs significantly reduced cell viability of primary human CML cells, as compared with either agent alone. Taken together, our findings suggest that SAHA exerts synergistically with S116836 at a non-toxic concentration to promote apoptosis in the CML, including those resistant to imatinib or dasatinib.
- Published
- 2014
- Full Text
- View/download PDF
18. PQJS380
- Author
-
Li Chen, Hongman Xue, Sheng Jiang, Chen Dong, Yiwu Yao, Chun Chen, Xiaohui Zhu, and Jingxuan Pan
- Subjects
Cancer Research ,Antineoplastic Agents ,Apoptosis ,Biology ,Pharmacology ,Pathogenesis ,Cell Line, Tumor ,Precursor cell ,medicine ,Humans ,Epigenetics ,Cell Proliferation ,Cell Cycle ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Small molecule ,Leukemia ,Oncology ,Cell culture ,Molecular Medicine ,Lymphoid Progenitor Cells ,Drug Screening Assays, Antitumor ,Azabicyclo Compounds ,Research Paper - Abstract
Acute lymphoblastic leukemia (ALL) is a malignant disorder of lymphoid progenitor cells that are committed to the B- or the T-cell lineage. The pathogenesis of ALL is heterogeneous and may be at least in part caused by genetic alterations. Although the modern sequencing technologies make it possible to rapidly discover novel genetic and epigenetic alterations and molecular targets for therapeutic intervention for ALL, conventional chemotherapy is still the most important therapeutic approach. Relapses and high morbidity and mortality remain major challenges particularly in adult patients with ALL. Therefore, development of novel chemotherapeutic agents remains in demand for ALL patients. In the course of seeking novel agents against ALL, we screened a library of small molecules and identified that PQJS380, a S-(E)-4-([7S,10S]-4-ethyl-7-isopropyl-2,5,8,12-tetraoxo-9-oxa-3,6,13,18-tetraaza-bicycle[13,2,1] octadec-1-en-10-yl)but-3-enyl octanethioate, showed potent anti-leukemia activity. PQJS380 inhibited the proliferation with IC 50 values of 14.25 nM and 5 nM in REH and NALM-6 cells, respectively. PQJS380 had 10-fold higher molar potency than the front-line ALL drugs Ara-C and VP-16. The median IC 50 value for leukemia blast cells from 17 patients with ALL was 52 nM. PQJS380 induced G 1-phase arrest in REH cells, and S-phase in NALM-6 cells, respectively. Treatment of PQJS380 led to apoptosis in ALL cell lines (REH and NALM-6) and primary ALL cells. Our data supported that PQJS380 may be a promising lead compound for ALL treatment even though the precise targets remain to be elucidated.
- Published
- 2013
- Full Text
- View/download PDF
19. Inhibitory effect of the anthelmintic drug pyrvinium pamoate on T315I BCR‑ABL‑positive CML cells
- Author
-
Yanli Jin, Jingxuan Pan, and Jing Zhang
- Subjects
0301 basic medicine ,Cancer Research ,Fusion Proteins, bcr-abl ,Chromosomal translocation ,Antineoplastic Agents ,Apoptosis ,Pharmacology ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Genetics ,medicine ,Humans ,neoplasms ,Molecular Biology ,Wnt Signaling Pathway ,Alleles ,Anthelmintics ,Oncogene ,Wnt signaling pathway ,Myeloid leukemia ,Cell cycle ,medicine.disease ,Leukemia ,030104 developmental biology ,Pyrimidines ,Oncology ,Amino Acid Substitution ,030220 oncology & carcinogenesis ,Mutation ,Molecular Medicine ,K562 Cells ,Tyrosine kinase ,K562 cells - Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder characterized by a chromosome translocation that generates the BCR‑ABL oncogene, which encodes a constitutively activated tyrosine kinase. Despite progress in controlling CML at the chronic phase by first and second generations of BCR‑ABL tyrosine kinase inhibitors (TKIs), effective drugs with good safety are not available for CML patients harboring T315I BCR‑ABL and those in advanced stages of CML. Therefore, there is an urgent requirement for the development of effective therapies against T315I BCR‑ABL. In the present study, it was demonstrated that pyrvinium pamoate, an anthelmintic drug approved by the Food and Drug Administration had potent inhibitory effects on growth and survival in CML cells with T315I BCR‑ABL. In addition, this agent was equally effective in inhibiting the Wnt/β‑catenin signaling in wild‑type and T315I BCR‑ABL CML cells. Thus, the clinical efficacy of pyrvinium pamoate in treating patients with CML bearing T315I BCR‑ABL should be further investigated.
- Published
- 2016
20. In vitro and in vivo anti-uveal melanoma activity of JSL-1, a novel HDAC inhibitor
- Author
-
Maoxing Liu, Sheng Jiang, Jingxuan Pan, Yun Wang, and Yanli Jin
- Subjects
0301 basic medicine ,Male ,Uveal Neoplasms ,Cancer Research ,Pathology ,Time Factors ,medicine.medical_treatment ,Apoptosis ,Histone Deacetylase 1 ,Mice, SCID ,Metastasis ,0302 clinical medicine ,Cell Movement ,Mice, Inbred NOD ,Depsipeptides ,Antineoplastic Combined Chemotherapy Protocols ,Cell Self Renewal ,Melanoma ,Wnt Signaling Pathway ,Bcl-2-Like Protein 11 ,Wnt signaling pathway ,Drug Synergism ,Mitochondria ,Tumor Burden ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Matrix Metalloproteinase 2 ,RNA Interference ,medicine.medical_specialty ,Antineoplastic Agents ,Biology ,Transfection ,Vinblastine ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Cell Proliferation ,Chemotherapy ,Dose-Response Relationship, Drug ,Cancer ,Aldehyde Dehydrogenase ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Cancer research - Abstract
Uveal melanoma (UM) is the most common intraocular malignant neoplasm in adults. Despite the availability of enucleation, radiation and chemotherapy, the prognosis of patients with metastasis remains poor. Therefore, novel effective therapies for patients with metastatic UM are urgently needed. In the present study, we demonstrated that JSL-1, a novel HDAC inhibitor, effectively inhibited the proliferation. JSL-1 induced apoptosis with increased expression of proapoptotic BH3-only protein BIM in UM cells. JSL-1 suppressed migration and invasion of UM cells with MMP-2 decreased. Furthermore, JSL-1 blocked the canonical Wnt/β-catenin pathway, impaired self-renewal capacity and decreased percentage of ALDH+ cells, thereby reflecting elimination of UM cancer stem-like cells (CSCs) which are believed seeds of metastasis. Importantly, JSL-1 potently inhibited the growth of uveal melanoma xenograft in NOD-SCID mice. These results suggested that JSL-1 may be a promising therapeutic agent for UM.
- Published
- 2016
21. Gas6/AXL Signaling Regulates Self-Renewal of Chronic Myelogenous Leukemia Stem Cells by Stabilizing β-Catenin
- Author
-
Yuhong Lu, Chang Liu, Danian Nie, Juan Li, Jingxuan Pan, Yanli Jin, Yangqiu Li, Jingfeng Zhou, and Xin Du
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,CD34 ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Cell Self Renewal ,neoplasms ,beta Catenin ,Cell Proliferation ,GAS6 ,Mesenchymal stem cell ,Receptor Protein-Tyrosine Kinases ,medicine.disease ,Flow Cytometry ,Xenograft Model Antitumor Assays ,Axl Receptor Tyrosine Kinase ,Gene Expression Regulation, Neoplastic ,Leukemia ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Imatinib Mesylate ,Neoplastic Stem Cells ,Intercellular Signaling Peptides and Proteins ,Stem cell ,Tyrosine kinase ,Chronic myelogenous leukemia ,Signal Transduction - Abstract
Purpose: Quiescent leukemia stem cells (LSC) are important resources of resistance and relapse in chronic myelogenous leukemia (CML). Thus, strategies eradicating CML LSCs are required for cure. In this study, we discovered that AXL tyrosine kinase was selectively overexpressed in primary CML CD34+ cells. However, the role of AXL and its ligand Gas6 secreted by stromal cells in the regulation of self-renewal capacity of LSCs has not been well investigated. Experimental Design: The function of CML CD34+ cells was evaluated by flow cytometer, CFC/replating, long-term culture-initiating cells (LTC-IC), CML mouse model driven by human BCR-ABL gene and NOD-scid-IL2Rg−/− (NSI) mice. Results: AXL was selectively overexpressed in primary CML CD34+ cells. AXL knockdown reduced the survival and self-renewal capacity of human CML CD34+ cells. Pharmacologic inhibition of AXL reduced the survival and self-renewal capacity of human CML LSCs in vitro and in long-term grafts in NSI mice. Human CML CD34+ cells conscripted bone marrow–derived stromal cells (BMDSC) and primary mesenchymal stem cells (MSC) to secrete Gas6 to form a paracrine loop that promoted self-renewal of LSCs. Suppression of AXL by shRNA and inhibitor prolonged survival of CML mice and reduced the growth of LSCs in mice. Gas6/AXL ligation stabilizes β-catenin in an AKT-dependent fashion in human CML CD34+ cells. Conclusions: Our findings improve the understanding of LSC regulation and validate Gas6/AXL as a pair of therapeutic targets to eliminate CML LSCs. Clin Cancer Res; 23(11); 2842–55. ©2016 AACR.
- Published
- 2016
22. Abstract 1980: S116836 overcomes BCR-ABL T315I mutation mediated imatinib resistance in chronic myeloid leukemia
- Author
-
Brian J. Druker, Jingxuan Pan, Ke Ding, Zhe-Sheng Chen, Pranav Gupta, Anna Maria Barbuti, and Guan-Nan Zhang
- Subjects
Cancer Research ,education.field_of_study ,Oncogene ,business.industry ,Cell ,Population ,Cancer ,Myeloid leukemia ,Imatinib ,Cell cycle ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Apoptosis ,hemic and lymphatic diseases ,Cancer research ,Medicine ,business ,education ,neoplasms ,medicine.drug - Abstract
Chronic Myeloid Leukemia (CML) is largely caused by the Philadelphia (Ph) chromosome carrying the Break point Cluster Region-Abelson (BCR-ABL) oncogene. Imatinib is a BCR-ABL-targeted therapy and considered the standard of care in CML management. Resistance to imatinib therapy often develops because of mutations in the BCR-ABL kinase domain. In this study, we evaluated S116836, a novel BCR-ABL inhibitor, for its anti-cancer activity against BCR-ABL expressing BaF3 cells. S116836 shows potent activity against wild-type and T315I mutated BaF3 cells as compared with imatinib. S116836 inhibited the phosphorylation of BCR-ABL and its downstream signaling in BaF3/WT and BaF3/T315I cells. S116836 inhibited the mRNA expression of BCR-ABL in BaF3/WT and BaF3/T315I cells. Mechanistically, S116836 increased the cell population in the G0/G1 phase of the cell cycle, induced apoptosis and elevated ROS production in both BaF3/WT and BaF3/T315I cells. Moreover, S116836 inhibited tumor growth in BaF3/WT and BaF3/T315I tumor xenografts. Taken together, our results indicate that S116836 exhibits anti-proliferative effects and inhibits the imatinib-resistant T315I BCR-ABL mutation. S116836 may be a novel drug candidate for overcoming the resistance to imatinib in CML patients. Keywords: S116836, Chronic myeloid leukemia, BCR-ABL, T315I, Resistance Citation Format: Pranav Gupta, Guannan Zhang, Anna Maria Barbuti, Ke Ding, Jingxuan Pan, Brian J. Druker, Zhe-Sheng Chen. S116836 overcomes BCR-ABL T315I mutation mediated imatinib resistance in chronic myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1980.
- Published
- 2018
- Full Text
- View/download PDF
23. The BH3-mimetic GX15-070 induces autophagy, potentiates the cytotoxicity of carboplatin and 5-fluorouracil in esophageal carcinoma cells
- Author
-
Qi Chen, Srdan Verstovsek, Chao Cheng, Yanli Jin, Jingxuan Pan, and Qi Cao
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Indoles ,Esophageal Neoplasms ,medicine.medical_treatment ,Carboplatin ,Targeted therapy ,chemistry.chemical_compound ,Cell Line, Tumor ,Internal medicine ,Autophagy ,medicine ,Carcinoma ,Humans ,Pyrroles ,Esophagus ,Cytotoxicity ,Cell Proliferation ,Osteosarcoma ,business.industry ,Drug Synergism ,Esophageal cancer ,medicine.disease ,medicine.anatomical_structure ,chemistry ,Cancer research ,Drug Therapy, Combination ,Fluorouracil ,business ,Signal Transduction - Abstract
Despite improvements in both surgical techniques and radio- and chemo-therapy regimens, the prognosis of esophageal cancer is poor. In pursuit of novel effective strategy, this study examined the effect of the BH3-mimetic GX15-070 on esophageal carcinoma cells. We discovered that GX15-070 inhibited the growth of esophageal cancer cells. There was synergism between GX15-070 and carboplatin or 5-fluorouracil. GX15-070 induced autophagy in esophagus cancer cell line EC9706 and osteosarcoma cancer cell line U2OS. 3-methyladenine and chloroquine, inhibitors of autophagy with distinct mechanisms, potentiated the cytotoxicity of GX15-070. In conclusion, GX15-070 inhibits growth of esophageal cancer cells.
- Published
- 2010
- Full Text
- View/download PDF
24. RNA polymerase – An important molecular target of triptolide in cancer cells
- Author
-
Jingxuan Pan
- Subjects
Cancer Research ,Inflammation ,Pharmacology ,Biology ,chemistry.chemical_compound ,Neoplasms ,RNA polymerase ,medicine ,Animals ,Humans ,Cytotoxicity ,Cancer ,DNA-Directed RNA Polymerases ,Oncogenes ,Phenanthrenes ,Triptolide ,medicine.disease ,biology.organism_classification ,In vitro ,Oncology ,chemistry ,Cancer cell ,Epoxy Compounds ,Tripterygium wilfordii ,Diterpenes ,medicine.symptom ,Cell Division - Abstract
Triptolide, a diterpenoid triepoxide, is the key biological component of Tripterygium wilfordii Hook. f. which was used in traditional Chinese medicine for centuries to treat inflammation and autoimmune diseases. Triptolide has shown potent activity in not only anti-inflammation and immune modulation, but also antiproliferative and proapoptotic activity in many different types of cancer cells. However, for a long time, the precise molecular target(s) of triptolide have remained elusive. Recently, several groups discovered that triptolide inhibited the activity of RNA polymerase. This review will focus on these breakthrough findings about the molecular target of triptolide and its implications for targeted-cancer therapeutics.
- Published
- 2010
- Full Text
- View/download PDF
25. Sustained c-Jun-NH2-Kinase Activity Promotes Epithelial-Mesenchymal Transition, Invasion, and Survival of Breast Cancer Cells by Regulating Extracellular Signal-Regulated Kinase Activation
- Author
-
Jinhua Wang, Jingxuan Pan, Xiaojiang Cui, Armando E. Giuliano, Isere Kuiatse, and Adrian V. Lee
- Subjects
MAPK/ERK pathway ,Cancer Research ,Paclitaxel ,Cell Survival ,Antineoplastic Agents ,Breast Neoplasms ,Biology ,Article ,Mesoderm ,Mice ,Cell Movement ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Cell Lineage ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Kinase activity ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Kinase ,Carcinoma ,c-jun ,JNK Mitogen-Activated Protein Kinases ,Cancer ,Epithelial Cells ,Cell Dedifferentiation ,medicine.disease ,Up-Regulation ,Cell biology ,Enzyme Activation ,Transcription Factor AP-1 ,Cell Transformation, Neoplastic ,Oncology ,Cancer cell ,Insulin Receptor Substrate Proteins ,Cancer research ,Female ,Proto-Oncogene Proteins c-fos - Abstract
The c-Jun NH2-terminus kinase (JNK) mediates stress-induced apoptosis and the cytotoxic effect of anticancer therapies. Paradoxically, recent clinical studies indicate that elevated JNK activity in human breast cancer is associated with poor prognosis. Here, we show that overexpression of a constitutively active JNK in human breast cancer cells did not cause apoptosis, but actually induced cell migration and invasion, a morphologic change associated with epithelial-mesenchymal transition (EMT), expression of mesenchymal-specific markers vimentin and fibronectin, and activity of activator protein transcription factors. Supporting this observation, mouse mammary tumor cells that have undergone EMT showed upregulated JNK activity, and the EMT was reversed by JNK inhibition. Sustained JNK activity enhanced insulin receptor substrate-2–mediated ERK activation, which in turn increased c-Fos expression and activator protein activity. In addition, hyperactive JNK attenuated the apoptosis of breast cancer cells treated by the chemotherapy drug paclitaxel, which is in contrast to the requirement for inducible JNK activity in response to cytotoxic chemotherapy. Blockade of extracellular signal-regulated kinase activity diminished hyperactive JNK-induced cell invasion and survival. Our data suggest that the role of JNK changes when its activity is elevated persistently above the basal levels associated with cell apoptosis, and that JNK activation may serve as a marker of breast cancer progression and resistance to cytotoxic drugs. Mol Cancer Res; 8(2); 266–77
- Published
- 2010
- Full Text
- View/download PDF
26. Triptolide Inhibits Bcr-Abl Transcription and Induces Apoptosis in STI571-resistant Chronic Myelogenous Leukemia Cells Harboring T315I Mutation
- Author
-
Waiyi Zou, Qin Zheng, Jingxuan Pan, Yingrong Lai, Qi Chen, Yanli Jin, Hui Zhang, Xianping Shi, Zhongzheng Lu, and Chao Cheng
- Subjects
Male ,Cancer Research ,Transcription, Genetic ,Fusion Proteins, bcr-abl ,Apoptosis ,Piperazines ,Tyrosine-kinase inhibitor ,Immunoenzyme Techniques ,Mice ,chemistry.chemical_compound ,Nude mouse ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,Tumor Stem Cell Assay ,Membrane Potential, Mitochondrial ,Mice, Inbred BALB C ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Cycle ,Middle Aged ,Protein-Tyrosine Kinases ,Flow Cytometry ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,Benzamides ,Imatinib Mesylate ,Female ,Diterpenes ,Signal Transduction ,medicine.drug ,Adult ,Adolescent ,Tripterygium ,medicine.drug_class ,Blotting, Western ,Mice, Nude ,Philadelphia chromosome ,Young Adult ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Survivin ,medicine ,Animals ,Humans ,RNA, Messenger ,Antineoplastic Agents, Alkylating ,Protein Kinase Inhibitors ,neoplasms ,Aged ,Cell Proliferation ,Imatinib ,Phenanthrenes ,Triptolide ,biology.organism_classification ,medicine.disease ,Xenograft Model Antitumor Assays ,Pyrimidines ,Imatinib mesylate ,chemistry ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,Epoxy Compounds ,Myeloid Cell Leukemia Sequence 1 Protein ,Reactive Oxygen Species ,Proto-Oncogene Proteins c-akt ,Chronic myelogenous leukemia - Abstract
Purpose: Resistance to STI571 is an emerging problem for patients with chronic myelogenous leukemia (CML). Mutation in the kinase domain of Bcr-Abl is the predominant mechanism of the acquired resistance to STI571. In the present study, we investigated the effect of triptolide on cell survival or apoptosis in CML cells bearing Bcr-Abl-T315I or wild-type Bcr-Abl. Experimental Design: CML cell lines (KBM5 versus KBM5-T315I, BaF3-Bcr-Abl versus BaF3-Bcr-Abl-T315I) and primary cells from CML patients with clinical resistance to STI571 were treated with triptolide, and analyzed in terms of growth, apoptosis, and signal transduction. Nude mouse xenograft model was also used to evaluate the antitumor activity. Results: Triptolide potently down-regulated the mRNA and protein levels of Bcr-Abl independently of the caspase or proteosome activation in CML cells. It induced mitochondrial-dependent apoptosis in Bcr-Abl-T315I CML cells and primary cells from CML patients with clinical resistance to STI571. Additionally, triptolide inhibited the growth of STI571-sensitive KBM5 and STI571-resistant KBM5-T315I CML cells in nude mouse xenografts. Triptolide also down-regulated the expression of survivin, Mcl-1, and Akt in CML cells, which suggests that it may have multiple targets. Conclusions: These findings suggest that triptolide is a promising agent to overcome STI571-resistant CML cells, and warrant a clinical trial of triptolide derivatives for CML with Bcr-Abl-T315I mutation.
- Published
- 2009
- Full Text
- View/download PDF
27. Recent Advances in Understanding the Antineoplastic Mechanisms of Farnesyltransferase Inhibitors
- Author
-
Sai Ching Jim Yeung and Jingxuan Pan
- Subjects
Cancer Research ,Farnesyltranstransferase ,biology ,Farnesylation Inhibition ,Farnesyltransferase ,RHOB ,Antineoplastic Agents ,Cell cycle ,medicine.disease_cause ,Oncology ,Biochemistry ,Cancer cell ,biology.protein ,Cancer research ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Carcinogenesis ,RHEB - Abstract
Farnesyltransferase (FTase) inhibitors (FTI) have broad antineoplastic actions targeting both cancer cells and mesenchymal cells involved in tumor angiogenesis. The small GTPases H-Ras, Rheb, and RhoB and the centromere proteins CENP-E and CENP-F are relevant targets of farnesylation inhibition; however, their relative importance in the antineoplastic effect of FTIs may vary in different cell types at different stages of the cell cycle and at different stages in oncogenesis. Three recent studies argue that Ras-independent and perhaps even FTase-independent properties are important to the antineoplastic action of this class of drugs. In mice, genetic ablation of FTase does not abolish the oncogenic activity of Ras, limiting the original conception of FTIs as an effective means to target Ras in cancer cells. FTase may not be the sole molecular target of these agents, and one study has suggested that FTIs act by targeting geranylgeranyl transferase II. Lastly, we have obtained evidence that induction of reactive oxygen species and reactive oxygen species–mediated DNA damage by FTIs may be critical for their antineoplastic action as a class. Together, these findings may alter thinking about how to apply FTIs in the clinic.
- Published
- 2005
- Full Text
- View/download PDF
28. Farnesyltransferase Inhibitors Induce DNA Damage via Reactive Oxygen Species in Human Cancer Cells
- Author
-
Zhi-Xiang Xu, Miaorong She, Jingxuan Pan, Lily Sun, and Sai Ching J. Yeung
- Subjects
Cancer Research ,Polyunsaturated Alkamides ,DNA repair ,DNA damage ,RHOB ,Farnesyltransferase ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,DNA-Activated Protein Kinase ,Polyenes ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Cell Line, Tumor ,Neoplasms ,medicine ,Farnesyltranstransferase ,Humans ,Enzyme Inhibitors ,Nuclear protein ,rhoB GTP-Binding Protein ,Alkyl and Aryl Transferases ,biology ,Tumor Suppressor Proteins ,Nuclear Proteins ,DNA, Neoplasm ,HCT116 Cells ,DNA-Binding Proteins ,Non-homologous end joining ,Oncology ,Apoptosis ,biology.protein ,Cancer research ,Reactive Oxygen Species ,Oxidative stress ,DNA Damage - Abstract
Farnesyltransferase inhibitors (FTIs) possess antitumor activity. Based on recent findings, we hypothesized that FTIs induce reactive oxygen species (ROS) that damage DNA, leading to DNA damage responses. To test this hypothesis, we investigated the effects of FTIs on the generation of ROS, DNA double-strand breaks (DSB), DNA damage responses, and RhoB, and the effects of quenching ROS on these FTI effects. We evaluated four FTIs in human cancer cell lines of different tissue origins. We found that FTIs induced ROS and DSBs. Suppressing expression of the β-subunit of farnesyltransferase with siRNA did not induce ROS, but slightly attenuated the ROS induced by FTIs. N-acetyl-l-cysteine (NAC), but not caspase inhibitors, blocked FTI-induced DSBs, suggesting that the DSBs were caused by ROS and did not result from apoptosis. The DSBs led to DNA damage responses. H2AX became phosphorylated and formed nuclear foci. The DNA-damage-sensing molecules involved were probably ataxia-telangiectasia mutated protein (ATM) and DNA-dependent protein kinase (DNA-PK) but not ATM- and Rad3-related protein (ATR). Key components of the homologous recombination and nonhomologous end joining repair pathways (DNA-PK, BRCA1, and NBS1) underwent phosphorylation and formed nuclear foci. RhoB, a mediator of the antineoplastic effect of FTIs and a protein inducible by DNA damage, was increased by FTIs. This increase was blocked by NAC. We concluded that FTIs induced oxidative DNA damage by inducing ROS and initiated DNA damage responses, including RhoB induction, and there was a complex relationship among FTIs, farnesyltransferase, ROS, and RhoB. Our data also imply that inhibitors of DNA repair may accentuate the clinical efficacy of FTIs.
- Published
- 2005
- Full Text
- View/download PDF
29. Cytokine-induced apoptosis inhibitor 1 inhibits the growth and proliferation of multiple myeloma
- Author
-
Juan Li, Xiaobo Wang, and Jingxuan Pan
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_treatment ,Cell ,Down-Regulation ,Mice, Nude ,Biology ,Biochemistry ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Insulin-Like Growth Factor I ,Molecular Biology ,Cell Proliferation ,Mice, Inbred BALB C ,Cell growth ,Growth factor ,Cell Cycle ,Intracellular Signaling Peptides and Proteins ,Transfection ,Cell cycle ,Middle Aged ,Molecular biology ,Cell biology ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,Cell culture ,Molecular Medicine ,Female ,Bone marrow ,Multiple Myeloma ,A431 cells - Abstract
The present study investigated the differential expression of cytokine-induced apoptosis inhibitor 1 (CIAPIN1) in human multiple myeloma (MM) bone marrow tissue and adjacent healthy bone marrow tissue. In addition, the effect of a transduced CIAPIN1 gene on the growth of the RPMI-8226 human MM cell line was investigated. CIAPIN1 protein expression was detected in 32 samples of paraffin-embedded MM and adjacent healthy bone marrow tissue using immunohistochemistry. The CIAPIN1 gene (Ad-CIAPIN1, small interfering RNA) was inserted into a lentiviral vector and transfected into the RPMI-8226 human MM cell line. The expression of target proteins CIAPIN1 and insulin-like growth factor 1 U (IGF-1), cell cycle-regulatory proteins and functional proteins was detected using western blotting. MTT and soft agar colony formation assays were conducted, and cellular tumorigenicity in nude mice was assessed, in order to investigate the proliferative capacity of cells in vitro and in vivo. Flow cytometry was applied in order to analyze changes in the cell cycle and cell apoptosis. CIAPIN1 expression was significantly reduced in cells from the 32 MM samples compared with those from healthy bone marrow (P
- Published
- 2014
30. Ponatinib induces apoptosis in imatinib-resistant human mast cells by dephosphorylating mutant D816V KIT and silencing β-catenin signaling
- Author
-
Ke Ding, Jingxuan Pan, and Bei Jin
- Subjects
Male ,Cancer Research ,Drug Resistance ,Antineoplastic Agents ,Apoptosis ,Cell Line ,chemistry.chemical_compound ,Mice ,Nude mouse ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Point Mutation ,Gene Silencing ,Mast Cells ,Systemic mastocytosis ,Phosphorylation ,Protein Kinase Inhibitors ,Wnt Signaling Pathway ,beta Catenin ,Cell Proliferation ,biology ,Kinase ,Ponatinib ,Imidazoles ,Cytochromes c ,Imatinib ,biology.organism_classification ,medicine.disease ,Mast cell leukemia ,Xenograft Model Antitumor Assays ,Pyridazines ,Disease Models, Animal ,Proto-Oncogene Proteins c-kit ,Oncology ,chemistry ,Gene Expression Regulation ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,Signal transduction ,Apoptosis Regulatory Proteins ,TCF Transcription Factors ,Tyrosine kinase ,Mastocytosis ,medicine.drug ,Protein Binding ,Signal Transduction - Abstract
Gain-of-function mutations of membrane receptor tyrosine kinase KIT, especially gatekeeper D816V point mutation in KIT, render kinase autoactivation, disease progression, and poor prognosis. D816V KIT is found in approximately 80% of the patients with systemic mastocytosis, and is resistant to the first and second generations of tyrosine kinase inhibitors (TKI). The purpose of this investigation was aimed at exploring whether ponatinib (AP24534), a novel effective TKI against T315I Bcr-Abl, was active against D816V KIT. We discovered that ponatinib abrogated the phosphorylation of KIT harboring either V560G (sensitive to imatinib) or D816V mutation (resistant to imatinib) and the downstream signaling transduction. Ponatinib inhibited the growth of D816V KIT–expressing cells in culture and nude mouse xenografted tumor. Ponatinib triggered apoptosis by inducing the release of cytochrome c and AIF, downregulation of Mcl-1. Furthermore, ponatinib abrogated the phosphorylation of β-catenin at the site Y654, suppressed the translocation of β-catenin, and inhibited the transcription and DNA binding of TCF and the expression of its targets (e.g., AXIN2, c-MYC, and CCND1). Moreover, ponatinib was highly active against xenografted D816V KIT tumors in nude mice and significantly prolonged the survival of mice with aggressive systemic mastocytosis or mast cell leukemia by impeding the expansion and infiltration of mast cells with imatinib-resistant D814Y KIT. Our findings warrant a clinical trial of ponatinib in patients with systemic mastocytosis harboring D816V KIT. Mol Cancer Ther; 13(5); 1217–30. ©2014 AACR.
- Published
- 2014
31. Sensitivity of human cells bearing oncogenic mutant kit isoforms to the novel tyrosine kinase inhibitor INNO-406
- Author
-
Srdan Verstovsek, Jingxuan Pan, Hagop M. Kantarjian, Alfonso Quintás-Cardama, Jorge E. Cortes, and Taghi Manshouri
- Subjects
Cancer Research ,medicine.drug_class ,Apoptosis ,Biology ,Piperazines ,Tyrosine-kinase inhibitor ,medicine ,Humans ,Protein Isoforms ,Mast Cells ,Phosphorylation ,Protein Kinase Inhibitors ,Cell Proliferation ,Cell growth ,Imatinib ,General Medicine ,Protein-Tyrosine Kinases ,Mast cell ,Proto-Oncogene Proteins c-kit ,Pyrimidines ,medicine.anatomical_structure ,Imatinib mesylate ,Oncology ,Protein kinase domain ,Benzamides ,Mutation ,Imatinib Mesylate ,Cancer research ,medicine.drug - Abstract
The activity of the novel tyrosine kinase inhibitor INNO-406 against human cells with mutated KIT was investigated. Human mast cell (HMC)-1.1 cells with juxtamembrane domain mutation V560G, and HMC-1.2 cells with both V560G and the kinase domain mutation D816V, were treated with INNO-406 (0.02-5.00 microM) or imatinib for 72 h. INNO-406 and imatinib were equipotent against HMC-1 cells regarding cell proliferation (IC50 51 nM and 75 nM, respectively), inhibition of KIT phosphorylation, and induction of apoptosis. In contrast, neither drug was effective against HMC-1.2 cells at the dose range tested. The present results suggest clinical potential for INNO-406 in KIT V560G-expressing malignancies.
- Published
- 2007
- Full Text
- View/download PDF
32. The conformational control inhibitor of tyrosine kinases DCC-2036 is effective for imatinib-resistant cells expressing T674I FIP1L1-PDGFRα
- Author
-
Xiaoke Shi, Jingxuan Pan, and Yingying Shen
- Subjects
Receptor, Platelet-Derived Growth Factor alpha ,Oncogene Proteins, Fusion ,Cancer Treatment ,lcsh:Medicine ,Apoptosis ,Piperazines ,Hematologic Cancers and Related Disorders ,Mice ,Nude mouse ,Molecular cell biology ,RNA interference ,hemic and lymphatic diseases ,Signaling in Cellular Processes ,Phosphorylation ,lcsh:Science ,Apoptotic Signaling ,Multidisciplinary ,biology ,Bcl-2-Like Protein 11 ,Protein translation ,Cell Death ,Gene Expression Regulation, Leukemic ,Mechanisms of Signal Transduction ,Antiapoptotic Signaling ,Hematology ,Signaling Cascades ,Tumor Burden ,Oncology ,Benzamides ,Imatinib Mesylate ,Quinolines ,Medicine ,Signal transduction ,Tyrosine kinase ,medicine.drug ,Signal Transduction ,Research Article ,Drugs and Devices ,Phosphoinositide Signal Transduction ,Signaling Pathways ,Cell Growth ,Growth factor receptor ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,Proto-Oncogene Proteins ,Leukemias ,medicine ,Animals ,Humans ,Codon ,Protein Kinase Inhibitors ,Biology ,Cell Proliferation ,mRNA Cleavage and Polyadenylation Factors ,Cell growth ,fungi ,lcsh:R ,Membrane Proteins ,Cancers and Neoplasms ,Imatinib ,Chemotherapy and Drug Treatment ,biology.organism_classification ,Molecular biology ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Imatinib mesylate ,Pyrimidines ,Cell culture ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,lcsh:Q ,Gene expression ,Apoptosis Regulatory Proteins - Abstract
The cells expressing the T674I point mutant of FIP1-like-1-platelet-derived growth factor receptor alpha (FIP1L1-PDGFRα) in hypereosinophilics syndrome (HES) are resistant to imatinib and some second-generation tyrosine kinase inhibitors (TKIs). There is a desperate need to develop therapy to combat this acquired drug resistance. DCC-2036 has been synthesized as a third-generation TKI to combat especially the Bcr-Abl T315I mutant in chronic myeloid leukemia. This study evaluated the effect of DCC-2036 on FIP1L1-PDGFRα-positive cells, including the wild type (WT) and the T674I mutant. The in vitro effects of DCC-2036 on the PDGFRα signal pathways, proliferation, cell cycling and apoptosis of FIP1L1-PDGFRα-positive cells were investigated, and a nude mouse xenograft model was employed to assess the in vivo antitumor activity. We found that DCC-2036 decreased the phosphorylated levels of PDGFRα and its downstream targets without apparent effects on total protein levels. DCC-2036 inhibited proliferation, and induced apoptosis with MEK-dependent up-regulation of the pro-apoptotic protein Bim in FIP1L1-PDGFRα-positive cells. DCC-2036 also exhibited in vivo antineoplastic activity against cells with T674I FIP1L1-PDGFRα. In summary, FIP1L1-PDGFRα-positive cells are sensitive to DCC-2036 regardless of their sensitivity to imatinib. DCC-2036 may be a potential compound to treat imatinib-resistant HES.
- Published
- 2013
33. Cyclin-dependent kinase 7/9 inhibitor SNS-032 abrogates FIP1-like-1 platelet-derived growth factor receptor α and bcr-abl oncogene addiction in malignant hematologic cells
- Author
-
Yongbin Wu, Sai Ching J. Yeung, Chun Chen, Jingxuan Pan, Ke Ding, Xianping Shi, Bei Jin, and Xiaoyong Sun
- Subjects
Male ,Cancer Research ,Receptor, Platelet-Derived Growth Factor alpha ,Oncogene Proteins, Fusion ,Blotting, Western ,Fusion Proteins, bcr-abl ,Mice, Nude ,Apoptosis ,Cell Line ,Mice ,Growth factor receptor ,Cyclin-dependent kinase ,hemic and lymphatic diseases ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Animals ,Humans ,Phosphorylation ,Oxazoles ,Cell Proliferation ,mRNA Cleavage and Polyadenylation Factors ,Mice, Inbred BALB C ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Cycle ,Imatinib ,Oncogene Addiction ,Cyclin-Dependent Kinase 9 ,Xenograft Model Antitumor Assays ,Cyclin-Dependent Kinases ,Thiazoles ,Oncology ,biology.protein ,Cancer research ,Cyclin-dependent kinase 9 ,Female ,Signal transduction ,Tyrosine kinase ,Platelet-derived growth factor receptor ,Cyclin-Dependent Kinase-Activating Kinase ,medicine.drug ,Signal Transduction - Abstract
Purpose: The “gate-keeper” mutations T674I platelet—derived growth factor receptor α (PDGFRα) in hypereosinophilic syndrome (HES) and T315I Bcr-Abl in chronic myeloid leukemia (CML) are resistant to imatinib and the second-generation small-molecule tyrosine kinase inhibitors (TKI). However, to combat acquired resistance to imatinib, an alternative approach is to decrease the expression of the addicted gene to efficiently kill resistant malignant hematologic cells. The purpose of this study was to evaluate the strategy of shutting down the transcription and expression of FIP1-like-1 (FIP1L1)–PDGFRα and Bcr-Abl with SNS-032, an inhibitor of cyclin-dependent kinase 7 (CDK7) and CDK9 in phase I clinical trials. Experimental Design: The effects of SNS-032 on PDGFRα and Bcr-Abl signaling pathways, apoptosis, and cell cycling were analyzed in TKI-resistant cells of HES and CML. The in vivo antitumor activity of SNS-032 was assessed with xenografted BaF3-T674I FIP1L1-PDGFRα and KBM5-T315I Bcr-Abl cells in nude mouse models. Results: SNS-032 inhibited the phosphorylation on Ser5 and Ser2 of RNA polymerase II. SNS-032 decreased both the mRNA and protein levels of FIP1L1-PDGFRα and Bcr-Abl and inhibited the proliferation of malignant cells expressing FIP1L1-PDGFRα or Bcr-Abl. It also decreased the phosphorylation of downstream molecules. It induced apoptosis by triggering both the mitochondrial pathway and the death receptor pathway. Conclusions: This CDK7/9 inhibitor potently inhibits FIP1L1-PDGFRα–positive HES cells and Bcr-Abl–positive CML cells regardless of their sensitivity to imatinib. SNS-032 may have potential in treating hematologic malignancy by abrogating oncogene addiction. Clin Cancer Res; 18(7); 1966–78. ©2012 AACR.
- Published
- 2012
34. Niclosamide, an old antihelminthic agent, demonstrates antitumor activity by blocking multiple signaling pathways of cancer stem cells
- Author
-
Jingxuan Pan, Ke Ding, and Cheng-Yan Wang
- Subjects
Drug ,STAT3 Transcription Factor ,cancer stem cells ,media_common.quotation_subject ,Antineoplastic Agents ,mTORC1 ,Review ,Biology ,Pharmacology ,Mechanistic Target of Rapamycin Complex 1 ,Cancer stem cell ,Cell Movement ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,cancer ,Neoplasm Metastasis ,Wnt Signaling Pathway ,Niclosamide ,media_common ,Cell Proliferation ,Tapeworm infection ,Receptors, Notch ,TOR Serine-Threonine Kinases ,Wnt signaling pathway ,NF-kappa B ,Antihelminthic Agent ,medicine.disease ,Oncology ,Multiprotein Complexes ,Cancer cell ,Neoplastic Stem Cells ,Reactive Oxygen Species ,signal transduction ,medicine.drug - Abstract
Niclosamide, an oral antihelminthic drug, has been used to treat tapeworm infection for about 50 years. Niclosamide is also used as a molluscicide for water treatment in schistosomiasis control programs. Recently, several groups have independently discovered that niclosamide is also active against cancer cells, but its precise mechanism of antitumor action is not fully understood. Evidence supports that niclosamide targets multiple signaling pathways (NF-κB, Wnt/β-catenin, Notch, ROS, mTORC1, and Stat3), most of which are closely involved with cancer stem cells. The exciting advances in elucidating the antitumor activity and the molecular targets of this drug will be discussed. A method for synthesizing a phosphate pro-drug of niclosamide is provided. Given its potential antitumor activity, clinical trials for niclosamide and its derivatives are warranted for cancer treatment.
- Published
- 2012
35. Pristimerin induces apoptosis in imatinib-resistant chronic myelogenous leukemia cells harboring T315I mutation by blocking NF-κB signaling and depleting Bcr-Abl
- Author
-
Zhongzheng Lu, Chun Chen, Jingxuan Pan, Juan Li, Yanli Jin, and Qi Cao
- Subjects
Male ,Cancer Research ,Transcription, Genetic ,Fusion Proteins, bcr-abl ,Fluorescent Antibody Technique ,Gene Expression ,Apoptosis ,Electrophoretic Mobility Shift Assay ,Cell Separation ,Piperazines ,Mice ,hemic and lymphatic diseases ,RNA, Small Interfering ,Child ,NF-kappa B ,Middle Aged ,Flow Cytometry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunohistochemistry ,Leukemia ,Oncology ,Benzamides ,Imatinib Mesylate ,Molecular Medicine ,Signal transduction ,Pentacyclic Triterpenes ,Tyrosine kinase ,Signal Transduction ,medicine.drug ,Adult ,Adolescent ,Mice, Nude ,Antineoplastic Agents ,Biology ,Transfection ,lcsh:RC254-282 ,Young Adult ,Myelogenous ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Animals ,Humans ,Point Mutation ,neoplasms ,Cell Proliferation ,Research ,Correction ,Imatinib ,medicine.disease ,Xenograft Model Antitumor Assays ,Molecular biology ,Fusion protein ,Triterpenes ,Enzyme Activation ,Pyrimidines ,Imatinib mesylate ,Drug Resistance, Neoplasm ,Cancer research ,Chronic myelogenous leukemia - Abstract
Background Chronic myelogenous leukemia (CML) is characterized by the chimeric tyrosine kinase Bcr-Abl. Bcr-Abl-T315I is the notorious point mutation that causes resistance to imatinib and the second generation tyrosine kinase inhibitors, leading to poor prognosis. CML blasts have constitutive p65 (RelA NF-κB) transcriptional activity, and NF-κB may be a potential target for molecular therapies in CML that may also be effective against CML cells with Bcr-Abl-T315I. Results In this report, we discovered that pristimerin, a quinonemethide triterpenoid isolated from Celastraceae and Hippocrateaceae, inhibited growth and induced apoptosis in CML cells, including the cells harboring Bcr-Abl-T315I mutation. Additionally, pristimerin inhibited the growth of imatinib-resistant Bcr-Abl-T315I xenografts in nude mice. Pristimerin blocked the TNFα-induced IκBα phosphorylation, translocation of p65, and expression of NF-κB-regulated genes. Pristimerin inhibited two steps in NF-κB signaling: TAK1→IKK and IKK→IκBα. Pristimerin potently inhibited two pairs of CML cell lines (KBM5 versus KBM5-T315I, 32D-Bcr-Abl versus 32D-Bcr-Abl-T315I) and primary cells from a CML patient with acquired resistance to imatinib. The mRNA and protein levels of Bcr-Abl in imatinib-sensitive (KBM5) or imatinib-resistant (KBM5-T315I) CML cells were reduced after pristimerin treatment. Further, inactivation of Bcr-Abl by imatinib pretreatment did not abrogate the TNFα-induced NF-κB activation while silencing p65 by siRNA did not affect the levels of Bcr-Abl, both results together indicating that NF-κB inactivation and Bcr-Abl inhibition may be parallel independent pathways. Conclusion To our knowledge, this is the first report to show that pristimerin is effective in vitro and in vivo against CML cells, including those with the T315I mutation. The mechanisms may involve inhibition of NF-κB and Bcr-Abl. We concluded that pristimerin could be a lead compound for further drug development to overcome imatinib resistance in CML patients.
- Published
- 2010
36. Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-kappaB pathway and generation of reactive oxygen species
- Author
-
Yongbin Wu, Chun Chen, Juan Li, Jingxuan Pan, Ke Ding, Xiaoyong Sun, Yanli Jin, Zhongzheng Lu, Xin Du, and Jing Zhou
- Subjects
Adult ,Male ,Cancer Research ,Myeloid ,Transcription, Genetic ,Daunorubicin ,Antineoplastic Agents ,HL-60 Cells ,Biology ,Protein Serine-Threonine Kinases ,Mice ,NF-KappaB Inhibitor alpha ,Genes, Reporter ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,neoplasms ,Niclosamide ,Mice, Inbred BALB C ,Tumor Necrosis Factor-alpha ,NF-kappa B ,Transcription Factor RelA ,Drug Synergism ,DNA, Neoplasm ,U937 Cells ,medicine.disease ,MAP Kinase Kinase Kinases ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,Apoptosis ,Immunology ,Cancer research ,Cytarabine ,Neoplastic Stem Cells ,Tumor necrosis factor alpha ,I-kappa B Proteins ,Stem cell ,Reactive Oxygen Species ,medicine.drug - Abstract
NF-κB may be a potential therapeutic target for acute myelogenous leukemia (AML) because NF-κB activation is found in primitive human AML blast cells. In this report, we initially discovered that the potent antineoplastic effect of niclosamide, a Food and Drug Administration–approved antihelminthic agent, was through inhibition of the NF-κB pathway in AML cells. Niclosamide inhibited the transcription and DNA binding of NF-κB. It blocked tumor necrosis factor–induced IκBα phosphorylation, translocation of p65, and expression of NF-κB–regulated genes. Niclosamide inhibited the steps TAK1→IκB kinase (IKK) and IKK→IκBα. Niclosamide also increased the levels of reactive oxygen species (ROS) in AML cells. Quenching ROS by the glutathione precursor N-acetylcysteine attenuated niclosamide-induced apoptosis. Our results together suggest that niclosamide inhibited the NF-κB pathway and increased ROS levels to induce apoptosis in AML cells. On translational study of the efficacy of niclosamide against AML, niclosamide killed progenitor/stem cells from AML patients but spared those from normal bone marrow. Niclosamide was synergistic with the frontline chemotherapeutic agents cytarabine, etoposide, and daunorubicin. It potently inhibited the growth of AML cells in vitro and in nude mice. Our results support further investigation of niclosamide in clinical trials of AML patients. Cancer Res; 70(6); 2516–27
- Published
- 2010
37. GDP366, a novel small molecule dual inhibitor of survivin and Op18, induces cell growth inhibition, cellular senescence and mitotic catastrophe in human cancer cells
- Author
-
Zhongzheng Lu, Yanli Jin, Xianping Shi, Jingxuan Pan, Ke Ding, Deping Wang, and Jin Zhang
- Subjects
Male ,Cancer Research ,Telomerase ,Survivin ,Mice, Nude ,Stathmin ,Antineoplastic Agents ,Inhibitor of Apoptosis Proteins ,Mice ,Nude mouse ,medicine ,Animals ,Humans ,Mitotic catastrophe ,Cellular Senescence ,Pharmacology ,Mice, Inbred BALB C ,biology ,Cell growth ,Phenylurea Compounds ,Cancer ,biology.organism_classification ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,HCT116 Cells ,Xenograft Model Antitumor Assays ,Cell biology ,Oncology ,Apoptosis ,biology.protein ,Cancer research ,Molecular Medicine ,Colorectal Neoplasms ,Microtubule-Associated Proteins ,HeLa Cells - Abstract
Accumulating evidence indicates that survivin plays a pivotal role in not only cell survival but also cell cycle progression. Op18/stathmin is an oncoprotein that regulates microtubule stabilization. Both survivin and Op18 have been proposed as therapeutic targets for cancer. However, few small molecule inhibitors of survivin and Op18 have been reported. In this study, we have identified a novel small molecule compound (GDP366) which potently and selectively inhibited the expression of both survivin and Op18. It decreased both the mRNA and protein levels of survivin and Op18. This inhibitory effect was not dependent on the status of p53 and p21 although GDP366 potently increased p53 and p21 levels. GDP366 significantly inhibited the growth of tumor cells in vitro and in vivo (nude mouse model) without rapid induction of apoptosis. GDP366 induced polyploidy in multiple types of cancer cell lines. GDP366 increased chromosomal instability, and induced cellular senescence by inhibiting telomerase activity. We conclude that GDP366 is a novel dual inhibitor of survivin and Op18. Our results warrant further translational evaluation of this compound.
- Published
- 2010
38. Triptolide inhibits Jak2 transcription and induces apoptosis in human myeloproliferative disorder cells bearing Jak2V617F through caspase-3-mediated cleavage of Mcl-1
- Author
-
Yanli Jin, Zhongzheng Lu, Jingxuan Pan, Yongbin Wu, and Qi Chen
- Subjects
Cancer Research ,Transcription, Genetic ,Caspase 3 ,Apoptosis ,Biology ,chemistry.chemical_compound ,Myeloproliferative Disorders ,Transcription (biology) ,Cell Line, Tumor ,Survivin ,Humans ,RNA, Messenger ,Cycloheximide ,DNA Primers ,Reverse Transcriptase Polymerase Chain Reaction ,Anti-Inflammatory Agents, Non-Steroidal ,Cell Cycle ,Triptolide ,Janus Kinase 2 ,Phenanthrenes ,Oncogene Addiction ,Flow Cytometry ,Caspase Inhibitors ,Oncology ,chemistry ,Amino Acid Substitution ,Proto-Oncogene Proteins c-bcl-2 ,Cell culture ,Cancer research ,Epoxy Compounds ,Myeloid Cell Leukemia Sequence 1 Protein ,Leukemia, Erythroblastic, Acute ,Diterpenes ,Signal Transduction - Abstract
The discovery of oncogene addiction in myeloproliferative disorders (MPDs) driven by the gain-of-function mutant Jak2V617F has attracted intense interest in targeted therapy for MPDs. In this report, we demonstrate that triptolide potently downregulated the transcription of Jak2 by inhibiting the activity of RNA polymerase. Triptolide inhibited the in vitro and in vivo growth of tumor cells harboring Jak2V617F. Triptolide induced abundant apoptosis with a prominent decline of Bcl-2, Bcl-X(L), survivin and Mcl-1. As well, triptolide induced caspase-3-dependent Mcl-1 cleavage, which may potentiate apoptosis. These findings suggest that triptolide is a promising agent to kill Jak2V617F-harboring cells.
- Published
- 2009
39. Triptolide abrogates oncogene FIP1L1-PDGFRalpha addiction and induces apoptosis in hypereosinophilic syndrome
- Author
-
Jingxuan Pan, Yanli Jin, Qi Chen, Bo Chen, and Zhongzheng Lu
- Subjects
Cancer Research ,Receptor, Platelet-Derived Growth Factor alpha ,Oncogene Proteins, Fusion ,medicine.drug_class ,Cell Survival ,Apoptosis ,Biology ,Tyrosine-kinase inhibitor ,Cell Line ,chemistry.chemical_compound ,Growth factor receptor ,Hypereosinophilic Syndrome ,medicine ,Humans ,RNA, Messenger ,Phosphorylation ,STAT3 ,Protein kinase B ,mRNA Cleavage and Polyadenylation Factors ,Cell Cycle ,General Medicine ,Triptolide ,Phenanthrenes ,Antineoplastic Agents, Phytogenic ,Oncology ,chemistry ,Cancer cell ,Cancer research ,biology.protein ,Epoxy Compounds ,Signal transduction ,Diterpenes ,Tyrosine kinase - Abstract
The pathogenesis of hypereosinophilic syndrome (HES) in some patients is highly dependent on FIP1-Like-1 (FIP1L1)-platelet-derived growth factor receptor alpha (PDGFRalpha), which can generate sustained activation signaling to maintain a cell malignant phenotype. HES usually shows good response to the tyrosine kinase inhibitor imatinib, but mutations in FIP1L1-PDGFRalpha (e.g. T674I) can confer acquired resistance to imatinib. An alternative therapeutic strategy other than with tyrosine kinase inhibitors is needed to overcome acquired drug resistance. We hypothesized that switching off the crucial chimeric oncoprotein FIP1L1-PDGFRalpha on which HES cells depend, should have deleterious effects on the cancer cells. We used low concentrations of triptolide, a transcription inhibitor, to shut down the expression of FIP1L1-PDGFRalpha. EOL-1 cells and BaF3 cells expressing wild-type or T674I FIP1L1-PDGFRalpha were treated with triptolide, and signaling pathways, cell cycling, and apoptosis were analyzed by RT-PCR, immunoblotting, and flow cytometry, respectively. The results revealed that at nanomolar concentrations triptolide decreased the levels of mRNA and protein of FIP1L1-PDGFRalpha and the growth of the neoplastic cells, regardless of the mutational status of PDGFRalpha. Triptolide also downregulated the signaling molecules Stat3, Akt, and Erk1/2, which are downstream from PDGFRalpha, and induced G1 cell-cycle arrest. Triptolide time- and dose-dependently induced apoptosis by decreasing the anti-apoptotic proteins Mcl-1 and Bcl-X(L),triggering the intrinsic apoptotic pathway. In conclusion, triptolide has potent activity against malignant cells in HES bearing FIP1L1-PDGFRalpha, regardless of its mutational status that confer acquired resistance to imatinib. Our results suggest that triptolide may be a promising agent in the treatment of HES.
- Published
- 2009
40. Activity of triptolide against human mast cells harboring the kinase domain mutant KIT
- Author
-
Qi Chen, Zhongzheng Lu, Qin Zheng, Yingrong Lai, Jingxuan Pan, Xianping Shi, Chao Cheng, and Yanli Jin
- Subjects
Male ,Cancer Research ,Down-Regulation ,Mice, Nude ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Mice ,Nude mouse ,medicine ,Animals ,Humans ,Mast Cells ,RNA, Messenger ,Systemic mastocytosis ,STAT3 ,Protein kinase B ,Antineoplastic Agents, Alkylating ,biology ,General Medicine ,Triptolide ,Phenanthrenes ,biology.organism_classification ,Mast cell ,medicine.disease ,XIAP ,Proto-Oncogene Proteins c-kit ,medicine.anatomical_structure ,Oncology ,chemistry ,Mutation ,biology.protein ,Cancer research ,Epoxy Compounds ,Diterpenes - Abstract
Gain-of-function mutations of the receptor tyrosine kinase KIT can cause systemic mastocytosis (SM) and gastrointestinal stromal tumors. Most of the constitutively active KIT can be inhibited by imatinib; D816V KIT cannot. In this study, we investigated the activity of triptolide, a diterpenoid isolated from the Chinese herb Tripterygium wilfordii Hook. f., in cells expressing mutant KIT, including D816V KIT. Imatinib-sensitive HMC-1.1 cells harboring the mutation V560G in the juxtamembrane domain of KIT, imatinib-resistant HMC-1.2 cells harboring both V560G and D816V mutations, and murine P815 cells, were treated with triptolide, and analyzed in terms of growth, apoptosis, and signal transduction. The in vivo antitumor activity was evaluated by using the nude mouse xenograft model. Our results demonstrated that triptolide potently inhibits the growth of both human and murine mast cells harboring not only imatinib-sensitive KIT mutation but also imatinib-resistant D816V KIT. Triptolide markedly inhibited KIT mRNA levels and strikingly reduced the levels of phosphorylated and total Stat3, Akt, and Erk1/2, downstream targets of KIT. Triptolide triggered apoptosis by inducing depolarization of mitochondrial potential and release of cytochrome c, downregulation of Mcl-1 and XIAP. Furthermore, triptolide significantly abrogated the growth of imatinib-resistant HMC-1.2 cell xenografts in nude mice and decreased KIT expression in xenografts. Our data demonstrate that triptolide inhibits imatinib-resistant mast cells harboring D816V KIT. Further investigation of triptolide for treatment of human neoplasms driven by gain-of-function KIT mutations is warranted.
- Published
- 2009
41. Autophagy induced by farnesyltransferase inhibitors in cancer cells
- Author
-
Ruiying Zhao, Lily Sun, Chun Hui Su, Jingxuan Pan, Mong Hong Lee, Zhi-Xiang Xu, Bo Chen, and Sai Ching J. Yeung
- Subjects
Cancer Research ,Programmed cell death ,Polyunsaturated Alkamides ,Pyridines ,Farnesyltransferase ,Apoptosis ,Polyenes ,chemistry.chemical_compound ,Microscopy, Electron, Transmission ,Piperidines ,Cell Line, Tumor ,Autophagy ,Farnesyltranstransferase ,Humans ,Lonafarnib ,Enzyme Inhibitors ,RNA, Small Interfering ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Pharmacology ,biology ,Dose-Response Relationship, Drug ,TOR Serine-Threonine Kinases ,Cell biology ,Pancreatic Neoplasms ,Oncology ,chemistry ,Microscopy, Fluorescence ,Cancer cell ,biology.protein ,Cancer research ,Molecular Medicine ,Protein Kinases ,RHEB ,Signal Transduction - Abstract
The mechanisms of action of farnesyltransferase inhibitors (FTIs) involve Rheb and the phosphatidylinositide 3-kinase/Akt/mammalian target of rapamycin (mTOR) pathway. mTOR in particular plays a key role in the regulation of autophagy. Collectively, the literature suggests that FTIs very likely induce autophagy, but thus far there have been no reports that FTIs affect this process relevant to cancer cell biology. We hypothesized that FTIs can induce autophagy. In this study, we found that the FTIs manumycin A, FTI-276, and lonafarnib induced autophagy in two human cancer cell lines. We also found that neither inhibition of apoptosis with a pan-caspase inhibitor nor inhibition of autophagy increased the number of clones of lonafarnib-treated U2OS osteosarcoma cells that formed in soft agar. Although whether autophagy is a cell death or cell survival mechanism after FTI treatment remains unresolved, our data show that cancer cells apparently can shift between apoptosis and autophagy once they are committed to die after FTI treatment.
- Published
- 2008
42. The novel tyrosine kinase inhibitor EXEL-0862 induces apoptosis in human FIP1L1-PDGFR-alpha-expressing cells through caspase-3-mediated cleavage of Mcl-1
- Author
-
Alfonso Quintás-Cardama, A Tefferi, Taghi Manshouri, Hagop M. Kantarjian, Francis J. Giles, Jorge E. Cortes, Jingxuan Pan, Srdan Verstovsek, and Peter Lamb
- Subjects
Cancer Research ,Receptor, Platelet-Derived Growth Factor alpha ,Oncogene Proteins, Fusion ,medicine.drug_class ,Drug Evaluation, Preclinical ,Caspase 3 ,Apoptosis ,Tyrosine-kinase inhibitor ,Piperazines ,Hypereosinophilic Syndrome ,medicine ,Tumor Cells, Cultured ,Humans ,Enzyme Inhibitors ,neoplasms ,mRNA Cleavage and Polyadenylation Factors ,ABL ,biology ,Kinase ,Hematology ,Protein-Tyrosine Kinases ,Neoplasm Proteins ,Imatinib mesylate ,Pyrimidines ,Oncology ,Proto-Oncogene Proteins c-bcl-2 ,Drug Resistance, Neoplasm ,Benzamides ,cardiovascular system ,biology.protein ,Cancer research ,Imatinib Mesylate ,Phosphorylation ,Myeloid Cell Leukemia Sequence 1 Protein ,biological phenomena, cell phenomena, and immunity ,Tyrosine kinase ,Platelet-derived growth factor receptor - Abstract
The FIP1-like-1 (FIP1L1)-platelet-derived growth factor receptor-alpha (FIP1L1-PDGFR-alpha) fusion kinase causes hypereosinophilic syndrome (HES) in a defined subset of patients. Imatinib mesylate is a potent inhibitor of ABL but also of PDGFR-alpha, and has been associated with durable hematologic responses in patients with HES. However, development of mutations in the tyrosine kinase domain may hamper the activity of tyrosine kinase inhibitors (TKIs), which suggests that novel agents are warranted to prevent or overcome resistance. We evaluated the efficacy of the novel TKI EXEL-0862 in FIP1L1-PDGFR-alpha-expressing cell lines and in cells from a patient with HES harboring the FIP1L1-PDGFR-alpha gene. EXEL-0862 inhibited the proliferation of EOL-1 and imatinib-resistant T674I FIP1L1-PDGFR-alpha-expressing cells and resulted in potent inhibition of the phosphorylation of PDGFR-alpha and downstream proteins STAT3 and Erk1/2, both in vitro and ex vivo. Moreover, EXEL-0862 induced apoptotic death in EOL-1 cells and imatinib-resistant T674I FIP1L1-PDGFR-alpha-expressing cells, and resulted in significant downregulation of the antiapoptotic protein Mcl-1 through a caspase-dependent mechanism. Our data establish EXEL-0862 as a solid candidate for the targeted treatment of patients with FIP1L1-PDGFR-alpha-positive HES.
- Published
- 2007
43. Tenovin-6-mediated inhibition of SIRT1/2 induces apoptosis in acute lymphoblastic leukemia (ALL) cells and eliminates ALL stem/progenitor cells
- Author
-
Bei Jin, Xin Du, Yanli Jin, Qi Cao, Jingxuan Pan, and Chun Chen
- Subjects
p53 ,Adult ,Male ,Cancer Research ,Adolescent ,Cell Survival ,Cell ,Apoptosis ,Antineoplastic Agents ,Biology ,Acute lymphoblastic leukemia ,Inhibitor of apoptosis ,Tenovin-6 ,Targeted therapy ,Young Adult ,Sirtuin 2 ,Sirtuin 1 ,Cell Line, Tumor ,medicine ,Genetics ,Humans ,Viability assay ,Progenitor cell ,Stem/progenitor cells ,Child ,Wnt Signaling Pathway ,Cell Proliferation ,Cell growth ,Cell Cycle ,Infant ,Acetylation ,β-catenin ,Middle Aged ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.anatomical_structure ,Oncology ,Cell culture ,Child, Preschool ,Benzamides ,Cancer research ,Neoplastic Stem Cells ,Lymphoid Progenitor Cells ,Epigenetics ,Female ,Stem cell ,Tumor Suppressor Protein p53 ,SIRT1 inhibitor ,Research Article - Abstract
Background Acute lymphoblastic leukemia (ALL) is a heterogeneous group of malignant disorders derived from B- or T-cell lymphoid progenitor cells. ALL often is refractory to or relapses after treatment; thus, novel targeted therapy for ALL is urgently needed. In the present study, we initially found that the level of SIRT1, a class III histone deacetylase, was higher in primary ALL cells from patients than in peripheral blood mononuclear cells from healthy individuals. But it is not clear whether inhibition of SIRT1 by its selective small molecule inhibitor Tenovin-6 is effective against ALL cells. Methods We tested the effect of Tenovin-6 on ALL cell lines (REH and NALM-6) and primary cells from 41 children with ALL and 2 adult patients with ALL. The effects of Tenovin-6 on cell viability were determined by MTS assay; colony-forming assays were determined by soft agar in ALL cell lines and methylcellulose medium in normal bone marrow cells and primary ALL blast cells; cell apoptosis and cell cycling were examined by flow cytometry; the signaling pathway was determined by Western blotting; ALL stem/progenitor cells were seperated by using MACS MicroBead kit. Results The results showed that Tenovin-6 treatment activated p53, potently inhibited the growth of pre-B ALL cells and primary ALL cells, and sensitized ALL cells to frontline chemotherapeutic agents etoposide and cytarabine. Tenovin-6 induced apoptosis in REH and NALM-6 cells and primary ALL cells and diminished expression of Mcl-1 and X-linked inhibitor of apoptosis protein (XIAP) in such cells. Furthermore, inhibition of SIRT1 by Tenovin-6 inhibited the Wnt/β-catenin signaling pathway and eliminated ALL stem/progenitor (CD133 + CD19-) cells. Conclusion Our results indicate that Tenovin-6 may be a promising targeted therapy for ALL and clinical trials are warranted to investigate its efficacy in ALL patients.
- Full Text
- View/download PDF
44. Ponatinib efficiently kills imatinib-resistant chronic eosinophilic leukemia cells harboring gatekeeper mutant T674I FIP1L1-PDGFRα: roles of Mcl-1 and β-catenin
- Author
-
Honglin Li, Chengyan Wang, Yanli Jin, Jingxuan Pan, Ke Ding, Xiaoke Shi, and Mengzhu Xue
- Subjects
PDGFRα ,Cancer Research ,medicine.drug_class ,Resistance ,Tyrosine kinase inhibitor ,Apoptosis ,Biology ,Tyrosine-kinase inhibitor ,chemistry.chemical_compound ,hemic and lymphatic diseases ,medicine ,Chronic eosinophilic leukemia ,Research ,Ponatinib ,Imatinib ,Tyrosine phosphorylation ,medicine.disease ,Dasatinib ,Nilotinib ,chemistry ,Oncology ,T674I mutant ,Cancer research ,Molecular Medicine ,Tyrosine kinase ,medicine.drug - Abstract
Background T674I FIP1L1-PDGFRα in a subset of chronic eosinophilic leukemia (CEL) is a gatekeeper mutation that is resistant to many tyrosine kinase inhibitors (TKIs) (e.g., imatinib, nilotinib and dasatinib), similar to T315I Bcr-Abl. Therefore, novel TKIs effective against T674I FIP1L1-PDGFRα are needed. Ponatinib (AP24534) is a novel orally bioavailable TKI against T315I Bcr-Abl, but it is not clear whether ponatinib is effective against T674I FIP1L1-PDGFRα. The purpose of this study was to examine the effect of ponatinib on T674I FIP1L1-PDGFRα. Methods Molecular docking analysis in silico was performed. The effects of ponatinib on PDGFRα signaling pathways, apoptosis and cell cycling were examined in EOL-1, BaF3 cells expressing either wild type (WT) or T674I FIP1L1-PDGFRα. The in vivo antitumor activity of ponatinib was evaluated with xenografted BaF3-T674I FIP1L1-PDGFRα cells in nude mice models. Results Molecular docking analysis revealed that ponatinib could bind to the DFG (Asp-Phe-Gly)-out state of T674I PDGFRα. Ponatinib potently inhibited the phosphorylation of WT and T674I FIP1L1-PDGFRα and their downstream signaling molecules (e.g., Stat3, Stat5). Ponatinib strikingly inhibited the growth of both WT and T674I FIP1L1-PDGFRα-carrying CEL cells (IC50: 0.004–2.5 nM). It induced apoptosis in CEL cells with caspase-3-dependent cleavage of Mcl-1, and inhibited tyrosine phosphorylation of β-catenin to decrease its stability and pro-survival functions. In vivo, ponatinib abrogated the growth of xenografted BaF3-T674I FIP1L1-PDGFRα cells in nude mice. Conclusions Ponatinib is a pan-FIP1L1-PDGFRα inhibitor, and clinical trials are warranted to investigate its efficacy in imatinib-resistant CEL.
- Full Text
- View/download PDF
45. Overcoming Gleevec-resistance by blocking oncogene addiction in malignant hematologic cells
- Author
-
Jingxuan Pan
- Subjects
Cancer Research ,medicine.drug_class ,Pharmacology ,lcsh:RC254-282 ,Receptor tyrosine kinase ,Tyrosine-kinase inhibitor ,chemistry.chemical_compound ,hemic and lymphatic diseases ,medicine ,Molecular Biology ,biology ,lcsh:RC633-647.5 ,business.industry ,Ponatinib ,Imatinib ,lcsh:Diseases of the blood and blood-forming organs ,Hematology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dasatinib ,Imatinib mesylate ,chemistry ,Nilotinib ,Oncology ,Meeting Abstract ,biology.protein ,business ,Tyrosine kinase ,medicine.drug - Abstract
In some types of tumors, malignant cells are highly dependent on the constitutive activation of a certain protein encoded by oncogene, despite existence of additional carcinogenic genetic changes. This phenomenon is referred to oncogene addiction. Typical examples include cytoplasmic tyrosine kinase Bcr-Abl in chronic myeloid leukemia (CML), receptor tyrosine kinase KIT in systemic mastocytosis and gastrointestinal stromal tumors (GISTs), and PDGFRα in hypereosinophilic syndrome (HES). In 2001, the approval of Gleevec (STI571, imatinib mesylate, Norvartis) by FDA (Food and Drug Administration, USA) initiated a revolutionary targeted therapy against cancer with small molecule tyrosine kinase inhibitors. Gleevec blocks the signaling pathway of tyrosine kinase by competitively occupying the ATP-binding pocket of Bcr-Abl, KIT and PDGFRα, and therefore kills these oncogene-addicted tumor cells. Patients with CML and HES have gained much better prognosis with the treatment of Gleevec. However, in some patients (particularly in accelerating and blast crisis phases), relapse due to resistance to Gleevec is an emerging problem. Acquired point mutations within the target genes (Bcr-Abl, KIT and PDGFRα) are a major mechanism of resistance to Gleevec in some patients with hematologic malignance. The mutations are believed to block the binding of Gleevec to ATP binding pockets of these tyrosine kinases. In this case, novel tyrosine kinase inhibitor such as nilotinib and dasatinib (also called the second-generation of tyrosine kinase inhibitor) have been shown activity against Gleevec-resistant patients bearing some point mutations but the “gate-keeper” mutations (e.g., T315I Bcr-Abl, T674I PDGFRα). Therefore, development of more novel small molecule tyrosine kinase inhibitors is still needed. This talk covered the advance in the field of overcoming Gleevec resistance in terms of novel compounds and strategeies. Pan J et al reported that EXEL-0862 is effective against Gleevec-resistant D816V KIT and T674I PDGFRα (2007). Recently, in vitro and animal data supported that several novel tyrosine kinase inhibitors including (but not limited) AP24534 (ponatinib) and DCC-2036 have been demonstrated effective against T315I Bcr-Abl. However, the efficacy and safety of these pounds (EXEL-0862, AP24534 and DCC-2036) in patients remains to be defined. An alternative approach for overcoming Gleevec-resistance is to decrease the expression of “addicted” oncogenes, which are driving forces of the tumor cells, to kill the malignant cells. Our group discovered several compounds which are effective against Gleevec-resistant tumor cells regardless of resistance to imatinib. The compounds kill cells harboring gate-keeper mutants of tyrosine kinases by lowering the expression of the oncoproteins (Bcr-Abl, KIT and PDGFRα). Examples include triptolide, pristimerin and SNS-032 (transcription inhibitors), homoharringtonine (a translation inhibitor), and celastrol (a hsp90 inhibitor). In summary, Gleevec-resistance remains a challenge in leukemia. The findings from us and others suggest that several aforementioned compounds are promising agents to overcome Gleevec resistance, and warrant clinical trials.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.