1
|
Schulz B, Leitner E, Schreiber T, Lindner T, Schwarz R, Aboutara N, Ma Y, Escobar HM, Palme R, Hinz B, Vollmar B, Zechner D. Sex Matters-Insights from Testing Drug Efficacy in an Animal Model of Pancreatic Cancer. Cancers (Basel) 2024; 16:1901. [PMID: 38791980 PMCID: PMC11120498 DOI: 10.3390/cancers16101901] [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: 04/27/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Preclinical studies rarely test the efficacy of therapies in both sexes. The field of oncology is no exception in this regard. In a model of syngeneic, orthotopic, metastasized pancreatic ductal adenocarcinoma we evaluated the impact of sex on pathological features of this disease as well as on the efficacy and possible adverse side effects of a novel, small molecule-based therapy inhibiting KRAS:SOS1, MEK1/2 and PI3K signaling in male and female C57BL/6J mice. Male mice had less tumor infiltration of CD8-positive cells, developed bigger tumors, had more lung metastasis and a lower probability of survival compared to female mice. These more severe pathological features in male animals were accompanied by higher distress at the end of the experiment. The evaluated inhibitors BI-3406, trametinib and BKM120 showed synergistic effects in vitro. This combinatorial therapy reduced tumor weight more efficiently in male animals, although the drug concentrations were similar in the tumors of both sexes. These results underline the importance of sex-specific preclinical research and at the same time provide a solid basis for future studies with the tested compounds.
Collapse
Affiliation(s)
- Benjamin Schulz
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| | - Emily Leitner
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| | - Tim Schreiber
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| | - Tobias Lindner
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Rico Schwarz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Nadine Aboutara
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Yixuan Ma
- Department of Medicine Clinic III, Hematology, Oncology and Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Hugo Murua Escobar
- Department of Medicine Clinic III, Hematology, Oncology and Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Rupert Palme
- Experimental Endocrinology, Department of Biological Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| | - Dietmar Zechner
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| |
Collapse
|
2
|
Hu H, Ng TS, Kang M, Scott E, Li R, Quintana JM, Matvey D, Vantaku VR, Weissleder R, Parangi S, Miller MA. Thyroid Cancers Exhibit Oncogene-Enhanced Macropinocytosis that Is Restrained by IGF1R and Promote Albumin-Drug Conjugate Response. Clin Cancer Res 2023; 29:3457-3470. [PMID: 37289199 PMCID: PMC10527034 DOI: 10.1158/1078-0432.ccr-22-2976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/22/2022] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
PURPOSE Oncogene-driven macropinocytosis fuels nutrient scavenging in some cancer types, yet whether this occurs in thyroid cancers with prominent MAPK-ERK and PI3K pathway mutations remains unclear. We hypothesized that understanding links between thyroid cancer signaling and macropinocytosis might uncover new therapeutic strategies. EXPERIMENTAL DESIGN Macropinocytosis was assessed across cells derived from papillary thyroid cancer (PTC), follicular thyroid cancer (FTC), non-malignant follicular thyroid, and aggressive anaplastic thyroid cancer (ATC), by imaging fluorescent dextran and serum albumin. The impacts of ectopic BRAFV600E and mutant RAS, genetic PTEN silencing, and inhibitors targeting RET, BRAF, and MEK kinases were quantified. BrafV600E p53-/- ATC tumors in immunocompetent mice were used to measure efficacy of an albumin-drug conjugate comprising microtubule-destabilizing monomethyl auristatin E (MMAE) linked to serum albumin via a cathepsin-cleavable peptide (Alb-vc-MMAE). RESULTS FTC and ATC cells showed greater macropinocytosis than non-malignant and PTC cells. ATC tumors accumulated albumin at 8.8% injected dose per gram tissue. Alb-vc-MMAE, but not MMAE alone, reduced tumor size by >90% (P < 0.01). ATC macropinocytosis depended on MAPK/ERK activity and nutrient signaling, and increased by up to 230% with metformin, phenformin, or inhibition of IGF1Ri in monoculture but not in vivo. Macrophages also accumulated albumin and express the cognate IGF1R ligand, IGF1, which reduced ATC responsiveness to IGF1Ri. CONCLUSIONS These findings identify regulated oncogene-driven macropinocytosis in thyroid cancers and demonstrate the potential of designing albumin-bound drugs to efficiently treat them.
Collapse
Affiliation(s)
- Huiyu Hu
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
- Department of General Surgery, Xiangya Hospital, Central South University, China
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Mikyung Kang
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Ella Scott
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Jeremy M. Quintana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Dylan Matvey
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Venkata R. Vantaku
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
- Department of Systems Biology, Harvard Medical School, United States
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| |
Collapse
|
3
|
Basu D, Pal R, Sarkar M, Barma S, Halder S, Roy H, Nandi S, Samadder A. To Investigate Growth Factor Receptor Targets and Generate Cancer Targeting Inhibitors. Curr Top Med Chem 2023; 23:2877-2972. [PMID: 38164722 DOI: 10.2174/0115680266261150231110053650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024]
Abstract
Receptor tyrosine kinase (RTK) regulates multiple pathways, including Mitogenactivated protein kinases (MAPKs), PI3/AKT, JAK/STAT pathway, etc. which has a significant role in the progression and metastasis of tumor. As RTK activation regulates numerous essential bodily processes, including cell proliferation and division, RTK dysregulation has been identified in many types of cancers. Targeting RTK is a significant challenge in cancer due to the abnormal upregulation and downregulation of RTK receptors subfamily EGFR, FGFR, PDGFR, VEGFR, and HGFR in the progression of cancer, which is governed by multiple RTK receptor signalling pathways and impacts treatment response and disease progression. In this review, an extensive focus has been carried out on the normal and abnormal signalling pathways of EGFR, FGFR, PDGFR, VEGFR, and HGFR and their association with cancer initiation and progression. These are explored as potential therapeutic cancer targets and therefore, the inhibitors were evaluated alone and merged with additional therapies in clinical trials aimed at combating global cancer.
Collapse
Affiliation(s)
- Debroop Basu
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Riya Pal
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, IndiaIndia
| | - Maitrayee Sarkar
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Soubhik Barma
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Sumit Halder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Harekrishna Roy
- Nirmala College of Pharmacy, Vijayawada, Guntur, Andhra Pradesh, India
| | - Sisir Nandi
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur, 244713, India
| | - Asmita Samadder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| |
Collapse
|
4
|
Uesato N, Kitagawa Y, Matsuo Y, Miyagawa N, Inagaki K, Kakefuda R, Yamaguchi T, Hata T, Ikegashira K, Matsushita M. Therapeutic Effect of Colony Stimulating Factor 1 Receptor Kinase Inhibitor, JTE-952, on Methotrexate-Refractory Pathology in a Rat Model of Rheumatoid Arthritis. Biol Pharm Bull 2023; 46:1223-1230. [PMID: 37661402 DOI: 10.1248/bpb.b23-00148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by inflammation and the destruction of bone and cartilage in affected joints. One of the unmet medical needs in the treatment of RA is to effectively prevent the structural destruction of joints, especially bone, which progresses because of resistance to conventional drugs that mainly have anti-inflammatory effects, and directly leads to a decline in the QOL of patients. We previously developed a novel and orally available type II kinase inhibitor of colony-stimulating factor-1 receptor (CSF1R), JTE-952. CSF1R is specifically expressed by monocytic-lineage cells, including bone-resorbing osteoclasts, and is important for promoting the differentiation and proliferation of osteoclasts. In the present study, we investigated the therapeutic effect of JTE-952 on methotrexate (MTX)-refractory joint destruction in a clinically established adjuvant-induced arthritis rat model. JTE-952 did not suppress paw swelling under inflammatory conditions, but it inhibited the destruction of joint structural components including bone and cartilage in the inflamed joints. In addition, decreased range of joint motion and mechanical hyperalgesia after disease onset were suppressed by JTE-952. These results suggest that JTE-952 is expected to prevent the progression of the structural destruction of joints and its associated effects on joint motion and pain by inhibiting CSF1/CSF1R signaling in RA pathology, which is resistant to conventional disease-modifying anti-rheumatic drugs such as MTX.
Collapse
Affiliation(s)
- Naofumi Uesato
- Central Pharmaceutical Research Institute, Japan Tobacco Inc
| | | | - Yushi Matsuo
- Central Pharmaceutical Research Institute, Japan Tobacco Inc
| | - Naoki Miyagawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc
| | - Koji Inagaki
- Central Pharmaceutical Research Institute, Japan Tobacco Inc
| | - Reina Kakefuda
- Central Pharmaceutical Research Institute, Japan Tobacco Inc
| | | | - Takahiro Hata
- Central Pharmaceutical Research Institute, Japan Tobacco Inc
| | | | | |
Collapse
|
5
|
Yu H, Wu Z, Bao X, Tang X, Zhang J, Zhang Y, Hu M. A sustained-release Trametinib bio-multifunction hydrogel inhibits orthodontically induced inflammatory root resorption. RSC Adv 2022; 12:16444-16453. [PMID: 35754868 PMCID: PMC9168831 DOI: 10.1039/d2ra00763k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 05/13/2022] [Indexed: 11/21/2022] Open
Abstract
Orthodontic tooth movement (OTM) is a bone reconstruction process. In most cases, OTM could induce root resorption as a common side effect, called orthodontically induced inflammatory root resorption (OIIRR). OIIRR affects tooth health and interferes with the stability of orthodontic treatment. Osteoclasts, which perform bone resorption in OTM, attack cementum, causing OIIRR. Many signaling pathways are involved in the maturation and differentiation of osteoclasts, among which the ERK1/2 is one of the important pathways. In this experiment, we added Trametinib (Tra), a specific inhibitor of ERK1/2, to catechol-modified chitosan (CHI-C) and oxidized dextran (ODex) to form a CCOD-Trametinib composite hydrogel (CCOD-Tra) to prevent OIIRR. CCOD-Tra exhibited good biocompatibility, injectability, strong adhesion, good hemostatic function and sustained release of Tra. We performed local injection of CCOD-Tra into the periodontal tissues of rats. CCOD-Tra firmly adhered to the periodontal tissues and then released Tra to establish a good biological environment and maintain a drug concentration at a high level around the roots for a long time. H&E, TRAP, immunochemistry staining and micro-CT indicated that CCOD-Tra had a good effect in terms of preventing OIIRR. Cell experiments showed that CCOD-Tra reduced the expression of TRAP, MMP-9 and C-FOS in osteoclast cells through the ERK1/2 signaling pathway to inhibit the differentiation and maturation of osteoclasts. Based on the above results, we concluded that CCOD-Tra had the ability to prevent OIIRR, the high adhesion and injectability of CCOD may provide better therapeutic ideas for clinical prevention of OIIRR.
Collapse
Affiliation(s)
- Hang Yu
- Department of Orthodontics, Hospital of Stomatology, Jilin University No. 1500 Qinghua Road, ChaoYang District Changchun Jilin P. R. China +86 431 88975348 +86 431 85579371 +86 13504484365
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling (School and Hospital of Stomatology, Jilin University) P. R. China
| | - Zhina Wu
- Department of Orthodontics, Hospital of Stomatology, Jilin University No. 1500 Qinghua Road, ChaoYang District Changchun Jilin P. R. China +86 431 88975348 +86 431 85579371 +86 13504484365
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling (School and Hospital of Stomatology, Jilin University) P. R. China
| | - Xingfu Bao
- Department of Orthodontics, Hospital of Stomatology, Jilin University No. 1500 Qinghua Road, ChaoYang District Changchun Jilin P. R. China +86 431 88975348 +86 431 85579371 +86 13504484365
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling (School and Hospital of Stomatology, Jilin University) P. R. China
| | - Xiaoduo Tang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University Changchun 130012 P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Junhu Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University Changchun 130012 P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University Changchun 130021 P. R. China
| | - Yi Zhang
- Department of Orthodontics, Hospital of Stomatology, Jilin University No. 1500 Qinghua Road, ChaoYang District Changchun Jilin P. R. China +86 431 88975348 +86 431 85579371 +86 13504484365
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling (School and Hospital of Stomatology, Jilin University) P. R. China
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University No. 1500 Qinghua Road, ChaoYang District Changchun Jilin P. R. China +86 431 88975348 +86 431 85579371 +86 13504484365
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling (School and Hospital of Stomatology, Jilin University) P. R. China
| |
Collapse
|
6
|
Tenshin H, Teramachi J, Ashtar M, Hiasa M, Inoue Y, Oda A, Tanimoto K, Shimizu S, Higa Y, Harada T, Oura M, Sogabe K, Hara T, Sumitani R, Maruhashi T, Sebe M, Tsutsumi R, Sakaue H, Endo I, Matsumoto T, Tanaka E, Abe M. TGF‐β‐activated kinase‐1 inhibitor LL‐Z1640‐2 reduces joint inflammation and bone destruction in mouse models of rheumatoid arthritis by inhibiting NLRP3 inflammasome, TACE, TNF‐α and RANKL expression. Clin Transl Immunology 2022; 11:e1371. [PMID: 35079379 PMCID: PMC8770968 DOI: 10.1002/cti2.1371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 10/29/2021] [Accepted: 01/06/2022] [Indexed: 02/03/2023] Open
Affiliation(s)
- Hirofumi Tenshin
- Department of Orthodontics and Dentofacial Orthopedics Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Jumpei Teramachi
- Department of Oral Function and Anatomy, Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama University Okayama Japan
| | - Mohannad Ashtar
- Department of Orthodontics and Dentofacial Orthopedics Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Masahiro Hiasa
- Department of Orthodontics and Dentofacial Orthopedics Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Yusuke Inoue
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Asuka Oda
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and Dentofacial Orthopedics Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - So Shimizu
- Department of Orthodontics and Dentofacial Orthopedics Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Yoshiki Higa
- Department of Orthodontics and Dentofacial Orthopedics Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Takeshi Harada
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Masahiro Oura
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Kimiko Sogabe
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Tomoyo Hara
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Ryohei Sumitani
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Tomoko Maruhashi
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Mayu Sebe
- Department of Nutrition and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Itsuro Endo
- Department of Bioregulatory Sciences Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences Tokushima University Tokushima Japan
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Masahiro Abe
- Department of Hematology, Endocrinology and Metabolism Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| |
Collapse
|
7
|
Zhu J, Wang J, Wang X, Gao M, Guo B, Gao M, Liu J, Yu Y, Wang L, Kong W, An Y, Liu Z, Sun X, Huang Z, Zhou H, Zhang N, Zheng R, Xie Z. Prediction of drug efficacy from transcriptional profiles with deep learning. Nat Biotechnol 2021; 39:1444-1452. [PMID: 34140681 DOI: 10.1038/s41587-021-00946-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 05/06/2021] [Indexed: 02/05/2023]
Abstract
Drug discovery focused on target proteins has been a successful strategy, but many diseases and biological processes lack obvious targets to enable such approaches. Here, to overcome this challenge, we describe a deep learning-based efficacy prediction system (DLEPS) that identifies drug candidates using a change in the gene expression profile in the diseased state as input. DLEPS was trained using chemically induced changes in transcriptional profiles from the L1000 project. We found that the changes in transcriptional profiles for previously unexamined molecules were predicted with a Pearson correlation coefficient of 0.74. We examined three disorders and experimentally tested the top drug candidates in mouse disease models. Validation showed that perillen, chikusetsusaponin IV and trametinib confer disease-relevant impacts against obesity, hyperuricemia and nonalcoholic steatohepatitis, respectively. DLEPS can generate insights into pathogenic mechanisms, and we demonstrate that the MEK-ERK signaling pathway is a target for developing agents against nonalcoholic steatohepatitis. Our findings suggest that DLEPS is an effective tool for drug repurposing and discovery.
Collapse
Affiliation(s)
- Jie Zhu
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China.,Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Jingxiang Wang
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, China
| | - Xin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Mingjing Gao
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, China
| | - Bingbing Guo
- Department of Anatomy, Histology and Embryology, Neuroscience Research Institute, Health Science Center, Peking University, Beijing, China
| | - Miaomiao Gao
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, Neuroscience Research Institute, Health Science Center, Peking University, Beijing, China
| | - Yanqiu Yu
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China
| | - Liang Wang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Weikaixin Kong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Health Science Center, Peking University, Beijing, China
| | - Yongpan An
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Zurui Liu
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, China
| | - Xinpei Sun
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Health Science Center, Peking University, Beijing, China
| | - Hong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.
| | - Ning Zhang
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China.
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, Neuroscience Research Institute, Health Science Center, Peking University, Beijing, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China. .,Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, China.
| |
Collapse
|
8
|
Kurimoto T, Tamai I, Nakagawa T, Miyai A, Yamamoto Y, Kosugi Y, Deai K, Hata T, Ohta T, Matsushita M, Yamada T. JTP-117968, a novel selective glucocorticoid receptor modulator, exhibits significant anti-inflammatory effect while maintaining bone mineral density in mice. Eur J Pharmacol 2021; 895:173880. [PMID: 33476654 DOI: 10.1016/j.ejphar.2021.173880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/25/2020] [Accepted: 01/12/2021] [Indexed: 01/10/2023]
Abstract
Classic glucocorticoids have been prescribed for various inflammatory diseases, such as rheumatoid arthritis, due to their outstanding anti-inflammatory effects. However, glucocorticoids cause numerous unwanted side effects, including osteoporosis and diabetes. Hence, selective glucocorticoid receptor modulators (SGRMs), which retain anti-inflammatory effects with minimized side effects, are among the most anticipated drugs in the clinical field. The assumption is that there are two major mechanisms of action via glucocorticoid receptors, transrepression (TR) and transactivation (TA). In general, anti-inflammatory effects of glucocorticoids are largely due to TR, while the side effects associated with glucocorticoids are mostly mediated through TA. We previously reported that JTP-117968, a novel SGRM, maintained partial TR activity while remarkably reducing the TA activity. In this study, we investigated the anti-inflammatory effect of JTP-117968 on a lipopolysaccharide (LPS) challenge model and collagen-induced arthritis (CIA) model in mice. Meanwhile, we tested the effect of JTP-117968 on the bone mineral density (BMD) in mouse femur to evaluate the side effect. Based on the evaluation, JTP-117968 reduced the plasma levels of tumor necrosis factor α induced by LPS challenge in mice significantly. Remarkably, CIA development was suppressed by JTP-117968 comparably with prednisolone and PF-802, an active form of fosdagrocorat that has been developed clinically as an orally available SGRM. Strikingly, the side effect of JTP-117968 on mouse femoral BMD was much lower than those of PF-802 and prednisolone. Therefore, JTP-117968 has attractive potential as a new therapeutic option against inflammatory diseases with minimized side effects compared to classic glucocorticoids.
Collapse
Affiliation(s)
- Takafumi Kurimoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan; Faculty of Agriculture, Department of Agrobiology, Niigata University, Niigata, Japan.
| | - Isao Tamai
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Takashi Nakagawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Atsuko Miyai
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Yasuo Yamamoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Yoshinori Kosugi
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Katsuya Deai
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Takahiro Hata
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Takeshi Ohta
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | | | - Takahisa Yamada
- Faculty of Agriculture, Department of Agrobiology, Niigata University, Niigata, Japan
| |
Collapse
|
9
|
Zhang ZQ, Hu XS, Lu YC, Zhang JP, Li WY, Zhang WY, Feng W, Ding DF, Xu JG. MEK1/2 Inhibitor (GDC0623) Promotes Osteogenic Differentiation of Primary Osteoblasts Inhibited by IL-1 β through the MEK-Erk1/2 and Jak/Stat3 Pathways. Int J Endocrinol 2021; 2021:5720145. [PMID: 34976051 PMCID: PMC8716208 DOI: 10.1155/2021/5720145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/02/2021] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE We evaluated the effects and mechanisms of GDC0623 on osteogenic differentiation of osteoblasts induced by IL-1β. Methodology. Osteoblasts were treated with 20 ng/ml IL-1β and 0.1 µM GDC0623. Cell proliferation levels were evaluated by the cell counting kit 8 (CCK8), EdU assay, and western blotting [proliferating cell nuclear antigen (PCNA) and Cyclin D1]. Osteoblasts were cultured in an osteogenic induction medium for 1-3 weeks after which their differentiations were assessed by alkaline phosphatase (ALP) staining, Alizarin Red staining, calcium concentration, immunocytochemistry staining, real-time quantitative PCR (RT-qPCR), and immunofluorescence staining. The osteogenesis-associated mechanisms were further evaluated by western blotting using appropriate antibodies. RESULTS Relative to the control group, IL-1β induced the rapid proliferation of osteoblasts and suppressed their osteogenic differentiations by upregulating the activities of MEK-Erk1/2 as well as Jak-Stat3 pathways and by elevating MMP13 and MMP9 levels. However, blocking of the MEK-Erk1/2 signaling pathway by GDC0623 treatment reversed these effects. CONCLUSION Inhibition of Jak-Stat3 pathway by C188-9 downregulated the expression levels of MMP9 and MMP13, activated MEK-Erk1/2 pathway, and inhibited osteogenic differentiation.
Collapse
Affiliation(s)
- Zeng-Qiao Zhang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Shen Hu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ye-Chen Lu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun-Peng Zhang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Yao Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei-Yang Zhang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Wei Feng
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dao-Fang Ding
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian-Guang Xu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
10
|
Arthralgia Induced by BRAF Inhibitor Therapy in Melanoma Patients. Cancers (Basel) 2020; 12:cancers12103004. [PMID: 33081201 PMCID: PMC7602871 DOI: 10.3390/cancers12103004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/05/2020] [Accepted: 10/15/2020] [Indexed: 11/16/2022] Open
Abstract
Simple Summary BRAF inhibitors (BRAFi) are standard of care for BRAF-mutated metastatic melanoma (MM). One of the most common side effects is arthralgia, for which a high incidence has been described, but whose clinical presentation and management have not yet been characterized. The aim of this retrospective study was to assess the patterns and clinical course of this drug-induced joint pain and to discuss a potential pathogenesis based on our clinical findings. In our cohort of patients treated with BRAFi between 2010 and 2018, 48 of 154 (31%) patients suffered from new-onset joint pain, which primarily affected small joints with a symmetrical pattern, as can be observed in patients affected by rheumatoid arthritis, the most frequent rheumatic and musculoskeletal disease. Most cases were sufficiently treated by non-steroidal anti-inflammatory drugs; however, some patients required dose reduction or permanent discontinuation of the BRAFi. Interestingly, we found that the occurrence of arthralgia was associated with better tumor control. Abstract Introduction: BRAF inhibitors (BRAFi), commonly used in BRAF-mutated metastatic melanoma (MM) treatment, frequently cause arthralgia. Although this is one of the most common side effects, it has not been characterized yet. Methods: We retrospectively included all patients treated with BRAFi +/− MEK inhibitors (MEKi) for MM at the National Center for Tumor Diseases (Heidelberg) between 2010 and 2018 and reviewed patient charts for the occurrence and management of arthralgia. The evaluation was supplemented by an analysis of frozen sera. Results: We included 154 patients (63% males); 31% (48/154) of them reported arthralgia with a median onset of 21 days after the start of the therapy. Arthralgia mostly affected small joints (27/36, 75%) and less frequently large joints (19/36, 53%). The most commonly affected joints were in fingers (19/36, 53%), wrists (16/36, 44%), and knees (12/36, 33%). In 67% (24/36) of the patients, arthralgia occurred with a symmetrical polyarthritis, mainly of small joints, resembling the pattern typically observed in patients affected by rheumatoid arthritis (RA), for which a role of the MAPK signaling pathway was previously described. Patients were negative for antinuclear antibodies, anti-citrullinated protein antibodies, and rheumatoid factor; arthritis was visible in 10 of 13 available PET–CT scans. The development of arthralgia was linked to better progression-free survival and overall survival. Conclusion: Arthralgia is a common side effect in patients receiving BRAFi +/− MEKi therapy and often presents a clinical pattern similar to that observed in RA patients. Its occurrence was associated with longer-lasting tumor control.
Collapse
|
11
|
Wang X, Wu F, Wang H, Duan X, Huang R, Tuersuntuoheti A, Su L, Yan S, Zhao Y, Lu Y, Li K, Yao J, Luo Z, Guo L, Liu J, Chen X, Lu Y, Hu H, Li X, Bao M, Bi X, Du B, Miao S, Cai J, Wang L, Zhou H, Ying J, Song W, Zhao H. PDCD6 cooperates with C-Raf to facilitate colorectal cancer progression via Raf/MEK/ERK activation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:147. [PMID: 32746883 PMCID: PMC7398064 DOI: 10.1186/s13046-020-01632-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023]
Abstract
Background Colorectal cancer (CRC) is one of the most common malignancies, and it’s expected that the CRC burden will substantially increase in the next two decades. New biomarkers for targeted treatment and associated molecular mechanism of tumorigenesis remain to be explored. In this study, we investigated whether PDCD6 plays an oncogenic role in colorectal cancer and its underlying mechanism. Methods Programmed cell death protein 6 (PDCD6) expression in CRC samples were analyzed by immunohistochemistry and immunofluorescence. The prognosis between PDCD6 and clinical features were analyzed. The roles of PDCD6 in cellular proliferation and tumor growth were measured by using CCK8, colony formation, and tumor xenograft in nude mice. RNA-sequence (RNA-seq), Mass Spectrum (MS), Co-Immunoprecipitation (Co-IP) and Western blot were utilized to investigate the mechanism of tumor progression. Immunohistochemistry (IHC) and quantitative real-time PCR (qRT-PCR) were performed to determine the correlation of PDCD6 and MAPK pathway. Results Higher expression levels of PDCD6 in tumor tissues were associated with a poorer prognosis in patients with CRC. Furthermore, PDCD6 increased cell proliferation in vitro and tumor growth in vivo. Mechanistically, RNA-seq showed that PDCD6 could affect the activation of the MAPK signaling pathway. PDCD6 interacted with c-Raf, resulting in the activation of downstream c-Raf/MEK/ERK pathway and the upregulation of core cell proliferation genes such as MYC and JUN. Conclusions These findings reveal the oncogenic effect of PDCD6 in CRC by activating c-Raf/MEK/ERK pathway and indicate that PDCD6 might be a potential prognostic indicator and therapeutic target for patients with colorectal cancer.
Collapse
Affiliation(s)
- Xiaojuan Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.,State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, TsinghuaUniversity, Beijing, 100084, China
| | - Fan Wu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Han Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Xiaoyuan Duan
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Rong Huang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Amannisa Tuersuntuoheti
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Luying Su
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Shida Yan
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuechao Zhao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Yan Lu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Kai Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Jinjie Yao
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiwen Luo
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Guo
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianmei Liu
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiao Chen
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yalan Lu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Hanjie Hu
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xingchen Li
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Mandula Bao
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinyu Bi
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Boyu Du
- Department of Medical Biology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Shiying Miao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Linfang Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Haitao Zhou
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China. .,Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Wei Song
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
| | - Hong Zhao
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China. .,Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
12
|
Takahagi A, Shindo T, Chen-Yoshikawa TF, Yoshizawa A, Gochi F, Miyamoto E, Saito M, Tanaka S, Motoyama H, Aoyama A, Takaori-Kondo A, Date H. Trametinib Attenuates Delayed Rejection and Preserves Thymic Function in Rat Lung Transplantation. Am J Respir Cell Mol Biol 2020; 61:355-366. [PMID: 30849233 DOI: 10.1165/rcmb.2018-0188oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Delayed immunological rejection after human lung transplantation causes chronic lung allograft dysfunction, which is associated with high mortality. Delayed rejection may be attributable to indirect alloantigen presentation by host antigen-presenting cells; however, its pathophysiology is not fully understood. The mitogen-activated protein kinase pathway is activated in T cells upon stimulation, and we previously showed that the MEK inhibitor, trametinib, suppresses graft-versus-host disease after murine bone marrow transplantation. We investigated whether trametinib suppresses graft rejection after two types of rat lung transplantation and analyzed its immunological mode of action. Major histocompatibility complex-mismatched transplantation from brown Norway rats into Lewis rats and minor histocompatibility antigen-mismatched transplantation from Fischer 344 rats into Lewis rats were performed. Cyclosporine (CsA) and/or trametinib were administered alone or consecutively. Acute and delayed rejection, lymphocyte infiltration, and pulmonary function were evaluated. Administration of trametinib after CsA suppressed delayed rejection, reduced inflammatory cell infiltration and fibrosis within the graft, and preserved pulmonary functions at Day 28. Trametinib suppressed functional differentiation of T and B cells in the periphery but preserved thymic T cell differentiation. Donor B cells within the graft disappeared by Day 14, indicating that delayed graft rejection at Day 28 was mainly due to indirect presentation by host antigen-presenting cells. Finally, trametinib administration without CsA preconditioning suppressed rejection after minor histocompatibility antigen-mismatched transplantation. Trametinib attenuates delayed rejection upon major histocompatibility complex-mismatched transplantation by suppressing indirect presentation and is a promising candidate to treat chronic lung allograft dysfunction in humans.
Collapse
Affiliation(s)
| | - Takero Shindo
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan; and
| | | | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | | | | | | | | | | | | | - Akifumi Takaori-Kondo
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan; and
| | | |
Collapse
|
13
|
Trametinib alleviates lipopolysaccharide-induced acute lung injury by inhibiting the MEK-ERK-Egr-1 pathway. Int Immunopharmacol 2020; 80:106152. [PMID: 31926447 DOI: 10.1016/j.intimp.2019.106152] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/09/2019] [Accepted: 12/22/2019] [Indexed: 01/15/2023]
Abstract
Acute lung injury (ALI) is a devastating clinical disorder with a high mortality rate and for which there is no effective treatment. The main characteristic of ALI is uncontrolled inflammation, and macrophages play a critical role in the development of this disorder. Trametinib, an inhibitor of MAPK/ERK kinase (MEK) activity that possesses anti-inflammatory properties, has been approved for clinical use. Herein, the influence of trametinib and its underlying mechanism were investigated using a lipopolysaccharide (LPS)-induced murine ALI model. We found that trametinib treatment prevented the LPS-facilitated expression of proinflammatory mediators in macrophages, and this anti-inflammatory action was closely correlated with suppression of the MEK-ERK-early growth response (Egr)-1 pathway. Furthermore, trametinib treatment alleviated LPS-induced ALI in mice, and attenuated edema, proinflammatory mediator production, and neutrophil infiltration. Trametinib pretreatment also attenuated the MEK-ERK-Egr-1 pathway in lung tissues. In conclusion, these data demonstrate that trametinib pretreatment suppresses inflammation in LPS-activated macrophages in vitro and protects against murine ALI established by LPS administration in vivo through inhibition of the MEK-ERK-Egr-1 pathway. Therefore, trametinib might have therapeutic potential for ALI.
Collapse
|
14
|
Chan KK, Bass AR. Checkpoint inhibitor-induced polymyalgia rheumatica controlled by cobimetinib, a MEK 1/2 inhibitor. Ann Rheum Dis 2019; 78:e70. [PMID: 29760158 DOI: 10.1136/annrheumdis-2018-213672] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 04/30/2018] [Indexed: 11/04/2022]
Affiliation(s)
- Karmela Kim Chan
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Anne R Bass
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
15
|
Li CC, Wang XJ, Wang HCR. Repurposing host-based therapeutics to control coronavirus and influenza virus. Drug Discov Today 2019; 24:726-736. [PMID: 30711575 PMCID: PMC7108273 DOI: 10.1016/j.drudis.2019.01.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/11/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022]
Abstract
Drug repositioning is a cost- and time-efficient approach for new indications. Targeting host machineries, used by viruses, could develop broad-spectrum antivirals. Repurposing existing drugs could efficiently identify antiviral agents.
The development of highly effective antiviral agents has been a major objective in virology and pharmaceutics. Drug repositioning has emerged as a cost-effective and time-efficient alternative approach to traditional drug discovery and development. This new shift focuses on the repurposing of clinically approved drugs and promising preclinical drug candidates for the therapeutic development of host-based antiviral agents to control diseases caused by coronavirus and influenza virus. Host-based antiviral agents target host cellular machineries essential for viral infections or innate immune responses to interfere with viral pathogenesis. This review discusses current knowledge, prospective applications and challenges in the repurposing of clinically approved and preclinically studied drugs for newly indicated antiviral therapeutics.
Collapse
Affiliation(s)
- Cui-Cui Li
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiao-Jia Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| | - Hwa-Chain Robert Wang
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, USA.
| |
Collapse
|
16
|
Ke XX, Pang Y, Chen K, Zhang D, Wang F, Zhu S, Mao J, Hu X, Zhang G, Cui H. Knockdown of arsenic resistance protein 2 inhibits human glioblastoma cell proliferation through the MAPK/ERK pathway. Oncol Rep 2018; 40:3313-3322. [PMID: 30542699 PMCID: PMC6196630 DOI: 10.3892/or.2018.6777] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/27/2018] [Indexed: 12/20/2022] Open
Abstract
It is generally known that glioblastoma is the most common primary malignant brain tumor and that it is highly aggressive and deadly. Although surgical and pharmacological therapies have made long‑term progress, glioblastoma remains extremely lethal and has an uncommonly low survival rate. Therefore, further elucidation of the molecular mechanisms of glioblastoma initiation and its pathological processes are urgent. Arsenic resistance protein 2 (Ars2) is a highly conserved gene, and it has been found to play an important role in microRNA biosynthesis and cell proliferation in recent years. Furthermore, absence of Ars2 results in developmental death in Drosophila, zebrafish and mice. However, there are few studies on the role of Ars2 in regulating tumor development, and the mechanism of its action is mostly unknown. In the present study, we revealed that Ars2 is involved in glioblastoma proliferation and we identified a potential mechanistic role for it in cell cycle control. Our data demonstrated that Ars2 knockdown significantly repressed the proliferation and tumorigenesis abilities of glioblastoma cells in vitro and in vivo. Further investigation clarified that Ars2 deficiency inhibited the activation of the MAPK/ERK pathway, leading to cell cycle arrest in the G1 phase, resulting in suppression of cell proliferation. These findings support the conclusion that Ars2 is a key regulator of glioblastoma progression.
Collapse
Affiliation(s)
- Xiao-Xue Ke
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
| | - Yi Pang
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing 404110, P.R. China
| | - Kuijun Chen
- Department 6 of The Research Institute of Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Dunke Zhang
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
| | - Feng Wang
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
| | - Shunqin Zhu
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
| | - Jingxin Mao
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
| | - Xiaosong Hu
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
| | - Guanghui Zhang
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
| | - Hongjuan Cui
- Cell Biology Laboratory, State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, P.R. China
| |
Collapse
|
17
|
Bao CX, Chen HX, Mou XJ, Zhu XK, Zhao Q, Wang XG. RETRACTED: GZMB gene silencing confers protection against synovial tissue hyperplasia and articular cartilage tissue injury in rheumatoid arthritis through the MAPK signaling pathway. Biomed Pharmacother 2018; 103:346-354. [PMID: 29669300 DOI: 10.1016/j.biopha.2018.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/03/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022] Open
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. An Expression of Concern for this article was previously published while an investigation was conducted (see related editorial: https://doi.org/10.1016/j.biopha.2022.113812). This retraction notice supersedes the Expression of Concern published earlier. Concern was raised about the reliability of the Western blot data in Figure 7C, which appear to contain a similar phenotype to those found in other publications, as detailed here: https://pubpeer.com/publications/7DD2DDC979F8CE2B00555332B01F81; and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. The journal requested the corresponding author comment on these concerns and provide the associated raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Cui-Xia Bao
- Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai 264000, PR China
| | - Hai-Xia Chen
- Clinical Laboratory, Yeda Hospital, Yantai 264000, PR China
| | - Xue-Jie Mou
- Clinical Laboratory, Yantai Taocun Central Hospital, Yantai 265301, PR China
| | - Xiang-Kui Zhu
- Department of Radiology, Yantai Oral Hospital, Yantai 264000, PR China
| | - Qi Zhao
- Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai 264000, PR China
| | - Xin-Guang Wang
- Department of Blood Transfusion, Yeda Hospital, Yantai 264000, PR China.
| |
Collapse
|
18
|
Song SC, An YM, Shin JH, Chung MJ, Seo JG, Kim E. Beneficial effects of a probiotic blend on gastrointestinal side effects induced by leflunomide and amlodipine in a rat model. Benef Microbes 2017; 8:801-808. [DOI: 10.3920/bm2016.0231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Patients with metabolic disorders frequently suffer from side effects induced by long-term oral medications. The present study using a rat model system indicated that leflunomide (LF) and amlodipine (AMD), the active ingredients contained in the medications for rheumatoid arthritis and hypertension, respectively, appeared to induce various bowel problems including constipation and inflammation. In the small and large intestine, LF increased the expression of tumour necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 compared to the null control and AMD increased the expression of both TNF-α and IL-1β, although its effect on IL-6 was only increased in the large intestine. It is noteworthy that the probiotic blend tested was found to alleviate intestinal complications caused by LF and AMD. Analysis of the gut microbiota revealed that AMD induced compositional changes in the gut microbiota. Namely, members of the phylum Bacteroidetes, which constituted only about 0.3% of the microbiota in the null control, made up more than 10% of the total composition in the AMD-administered rats. Interestingly, the probiotic blend was also found to normaliSe the gut microbiota.
Collapse
Affiliation(s)
- S.-C. Song
- Department of Systems Biology, Yonsei University, 323, SB140, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Y.-M. An
- Department of Systems Biology, Yonsei University, 323, SB140, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - J.-H. Shin
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - M.-J. Chung
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - J.-G. Seo
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - E. Kim
- Department of Systems Biology, Yonsei University, 323, SB140, Yonsei-ro 50, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
19
|
Allegrezza MJ, Rutkowski MR, Stephen TL, Svoronos N, Perales-Puchalt A, Nguyen JM, Payne KK, Singhal S, Eruslanov EB, Tchou J, Conejo-Garcia JR. Trametinib Drives T-cell-Dependent Control of KRAS-Mutated Tumors by Inhibiting Pathological Myelopoiesis. Cancer Res 2017; 76:6253-6265. [PMID: 27803104 DOI: 10.1158/0008-5472.can-16-1308] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/19/2016] [Indexed: 12/11/2022]
Abstract
Targeted therapies elicit seemingly paradoxical and poorly understood effects on tumor immunity. Here, we show that the MEK inhibitor trametinib abrogates cytokine-driven expansion of monocytic myeloid-derived suppressor cells (mMDSC) from human or mouse myeloid progenitors. MEK inhibition also reduced the production of the mMDSC chemotactic factor osteopontin by tumor cells. Together, these effects reduced mMDSC accumulation in tumor-bearing hosts, limiting the outgrowth of KRas-driven breast tumors, even though trametinib largely failed to directly inhibit tumor cell proliferation. Accordingly, trametinib impeded tumor progression in vivo through a mechanism requiring CD8+ T cells, which was paradoxical given the drug's reported ability to inhibit effector lymphocytes. Confirming our observations, adoptive transfer of tumor-derived mMDSC reversed the ability of trametinib to control tumor growth. Overall, our work showed how the effects of trametinib on immune cells could partly explain its effectiveness, distinct from its activity on tumor cells themselves. More broadly, by providing a more incisive view into how MEK inhibitors may act against tumors, our findings expand their potential uses to generally block mMDSC expansion, which occurs widely in cancers to drive their growth and progression. Cancer Res; 76(21); 6253-65. ©2016 AACR.
Collapse
Affiliation(s)
- Michael J Allegrezza
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Melanie R Rutkowski
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Tom L Stephen
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Nikolaos Svoronos
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Alfredo Perales-Puchalt
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Jenny M Nguyen
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Kyle K Payne
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evgeniy B Eruslanov
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Julia Tchou
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jose R Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania.
| |
Collapse
|
20
|
Peres RS, Santos GB, Cecilio NT, Jabor VAP, Niehues M, Torres BGS, Buqui G, Silva CHTP, Costa TD, Lopes NP, Nonato MC, Ramalho FS, Louzada-Júnior P, Cunha TM, Cunha FQ, Emery FS, Alves-Filho JC. Lapachol, a compound targeting pyrimidine metabolism, ameliorates experimental autoimmune arthritis. Arthritis Res Ther 2017; 19:47. [PMID: 28270195 PMCID: PMC5341405 DOI: 10.1186/s13075-017-1236-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 01/13/2017] [Indexed: 12/29/2022] Open
Abstract
Background The inhibition of pyrimidine biosynthesis by blocking the dihydroorotate dehydrogenase (DHODH) activity, the prime target of leflunomide (LEF), has been proven to be an effective strategy for rheumatoid arthritis (RA) treatment. However, a considerable proportion of RA patients are refractory to LEF. Here, we investigated lapachol (LAP), a natural naphthoquinone, as a potential DHODH inhibitor and addressed its immunosuppressive properties. Methods Molecular flexible docking studies and bioactivity assays were performed to determine the ability of LAP to interact and inhibit DHODH. In vitro studies were conducted to assess the antiproliferative effect of LAP using isolated lymphocytes. Finally, collagen-induced arthritis (CIA) and antigen-induced arthritis (AIA) models were employed to address the anti-arthritic effects of LAP. Results We found that LAP is a potent DHODH inhibitor which had a remarkable ability to inhibit both human and murine lymphocyte proliferation in vitro. Importantly, uridine supplementation abrogated the antiproliferative effect of LAP, supporting that the pyrimidine metabolic pathway is the target of LAP. In vivo, LAP treatment markedly reduced CIA and AIA progression as evidenced by the reduction in clinical score, articular tissue damage, and inflammation. Conclusions Our findings propose a binding model of interaction and support the ability of LAP to inhibit DHODH, decreasing lymphocyte proliferation and attenuating the severity of experimental autoimmune arthritis. Therefore, LAP could be considered as a potential immunosuppressive lead candidate with potential therapeutic implications for RA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1236-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raphael S Peres
- Department of Pharmacology, Ribeirão Preto Medical School, Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Avenida Bandeirantes, 3900, Ribeirão Preto, São Paulo, CEP: 14049-900, Brazil
| | - Gabriela B Santos
- Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, CEP: 14040-903, Brazil
| | - Nerry T Cecilio
- Department of Pharmacology, Ribeirão Preto Medical School, Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Avenida Bandeirantes, 3900, Ribeirão Preto, São Paulo, CEP: 14049-900, Brazil
| | - Valquíria A P Jabor
- NPPNS, Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, Brazil
| | - Michael Niehues
- NPPNS, Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, Brazil
| | - Bruna G S Torres
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Sarmento Leite 521, Porto Alegre, Brazil
| | - Gabriela Buqui
- NPPNS, Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, Brazil
| | - Carlos H T P Silva
- Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, CEP: 14040-903, Brazil
| | - Teresa Dalla Costa
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Sarmento Leite 521, Porto Alegre, Brazil
| | - Norberto P Lopes
- NPPNS, Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, Brazil
| | - Maria C Nonato
- NPPNS, Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, Brazil
| | - Fernando S Ramalho
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Paulo Louzada-Júnior
- Department of Internal Medicine, Ribeirão Preto Medical School, Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Avenida Bandeirantes, 3900, Ribeirão Preto, São Paulo, CEP: 14049-900, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Avenida Bandeirantes, 3900, Ribeirão Preto, São Paulo, CEP: 14049-900, Brazil
| | - Flavio S Emery
- Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/n, Ribeirão Preto, CEP: 14040-903, Brazil.
| | - Jose C Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Avenida Bandeirantes, 3900, Ribeirão Preto, São Paulo, CEP: 14049-900, Brazil.
| |
Collapse
|
21
|
Targeted Therapy and Immunosuppression in the Tumor Microenvironment. Trends Cancer 2016; 3:19-27. [PMID: 28718424 DOI: 10.1016/j.trecan.2016.11.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/21/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023]
Abstract
Small-molecule inhibitors offer great promise for targeting pathways that are specifically deregulated in different tumors. However, such 'targeted' therapies also elicit poorly understood effects on protective antitumor immunity. Given the emerging relevance of immunotherapies that boost pre-existing T cell responses, understanding how different immune cells are affected by small-molecule inhibitors could lead to more-effective interventions, alone or combined with immunotherapy. This review discusses the growing array of activities elicited by multiple 'targeted' inhibitors on antitumor immunity, underscoring the complex effects resulting from diverse activities on different immune cell types in vivo, and the need to conduct mechanistic research that identifies drugs performing well not only in immunocompromised mice but also in the presence of spontaneous or therapeutic antitumor immunity.
Collapse
|
22
|
Itamura H, Shindo T, Tawara I, Kubota Y, Kariya R, Okada S, Komanduri KV, Kimura S. The MEK inhibitor trametinib separates murine graft-versus-host disease from graft-versus-tumor effects. JCI Insight 2016; 1:e86331. [PMID: 27699218 PMCID: PMC5033881 DOI: 10.1172/jci.insight.86331] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/31/2016] [Indexed: 01/21/2023] Open
Abstract
The efficacy of allogeneic hematopoietic stem cell transplantation for hematologic malignancies is limited by the difficulty in suppressing graft-versus-host disease (GVHD) without compromising graft-versus-tumor (GVT) effects. We previously showed that RAS/MEK/ERK signaling depends on memory differentiation in human T cells, which confers susceptibility to selective inhibition of naive T cells. Actually, antineoplastic MEK inhibitors selectively suppress alloreactive T cells, sparing virus-specific T cells in vitro. Here, we show that trametinib, a MEK inhibitor clinically approved for melanoma, suppresses GVHD safely without affecting GVT effects in vivo. Trametinib prolonged survival of GVHD mice and attenuated GVHD symptoms and pathology in the gut and skin. It inhibited ERK1/2 phosphorylation and expansion of donor T cells, sparing Tregs and B cells. Although high-dose trametinib inhibited myeloid cell engraftment, low-dose trametinib suppressed GVHD without severe adverse events. Notably, trametinib facilitated the survival of mice transplanted with allogeneic T cells and P815 tumor cells with no residual P815 cells observed in the livers and spleens, whereas tacrolimus resulted in P815 expansion. These results confirm that trametinib selectively suppresses GVHD-inducing T cells while sparing antitumor T cells in vivo, which makes it a promising candidate for translational studies aimed at preventing or treating GVHD.
Collapse
Affiliation(s)
- Hidekazu Itamura
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| | - Takero Shindo
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| | - Isao Tawara
- Department of Hematology/Oncology, Mie University School of Medicine, Tsu, Japan
| | - Yasushi Kubota
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| | - Ryusho Kariya
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Krishna V. Komanduri
- Adult Stem Cell Transplant Program and Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shinya Kimura
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| |
Collapse
|
23
|
Allegrezza MJ, Rutkowski MR, Stephen TL, Svoronos N, Tesone AJ, Perales-Puchalt A, Nguyen JM, Sarmin F, Sheen MR, Jeng EK, Tchou J, Wong HC, Fiering SN, Conejo-Garcia JR. IL15 Agonists Overcome the Immunosuppressive Effects of MEK Inhibitors. Cancer Res 2016; 76:2561-72. [PMID: 26980764 DOI: 10.1158/0008-5472.can-15-2808] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/01/2016] [Indexed: 11/16/2022]
Abstract
Many signal transduction inhibitors are being developed for cancer therapy target pathways that are also important for the proper function of antitumor lymphocytes, possibly weakening their therapeutic effects. Here we show that most inhibitors targeting multiple signaling pathways have especially strong negative effects on T-cell activation at their active doses on cancer cells. In particular, we found that recently approved MEK inhibitors displayed potent suppressive effects on T cells in vitro However, these effects could be attenuated by certain cytokines that can be administered to cancer patients. Among them, clinically available IL15 superagonists, which can activate PI3K selectively in T lymphocytes, synergized with MEK inhibitors in vivo to elicit potent and durable antitumor responses, including by a vaccine-like effect that generated resistance to tumor rechallenge. Our work identifies a clinically actionable approach to overcome the T-cell-suppressive effects of MEK inhibitors and illustrates how to reconcile the deficiencies of signal transduction inhibitors, which impede desired immunologic effects in vivo Cancer Res; 76(9); 2561-72. ©2016 AACR.
Collapse
Affiliation(s)
- Michael J Allegrezza
- Tumor Microenvironment and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Melanie R Rutkowski
- Tumor Microenvironment and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Tom L Stephen
- Tumor Microenvironment and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Nikolaos Svoronos
- Tumor Microenvironment and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Amelia J Tesone
- Tumor Microenvironment and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | | | - Jenny M Nguyen
- Tumor Microenvironment and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Fahmida Sarmin
- Tumor Microenvironment and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Mee R Sheen
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Emily K Jeng
- Research & Development, Altor BioScience Corporation, Miramar, Florida
| | - Julia Tchou
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania. Rena Rowan Breast Center, University of Pennsylvania, Philadelphia, Pennsylvania. Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hing C Wong
- Research & Development, Altor BioScience Corporation, Miramar, Florida
| | - Steven N Fiering
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Jose R Conejo-Garcia
- Tumor Microenvironment and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania.
| |
Collapse
|
24
|
Tanimoto A, Shinozaki Y, Nozawa K, Kimoto Y, Amano W, Matsuo A, Yamaguchi T, Matsushita M. Improvement of spontaneous locomotor activity with JAK inhibition by JTE-052 in rat adjuvant-induced arthritis. BMC Musculoskelet Disord 2015; 16:339. [PMID: 26546348 PMCID: PMC4636776 DOI: 10.1186/s12891-015-0802-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 11/03/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to joint destruction, disability, and decreased quality of life (QOL). Inhibition of Janus kinase (JAK) signaling ameliorates articular inflammation and joint destruction in animal models of RA, but its effects on behaviors indicating well-being are poorly understood. In this study, we evaluated the effect of JAK inhibition on spontaneous locomotor activity in rats with adjuvant-induced arthritis, a rodent model of RA. METHODS Arthritis was induced in male Lewis rats by a single subcutaneous injection of Freund's complete adjuvant. The novel JAK inhibitor JTE-052 was orally administered for 7 days after the onset of arthritis. RESULTS Induction of arthritis suppressed the spontaneous locomotor activity of the rats. Administration of JTE-052 completely improved the spontaneous locomotor activity, with partial reductions in articular inflammation and joint destruction. Hyperalgesia and motor functions were also improved, but the efficacy was not complete. However, serum interleukin (IL)-6 levels were completely decreased at 4 h after administration of the first dose of JTE-052. CONCLUSIONS This study demonstrated that JAK inhibition improved the spontaneous locomotor activity of rats with adjuvant-induced arthritis, in association with amelioration of pain and physical dysfunction as a consequence of suppression of joint inflammation. Moreover, although further studies are needed, there was possible participation of IL-6 downregulation in the improvement of locomotor activity by JAK inhibition.
Collapse
Affiliation(s)
- Atsuo Tanimoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan.
| | - Yuichi Shinozaki
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan.
| | - Keisuke Nozawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan.
| | - Yukari Kimoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan.
| | - Wataru Amano
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan.
| | - Akira Matsuo
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan.
| | - Takayuki Yamaguchi
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan.
| | - Mutsuyoshi Matsushita
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan.
| |
Collapse
|
25
|
Smith JA, Stallons LJ, Collier JB, Chavin KD, Schnellmann RG. Suppression of mitochondrial biogenesis through toll-like receptor 4-dependent mitogen-activated protein kinase kinase/extracellular signal-regulated kinase signaling in endotoxin-induced acute kidney injury. J Pharmacol Exp Ther 2015; 352:346-57. [PMID: 25503387 PMCID: PMC4293437 DOI: 10.1124/jpet.114.221085] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/10/2014] [Indexed: 12/26/2022] Open
Abstract
Although disruption of mitochondrial homeostasis and biogenesis (MB) is a widely accepted pathophysiologic feature of sepsis-induced acute kidney injury (AKI), the molecular mechanisms responsible for this phenomenon are unknown. In this study, we examined the signaling pathways responsible for the suppression of MB in a mouse model of lipopolysaccharide (LPS)-induced AKI. Downregulation of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), a master regulator of MB, was noted at the mRNA level at 3 hours and protein level at 18 hours in the renal cortex, and was associated with loss of renal function after LPS treatment. LPS-mediated suppression of PGC-1α led to reduced expression of downstream regulators of MB and electron transport chain proteins along with a reduction in renal cortical mitochondrial DNA content. Mechanistically, Toll-like receptor 4 (TLR4) knockout mice were protected from renal injury and disruption of MB after LPS exposure. Immunoblot analysis revealed activation of tumor progression locus 2/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (TPL-2/MEK/ERK) signaling in the renal cortex by LPS. Pharmacologic inhibition of MEK/ERK signaling attenuated renal dysfunction and loss of PGC-1α, and was associated with a reduction in proinflammatory cytokine (e.g., tumor necrosis factor-α [TNF-α], interleukin-1β) expression at 3 hours after LPS exposure. Neutralization of TNF-α also blocked PGC-1α suppression, but not renal dysfunction, after LPS-induced AKI. Finally, systemic administration of recombinant tumor necrosis factor-α alone was sufficient to produce AKI and disrupt mitochondrial homeostasis. These findings indicate an important role for the TLR4/MEK/ERK pathway in both LPS-induced renal dysfunction and suppression of MB. TLR4/MEK/ERK/TNF-α signaling may represent a novel therapeutic target to prevent mitochondrial dysfunction and AKI produced by sepsis.
Collapse
Affiliation(s)
- Joshua A Smith
- Department of Drug Discovery and Biomedical Sciences (J.A.S., L.J.S., J.B.C., R.G.S.) and Division of Transplant Surgery, Department of Surgery (K.D.C.), Medical University of South Carolina, Charleston, South Carolina ; and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| | - L Jay Stallons
- Department of Drug Discovery and Biomedical Sciences (J.A.S., L.J.S., J.B.C., R.G.S.) and Division of Transplant Surgery, Department of Surgery (K.D.C.), Medical University of South Carolina, Charleston, South Carolina ; and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| | - Justin B Collier
- Department of Drug Discovery and Biomedical Sciences (J.A.S., L.J.S., J.B.C., R.G.S.) and Division of Transplant Surgery, Department of Surgery (K.D.C.), Medical University of South Carolina, Charleston, South Carolina ; and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| | - Kenneth D Chavin
- Department of Drug Discovery and Biomedical Sciences (J.A.S., L.J.S., J.B.C., R.G.S.) and Division of Transplant Surgery, Department of Surgery (K.D.C.), Medical University of South Carolina, Charleston, South Carolina ; and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| | - Rick G Schnellmann
- Department of Drug Discovery and Biomedical Sciences (J.A.S., L.J.S., J.B.C., R.G.S.) and Division of Transplant Surgery, Department of Surgery (K.D.C.), Medical University of South Carolina, Charleston, South Carolina ; and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| |
Collapse
|
26
|
Myelosuppressive and hepatotoxic potential of leflunomide and methotrexate combination in a rat model of rheumatoid arthritis. Pharmacol Rep 2014; 67:102-14. [PMID: 25560583 DOI: 10.1016/j.pharep.2014.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/09/2014] [Accepted: 08/01/2014] [Indexed: 11/20/2022]
Abstract
BACKGROUND Safety of the combination of leflunomide and methotrexate was examined in several studies with inconclusive results. The present study was designed to compare the efficacy and safety of the combination of leflunomide and methotrexate in adjuvant-induced arthritis (AIA) in rats focusing on immunosuppressive and hepatotoxic effects. METHODS Eighty four rats were divided into seven groups. Group 1: Sham control, group 2: the vehicle control, group 3: methotrexate group, group 4-5: leflunomide (5 and 10mg/kg/day) groups, group 6-7: combination 1 and 2 [methotrexate+leflunomide (5 and 10mg/kg/day)] groups, respectively. RESULTS The current results indicated that combination therapies improved the ankle circumference and clinical scores compared to monotherapies; histopathological examination confirmed these findings. The myelosuppressive effect of leflunomide (10mg/kg/day) was comparable to that produced by methotrexate as indicated by the complete blood count and bone marrow cellularity; however their combination resulted in greater toxicity. Furthermore, methotrexate greatly affected the splenic histopathology compared to leflunomide and the combination therapy produced a greater effect compared to leflunomide not methotrexate. Differently, assessment of the hepatotoxic potential of the two drugs highlighted that leflunomide induced a dose-dependent increase in the fibrosis score which was higher in their magnitude than that induced by methotrexate. Leflunomide (10mg/kg/day) and combination 2 groups showed the greatest degree of liver fibrosis. CONCLUSIONS In rats with AIA, current drug combinations provided higher therapeutic benefit compared to monotherapies, however, greater toxicities were observed. Therefore, continuous monitoring of hematologic parameters and liver function will be recommended in clinical settings.
Collapse
|
27
|
Vella LJ, Andrews MC, Behren A, Cebon J, Woods K. Immune consequences of kinase inhibitors in development, undergoing clinical trials and in current use in melanoma treatment. Expert Rev Clin Immunol 2014; 10:1107-23. [PMID: 24939732 DOI: 10.1586/1744666x.2014.929943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Metastatic malignant melanoma is a frequently fatal cancer. In recent years substantial therapeutic progress has occurred with the development of targeted kinase inhibitors and immunotherapeutics. Targeted therapies often result in rapid clinical benefit however responses are seldom durable. Immune therapies can result in durable disease control but responses may not be immediate. Optimal cancer therapy requires both rapid and durable cancer control and this can likely best be achieved by combining targeted therapies with immunotherapeutics. To achieve this, a detailed understanding of the immune consequences of the various kinase inhibitors, in development, clinical trial and currently used to treat melanoma is required.
Collapse
Affiliation(s)
- Laura J Vella
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immuno-biology Laboratory, Heidelberg, VIC 3084, Australia
| | | | | | | | | |
Collapse
|
28
|
Vella LJ, Pasam A, Dimopoulos N, Andrews M, Knights A, Puaux AL, Louahed J, Chen W, Woods K, Cebon JS. MEK Inhibition, Alone or in Combination with BRAF Inhibition, Affects Multiple Functions of Isolated Normal Human Lymphocytes and Dendritic Cells. Cancer Immunol Res 2014; 2:351-60. [DOI: 10.1158/2326-6066.cir-13-0181] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|