1
|
Mantzourani C, Vasilakaki S, Gerogianni VE, Kokotos G. The discovery and development of transmembrane serine protease 2 (TMPRSS2) inhibitors as candidate drugs for the treatment of COVID-19. Expert Opin Drug Discov 2022; 17:231-246. [PMID: 35072549 PMCID: PMC8862169 DOI: 10.1080/17460441.2022.2029843] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused the devastating pandemic named coronavirus disease 2019 (COVID-19). Unfortunately, the discovery of antiviral agents to combat COVID-19 is still an unmet need. Transmembrane serine protease 2 (TMPRSS2) is an important mediator in viral infection and thus, TMPRRS2 inhibitors may be attractive agents for COVID-19 treatment. AREAS COVERED This review article discusses the role of TMPRSS2 in SARS-CoV-2 cell entry and summarizes the inhibitors of TMPRSS2 and their potential anti-SARS activity. Two known TMPRSS2 inhibitors, namely camostat and nafamostat, approved drugs for the treatment of pancreatitis, are under clinical trials as potential drugs against COVID-19. EXPERT OPINION Due to the lack of the crystal structure of TMPRSS2, homology models have been developed to study the interactions of known inhibitors, including repurposed drugs, with the enzyme. However, novel TMPRSS2 inhibitors have been identified through high-throughput screening, and appropriate assays studying their in vitro activity have been set up. The discovery of TMPRSS2's crystal structure will facilitate the rational design of novel inhibitors and in vivo studies and clinical trials will give a clear answer if TMPRSS2 inhibitors could be a new weapon against COVID-19.
Collapse
Affiliation(s)
- Christiana Mantzourani
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Vasilakaki
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Athens, Greece
| | - Velisaria-Eleni Gerogianni
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Athens, Greece
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
2
|
Yang Q, Zhang S, Wu S, Yao B, Wang L, Li Y, Peng H, Huang M, Bi Q, Xiong P, Li L, Deng Y, Deng Y. Identification of nafamostat mesylate as a selective stimulator of NK cell IFN-γ production via metabolism-related compound library screening. Immunol Res 2022; 70:354-364. [PMID: 35167033 PMCID: PMC8852993 DOI: 10.1007/s12026-022-09266-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
Natural killer (NK) cells play important roles in controlling virus-infected and malignant cells. The identification of new molecules that can activate NK cells may effectively improve the antiviral and antitumour activities of these cells. In this study, by using a commercially available metabolism-related compound library, we initially screened the capacity of compounds to activate NK cells by determining the ratio of interferon-gamma (IFN-γ)+ NK cells by flow cytometry after the incubation of peripheral blood mononuclear cells (PBMCs) with IL-12 or IL-15 for 18 h. Our data showed that eight compounds (nafamostat mesylate (NM), loganin, fluvastatin sodium, atorvastatin calcium, lovastatin, simvastatin, rosuvastatin calcium, and pitavastatin calcium) and three compounds (NM, elesclomol, and simvastatin) increased the proportions of NK cells and CD3+ T cells that expressed IFN-γ among PBMCs cultured with IL-12 and IL-15, respectively. When incubated with enriched NK cells (purity ≥ 80.0%), only NM enhanced NK cell IFN-γ production in the presence of IL-12 or IL-15. When incubated with purified NK cells (purity ≥ 99.0%), NM promoted NK cell IFN-γ secretion in the presence or absence of IL-18. However, NM showed no effect on NK cell cytotoxicity. Collectively, our study identifies NM as a selective stimulator of IFN-γ production by NK cells, providing a new strategy for the prevention and treatment of infection or cancer in select populations.
Collapse
Affiliation(s)
- Qinglan Yang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China.,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China
| | - Shuju Zhang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China.,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China
| | - Shuting Wu
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China.,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China
| | - Baige Yao
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China.,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Lili Wang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China.,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China
| | - Yana Li
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China.,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China
| | - Hongyan Peng
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China.,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China
| | - Minghui Huang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China.,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China
| | - Qinghua Bi
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Peiwen Xiong
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China.,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China
| | - Liping Li
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China. .,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China.
| | - Yafei Deng
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, 410007, China. .,Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, 410007, China.
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
3
|
NF-κB in Gastric Cancer Development and Therapy. Biomedicines 2021; 9:biomedicines9080870. [PMID: 34440074 PMCID: PMC8389569 DOI: 10.3390/biomedicines9080870] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is considered one of the most common causes of cancer-related death worldwide and, thus, a major health problem. A variety of environmental factors including physical and chemical noxae, as well as pathogen infections could contribute to the development of gastric cancer. The transcription factor nuclear factor kappa B (NF-κB) and its dysregulation has a major impact on gastric carcinogenesis due to the regulation of cytokines/chemokines, growth factors, anti-apoptotic factors, cell cycle regulators, and metalloproteinases. Changes in NF-κB signaling are directed by genetic alterations in the transcription factors themselves, but also in NF-κB signaling molecules. NF-κB actively participates in the crosstalk of the cells in the tumor micromilieu with divergent effects on the heterogeneous tumor cell and immune cell populations. Thus, the benefits/consequences of therapeutic targeting of NF-κB have to be carefully evaluated. In this review, we address recent knowledge about the mechanisms and consequences of NF-κB dysregulation in gastric cancer development and therapy.
Collapse
|
4
|
Ghali GZ, Ghali MGZ. Nafamostat mesylate attenuates the pathophysiologic sequelae of neurovascular ischemia. Neural Regen Res 2020; 15:2217-2234. [PMID: 32594033 PMCID: PMC7749469 DOI: 10.4103/1673-5374.284981] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nafamostat mesylate, an apparent soi-disant panacea of sorts, is widely used to anticoagulate patients undergoing hemodialysis or cardiopulmonary bypass, mitigate the inflammatory response in patients diagnosed with acute pancreatitis, and reverse the coagulopathy of patients experiencing the commonly preterminal disseminated intravascular coagulation in the Far East. The serine protease inhibitor nafamostat mesylate exhibits significant neuroprotective effects in the setting of neurovascular ischemia. Nafamostat mesylate generates neuroprotective effects by attenuating the enzymatic activity of serine proteases, neuroinflammatory signaling cascades, and the endoplasmic reticulum stress responses, downregulating excitotoxic transient receptor membrane channel subfamily 7 cationic currents, modulating the activity of intracellular signal transduction pathways, and supporting neuronal survival (brain-derived neurotrophic factor/TrkB/ERK1/2/CREB, nuclear factor kappa B. The effects collectively reduce neuronal necrosis and apoptosis and prevent ischemia mediated disruption of blood-brain barrier microarchitecture. Investigational clinical applications of these compounds may mitigate ischemic reperfusion injury in patients undergoing cardiac, hepatic, renal, or intestinal transplant, preventing allograft rejection, and treating solid organ malignancies. Neuroprotective effects mediated by nafamostat mesylate support the wise conduct of randomized prospective controlled trials in Western countries to evaluate the clinical utility of this compound.
Collapse
Affiliation(s)
- George Zaki Ghali
- United States Environmental Protection Agency, Arlington, VA; Department of Toxicology, Purdue University, West Lafayette, IN, USA
| | - Michael George Zaki Ghali
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
5
|
Chen X, Xu Z, Zeng S, Wang X, Liu W, Qian L, Wei J, Yang X, Shen Q, Gong Z, Yan Y. The Molecular Aspect of Antitumor Effects of Protease Inhibitor Nafamostat Mesylate and Its Role in Potential Clinical Applications. Front Oncol 2019; 9:852. [PMID: 31552177 PMCID: PMC6733886 DOI: 10.3389/fonc.2019.00852] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/19/2019] [Indexed: 02/05/2023] Open
Abstract
Nafamostat mesylate (NM), a synthetic serine protease inhibitor first placed on the market by Japan Tobacco in 1986, has been approved to treat inflammatory-related diseases, such as pancreatitis. Recently, an increasing number of studies have highlighted the promising effects of NM in inhibiting cancer progression. Alone or in combination treatments, studies have shown that NM attenuates various malignant tumors, including pancreatic, colorectal, gastric, gallbladder, and hepatocellular cancers. In this review, based on several activating pathways, including the canonical Nuclear factor-κB (NF-κB) signaling pathway, tumor necrosis factor receptor-1 (TNFR1) signaling pathway, and tumorigenesis-related tryptase secreted by mast cells, we summarize the anticancer properties of NM in existing studies both in vitro and in vivo. In addition, the efficacy and side effects of NM in cancer patients are summarized in detail. To further clarify NM's antitumor activities, clinical trials devoted to validating the clinical applications and underlying mechanisms are needed in the future.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wanli Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Long Qian
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wei
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xue Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qiuying Shen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
6
|
Prevention of early liver metastasis after pancreatectomy by perioperative administration of a nuclear factor-κB inhibitor in mice. Surgery 2019; 166:991-996. [PMID: 31353078 DOI: 10.1016/j.surg.2019.05.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 05/08/2019] [Accepted: 05/27/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Liver metastasis is a common problem after pancreatectomy for pancreatic cancer. In pancreatic cancer cells, nuclear factor-κB is activated constitutively. Nuclear factor-κB activates matrix metalloproteinase-2/9, which plays an important role in cancer metastasis. Because the serine protease inhibitor FUT-175 suppresses nuclear factor-κB, we hypothesized that perioperative treatment with FUT-175 for pancreatic cancer may help to prevent liver metastasis. METHODS We compared in vitro cell viability, cell invasiveness, nuclear factor-κB signaling, and the expression levels of matrix metalloproteinase signals between the control group (C group) and the FUT-175 group (F group) using the murine pancreatic cancer cells PAN02. In addition, we evaluated the in vivo effect of pretreatment with FUT-175 using an established model of liver metastasis in mice. Metastatic liver lesions were assessed with magnetic resonance imaging. Liver recurrence and overall survival were evaluated. Also, the antimetastatic effect of systemic administration of FUT-175 was examined. RESULTS FUT-175 did not suppress the cell viability of PAN02 cells at or after 24 hours of treatment (P > .05); however, cell invasion was suppressed in the F group compared with the C group (P < .05). The levels of nuclear factor-κB activation, membrane type-1 (MT-1) matrix metalloproteinase (MMP)/matrix metalloproteinase-14 (MMP-14), and matrix metalloproteinase-2/9 (MMP-2/9) were lower in the F group compared with the C group. In vivo, both disease-free and overall survivals were prolonged in the F group compared with the C group. Systemic administration was also effective in suppressing the number of metastases. CONCLUSION Perioperative treatment with FUT-175 may help to prevent early liver metastasis after pancreatectomy for pancreatic cancer.
Collapse
|
7
|
Han X, Wang N, Li J, Wang Y, Wang R, Chang J. Identification of nafamostat mesilate as an inhibitor of the fat mass and obesity-associated protein (FTO) demethylase activity. Chem Biol Interact 2019; 297:80-84. [DOI: 10.1016/j.cbi.2018.10.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/12/2018] [Accepted: 10/24/2018] [Indexed: 12/25/2022]
|
8
|
Sugano H, Shirai Y, Horiuchi T, Saito N, Shimada Y, Eto K, Uwagawa T, Ohashi T, Yanaga K. Nafamostat Mesilate Enhances the Radiosensitivity and Reduces the Radiation-Induced Invasive Ability of Colorectal Cancer Cells. Cancers (Basel) 2018; 10:E386. [PMID: 30336548 PMCID: PMC6210678 DOI: 10.3390/cancers10100386] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022] Open
Abstract
Neoadjuvant chemoradiotherapy followed by radical surgery is the standard treatment for patients with locally advanced low rectal cancer. However, several studies have reported that ionizing radiation (IR) activates nuclear factor kappa B (NF-κB) that causes radioresistance and induces matrix metalloproteinase (MMP)-2/-9, which promote tumor migration and invasion. Nafamostat mesilate (FUT175), a synthetic serine protease inhibitor, enhances the chemosensitivity to cytotoxic agents in digestive system cancer cells by inhibiting NF-κB activation. Therefore, we evaluated the combined effect of IR and FUT175 on cell proliferation, migration and invasion of colorectal cancer (CRC) cells. IR-induced upregulation of intranuclear NF-κB, FUT175 counteracted this effect. Moreover, the combination treatment suppressed cell viability and induced apoptosis. Similar effects were also observed in xenograft tumors. In addition, FUT175 prevented the migration and invasion of cancer cells caused by IR by downregulating the enzymatic activity of MMP-2/-9. In conclusion, FUT175 enhances the anti-tumor effect of radiotherapy through downregulation of NF-κB and reduces IR-induced tumor invasiveness by directly inhibiting MMP-2/-9 in CRC cells. Therefore, the use of FUT175 during radiotherapy might improve the efficacy of radiotherapy in patients with CRC.
Collapse
Affiliation(s)
- Hiroshi Sugano
- Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
- Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Yoshihiro Shirai
- Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
- Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Takashi Horiuchi
- Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
- Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Nobuhiro Saito
- Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
- Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Yohta Shimada
- Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Ken Eto
- Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Tadashi Uwagawa
- Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
- Division of Medical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Toya Ohashi
- Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Katsuhiko Yanaga
- Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| |
Collapse
|
9
|
Haruki K, Shiba H, Shimada Y, Shirai Y, Iwase R, Fujiwara Y, Uwagawa T, Ohashi T, Yanaga K. Glycogen synthase kinase-3β activity plays a key role in the antitumor effect of nafamostat mesilate in pancreatic cancer cells. Ann Gastroenterol Surg 2017; 2:65-71. [PMID: 29863120 PMCID: PMC5868870 DOI: 10.1002/ags3.12025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/19/2017] [Indexed: 12/26/2022] Open
Abstract
Pancreatic cancer is often resistant to chemotherapy. We previously showed the efficacy of combination treatment using gemcitabine and nafamostat mesilate (FUT‐175) for patients with unresectable pancreatic cancer. However, the mechanisms that affect the sensitivity of FUT‐175 are not fully understood. The purpose of the present study was to clarify the mechanism of the sensitivity to FUT‐175, with a focus on the activity of glycogen synthase kinase‐3β (GSK‐3β). In vitro, we assessed sensitivity to FUT‐175 in human pancreatic cancer cell lines (PANC‐1 and MIAPaCa‐2) and difference of signaling in these cells by cell proliferation assay, Western blot analysis and microarray. Next, we assessed cell viability, apoptotic signal and nuclear factor‐kappa B (NF‐κB) activity in response to treatment with FUT‐175 alone and in combination with GSK‐3 inhibitor or protein phosphatase 2A (PP2A) by cell proliferation assay, Western blot analysis and enzyme‐linked immunosorbent assay. Phosphorylated GSK‐3β level was significantly higher in MIAPaCa‐2 (high sensitivity cell) than in PANC‐1 (low sensitivity cell). Cell viability and NF‐κB activity were significantly decreased by addition of GSK‐3 inhibitor to FUT‐175, and levels of cleaved caspase‐8 were increased by inhibition of GSK‐3. PP2A inhibitor increased the levels of phosphorylated GSK‐3β and sensitized both cell lines to FUT‐175 as measured by cell viability and apoptotic signal. The results indicate that GSK‐3β activity plays a key role in the antitumor effect of FUT‐175 in pancreatic cancer cells, and regulation of GSK‐3β by PP2A inhibition could be a novel therapeutic approach for pancreatic cancer.
Collapse
Affiliation(s)
- Koichiro Haruki
- Department of Surgery The Jikei University School of Medicine Tokyo Japan.,Division of Gene Therapy Research Center for Medical Science The Jikei University School of Medicine Tokyo Japan
| | - Hiroaki Shiba
- Department of Surgery The Jikei University School of Medicine Tokyo Japan
| | - Yohta Shimada
- Division of Gene Therapy Research Center for Medical Science The Jikei University School of Medicine Tokyo Japan
| | - Yoshihiro Shirai
- Department of Surgery The Jikei University School of Medicine Tokyo Japan.,Division of Gene Therapy Research Center for Medical Science The Jikei University School of Medicine Tokyo Japan
| | - Ryota Iwase
- Department of Surgery The Jikei University School of Medicine Tokyo Japan.,Division of Gene Therapy Research Center for Medical Science The Jikei University School of Medicine Tokyo Japan
| | - Yuki Fujiwara
- Department of Surgery The Jikei University School of Medicine Tokyo Japan
| | - Tadashi Uwagawa
- Department of Surgery The Jikei University School of Medicine Tokyo Japan
| | - Toya Ohashi
- Division of Gene Therapy Research Center for Medical Science The Jikei University School of Medicine Tokyo Japan
| | - Katsuhiko Yanaga
- Department of Surgery The Jikei University School of Medicine Tokyo Japan
| |
Collapse
|
10
|
Li Y, Zhou W, Tang K, Chen X, Feng Z, Chen J. Silencing Aurora A leads to re-sensitization of breast cancer cells to Taxol through downregulation of SRC-mediated ERK and mTOR pathways. Oncol Rep 2017; 38:2011-2022. [PMID: 28849180 PMCID: PMC5652950 DOI: 10.3892/or.2017.5908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/31/2017] [Indexed: 12/11/2022] Open
Abstract
While Taxol has been reported to improve the clinical survival of breast cancer patients, subsequently developed drug-resistance of the cancer cells limits its final efficacy and applications. Previous studies suggested that Aurora A is involved in the development of the Taxol-resistance of breast cancer. We established Taxol-resistant breast cancer MCF-7/T cells and xenograft models to explore the role of Aurora A in Taxol resistant ER-positive breast cancer. Compared with their parental MCF-7/C cells, the Taxol-resistant MCF-7/T cells exhibited enhanced colony formation, less cell death and higher invasive ability. The resistant cells presented overexpressed Aurora A, elevated phosphorylated SRC and upregulated Ras/Raf/ERK and Akt/mTOR pathways. Silencing of Aurora A reduced the activity of SRC and downregulated the ERK and Akt/mTOR pathways, which led to re-sensitization of the resistant MCF-7/T cells to Taxol in vitro. These results suggested that the activation of Aurora A and the subsequent upregulation of ERK and Akt through SRC induced Taxol-resistance in breast cancer cells, and inhibiting Aurora A and the related SRC/EKT/Akt pathway could restore the sensitivity of breast cancer cells to Taxol. These results might shed light on the development of strategies to circumvent Taxol-related chemoresistance in breast cancer clinical practice.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Wanqi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Ke Tang
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Zhiqiang Feng
- Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Jindong Chen
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
11
|
Sokolova O, Naumann M. NF-κB Signaling in Gastric Cancer. Toxins (Basel) 2017; 9:toxins9040119. [PMID: 28350359 PMCID: PMC5408193 DOI: 10.3390/toxins9040119] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/14/2017] [Accepted: 03/22/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is a leading cause of cancer death worldwide. Diet, obesity, smoking and chronic infections, especially with Helicobacter pylori, contribute to stomach cancer development. H. pylori possesses a variety of virulence factors including encoded factors from the cytotoxin-associated gene pathogenicity island (cagPAI) or vacuolating cytotoxin A (VacA). Most of the cagPAI-encoded products form a type 4 secretion system (T4SS), a pilus-like macromolecular transporter, which translocates CagA into the cytoplasm of the host cell. Only H. pylori strains carrying the cagPAI induce the transcription factor NF-κB, but CagA and VacA are dispensable for direct NF-κB activation. NF-κB-driven gene products include cytokines/chemokines, growth factors, anti-apoptotic factors, angiogenesis regulators and metalloproteinases. Many of the genes transcribed by NF-κB promote gastric carcinogenesis. Since it has been shown that chemotherapy-caused cellular stress could elicit activation of the survival factor NF-κB, which leads to acquisition of chemoresistance, the NF-κB system is recommended for therapeutic targeting. Research is motivated for further search of predisposing conditions, diagnostic markers and efficient drugs to improve significantly the overall survival of patients. In this review, we provide an overview about mechanisms and consequences of NF-κB activation in gastric mucosa in order to understand the role of NF-κB in gastric carcinogenesis.
Collapse
Affiliation(s)
- Olga Sokolova
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany.
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany.
| |
Collapse
|
12
|
Abstract
The development of drug resistance has largely limited the clinical outcome of anti-cancer treatment. Recent work has highlighted the involvement of non-coding RNAs, microRNAs (miRNAs), in cancer development. The present study aimed to investigate the role of miR-21 in the development of drug resistance to paclitaxel in gastric cancer cells. Our study found that the expression of miR-21 upregulated in the paclitaxel resistant cell line SGC7901/paclitaxel compared to its parental line SGC7901. Moreover, over-expression of miR-21 significantly decreased antiproliferative effects and apoptosis induced by paclitaxel, while knockdown of miR-21 dramatically increased antiproliferative effects and apoptosis induction by paclitaxel. Moreover, our results demonstrated that miR-21 may modulate the sensitivity to PTX, at least in part, by regulating the expression of P-glycoprotein.
Collapse
|
13
|
Lu YX, Ju HQ, Wang F, Chen LZ, Wu QN, Sheng H, Mo HY, Pan ZZ, Xie D, Kang TB, Chen G, Yun JP, Zeng ZL, Xu RH. Inhibition of the NF-κB pathway by nafamostat mesilate suppresses colorectal cancer growth and metastasis. Cancer Lett 2016; 380:87-97. [PMID: 27322737 DOI: 10.1016/j.canlet.2016.06.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/14/2016] [Accepted: 06/14/2016] [Indexed: 12/25/2022]
Abstract
Nafamostat mesilate is an anti-inflammatory drug that is usually used to treat pancreatitis. Recent studies show that it can suppress pancreatic cancer via inhibition of the nuclear factor κB (NF-κB) pathway. However, whether it has anti-tumor activity in some other cancer, including colorectal cancer (CRC), has not been investigated and remained unclear. Here, our study showed that nafamostat mesilate abrogated the constitutive NF-κB activation in CRC cells, which is mediated through inhibition of phosphorylation of IκBα and nuclear translocation of p65. Also, we found that nafamostat mesilate inhibited phosphorylation of Erk in CRC cells. Consistently, our study demonstrated that nafamostat mesilate inhibited the CRC cell proliferation, invasion and migration and induced mitochondria-dependent apoptosis. Furthermore, nafamostat mesilate could reverse oxaliplatin induced NF-κB and Erk activation in CRC cells, and enhance the sensitivity of CRC cells to oxaliplatin. Nafamostat mesilate combined with oxaliplatin repressed subcutaneous tumor growth and hepatic metastasis in vivo. Overall, our data suggest that nafamostat mesilate, a relatively non-toxic drug that targets NF-κB and Erk, may, in combination with oxaliplatin, represent a novel therapeutic strategy for CRC treatment.
Collapse
Affiliation(s)
- Yun-Xin Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Feng Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Le-Zong Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qi-Nian Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui Sheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hai-Yu Mo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhi-Zhong Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Tie-Bang Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Gong Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing-Ping Yun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhao-Lei Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
14
|
Takahashi R, Yokobori T, Osone K, Tatsuki H, Takada T, Suto T, Yajima R, Kato T, Fujii T, Tsutsumi S, Kuwano H, Asao T. Establishment of a novel method to evaluate peritoneal microdissemination and therapeutic effect using luciferase assay. Cancer Sci 2016; 107:341-6. [PMID: 26716425 PMCID: PMC4814254 DOI: 10.1111/cas.12872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 12/22/2015] [Accepted: 12/24/2015] [Indexed: 11/28/2022] Open
Abstract
Peritoneal dissemination is a major cause of recurrence in patients with malignant tumors in the peritoneal cavity. Effective anticancer agents and treatment protocols are necessary to improve outcomes in these patients. However, previous studies using mouse models of peritoneal dissemination have not detected any drug effect against peritoneal micrometastasis. Here we used the luciferase assay to evaluate peritoneal micrometastasis in living animals and established an accurate mouse model of early peritoneal microdissemination to evaluate tumorigenesis and drug efficacy. There was a positive correlation between luminescence intensity in in vivo luciferase assay and the extent of tumor dissemination evaluated by ex vivo luciferase assay and mesenteric weight. This model has advantages over previous models because optimal luciferin concentration without cell damage was validated and peritoneal microdissemination could be quantitatively evaluated. Therefore, it is a useful model to validate peritoneal micrometastasis formation and to evaluate drug efficacy without killing mice.
Collapse
Affiliation(s)
- Ryo Takahashi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Takehiko Yokobori
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan.,Department of Molecular Pharmacology and Oncology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Katsuya Osone
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Hironori Tatsuki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Takahiro Takada
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Toshinaga Suto
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Reina Yajima
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Toshihide Kato
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Takaaki Fujii
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Souichi Tsutsumi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Hiroyuki Kuwano
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Takayuki Asao
- Department of Oncology Clinical Development, Graduate School of Medicine, Gunma University, Maebashi, Japan
| |
Collapse
|
15
|
Shirai Y, Shiba H, Iwase R, Haruki K, Fujiwara Y, Furukawa K, Uwagawa T, Ohashi T, Yanaga K. Dual inhibition of nuclear factor kappa-B and Mdm2 enhance the antitumor effect of radiation therapy for pancreatic cancer. Cancer Lett 2015; 370:177-84. [PMID: 26546875 DOI: 10.1016/j.canlet.2015.10.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 10/28/2015] [Accepted: 10/28/2015] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Radiation therapy, alone or in combination with chemotherapy, is effective for patients with locally advanced and recurrent pancreatic cancer. Ionizing radiation induces cell cycle arrest and cell apoptosis through enhancement several signals such as p53, p21(Waf1/Cip1), and caspase. However, the therapeutic efficacy is attenuated by radiation-induced activation of NF-κB. Nafamostat mesilate, a synthetic serine protease inhibitor, inhibits NF-κB activation in pancreatic cancer. Therefore, we hypothesized that nafamostat mesilate inhibited radiation-induced activation of NF-κB and improves therapeutic outcome. RESULTS In combination group, NF-κB activation was significantly inhibited in comparison with that of radiation group. Nafamostat mesilate obviously down-regulated the expression levels of Mdm2 compared with control cells or irradiated cells. Consequently, p53 expression was stabilized inversely in correlation with Mdm2 protein expression level. The expression levels of p53, p21(Waf1/Cip1), cleaved caspase-3 and -8 were the highest in the combination group. Nafamostat mesilate enhanced ionizing radiation-induced cell apoptosis and G2/M cell cycle arrest. In combination group, cell proliferation and tumor growth were significantly slower than those in other groups. CONCLUSION Combination therapy of radiation with nafamostat mesilate exerts enhanced anti-tumor effect against human pancreatic cancer.
Collapse
Affiliation(s)
- Yoshihiro Shirai
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan; Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan.
| | - Hiroaki Shiba
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Ryota Iwase
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan; Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Koichiro Haruki
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuki Fujiwara
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Kenei Furukawa
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Tadashi Uwagawa
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Toya Ohashi
- Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Katsuhiko Yanaga
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Leporini C, Ammendola M, Marech I, Sammarco G, Sacco R, Gadaleta CD, Oakley C, Russo E, De Sarro G, Ranieri G. Targeting mast cells in gastric cancer with special reference to bone metastases. World J Gastroenterol 2015; 21:10493-10501. [PMID: 26457010 PMCID: PMC4588072 DOI: 10.3748/wjg.v21.i37.10493] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 06/15/2015] [Accepted: 08/25/2015] [Indexed: 02/06/2023] Open
Abstract
Bone metastases from gastric cancer (GC) are considered a relatively uncommon finding; however, they are related to poorer prognosis. Both primary GC and its metastatic progression rely on angiogenesis. Several lines of evidence from GC patients strongly support the involvement of mast cells (MCs) positive to tryptase (MCPT) in primary gastric tumor angiogenesis. Recently, we analyzed infiltrating MCs and neovascularization in bone tissue metastases from primary GC patients, and observed a significant correlation between infiltrating MCPT and angiogenesis. Such a finding suggested the involvement of peritumoral MCPT by infiltrating surrounding tumor cells, and in bone metastasis angiogenesis from primary GC. Thus, an MCPT-stimulated angiogenic process could support the development of metastases in bone tissue. From this perspective, we aim to review the hypothetical involvement of tumor-infiltrating, peritumoral MCPT in angiogenesis-mediated GC cell growth in the bone microenvironment and in tumor-induced osteoclastic bone resorption. We also focus on the potential use of MCPT targeting agents, such as MCs tryptase inhibitors (gabexate mesylate, nafamostat mesylate) or c-KitR tyrosine kinase inhibitors (imatinib, masitinib), as possible new anti-angiogenic and anti-resorptive strategies for the treatment of GC patients affected by bone metastases.
Collapse
|
17
|
Haruki K, Shiba H, Fujiwara Y, Furukawa K, Iwase R, Uwagawa T, Misawa T, Ohashi T, Yanaga K. Inhibition of nuclear factor-κB enhances the antitumor effect of tumor necrosis factor-α gene therapy for hepatocellular carcinoma in mice. Surgery 2013; 154:468-78. [PMID: 23972653 DOI: 10.1016/j.surg.2013.05.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Accepted: 05/28/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is often resistant to chemotherapy. Gene therapy using an adenoviral vector-expressing tumor necrosis factor (TNF)-α is a new therapeutic approach for chemoresistant malignancies. The efficacy of TNF-α, however, is limited, because it leads to the activation of nuclear factor (NF)-κB. We hypothesized that the NF-κB inhibitor nafamostat mesilate would enhance the antitumor effect of adenovirus vector-mediated TNF-α gene therapy for HCC. METHODS In vitro, we assessed the inhibitory effect of nafamostat mesilate on TNF-α-induced NF-κB activation and enhanced apoptosis in human HCC cell lines (Huh-7 and Hep3B). In vivo, we established a xenograft HCC model in mice by subcutaneous injection of Huh-7 and Hep3B cells. The animals received intraperitoneal (IP) injections of nafamostat mesilate 3 times a week (nafamostat mesilate group), intratumoral (IT) injections of the human TNF-α-expressing adenoviral vector (AxCAhTNF-α) once a week (TNF-α group), IT injections of AxCAhTNF-α once a week, or IP injections of nafamostat mesilate 3 times a week (combination group). RESULTS In the combination group, TNF-α-induced NF-κB activation was inhibited and TNF-α-induced apoptosis was enhanced in comparison with the other groups both in vitro and in vivo. In the combination group, tumor growth was significantly slower and the apoptotic cell numbers were significantly greater than those of the TNF-α group. CONCLUSION Inhibition of NF-κB by nafamostat mesilate enhances the antitumor effect of adenoviral vector-mediated TNF-α gene therapy for HCC in mice.
Collapse
Affiliation(s)
- Koichiro Haruki
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Combination chemotherapy of nafamostat mesylate with gemcitabine for gallbladder cancer targeting nuclear factor-κB activation. J Surg Res 2013; 184:605-12. [DOI: 10.1016/j.jss.2013.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/21/2013] [Accepted: 06/04/2013] [Indexed: 01/30/2023]
|