1
|
Chai YX, Ren JJ, Li YM, Bai YC, Zhang QQ, Zhao YZ, Yang X, Zhang XH, Zhang XS, Wu AX, Zhu YP, Sun YY. 5-Aminopyrazole Dimerization: Cu-Promoted Switchable Synthesis of Pyrazole-Fused Pyridazines and Pyrazines via Direct Coupling of C-H/N-H, C-H/C-H, and N-H/N-H Bonds. Molecules 2025; 30:381. [PMID: 39860249 PMCID: PMC11767409 DOI: 10.3390/molecules30020381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
A Cu-promoted highly chemoselective dimerization of 5-aminopyrazoles to produce pyrazole-fused pyridazines and pyrazines is reported. The protocol generates switchable products via the direct coupling of C-H/N-H, C-H/C-H and N-H/N-H bonds, with the merits of broad substrate scope and high functional group compatibility. Gram-scale experiments demonstrated the potential applications of this reaction. Moreover, the preliminary fluorescence results uncovered that dipyrazole-fused pyridazines and pyrazines may have some potential applications in materials chemistry.
Collapse
Affiliation(s)
- Yi-Xin Chai
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - Jun-Jie Ren
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - Yi-Ming Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - Yi-Cheng Bai
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - Qing-Qing Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - Yi-Zhen Zhao
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - Xue Yang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - Xiao-Han Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - Xin-Shuang Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| | - An-Xin Wu
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Yan-Ping Zhu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
- International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Yuan-Yuan Sun
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China; (Y.-X.C.); (J.-J.R.); (Y.-M.L.); (Y.-C.B.); (Q.-Q.Z.); (Y.-Z.Z.); (X.Y.); (X.-H.Z.); (X.-S.Z.)
| |
Collapse
|
2
|
Engür-Öztürk S, Kaya-Tİlkİ E, Cantürk Z, Dİkmen M. Enhanced angiogenesis of human umbilical vein endothelial cells via THP-1-derived M2c-like macrophages and treatment with proteasome inhibitors 'bortezomib and ixazomib'. APMIS 2024; 132:594-607. [PMID: 38775107 DOI: 10.1111/apm.13426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 05/07/2024] [Indexed: 07/11/2024]
Abstract
The leading cause of cancer-related death is lung cancer, with metastasis being the most common cause of death. To elucidate the role of macrophages in lung cancer and angiogenesis processes, we established an in vitro co-culture model of A549 or HUVEC with THP-1 cells that polarized to M2c macrophages with hydrocortisone. The proteasome inhibitors bortezomib and ixazomib were investigated for their effects on proliferation, invasion, migration, metastasis, and angiogenesis pathways. The effects of bortezomib and ixazomib on gene expression in gene panels, including crucial genes related to angiogenesis and proteasomes, were investigated after the co-culture model to determine these effects at the molecular level. In conclusion, bortezomib and ixazomib showed antiproliferative effects in both cells, as well as in M2c macrophage co-culture. M2c macrophages also increased invasion in A549 cells and both invasion and migration in HUVEC. mRNA expression upregulation, specifically in the NFKB and VEGF genes, supported the metastatic and angiogenic effects found in A549 and HUVEC with M2c macrophage co-culture. Additionally, bortezomib inhibited the VEGFB pathway in HUVEC and NFKB1 in A549 cells. The significant findings obtained as a result of this study will provide information regarding angiogenesis induced by M2 macrophages.
Collapse
Affiliation(s)
- Selin Engür-Öztürk
- Department of Pharmacy Services, Tavas Vocational School of Health Services, Pamukkale University, Denizli, Turkey
| | - Elif Kaya-Tİlkİ
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Zerrin Cantürk
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Miriş Dİkmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
3
|
Zafeiropoulou K, Kalampounias G, Alexis S, Anastasopoulos D, Symeonidis A, Katsoris P. Autophagy and oxidative stress modulation mediate Bortezomib resistance in prostate cancer. PLoS One 2024; 19:e0289904. [PMID: 38412186 PMCID: PMC10898778 DOI: 10.1371/journal.pone.0289904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 01/27/2024] [Indexed: 02/29/2024] Open
Abstract
Proteasome inhibitors such as Bortezomib represent an established type of targeted treatment for several types of hematological malignancies, including multiple myeloma, Waldenstrom's macroglobulinemia, and mantle cell lymphoma, based on the cancer cell's susceptibility to impairment of the proteasome-ubiquitin system. However, a major problem limiting their efficacy is the emergence of resistance. Their application to solid tumors is currently being studied, while simultaneously, a wide spectrum of hematological cancers, such as Myelodysplastic Syndromes show minimal or no response to Bortezomib treatment. In this study, we utilize the prostate cancer cell line DU-145 to establish a model of Bortezomib resistance, studying the underlying mechanisms. Evaluating the resulting resistant cell line, we observed restoration of proteasome chymotrypsin-like activity, regardless of drug presence, an induction of pro-survival pathways, and the substitution of the Ubiquitin-Proteasome System role in proteostasis by induction of autophagy. Finally, an estimation of the oxidative condition of the cells indicated that the resistant clones reduce the generation of reactive oxygen species induced by Bortezomib to levels even lower than those induced in non-resistant cells. Our findings highlight the role of autophagy and oxidative stress regulation in Bortezomib resistance and elucidate key proteins of signaling pathways as potential pharmaceutical targets, which could increase the efficiency of proteasome-targeting therapies, thus expanding the group of molecular targets for neoplastic disorders.
Collapse
Affiliation(s)
- Kalliopi Zafeiropoulou
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Georgios Kalampounias
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| | - Spyridon Alexis
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Daniil Anastasopoulos
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| | - Argiris Symeonidis
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Panagiotis Katsoris
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| |
Collapse
|
4
|
Arokia Femina T, Barghavi V, Archana K, Swethaa NG, Maddaly R. Non-uniformity in in vitro drug-induced cytotoxicity as evidenced by differences in IC 50 values - implications and way forward. J Pharmacol Toxicol Methods 2023; 119:107238. [PMID: 36521817 DOI: 10.1016/j.vascn.2022.107238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Cell lines have proven indispensable for in vitro experiments and their utility as experimental models range from understanding the fundamental cell functioning to drug discovery. One of the most common utility of cell lines is for in vitro drug testing. Drug testing involves determining the cytotoxic effects of the drugs and such a measurement is expressed as the IC50 values of drugs. Although determination of IC50 values of drugs on cell lines is one of the most common in vitro experimental approaches, a significant amount of variations can be observed in the results obtained from such studies. Although the variations in the IC50 values of a drug on different cells lines can and should vary, the non-uniformity of such results reported from different studies using a particular drug on a specific cell line is a matter of concern. We present the IC50 values of 5 most commonly used drugs 5-fluorouracil, bleomycin, cisplatin, doxorubicin and methotrexate obtained from several in vitro cell line-based studies. Some of the factors which contribute to the non-uniformity of the IC50 values for a particular drug from different studies are discussed as three types of factors, the biological, non-biological and human factors. Also, ways in which such variations can be reduced to obtain universally common, reliable results are presented.
Collapse
Affiliation(s)
- T Arokia Femina
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu 600116, India
| | - V Barghavi
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu 600116, India
| | - K Archana
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu 600116, India
| | - N G Swethaa
- Department of Biotechnology, Anna University, Guindy, Chennai 600 025, India
| | - Ravi Maddaly
- Department of Human Genetics, Faculty of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu 600116, India.
| |
Collapse
|
5
|
Yeh SJ, Chen BS. Systems Medicine Design based on Systems Biology Approaches and Deep Neural Network for Gastric Cancer. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3019-3031. [PMID: 34232888 DOI: 10.1109/tcbb.2021.3095369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Gastric cancer (GC) is the third leading cause of cancer death in the world. It is associated with the stimulation of microenvironment, aberrant epigenetic modification, and chronic inflammation. However, few researches discuss the GC molecular progression mechanisms from the perspective of the system level. In this study, we proposed a systems medicine design procedure to identify essential biomarkers and find corresponding drugs for GC. At first, we did big database mining to construct candidate protein-protein interaction network (PPIN) and candidate gene regulation network (GRN). Second, by leveraging the next-generation sequencing (NGS) data, we performed system modeling and applied system identification and model selection to obtain real genome-wide genetic and epigenetic networks (GWGENs). To make the real GWGENs easy to analyze, the principal network projection method was used to extract the core signaling pathways denoted by KEGG pathways. Subsequently, based on the identified biomarkers, we trained a deep neural network of drug-target interaction (DeepDTI) with supervised learning and filtered our candidate drugs considering drug regulation ability and drug sensitivity. With the proposed systematic strategy, we not only shed the light on the progression of GC but also suggested potential multiple-molecule drugs efficiently.
Collapse
|
6
|
Zhang W, Niu J, Ma Y, Yang X, Cao H, Guo H, Bao F, Haw A, Chen Y, Sun K. The Synergistic Antitumor Activity of Chidamide in Combination with Bortezomib on Gastric Cancer. Onco Targets Ther 2020; 13:3823-3837. [PMID: 32440150 PMCID: PMC7213427 DOI: 10.2147/ott.s240721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/25/2020] [Indexed: 12/24/2022] Open
Abstract
Purpose The aim of this study was to investigate the antitumor effect of chidamide in combination with bortezomib on gastric cancer cell lines. Materials and Methods First, the sensitivity and IC50 values of chidamide and bortezomib in several gastric cancer cell lines (MGC-803, BGC-823, SGC-7901, and MKN45) were measured using the CCK-8 assay. Then, the relatively insensitive gastric cancer cell lines (MGC-803 and BGC-823) were treated with low concentrations of chidamide alone, bortezomib alone, or chidamide and bortezomib combination to detect the effects on cell proliferation, apoptosis, migration, and invasion. Finally, the inhibitory effect of the combined chidamide and bortezomib treatment on MGC-803 cells was verified in vivo through tumor formation experiments in nude mice. Results Compared with low-dose chidamide or bortezomib alone, the low-dose drug combination significantly inhibited the proliferation, migration, and invasion of MGC-803 and BGC-823 cells and induced apoptosis of the cells. The effects of the low-dose chidamide and bortezomib combination reduced the growth on gastric cancer in vivo were investigated by using a subcutaneous tumor mouse model. Conclusion Our results suggest that the combination of chidamide and bortezomib can significantly reduce the proliferation, invasion, and migration of MGC-803 and BGC-823 cells, providing a framework for the clinical evaluation of combined therapies for gastric cancers.
Collapse
Affiliation(s)
- Wanjun Zhang
- Department of Hematology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, People's Republic of China
| | - Junwei Niu
- Department of Hematology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, People's Republic of China
| | - Yongcheng Ma
- Clinical Pharmacology Laboratory, Henan Provincial People's Hospital; Department of Pharmacy of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan 450003, People's Republic of China
| | - Xiawan Yang
- Department of Hematology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, People's Republic of China
| | - Huixia Cao
- Department of Nephrology, Henan Key Library for Kidney Disease and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, People's Republic of China
| | - Honggang Guo
- Department of Hematology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, People's Republic of China
| | - Fengchang Bao
- Department of Hematology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, People's Republic of China
| | - Ahmed Haw
- Department of Hematology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, People's Republic of China
| | - Yuqing Chen
- Department of Hematology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, People's Republic of China
| | - Kai Sun
- Department of Hematology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, People's Republic of China
| |
Collapse
|
7
|
Balakrishnan K, Panneerpandian P, Devanandan HJ, Sekar BT, Rayala SK, Ganesan K. Salt-mediated transcriptional and proteasomal dysregulations mimic the molecular dysregulations of stomach cancer. Toxicol In Vitro 2019; 61:104588. [PMID: 31279909 DOI: 10.1016/j.tiv.2019.104588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/21/2019] [Accepted: 07/01/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Karthik Balakrishnan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Ponmathi Panneerpandian
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Helen Jemimah Devanandan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Balaji T Sekar
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamilnadu, India
| | - Kumaresan Ganesan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India.
| |
Collapse
|
8
|
Henriques AC, Ribeiro D, Pedrosa J, Sarmento B, Silva PMA, Bousbaa H. Mitosis inhibitors in anticancer therapy: When blocking the exit becomes a solution. Cancer Lett 2018; 440-441:64-81. [PMID: 30312726 DOI: 10.1016/j.canlet.2018.10.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/12/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Abstract
Current microtubule-targeting agents (MTAs) remain amongst the most important antimitotic drugs used against a broad range of malignancies. By perturbing spindle assembly, MTAs activate the spindle assembly checkpoint (SAC), which induces mitotic arrest and subsequent apoptosis. However, besides toxic side effects and resistance, mitotic slippage and failure in triggering apoptosis in various cancer cells are limiting factors of MTAs efficacy. Alternative strategies to target mitosis without affecting microtubules have, thus, led to the identification of small molecules, such as those that target spindle Kinesins, Aurora and Polo-like kinases. Unfortunately, these so-called second-generation of antimitotics, encompassing mitotic blockers and mitotic drivers, have failed in clinical trials. Our recent understanding regarding the mechanisms of cell death during a mitotic arrest pointed out apoptosis as the main variable, providing an opportunity to control the cell fates and influence the effectiveness of antimitotics. Here, we provide an overview on the second-generation of antimitotics, and discuss possible strategies that exploit SAC activity, mitotic slippage/exit and apoptosis induction, in order to improve the efficacy of anticancer strategies that target mitosis.
Collapse
Affiliation(s)
- Ana C Henriques
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; INEB, Instituto Nacional de Engenharia Biomédica, Universidade Do Porto, Porto, Portugal
| | - Diana Ribeiro
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade Do Porto, Porto, Portugal
| | - Joel Pedrosa
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal
| | - Bruno Sarmento
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; INEB, Instituto Nacional de Engenharia Biomédica, Universidade Do Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação Em Saúde, Universidade Do Porto, Porto, Portugal
| | - Patrícia M A Silva
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal
| | - Hassan Bousbaa
- CESPU, Instituto de Investigação e Formação Avançada Em Ciências e Tecnologias da Saúde, Instituto Universitário de Ciências da Saúde, Gandra PRD, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade Do Porto, Porto, Portugal.
| |
Collapse
|
9
|
Leucovorin Enhances the Anti-cancer Effect of Bortezomib in Colorectal Cancer Cells. Sci Rep 2017; 7:682. [PMID: 28386133 PMCID: PMC5429730 DOI: 10.1038/s41598-017-00839-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/15/2017] [Indexed: 01/06/2023] Open
Abstract
Colorectal cancer is a major cancer type worldwide. 5-fluorouracil, often given with leucovorin, is the most commonly used drug in colorectal cancer chemotherapy, yet development of drug resistance to 5-fluorouracil in colorectal cancer cells is the primary cause of chemotherapy failure. Most patients receiving intravenous 5-fluorouracil develop side effects. Leucovorin, due to its vitamin-like profile, has few side-effects. Drug repurposing is the application of approved drugs to treat new indications. In this study, we performed a novel drug-repurposing screening to identify Food and Drug Administration-approved chemotherapeutic compounds possessing synergistic activity with leucovorin against colorectal cancer cells. We found that the combination of bortezomib and leucovorin enhanced caspase activation and increased apoptosis in colorectal cancer cells better than either agent alone. Further, the synergistic induction of apoptosis and inhibition of tumor growth were also observed in mouse colorectal cancer xenografts. These data support leucovorin enhances the anti-cancer effect of bortezomib and present this novel combinatorial treatment against colorectal cancer.
Collapse
|
10
|
Al-Batran SE, Werner D. Recent advances and future trends in the targeted therapy of metastatic gastric cancer. Expert Rev Gastroenterol Hepatol 2014; 8:555-69. [PMID: 24665840 DOI: 10.1586/17474124.2014.902304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The better understanding of the molecular mechanisms behind gastric cancer has led to the development of new therapeutic strategies that are likely to improve patient outcomes in the near future. Recently, targeting the HER2 and the VEGF pathways with trastuzumab and ramucirumab, respectively, have been found to improve survival, while directed therapies against a number of other pathways are under clinical evaluation. These include the hepatocyte growth factor and its receptor c-MET, the insulin-like growth factor 1, the fibroblast growth factor, the mammalian target of rapamycin (mTOR), the epidermal growth factor receptor, and other pathways, as well as relevant immunotherapeutic strategies. This article reviews recent advances and future trends of these concepts for gastric cancer and adenocarcinoma of the gastroesophageal junction.
Collapse
Affiliation(s)
- Salah-Eddin Al-Batran
- Krankenhaus Nordwest, UCT-University Cancer Center Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
11
|
BAY 11-7082, a nuclear factor-κB inhibitor, induces apoptosis and S phase arrest in gastric cancer cells. J Gastroenterol 2014; 49:864-74. [PMID: 23846545 DOI: 10.1007/s00535-013-0848-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/05/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Inhibitors of nuclear factor (NF)-κB pathway have shown potential anti-tumor activities. However, it is not fully elucidated in gastric cancer. METHODS Firstly, we screened the inhibitory effect of pharmacologic NF-κB inhibitors on cell viability of human gastric cancer cells via CCK-8 assay. Next, cell apoptosis, cell cycle distribution, and mitochondrial membrane potential after BAY 11-7082 treatment were detected by annexin V staining, propidium iodide staining, TUNEL, and JC-1 assays in human gastric cancer HGC-27 cells. Expression of regulatory factors for apoptosis and cell cycle were measured by western blot. Finally, human gastric cancer xenograft model was established to verify the anti-tumor effects of BAY 11-7082 in vivo. Cellular apoptosis and growth inhibition in subcutaneous tumor section were detected by TUNEL and immunohistochemistry assays. RESULTS BAY 11-7082 exhibited rapid and potent anti-tumor effects on gastric cancer cells in vitro within a panel of NF-κB inhibitors. BAY 11-7082 induced rapid apoptosis in HGC-27 cells through activating the mitochondrial pathway, as well as down-regulation of Bcl-2 and up-regulation of Bax. BAY 11-7082 also induced S phase arrest through suppressing Cyclin A and CDK-2 expression. Xenograft model confirmed the anti-tumor effects of BAY 11-7082 on apoptosis induction and growth inhibition in vivo. CONCLUSIONS Our results demonstrated that BAY 11-7082 presented the most rapid and potent anti-tumor effects within a panel of NF-κB inhibitors, and could induce cellular apoptosis and block cell cycle progression both in vitro and in vivo, thus providing basis for clinical application of BAY 11-7082 in gastric cancer cases.
Collapse
|
12
|
Kim JE, Lee JI, Jin DH, Lee WJ, Park GB, Kim S, Kim YS, Wu TC, Hur DY, Kim D. Sequential treatment of HPV E6 and E7-expressing TC-1 cells with bortezomib and celecoxib promotes apoptosis through p-p38 MAPK-mediated downregulation of cyclin D1 and CDK2. Oncol Rep 2014; 31:2429-37. [PMID: 24627094 DOI: 10.3892/or.2014.3082] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/27/2014] [Indexed: 11/05/2022] Open
Abstract
Interruption of the cell cycle is accompanied by changes in several related molecules that result in the activation of apoptosis. The present study was performed to verify the apoptotic effects of sequential treatment with bortezomib and celecoxib in TC-1 cells expressing the human papillomavirus (HPV) E6 and E7 proteins. In TC-1 cells sequentially treated with bortezomib and celecoxib, apoptosis was induced through decreased expression of signal transducer and activator of transcription-3 (STAT3), cyclin D1 and cyclin-dependent kinase (CDK) 2, which are major regulators of the G0/G1 cell cycle checkpoint. In addition, increased levels of p21, CHOP, BiP and p-p38 MAPK were identified in these cells. The treatment-induced apoptosis was effectively inhibited by treatment with SB203580, an inhibitor of p-p38. Moreover, the growth of tumors sequentially treated with bortezomib and celecoxib was retarded compared to the growth of tumors exposed to a single treatment with either bortezomib or celecoxib in vivo. We demonstrated that sequential treatment with bortezomib and celecoxib induced apoptosis via p-p38-mediated G0/G1 cell cycle arrest and endoplasmic reticulum (ER) stress. Sequential treatment with these two drugs could therefore be a useful therapy for cervical cancer.
Collapse
Affiliation(s)
- Jee-Eun Kim
- Department of Anatomy, Chung-Ang University, College of Medicine, Seoul, Republic of Korea
| | - Ji-In Lee
- Department of Anatomy, Chung-Ang University, College of Medicine, Seoul, Republic of Korea
| | - Dong-Hoon Jin
- Institute for Innovate Cancer Research, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Wang Jae Lee
- Department of Anatomy, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Ga Bin Park
- Department of Anatomy and Laboratory for Cancer Immunotherapy, Inje University, College of Medicine, Busan, Republic of Korea
| | - Seonghan Kim
- Department of Anatomy and Laboratory for Cancer Immunotherapy, Inje University, College of Medicine, Busan, Republic of Korea
| | - Yeong Seok Kim
- Department of Anatomy and Laboratory for Cancer Immunotherapy, Inje University, College of Medicine, Busan, Republic of Korea
| | - T-C Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Dae Young Hur
- Department of Anatomy and Laboratory for Cancer Immunotherapy, Inje University, College of Medicine, Busan, Republic of Korea
| | - Daejin Kim
- Department of Anatomy and Laboratory for Cancer Immunotherapy, Inje University, College of Medicine, Busan, Republic of Korea
| |
Collapse
|
13
|
Turkington RC, Purcell C, James CR, Millar J, Napier E, Law D, Gallagher R, Morris M, Wilson RH, Eatock MM. A phase I trial of bortezomib in combination with epirubicin, carboplatin and capecitabine (ECarboX) in advanced oesophagogastric adenocarcinoma. Invest New Drugs 2013; 32:250-60. [PMID: 23665866 DOI: 10.1007/s10637-013-9970-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 04/29/2013] [Indexed: 12/12/2022]
Abstract
PURPOSE The protease inhibitor bortezomib attenuates the action of NF-κB and has shown preclinical activity alone and in combination with chemotherapy. DESIGN A Phase I dose-escalation study was performed administering bortezomib (0.7, 1.0, 1.3 and 1.6 mg m(-2) on days 1 and 8 from cycle 2 onwards) in combination with Epirubicin 50 mg m(-2) intravenously on day 1, Carboplatin AUC 5 day 1 and Capecitabine 625 mg m(-2) BD days 1-21 every 21 days (VECarboX regimen), in patients with advanced oesophagogastric adenocarcinoma. The primary objective was to define the maximum tolerated dose (MTD) of Bortezomib when combined with ECarboX. RESULTS 18 patients received bortezomib 0.7 (n = 6), 1.0 (n = 3), 1.3 (n = 6) and 1.6 mg m(-2) (n = 3) and a protocol amendment reducing the capecitabine dose to 500 mg m(-2) BD was enacted due to myelotoxicity. Common treatment-related non-haematological adverse events of any grade were fatigue (83.3 %), anorexia (55.6 %), constipation (55.6 %) and nausea (55.6 %). Common Grade 3/4 haematological toxicities were neutropenia (77.8 %) and thrombocytopenia (44.4 %). Objective responses were achieved in 6 patients (33.3 %) and a further 5 patients (27.8 %) had stable disease for >8 weeks. CONCLUSIONS The addition of Bortezomib to ECarboX is well tolerated and response rates are comparable with standard chemotherapy.
Collapse
Affiliation(s)
- R C Turkington
- Northern Ireland Cancer Centre, Belfast City Hospital, Lisburn Road, Belfast, BT9 7AB, Northern Ireland, UK,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Yang X. Proteasome inhibitor bortezomi-induced the apoptosis of laryngeal squamous cell carcinoma Hep-2 cell line via disrupting redox equilibrium. Biomed Pharmacother 2012; 66:607-11. [DOI: 10.1016/j.biopha.2012.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 08/13/2012] [Indexed: 12/22/2022] Open
|
15
|
Zhang D, Fan D. New insights into the mechanisms of gastric cancer multidrug resistance and future perspectives. Future Oncol 2010; 6:527-37. [PMID: 20373867 DOI: 10.2217/fon.10.21] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is still the second leading cause of cancer death worldwide. Chemotherapy is one of the major treatment options for advanced gastric cancer. The efficacy of chemotherapy for gastric cancer is poor due to insensitivity and the development of multidrug resistance (MDR). Gastric cancer MDR involves a large number of molecules and complex mechanisms. Classical drug-resistant molecules, such as P-glycoprotein/ABCB1 and MRP1/ABCC1, have been found to play important roles in mediating MDR in some gastric cancers. In recent years, new molecules and mechanisms have been found to be associated with the development of gastric cancer MDR and might provide new targets for tackling gastric cancer MDR. Combined use of molecularly targeted therapy with chemotherapy may offer improved outcomes for gastric cancer patients and might provide new threads of hope for gastric cancer treatment.
Collapse
Affiliation(s)
- Dexin Zhang
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Fourth Military Medical University, 15 West Chang-Le Road, Xi'an 710032, People's Republic of China.
| | | |
Collapse
|
16
|
A multicenter, phase II study of bortezomib (PS-341) in patients with unresectable or metastatic gastric and gastroesophageal junction adenocarcinoma. Invest New Drugs 2010; 29:1475-81. [PMID: 20574790 DOI: 10.1007/s10637-010-9474-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 06/07/2010] [Indexed: 12/14/2022]
Abstract
PURPOSE The transcription factor nuclear factor-kB (NFkB) is implicated in gastric cancer carcinogenesis and survival, and its inhibition by proteosome inhibition is associated with preclinical gastric cancer anti-tumor activity. We examined the single agent efficacy of bortezomib, a selective proteasome inhibitor, in gastric adenocarcinoma. EXPERIMENTAL DESIGN We performed a phase II trial of bortezomib in patients with advanced gastric adenocarcinoma. Bortezomib 1.3 mg/m(2) was administered on days 1, 4, 8, and 11 every 21 days. The primary endpoint was objective response rate(RR); the null hypothesis was RR <1% versus the alternative ≥15%. One response in the first stage(15 patients) was required before proceeding with an additional 18 patients. If at least 2 or more responses out of 33 were observed, further study with bortezomib was warranted. Correlative studies evaluated pre-treatment tumor expression of NFkB, IkB, p53, p21, and cyclin D1. RESULTS We enrolled 16 patients (15 evaluable for response) from four institutions. No patients demonstrated an objective response(95% CI, 0-22%); one patient achieved stable disease. Fourteen out of 16 patients experienced ≥ grade 2 toxicity. The most common toxicity was fatigue in six patients (n = 4 grade 2, n = 2 grade 3). Seven patients experienced neuropathy (n = 5 grade 1, and 1 each grade 2 and 3). Seven (60%) had high cytoplasmic staining for NFkB. CONCLUSIONS Single agent bortezomib is inactive in metastatic gastric adenocarcinoma and should not be pursued. Future study of proteasome inhibition in gastric adenocarcinoma should be considered in combination with targeted inhibition of other non-overlapping oncogenic pathways as a potential rational approach.
Collapse
|
17
|
Ling X, Calinski D, Chanan-Khan AA, Zhou M, Li F. Cancer cell sensitivity to bortezomib is associated with survivin expression and p53 status but not cancer cell types. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:8. [PMID: 20096120 PMCID: PMC2826345 DOI: 10.1186/1756-9966-29-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 01/22/2010] [Indexed: 12/24/2022]
Abstract
Background Survivin is known playing a role in drug resistance. However, its role in bortezomib-mediated inhibition of growth and induction of apoptosis is unclear. There are conflicting reports for the effect of bortezomib on survivin expression, which lacks of a plausible explanation. Methods: In this study, we tested cancer cells with both p53 wild type and mutant/null background for the relationship of bortezomib resistance with survivin expression and p53 status using MTT assay, flow cytometry, DNA fragmentation, caspase activation, western blots and RNAi technology. Results We found that cancer cells with wild type p53 show a low level expression of survivin and are sensitive to treatment with bortezomib, while cancer cells with a mutant or null p53 show a high level expression of survivin and are resistant to bortezomib-mediated apoptosis induction. However, silencing of survivin expression utilizing survivin mRNA-specific siRNA/shRNA in p53 mutant or null cells sensitized cancer cells to bortezomib mediated apoptosis induction, suggesting a role for survivin in bortezomib resistance. We further noted that modulation of survivin expression by bortezomib is dependent on p53 status but independent of cancer cell types. In cancer cells with mutated p53 or p53 null, bortezomib appears to induce survivin expression, while in cancer cells with wild type p53, bortezomib downregulates or shows no significant effect on survivin expression, which is dependent on the drug concentration, cell line and exposure time. Conclusions Our findings, for the first time, unify the current inconsistent findings for bortezomib treatment and survivin expression, and linked the effect of bortezomib on survivin expression, apoptosis induction and bortezomib resistance in the relationship with p53 status, which is independent of cancer cell types. Further mechanistic studies along with this line may impact the optimal clinical application of bortezomib in solid cancer therapeutics.
Collapse
Affiliation(s)
- Xiang Ling
- Departments of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | | | |
Collapse
|
18
|
Hiss DC, Gabriels GA. Implications of endoplasmic reticulum stress, the unfolded protein response and apoptosis for molecular cancer therapy. Part II: targeting cell cycle events, caspases, NF-κB and the proteasome. Expert Opin Drug Discov 2009; 4:907-21. [PMID: 23480539 DOI: 10.1517/17460440903055032] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Endoplasmic reticulum stress (ERS), the unfolded protein response (UPR) and apoptosis signal transduction pathways are fundamental to normal cellular homeostasis and survival, but are exploited by cancer cells to promote the cancer phenotype. OBJECTIVE Collateral activation of ERS and UPR role players impact on cell growth, cell cycle arrest or apoptosis, genomic stability, tumour initiation and progression, tumour aggressiveness and drug resistance. An understanding of these processes affords promising prospects for specific cancer drug targeting of the ERS, UPR and apoptotic pathways. METHOD This review (Part II of II) brings forward the latest developments relevant to the molecular connections among cell cycle regulators, caspases, NF-κB, and the proteasome with ERS and UPR signalling cascades, their functions in apoptosis induction, apoptosis resistance and oncogenesis, and how these relationships can be exploited for targeted cancer therapy. CONCLUSION Overall, ERS, the UPR and apoptosis signalling cascades (the molecular therapeutic targets) and the development of drugs that attack these targets signify a success story in cancer drug discovery, but a more reductionist approach is necessary to determine the precise molecular switches that turn on antiapoptotic and pro-apoptotic programmes.
Collapse
Affiliation(s)
- Donavon C Hiss
- Head, Molecular Oncology Research Programme, University of the Western Cape, Department of Medical BioSciences, Bellville, 7535, South Africa +27 21 959 2334 ; +27 959 1563 ;
| | | |
Collapse
|
19
|
Gastric cancer in the era of molecularly targeted agents: current drug development strategies. J Cancer Res Clin Oncol 2009; 135:855-66. [PMID: 19363621 DOI: 10.1007/s00432-009-0583-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 03/26/2009] [Indexed: 12/15/2022]
Abstract
Gastric cancer is the second most common cause of cancer death worldwide with approximately one million cases diagnosed annually. Despite considerable improvements in surgical techniques, innovations in clinical diagnostics and the development of new chemotherapy regimens, the clinical outcome for patients with advanced gastric cancer and cancer of the GEJ is generally poor with 5-year survival rates ranging between 5 and 15%. The understanding of cancer relevant events has resulted in new therapeutic strategies, particularly in developing of new molecular targeted agents. These agents have the ability to target a variety of cancer relevant receptors and downstream pathways including the epidermal growth factor receptor (EGFR), the vascular endothelial growth factor receptor (VEGFR), the insulin-like growth factor receptor (IGFR), the c-Met pathway, cell-cycle pathways, and down-stream signalling pathways such as the Akt-PI3k-mTOR pathway. In the era of new molecularly targeted agents this review focuses on recent developments of targeting relevant pathways involved in gastric cancer and cancer of the GEJ.
Collapse
|
20
|
Targeting apoptosis as an approach for gastrointestinal cancer therapy. Drug Resist Updat 2009; 12:55-64. [PMID: 19278896 DOI: 10.1016/j.drup.2009.02.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Revised: 02/09/2009] [Accepted: 02/10/2009] [Indexed: 12/27/2022]
Abstract
Cancers in the gastrointestinal system account for a large proportion of malignancies and cancer-related deaths with gastric cancer and colorectal cancer being the most common ones. For those patients in whom surgical resection is not possible, other therapeutic approaches are necessary. Disordered apoptosis has been linked to cancer development and treatment resistance. Apoptosis occurs via extrinsic or intrinsic signaling each triggered and regulated by many different molecular pathways. In recent years, the selective induction of apoptosis in tumor cells has been increasingly recognized as a promising approach for cancer therapy. A detailed understanding of the molecular pathways involved in the regulation of apoptosis is essential for developing novel effective therapeutic approaches. Apoptosis can be induced by many different approaches including activating cell surface death receptors (for example, Fas, TRAIL and TNF receptors), inhibiting cell survival signaling (such as EGFR, MAPK and PI3K), altering apoptosis threshold by modulating pro-apoptotic and anti-apoptotic members of the Bcl-2 family, down-regulating anti-apoptosis proteins (such as XIAP, survivin and c-IAP2), and using other pro-apoptotic agents. In this review, the authors reviewed the currently reported apoptosis-targeting approaches in gastrointestinal cancers.
Collapse
|
21
|
Sarközi R, Perco P, Hochegger K, Enrich J, Wiesinger M, Pirklbauer M, Eder S, Rudnicki M, Rosenkranz AR, Mayer B, Mayer G, Schramek H. Bortezomib-induced survival signals and genes in human proximal tubular cells. J Pharmacol Exp Ther 2008; 327:645-56. [PMID: 18776064 DOI: 10.1124/jpet.108.142604] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Bortezomib has been introduced recently in the therapy of multiple myeloma (MM), a disease that is frequently associated with progressive renal failure. Because bortezomib-based therapy has been reported to lead to a rapid recovery of kidney function in patients with MM, we decided to study its direct effects in proximal tubular epithelial cells (PTCs) compared with glomerular mesangial cells (GMCs). After 24 h of stimulation, 50 nM bortezomib led to a 6.37-fold induction of apoptosis and markedly activated caspase-9 and -3 in GMCs but not in PTCs. In PTCs but not in GMCs, bortezomib led to a strong time-dependent degradation of IkappaB-alpha and to a long-lasting phosphorylation of both NF-kappaBp65 and extracellular signal-regulated kinase 1/2. Microarray analysis in bortezomib-treated PTCs revealed a time-dependent predominance of antiapoptotic genes compared with proapoptotic genes. Bortezomib (50 nM) induced heat shock protein (Hsp) 70 mRNA and protein levels in PTCs, whereas basal and bortezomib-stimulated Hsp70 protein expression was much weaker in GMCs. Moreover, bortezomib induced Bcl-2-associated athanogene (BAG) 3 mRNA and protein expression but inhibited BAG5 mRNA levels in PTCs. These data suggest that the reduced susceptibility of PTCs to bortezomib-induced cell apoptosis is because of cell type-specific effects of this compound on apoptosis/survival genes and pathways. The concept of bortezomib representing a blocker of both NF-kappaB activation and cell survival should be carefully examined in particular renal cell types.
Collapse
Affiliation(s)
- Rita Sarközi
- Division of Nephrology and Hypertension, Department of Internal Medicine IV, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Bortezomib, paclitaxel, and carboplatin as a first-line regimen for patients with metastatic esophageal, gastric, and gastroesophageal cancer: phase II results from the North Central Cancer Treatment Group (N044B). J Thorac Oncol 2008; 3:516-20. [PMID: 18449005 DOI: 10.1097/jto.0b013e31816de276] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE This study was undertaken to explore the response rate of a first-line, three-drug regimen that consisted of bortezomib, paclitaxel, and carboplatin in patients with metastatic adenocarcinoma of the esophagus, gastroesophageal junction, or gastric cardia. PATIENTS AND METHODS Patients with the above diagnosis and acceptable organ function were treated intravenously on a 21-day cycle with the following: bortezomib 1.2 mg/m on days 1, 4, and 8; paclitaxel 175 mg/m on day 2; and carboplatin with an area under the curve of 6 on day 2. Patients received indefinite treatment unless they manifested tumor progression or severe adverse events. All were monitored for tumor response as well as other clinical outcomes. RESULTS The cohort included 35 eligible patients with a median age of 59 years (range, 36-78) and an Eastern Cooperative Oncology Group performance score of 0, 1, and 2 in 60%, 34%, and 6% of patients, respectively. Although this regimen was well tolerated, the tumor response rate was lower than that anticipated at 23% (95% confidence interval: 10%, 40%), thereby prompting premature study closure. There were no complete responses. The median survival for the cohort was 8.9 months (95% confidence interval: 5.9, 12.8). CONCLUSION As prescribed in this trial and for this indication, this regimen does not merit further testing.
Collapse
|
23
|
Anti-proliferative and chemosensitizing effects of luteolin on human gastric cancer AGS cell line. Mol Cell Biochem 2008; 313:125-32. [PMID: 18398671 DOI: 10.1007/s11010-008-9749-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Accepted: 03/28/2008] [Indexed: 10/22/2022]
Abstract
To investigate the anti-proliferative and chemosensitizing effects of luteolin on human gastric cancer, gastric cancer AGS cells were treated with luteolin and/or other chemotherapeutic agents. Cell growth was assessed by MTT assay, cell cycle and apoptosis were assessed by flow-cytometric analysis, and the expression of major proteins regulating cell cycle and apoptosis was also detected. The results showed that luteolin inhibited the growth of gastric cancer cells in a dose- and time-dependent manner. Flow cytometry revealed that the percentage of cells at G2/M phase increased dose-dependently. The protein levels of Cdc2, Cyclin B1 and Cdc25C were reduced and p21/cip1 was up-regulated after the treatment with luteolin. Furthermore, luteolin induced apoptosis in gastric cancer AGS cells. Western blotting showed that luteolin treatment significantly increased the levels of pro-apoptotic proteins, including Caspase-3, 6, 9, Bax, and p53, and decreased the levels of anti-apoptotic protein Bcl-2, thus shifting the Bax/Bcl ratio in favor of apoptosis. It was also demonstrated that a combinational treatment of cisplatin and luteolin induced more effectively cell growth inhibition, compared to cisplatin treatment alone. These findings indicate the anti-proliferative and chemosensitizing effects of luteolin on human gastric cancer AGS cells and luteolin may be a promising candidate agent used in the treatment of gastric cancer.
Collapse
|