1
|
Zhang WY, Zheng XL, Coghi PS, Chen JH, Dong BJ, Fan XX. Revolutionizing adjuvant development: harnessing AI for next-generation cancer vaccines. Front Immunol 2024; 15:1438030. [PMID: 39206192 PMCID: PMC11349682 DOI: 10.3389/fimmu.2024.1438030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
With the COVID-19 pandemic, the importance of vaccines has been widely recognized and has led to increased research and development efforts. Vaccines also play a crucial role in cancer treatment by activating the immune system to target and destroy cancer cells. However, enhancing the efficacy of cancer vaccines remains a challenge. Adjuvants, which enhance the immune response to antigens and improve vaccine effectiveness, have faced limitations in recent years, resulting in few novel adjuvants being identified. The advancement of artificial intelligence (AI) technology in drug development has provided a foundation for adjuvant screening and application, leading to a diversification of adjuvants. This article reviews the significant role of tumor vaccines in basic research and clinical treatment and explores the use of AI technology to screen novel adjuvants from databases. The findings of this review offer valuable insights for the development of new adjuvants for next-generation vaccines.
Collapse
Affiliation(s)
- Wan-Ying Zhang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Xiao-Li Zheng
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Paolo Saul Coghi
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jun-Hui Chen
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bing-Jun Dong
- Gynecology Department, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, China
| | - Xing-Xing Fan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| |
Collapse
|
2
|
Kim KJ, Park JB, Lee SP, Kim HK, Kim YJ. Thalidomide and a Dipeptidyl Peptidase 4 Inhibitor in a Rat Model of Experimental Autoimmune Myocarditis. Korean Circ J 2023; 53:795-810. [PMID: 37880871 PMCID: PMC10751183 DOI: 10.4070/kcj.2023.0042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Myocarditis is a potentially fatal disease, but curative treatments have not yet been established. Myocardial inflammation is an important pathogenesis of this disease, and immunosuppressants such as methylprednisolone and immunoglobulin have been used for treatment; however, the effectiveness needs to be improved. Thalidomide and dipeptidyl peptidase (DPP) 4 inhibitors were recently investigated regarding their immunomodulatory properties. This study aimed to test whether thalidomide or a DPP4 inhibitor (evogliptin) can improve the effectiveness of myocarditis treatment using a rat model of experimental autoimmune myocarditis (EAM). METHODS Rats with or without myocarditis were administered thalidomide at 100 mg/kg/day and DPP4 inhibitor at 10 mg/kg/day orally. Measurement of echocardiography, serum inflammatory cytokines, myocardial histopathological examination, and immunohistochemical staining for leukocytes, macrophages, CD4+ T cells, and cytoskeleton were performed after 3 weeks, and the fibrosis area was measured after 3 and 6 weeks. RESULTS Thalidomide and DPP4 inhibitor did not reduce the severity of myocarditis compared with the EAM without treatment rats by comparing the echocardiographic data, myocardial CD4+, macrophages, neutrophil infiltrations, and the heart weight/body weight ratio in 3 weeks. The levels of inflammatory cytokines were not lower in the thalidomide and DPP4 inhibitor-treated group than in the untreated group in 3 weeks. In 6 weeks, thalidomide and DPP4 inhibitors did not reduce the fibrosis area compared to untreated groups. CONCLUSIONS Although thalidomide and the DPP4 inhibitor had an immunomodulatory effect and are used against inflammatory diseases, they did not ameliorate myocardial inflammation and fibrosis in this rat model of EAM.
Collapse
Affiliation(s)
- Kyung-Jin Kim
- Department of Internal Medicine, Ewha Womans University Medical Center, Ewha Womans University School of Medicine, Seoul, Korea
| | - Jun-Bean Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Pyo Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyung-Kwan Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yong-Jin Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
3
|
Lin JY, Liu HJ, Wu Y, Jin JM, Zhou YD, Zhang H, Nagle DG, Chen HZ, Zhang WD, Luan X. Targeted Protein Degradation Technology and Nanomedicine: Powerful Allies against Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207778. [PMID: 36693784 DOI: 10.1002/smll.202207778] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/08/2023] [Indexed: 05/04/2023]
Abstract
Targeted protein degradation (TPD) is an emerging therapeutic strategy with the potential of targeting undruggable pathogenic proteins. After the first proof-of-concept proteolysis-targeting chimeric (PROTAC) molecule was reported, the TPD field has entered a new era. In addition to PROTAC, numerous novel TPD strategies have emerged to expand the degradation landscape. However, their physicochemical properties and uncontrolled off-target side effects have limited their therapeutic efficacy, raising concerns regarding TPD delivery system. The combination of TPD and nanotechnology offers great promise in improving safety and therapeutic efficacy. This review provides an overview of novel TPD technologies, discusses their clinical applications, and highlights the trends and perspectives in TPD nanomedicine.
Collapse
Affiliation(s)
- Jia-Yi Lin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hai-Jun Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ye Wu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jin-Mei Jin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu-Dong Zhou
- Department of Chemistry and Biochemistry, College of Liberal Arts, University of Mississippi, University-1848, Boston, MA, 38677, USA
| | - Hong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dale G Nagle
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University-1848, Boston, MA, 38677, USA
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
4
|
Polydopamine-coated thalidomide nanocrystals promote DSS-induced murine colitis recovery through Macrophage M2 polarization together with the synergistic anti-inflammatory and anti-angiogenic effects. Int J Pharm 2022; 630:122376. [PMID: 36400133 DOI: 10.1016/j.ijpharm.2022.122376] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
High levels of proinflammatory cytokines, macrophage polarization status and immune-mediated angiogenesis play pivotal roles in the pathogenesis of inflammatory bowel disease (IBD). Thalidomide, an anti-inflammatory, immunomodulatory and antiangiogenic agent, is used off-label for treatment of IBD. The therapeutic potential of thalidomide is limited by its poor solubility and side effects associated with its systemic exposure. To address these issues and promote its therapeutic effects on IBD, thalidomide nanocrystals (Thali NCs) were prepared and coated with polydopamine (PDA), a potential macrophage polarization modulator, to form PDA coated Thali NCs (Thali@PDA). Thali@PDA possessed a high drug loading and displayed average particle size of 764.7 ± 50.30 nm. It showed a better anti-colitis effect than bare thalidomide nanocrystals at the same dose of thalidomide. Synergistic effects of polydopamine on anti-inflammatory and anti-angiogenic activities of thalidomide were observed. Furthermore, PDA coating could direct polarization of macrophages towards M2 phenotype, which boosted therapeutic effects of Thali@PDA on IBD. Upon repeated dosing of Thali@PDA for one week, symptoms of IBD in mice were significantly relieved, and histomorphology of the colitis colons were normalized. Key proinflammatory cytokine levels in the inflamed intestines were significantly decreased. Toxicity study also revealed that Thali@PDA is a safe formulation.
Collapse
|
5
|
Zhang Y, Zhang Y, Chen C, Cheng H, Deng X, Li D, Bai B, Yu Z, Deng Q, Guo J, Wen Z. Antibacterial activities and action mode of anti-hyperlipidemic lomitapide against Staphylococcus aureus. BMC Microbiol 2022; 22:114. [PMID: 35473561 PMCID: PMC9040290 DOI: 10.1186/s12866-022-02535-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/18/2022] [Indexed: 11/27/2022] Open
Abstract
Background The increasing emergence of multidrug-resistant Gram-positive bacterial infections necessitates new antibacterial agents with novel mechanisms of action that can be used to treat these infections. Lomitapide has been approved by FDA for years in reducing levels of low-density lipoprotein (LDL) in cases of familial hypercholesterolemia, whereas the antibacterial effect of lomitapide remains elusive. In this study, the inhibitory activities of lomitapide against Gram-positive bacteria were the first time explored. Quantitative proteomics analysis was then applied to investigate the mechanisms of action of lomitapide. Results The minimum inhibitory concentration (MIC) values of lomitapide against Gram-positive bacteria including both methicillin sensitive and resistant Staphylococcus aureus, Staphylococcus epidermidis, Enterococcus faecalis, Enterococcus faecium, and Streptococcus agalactiae were range 12.5–50 μM. Moreover, lomitapide also inhibited anti-biofilm activity against clinical S. aureus isolates. A total of 106 proteins with > 1.5-fold changes in expression were identified upon 1/2 × MIC lomitapide exposure, including 83 up-regulated proteins and 23 down-regulated proteins. Based on bioinformatics analysis, the expression of cell wall damage response proteins including two-component system VraS/VraR, lipoteichoic acid (LPA) D-alanylnation related proteins D-alanyl carrier protein (dltC) and carrier protein ligase (dltA), methionine sulfoxide reductases (mrsA1 and mrsB) were up-regulated. Moreover, the expression of SaeS and multiple fibrinogen-binding proteins (SAOUHSC_01110, FnBPB, SAOUHSC_02802, SdrC, SdrD) which were involved in the bacterial adhesion and biofilm formation, was inhibited by lomitapide. Furthermore, VraS/VraR deletion mutant (ΔvraSR) showed an enhanced lomitapide sensitivity phenotype. Conclusion Lomitapide displayed broad antimicrobial activities against Gram-positive bacteria. The antibacterial effect of lomitapide may be caused by cell wall destruction, while the anti-biofilm activity may be related to the inhibition of surface proteins. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-022-02535-9.
Collapse
Affiliation(s)
- Yufang Zhang
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.,Class of Biological Science, Futian District, Shenzhen College of International Education, No. 3 Antuoshan 6th Rd, Shenzhen, 518040, China
| | - Yiying Zhang
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Chengchun Chen
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Hang Cheng
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Xiangbin Deng
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Duoyun Li
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Bing Bai
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Qiwen Deng
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China. .,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.
| | - Jie Guo
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China. .,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.
| | - Zewen Wen
- Department of Infectious Diseases and Shenzhen Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and the 6th affiliated Hospital of Guangdong Medical University, Shenzhen, 518052, China. .,Quality Control Center of Hospital Infection Management of Shenzhen, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.
| |
Collapse
|
6
|
The ubiquitination-dependent and -independent functions of cereblon in cancer and neurological diseases. J Mol Biol 2022; 434:167457. [PMID: 35045330 DOI: 10.1016/j.jmb.2022.167457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/21/2021] [Accepted: 01/10/2022] [Indexed: 12/24/2022]
Abstract
Cereblon (CRBN) mediates the teratogenic effect of thalidomide in zebrafish, chicken, and humans. It additionally modulates the anti-myeloma effect of the immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide. IMiDs bind to CRBN and recruit neo-substrates for their ubiquitination and proteasome-mediated degradation, which significantly expands the application of proteolysis-targeting chimeras (PROTACs) for targeted drug discovery. However, the underlying molecular mechanisms by which CRBN mediates the teratogenicity and anti-myeloma effect of IMiDs are not fully elucidated. Furthermore, the normal physiological functions of endogenous CRBN have not been extensively studied, which precludes the thorough assessment of side effects of the CRBN ligand-based PROTACs in the treatment of cancer and neurological diseases. To advance our understanding of the diverse functions of CRBN, in this review, we will survey the ubiquitination-dependent and -independent functions of CRBN, summarize recent advances in the discovery of constitutive and neo-substrates of CRBN, and explore the molecular functions of CRBN in cancer treatment and in the development of neurological diseases. We will also discuss the potential future directions towards the identification of CRBN substrates and interacting proteins, and CRBN-ligand-based drug discovery in the treatment of cancer and neurological diseases.
Collapse
|
7
|
Zhang HX, Yuan J, Li RS. Thalidomide Mitigates Apoptosis via Endoplasmic Reticulum Stress in Diabetic Nephropathy. Endocr Metab Immune Disord Drug Targets 2021; 22:787-794. [PMID: 34967303 DOI: 10.2174/1871530322666211230115743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/14/2021] [Accepted: 11/24/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Previous studies have shown that endoplasmic reticulum (ER) stress is related to the apoptosis in the development of diabetic nephropathy (DN) and thalidomide (Thd) has renal-protective effects by suppressing inflammation and proliferation of MCs in DN. However, the effect of Thd on the apoptosis of MCs in DN remains largely unclear. The present research is designed to explore the effect of Thd on apoptosis in DN and the related mechanisms. OBJECTIVE The study is designed to examine the effect and mechanism of Thd on apoptosis in type 2 diabetic mice and high glucose (HG)-induced MCs. METHOD We first evaluated the ER stress markers and apoptosis-related proteins with the treatment of Thd in type 2 diabetic mice and MCs in vitro under HG conditions. MTT assay was used to assess cell viability. Additionally, we evaluated the effect of Thd treatment upon MC apoptosis through flow cytometry. Real-time polymerase chain reaction (RT-PCR) and Western blot were performed to evaluate genes and protein expression related to ER stress and apoptosis. RESULTS The levels of blood urea BUN, CREA, Urine albumin, and UACR in diabetic mice were significantly reduced after 8 weeks of intervention with Thd. And also, there were upregulated glucose-regulated protein 78 (GRP78), Caspase-12, and downregulated B-cell lymphoma 2 (Bcl-2) in glomeruli of DN mice. In vitro, compared with the HG group, MC apoptosis reduced dramatically with Thd treatment along with upregulation of Bcl-2 and downregulation of Bax. At the same time, ER stress markers GRP78, C/EBP homologous protein (CHOP), and Caspase-12 were also mitigated following the Thd treatment. CONCLUSION The present study indicates that Thd might reduce the ER stress in DN via downregulating of GRP78, CHOP, and Caspase12 expression, ultimately mitigating MCs apoptosis.
Collapse
Affiliation(s)
- Hong-Xia Zhang
- Department of Endocrinology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Jie Yuan
- Department of Radiology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Rong-Shan Li
- Department of Nephrology, Shanxi Provincial People's Hospital, Taiyuan, China
| |
Collapse
|
8
|
Pathogenesis and treatment of multiple myeloma bone disease. JAPANESE DENTAL SCIENCE REVIEW 2021; 57:164-173. [PMID: 34611468 PMCID: PMC8477206 DOI: 10.1016/j.jdsr.2021.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 02/02/2023] Open
Abstract
Multiple myeloma (Plasma cell myeloma), a malignancy of the plasma cells, exhibits tumor expansion preferentially in the bone marrow and the development of bone-destructive lesions. Multiple myeloma is still an incurable disease with changes in the bone marrow microenvironment in favor of the survival and proliferation of multiple myeloma cells and bone destruction. In this review, we described the recent findings on the regulators involved in the development of myeloma bone diseases, and succinctly summarize currently available therapeutic options and the development of novel bone modifying agents for myeloma treatment.
Collapse
|
9
|
Su X, You Z, Wang L, Hu L, Wong L, Ji B, Zhao B. SANE: A sequence combined attentive network embedding model for COVID-19 drug repositioning. Appl Soft Comput 2021; 111:107831. [PMID: 34456656 PMCID: PMC8381638 DOI: 10.1016/j.asoc.2021.107831] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/25/2021] [Accepted: 08/14/2021] [Indexed: 01/03/2023]
Abstract
The COVID-19 has now spread all over the world and causes a huge burden for public health and world economy. Drug repositioning has become a promising treatment strategy in COVID-19 crisis because it can shorten drug development process, reduce pharmaceutical costs and reposition approval drugs. Existing computational methods only focus on single information, such as drug and virus similarity or drug-virus network feature, which is not sufficient to predict potential drugs. In this paper, a sequence combined attentive network embedding model SANE is proposed for identifying drugs based on sequence features and network features. On the one hand, drug SMILES and virus sequence features are extracted by encoder-decoder in SANE as node initial embedding in drug-virus network. On the other hand, SANE obtains fields for each node by attention-based Depth-First-Search (DFS) to reduce noises and improve efficiency in representation learning and adopts a bottom-up aggregation strategy to learn node network representation from selected fields. Finally, a forward neural network is used for classifying. Experiment results show that SANE has achieved the performance with 81.98% accuracy and 0.8961 AUC value and outperformed state-of-the-art baselines. Further case study on COVID-19 indicates that SANE has a strong predictive ability since 25 of the top 40 (62.5%) drugs are verified by valuable dataset and literatures. Therefore, SANE is powerful to reposition drugs for COVID-19 and provides a new perspective for drug repositioning.
Collapse
Affiliation(s)
- Xiaorui Su
- Xinjiang Technical Institutes of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi 830011, China
| | - Zhuhong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710129, China
| | - Lei Wang
- Big Data and Intelligent Computing Research Center, Guangxi Academy of Science, Nanning, 530007, China
| | - Lun Hu
- Xinjiang Technical Institutes of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi 830011, China
| | - Leon Wong
- Big Data and Intelligent Computing Research Center, Guangxi Academy of Science, Nanning, 530007, China
| | - Boya Ji
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Bowei Zhao
- Xinjiang Technical Institutes of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi 830011, China
| |
Collapse
|
10
|
Campesi I, Racagni G, Franconi F. Just a Reflection: Does Drug Repurposing Perpetuate Sex-Gender Bias in the Safety Profile? Pharmaceuticals (Basel) 2021; 14:730. [PMID: 34451827 PMCID: PMC8402096 DOI: 10.3390/ph14080730] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 02/08/2023] Open
Abstract
Vaccines constitute a strategy to reduce the burden of COVID-19, but the treatment of COVID-19 is still a challenge. The lack of approved drugs for severe COVID-19 makes repurposing or repositioning of approved drugs a relevant approach because it occurs at lower costs and in a shorter time. Most preclinical and clinical tests, including safety and pharmacokinetic profiles, were already performed. However, infective and inflammatory diseases such as COVID-19 are linked with hypoalbuminemia and downregulation of both phase I and phase II drug-metabolizing enzymes and transporters, which can occur in modifications of pharmacokinetics and consequentially of safety profiles. This appears to occur in a sex- and gender-specific way because of the sex and gender differences present in the immune system and inflammation, which, in turn, reflect on pharmacokinetic parameters. Therefore, to make better decisions about drug dosage regimens and to increases the safety profile in patients suffering from infective and inflammatory diseases such as COVID-19, it is urgently needed to study repurposing or repositioning drugs in men and in women paying attention to pharmacokinetics, especially for those drugs that are previously scarcely evaluated in women.
Collapse
Affiliation(s)
- Ilaria Campesi
- Department of Biomedical Science, University of Sassari, 07100 Sassari, Italy
- National Laboratory of Pharmacology and Gender Medicine, National Institute of Biostructure and Biosystem, 07100 Sassari, Italy;
| | - Giorgio Racagni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy;
| | - Flavia Franconi
- National Laboratory of Pharmacology and Gender Medicine, National Institute of Biostructure and Biosystem, 07100 Sassari, Italy;
| |
Collapse
|
11
|
Barghout SH. Targeted Protein Degradation: An Emerging Therapeutic Strategy in Cancer. Anticancer Agents Med Chem 2021; 21:214-230. [PMID: 32275492 DOI: 10.2174/1871520620666200410082652] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/20/2020] [Accepted: 02/19/2020] [Indexed: 11/22/2022]
Abstract
Drug discovery in the scope of cancer therapy has been focused on conventional agents that nonselectively induce DNA damage or selectively inhibit the activity of key oncogenic molecules without affecting their protein levels. An emerging therapeutic strategy that garnered attention in recent years is the induction of Targeted Protein Degradation (TPD) of cellular targets by hijacking the intracellular proteolysis machinery. This novel approach offers several advantages over conventional inhibitors and introduces a paradigm shift in several pharmacological aspects of drug therapy. While TPD has been found to be the major mode of action of clinically approved anticancer agents such as fulvestrant and thalidomide, recent years have witnessed systematic endeavors to expand the repertoire of proteins amenable to therapeutic ablation by TPD. Such endeavors have led to three major classes of agents that induce protein degradation, including molecular glues, Proteolysis Targeting Chimeras (PROTACs) and Hydrophobic Tag (HyT)-based degraders. Here, we briefly highlight agents in these classes and key advances made in the field with a focus on clinical translation in cancer therapy.
Collapse
Affiliation(s)
- Samir H Barghout
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
12
|
Jourdan JP, Bureau R, Rochais C, Dallemagne P. Drug repositioning: a brief overview. J Pharm Pharmacol 2020; 72:1145-1151. [PMID: 32301512 PMCID: PMC7262062 DOI: 10.1111/jphp.13273] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/17/2020] [Accepted: 03/21/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Drug repositioning, that is, the use of a drug in an indication other than the one for which it was initially marketed, is a growing trend. Its origins lie mainly in the attrition experienced in recent years in the field of new drug discovery. KEY FINDINGS Despite some regulatory and economic challenges, drug repositioning offers many advantages, and a number of recent successes have confirmed both its public health benefits and its commercial value. The first examples of successful drug repositioning mainly came about through serendipity like acetylsalicylic acid, thalidomide, sildenafil or dimethylfumarate. CONCLUSION The history of great-repositioned drugs has given some solutions to various pathologies. Serendipity is not yet useful to find repositioning drugs. Drug repositioning is of growing interest. Nowadays, a more rational approach to the identification of drug candidates for repositioning is possible, especially using data mining.
Collapse
Affiliation(s)
- Jean-Pierre Jourdan
- UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ., Caen, France.,Pharmacy Department, CHRU de Caen, Caen, France
| | - Ronan Bureau
- UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ., Caen, France
| | - Christophe Rochais
- UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ., Caen, France
| | - Patrick Dallemagne
- UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ., Caen, France
| |
Collapse
|
13
|
Repurposing old drugs as new inhibitors of the ubiquitin-proteasome pathway for cancer treatment. Semin Cancer Biol 2019; 68:105-122. [PMID: 31883910 DOI: 10.1016/j.semcancer.2019.12.013] [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: 07/14/2019] [Revised: 10/30/2019] [Accepted: 12/15/2019] [Indexed: 12/25/2022]
Abstract
The ubiquitin-proteasome system (UPS) plays a central role in the degradation of cellular proteins. Targeting protein degradation has been validated as an effective strategy for cancer therapy since 2003. Several components of the UPS have been validated as potential anticancer targets, including 20S proteasomes, 19S proteasome-associated deubiquitinases (DUBs) and ubiquitin ligases (E3s). 20S proteasome inhibitors (such as bortezomib/BTZ and carfilzomib/CFZ) have been approved by the U.S. Food and Drug Administration (FDA) for the treatment of multiple myeloma (MM) and some other liquid tumors. Although survival of MM patients has been improved by the introduction of BTZ-based therapies, these clinical 20S proteasome inhibitors have several limitations, including emergence of resistance in MM patients, neuro-toxicities, and little efficacy in solid tumors. One of strategies to improve the current status of cancer treatment is to repurpose old drugs with UPS-inhibitory properties as new anticancer agents. Old drug reposition represents an attractive drug discovery approach compared to the traditional de novo drug discovery process which is time-consuming and costly. In this review, we summarize status of repurposed inhibitors of various UPS components, including 20S proteasomes, 19S-associated DUBs, and ubiquitin ligase E3s. The original and new mechanisms of action, molecular targets, and potential anticancer activities of these repurposed UPS inhibitors are reviewed, and their new uses including combinational therapies for cancer treatment are discussed.
Collapse
|
14
|
El Ati Z, Lamia R, Cherif J, Jbali H, Fatma LB, Mami I, Khedher R, Smaoui W, Krid M, Hamida FB, Beji S, Zouaghi MK. Thalidomide-induced bronchiolitis obliterans organizing pneumonia in a patient with multiple myeloma. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2019; 30:974-977. [PMID: 31464258 DOI: 10.4103/1319-2442.265477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Thalidomide, which is an angiogenesis inhibitor and immunomodulator that reduces tumor necrosis factor-alpha, has regained value in the treatment of multiple myeloma. Serious pulmonary complications due to thalidomide use remain relatively uncommon. We describe a case of bronchiolitis obliterans organizing pneumonia (BOOP) due to thalidomide. A 51-year-old man with IgG lambda myeloma was treated with thalidomide and dexamethasone. Seven days after the beginning of chemotherapy, the patient presented a fever and a persistent cough. Auscultation revealed crackles in both pulmonary bases. The chest X-ray showed a diffuse bilateral alveolar-interstitial syndrome. Computed tomography scan revealed bilateral pulmonary involvement, with bilateral interstitial alveolar infiltration and ground-glass pattern consolidations. Pulmonary infection, malignant tumor, and lung involvement of multiple myeloma were excluded through various tests. Thalidomide-induced BOOP was suspected, and the drug was withdrawn and replaced by Melphalan. The patient had complete resolution of his symptoms and radiologic pulmonary involvement on discontinuation of the drug. In the absence of other etiologies, physicians should be cognizant of this potential complication in patients receiving thalidomide who present with respiratory symptoms.
Collapse
Affiliation(s)
- Zohra El Ati
- Department of Hemodialysis, Tahar Sfar Hospital, Mahdia, Faculty of Medicine, Monastir University, Monastir, Tunisia
| | - Rais Lamia
- Department of Nephrology, La Rabta Hospital, Tunis, Tunisia
| | - Jouda Cherif
- Department of Pneumology, La Rabta Hospital, Tunis, Tunisia
| | - Hela Jbali
- Department of Nephrology, La Rabta Hospital, Tunis, Tunisia
| | | | - Ikram Mami
- Department of Nephrology, La Rabta Hospital, Tunis, Tunisia
| | - Rania Khedher
- Department of Nephrology, La Rabta Hospital, Tunis, Tunisia
| | - Wided Smaoui
- Department of Nephrology, La Rabta Hospital, Tunis, Tunisia
| | - Madiha Krid
- Department of Nephrology, La Rabta Hospital, Tunis, Tunisia
| | - Fethi Ben Hamida
- Department of Research Laboratory of Kidney Diseases (LR00SP01), Charles Nicolle Hospital, Faculty of Medicine, Tunis El Manar University, Tunis, Tunisia
| | - Soumaya Beji
- Department of Nephrology, La Rabta Hospital, Tunis, Tunisia
| | | |
Collapse
|
15
|
Li L, Wang L. Multiple Myeloma: What Do We Do About Immunodeficiency? J Cancer 2019; 10:1675-1684. [PMID: 31205523 PMCID: PMC6548011 DOI: 10.7150/jca.29993] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/30/2019] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematological malignancy. Immunodeficiency results in the incapability of immunity to eradicate both tumor cells and pathogens. Immunotherapies along with antibiotics and other anti-infectious agents are applied as substitutes for immunity in MM. Immunotherapies including monoclonal antibodies, immune checkpoints inhibitors, affinity- enhanced T cells, chimeric antigen receptor T cells and dendritic cell vaccines are revolutionizing MM treatment. By suppressing the pro-inflammatory milieu and pathogens, prophylactic and therapeutic antibiotics represent anti-tumor and anti-infection properties. It is expected that deeper understanding of infection, immunity and tumor physio-pathologies in MM will accelerate the optimization of combined therapies, thus improving prognosis in MM.
Collapse
Affiliation(s)
- Linrong Li
- Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Liang Wang
- Department of Hematology, ZhuJiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
|
17
|
Zhang HX, Yuan J, Li YF, Li RS. Thalidomide decreases high glucose-induced extracellular matrix protein synthesis in mesangial cells via the AMPK pathway. Exp Ther Med 2019; 17:927-934. [PMID: 30651882 DOI: 10.3892/etm.2018.6995] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/26/2018] [Indexed: 01/03/2023] Open
Abstract
A previous study demonstrated the renal-protective effect of thalidomide (Thd) in diabetic nephropathy rats through the activation of the adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the nuclear factor κB (NF-κB)/monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-β1/mothers against decapentaplegic homolog signaling pathways. The association between AMPK inactivation and high glucose (HG)-induced meningeal cell (MC) proliferation and extracellular matrix (ECM) accumulation via NF-κB and TGF-β1 signaling remains unknown. The aim of the current study was to demonstrate the effects of Thd on cell proliferation and ECM expression in HG-cultured MCs and the underlying mechanisms. HG-cultured human MCs were treated with Thd. Cell proliferation was measured by MTT assay and quantification of cell proliferation was based on the measurement of bromodeoxyuridine incorporation. The differences in TGF-β1, fibronectin and MCP-1 protein expression levels were detected via ELISA and western blot analysis. The AMPK signaling pathway was also examined by western blot analysis in MCs. Compound C, an AMPK inhibitor and AICAR (5-aminoimidazole-4-carboxamide 1β-D-ribofuranoside), an AMPK agonist, were used to analyze the functional role of AMPK in MCs. Cell proliferation was significantly decreased in HG-cultured MCs following treatment with high concentrations of Thd (100 and 200 µg/ml) for 24 h compared with the HG-cultured MC group. Thd suppressed the inflammatory processes in HG-induced MCs. These effects were partially mediated through the activation of AMPK and inhibition of the NF-κB/MCP-1 signaling pathways. Taken together, these results suggest that Thd may have therapeutic potential in diabetic renal injury via the AMPK signaling pathway.
Collapse
Affiliation(s)
- Hong-Xia Zhang
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Jie Yuan
- Department of Radiology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Ya-Feng Li
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Rong-Shan Li
- Department of Nephrology, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| |
Collapse
|
18
|
Hijji Y, Benjamin E, Jasinski JP, Butcher RJ. Crystal structure of the thalidomide analog (3a R*,7a S*)-2-(2,6-dioxopiperidin-3-yl)hexa-hydro-1 H-iso-indole-1,3(2 H)-dione. Acta Crystallogr E Crystallogr Commun 2018; 74:1595-1598. [PMID: 30443388 PMCID: PMC6218906 DOI: 10.1107/s2056989018014317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/10/2018] [Indexed: 11/23/2022]
Abstract
The title compound, C13H16N2O4, crystallizes in the monoclinic centrosymmetric space group, P21/c, with four mol-ecules in the asymmetric unit, thus there is no crystallographically imposed symmetry and it is a racemic mixture. The structure consists of a six-membered unsaturated ring bound to a five-membered pyrrolidine-2,5-dione ring N-bound to a six-membered piperidine-2,6-dione ring and thus has the same basic skeleton as thalidomide, except for the six-membered unsaturated ring substituted for the aromatic ring. In the crystal, the mol-ecules are linked into inversion dimers by R 2 2(8) hydrogen bonding involving the N-H group. In addition, there are bifurcated C-H⋯O inter-actions involving one of the O atoms on the pyrrolidine-2,5-dione with graph-set notation R 1 2(5). These inter-actions along with C-H⋯O inter-actions involving one of the O atoms on the piperidine-2,6-dione ring link the mol-ecules into a complex three-dimensional array. There is pseudomerohedral twinning present which results from a 180° rotation about the [100] reciprocal lattice direction and with a twin law of 1 0 0 0 0 0 0 [BASF 0.044 (1)].
Collapse
Affiliation(s)
- Yousef Hijji
- Department of Chemistry and Earth Sciences, Qatar University, Doha, Qatar
| | - Ellis Benjamin
- Department of Chemistry, Richard Stockton College of New Jersey, Galloway, NJ 08205, USA
| | - Jerry P Jasinski
- Department of Chemistry, Keene State College, 229 Main Street, Keene NH 03435, USA
| | - Ray J Butcher
- Department of Chemistry, Howard University, 525 College Street NW, Washington, DC 20059, USA
| |
Collapse
|
19
|
Collins I, Wang H, Caldwell JJ, Chopra R. Chemical approaches to targeted protein degradation through modulation of the ubiquitin-proteasome pathway. Biochem J 2017; 474:1127-1147. [PMID: 28298557 PMCID: PMC5350610 DOI: 10.1042/bcj20160762] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/04/2017] [Accepted: 01/16/2017] [Indexed: 12/11/2022]
Abstract
Manipulation of the ubiquitin-proteasome system to achieve targeted degradation of proteins within cells using chemical tools and drugs has the potential to transform pharmacological and therapeutic approaches in cancer and other diseases. An increased understanding of the molecular mechanism of thalidomide and its analogues following their clinical use has unlocked small-molecule modulation of the substrate specificity of the E3 ligase cereblon (CRBN), which in turn has resulted in the advancement of new immunomodulatory drugs (IMiDs) into the clinic. The degradation of multiple context-specific proteins by these pleiotropic small molecules provides a means to uncover new cell biology and to generate future drug molecules against currently undruggable targets. In parallel, the development of larger bifunctional molecules that bring together highly specific protein targets in complexes with CRBN, von Hippel-Lindau, or other E3 ligases to promote ubiquitin-dependent degradation has progressed to generate selective chemical compounds with potent effects in cells and in vivo models, providing valuable tools for biological target validation and with future potential for therapeutic use. In this review, we survey recent breakthroughs achieved in these two complementary methods and the discovery of new modes of direct and indirect engagement of target proteins with the proteasome. We discuss the experimental characterisation that validates the use of molecules that promote protein degradation as chemical tools, the preclinical and clinical examples disclosed to date, and the future prospects for this exciting area of chemical biology.
Collapse
Affiliation(s)
- Ian Collins
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, U.K
| | - Hannah Wang
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, U.K
| | - John J Caldwell
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, U.K
| | - Raj Chopra
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, U.K.
| |
Collapse
|
20
|
Computational Discovery of Putative Leads for Drug Repositioning through Drug-Target Interaction Prediction. PLoS Comput Biol 2016; 12:e1005219. [PMID: 27893735 PMCID: PMC5125559 DOI: 10.1371/journal.pcbi.1005219] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/21/2016] [Indexed: 12/23/2022] Open
Abstract
De novo experimental drug discovery is an expensive and time-consuming task. It requires the identification of drug-target interactions (DTIs) towards targets of biological interest, either to inhibit or enhance a specific molecular function. Dedicated computational models for protein simulation and DTI prediction are crucial for speed and to reduce the costs associated with DTI identification. In this paper we present a computational pipeline that enables the discovery of putative leads for drug repositioning that can be applied to any microbial proteome, as long as the interactome of interest is at least partially known. Network metrics calculated for the interactome of the bacterial organism of interest were used to identify putative drug-targets. Then, a random forest classification model for DTI prediction was constructed using known DTI data from publicly available databases, resulting in an area under the ROC curve of 0.91 for classification of out-of-sampling data. A drug-target network was created by combining 3,081 unique ligands and the expected ten best drug targets. This network was used to predict new DTIs and to calculate the probability of the positive class, allowing the scoring of the predicted instances. Molecular docking experiments were performed on the best scoring DTI pairs and the results were compared with those of the same ligands with their original targets. The results obtained suggest that the proposed pipeline can be used in the identification of new leads for drug repositioning. The proposed classification model is available at http://bioinformatics.ua.pt/software/dtipred/. The emergence of multi-resistant bacterial strains and the existing void in the discovery and development of new classes of antibiotics is a growing concern. Indeed, some bacterial strains are now resistant to last-line antibiotics and considered untreatable. Drug repositioning has been suggested as a strategy to minimize time and cost expenses until the drug reaches the market, compared to traditional drug design. Drug-target interactions (DTIs) are the basis of rational drug design and thus, we proposed a computational approach to predict DTIs solely based on the primary sequence of the protein and the simplified molecular-input line-entry system of the ligand. In addition, network metrics are used to identify vital putative drug-targets in bacteria. Molecular docking experiments were performed to compare the binding affinities between a given ligand and a putative drug-target, as well as with their original targets. According to the docking results, the predicted DTIs have better or similar binding activities than the ligand and their real target, indicating the validity of the proposed model.
Collapse
|
21
|
Abstract
This article discusses medications as socially embedded phenomena, using the class of psychoactive medications as a primary example. The analytical perspective is systemic, constructivist, and critical. We suggest that the ‘rational use of drugs’ paradigm fails to appreciate various legitimate rationalities motivating medication usages and is therefore inadequate to understand the place of medications in society. Medications have complex life cycles, with diverse actors, social systems, and institutions determining who uses what medications, how, when and why. Such understanding permits analyzing medications simultaneously as entities and representations. We outline recent changes in usage patterns of psychoactive medications (notably prescriptions to children), in pharmaceutical marketing practices (notably direct-to-consumer advertising), and in the construction of knowledge about drugs (notably the role of the Internet in legitimating consumers’ viewpoints). These changes indicate that medication life cycles evolve and mutate with social and technological change. These life cycles are viewed, then, as systems – part of other social, cultural, and economic systems, themselves in constant change. This perspective provides fertile ground to raise several research questions in order to understand better the nature of medications, their effects, and their place in society.
Collapse
|
22
|
Bakacak M, Ercan Ö, Köstü B, Bostancı MS, İnanç F, Yaylalı A, Serin S, Balakan O, Kıran G. The effects of thalidomide in a rat model of surgically-induced endometriosis. Turk J Obstet Gynecol 2015; 12:125-131. [PMID: 28913056 PMCID: PMC5558385 DOI: 10.4274/tjod.71601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/02/2015] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE The aim of the study was to analyze the anti-angiogenic role of thalidomide and to assess whether thalidomide had any influence on a rat model of surgically-induced endometriosis. MATERIALS AND METHODS Endometriosis was induced through surgical induction and homologous transplantation in 16 rats. The rats were randomly separated into two groups as thalidomide (n=8) and control (n=8) groups. Using oral gavage, 100 mg/kg thalidomide 0.5 ml was administered to the first group and saline 0.5 ml to the control group. Histopathologic findings and volume analysis of implants were evaluated after 4 weeks. Vascular endothelial growth factor-A (VEGF-A) and oxidative markers were run from the fluid through peritoneal lavage. RESULTS The average implant volume decreased significantly in the thalidomide administrated group after treatment (53.3 and 22.9 mm3 respectively, p=0.012). Significant differences observed in the histopathologic scores of the thalidomide group (3 and 1 respectively, p=0.012) were not observed in the control group. Significant decreases were observed in the levels of VEGF-A and myeloperoxidase (MPO) from oxidative markers (p=0.004, p=0.037, respectively). CONCLUSION Thalidomide provides volumetric and histopathologic recovery in implants particularly because the VEGF inhibition and anti-angiogenic effect, which suggests that it could be effective in the treatment of endometriosis.
Collapse
Affiliation(s)
- Murat Bakacak
- Sütçü İmam University Faculty of Medicine, Department of Obstetrics and Gynecology, Kahramanmaraş, Turkey
| | - Önder Ercan
- Sütçü İmam University Faculty of Medicine, Department of Obstetrics and Gynecology, Kahramanmaraş, Turkey
| | - Bülent Köstü
- Sütçü İmam University Faculty of Medicine, Department of Obstetrics and Gynecology, Kahramanmaraş, Turkey
| | - Mehmet Sühha Bostancı
- Sakarya University Faculty of Medicine, Department of Obstetrics and Gynecology, Sakarya, Turkey
| | - Fatma İnanç
- Sütçü İmam University Faculty of Medicine, Department of Biochemistry, Kahramanmaraş, Turkey
| | - Aslı Yaylalı
- Sütçü İmam University Faculty of Medicine, Department of Histology and Embryology, Kahramanmaraş, Turkey
| | - Salih Serin
- Sütçü İmam University Faculty of Medicine, Department of Obstetrics and Gynecology, Kahramanmaraş, Turkey
| | - Ozan Balakan
- Sütçü İmam University Faculty of Medicine, Department of Internal Medicine, Medical Oncology, Kahramanmaraş, Turkey
| | - Gürkan Kıran
- Sütçü İmam University Faculty of Medicine, Department of Obstetrics and Gynecology, Kahramanmaraş, Turkey
| |
Collapse
|
23
|
Ponomaryov Y, Krasikova V, Lebedev A, Chernyak D, Varacheva L, Chernobroviy A. Scalable and green process for the synthesis of anticancer drug lenalidomide. Chem Heterocycl Compd (N Y) 2015. [DOI: 10.1007/s10593-015-1670-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
24
|
Gureje O, Nortje G, Makanjuola V, Oladeji B, Seedat S, Jenkins R. The role of global traditional and complementary systems of medicine in treating mental health problems. Lancet Psychiatry 2015; 2:168-177. [PMID: 26052502 PMCID: PMC4456435 DOI: 10.1016/s2215-0366(15)00013-9] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/26/2014] [Indexed: 11/26/2022]
Abstract
Traditional and complementary systems of medicine (TCM) encompass a broad range of practices which are commonly embedded within contextual cultural milieu, reflecting community beliefs, experiences, religion and spirituality. Evidence from across the world, especially from low- and middle-income countries (LMIC), suggests that TCM is commonly used by a large number of persons with mental illness. Even though some overlap exists between the diagnostic approaches of TCM and conventional biomedicine (CB), there are major differences, largely reflecting differences in the understanding of the nature and etiology of mental disorders. However, treatment modalities employed by providers of TCM may sometimes fail to meet common understandings of human rights and humane care. Still, there are possibilities for collaboration between TCM and CB in the care of persons with mental illness. Research is required to clearly delineate the boundaries of such collaboration and to test its effectiveness in bringing about improved patient outcomes.
Collapse
Affiliation(s)
- Oye Gureje
- Department of Psychiatry, University of Ibadan, Ibadan, Nigeria
| | - Gareth Nortje
- Department of Psychiatry, Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | | | - Soraya Seedat
- Department of Psychiatry, Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Rachel Jenkins
- Health Service and Population Research Department, Institute of Psychiatry, King's College, London
| |
Collapse
|
25
|
Galluzzi L, Vacchelli E, Pedro JMBS, Buqué A, Senovilla L, Baracco EE, Bloy N, Castoldi F, Abastado JP, Agostinis P, Apte RN, Aranda F, Ayyoub M, Beckhove P, Blay JY, Bracci L, Caignard A, Castelli C, Cavallo F, Celis E, Cerundolo V, Clayton A, Colombo MP, Coussens L, Dhodapkar MV, Eggermont AM, Fearon DT, Fridman WH, Fučíková J, Gabrilovich DI, Galon J, Garg A, Ghiringhelli F, Giaccone G, Gilboa E, Gnjatic S, Hoos A, Hosmalin A, Jäger D, Kalinski P, Kärre K, Kepp O, Kiessling R, Kirkwood JM, Klein E, Knuth A, Lewis CE, Liblau R, Lotze MT, Lugli E, Mach JP, Mattei F, Mavilio D, Melero I, Melief CJ, Mittendorf EA, Moretta L, Odunsi A, Okada H, Palucka AK, Peter ME, Pienta KJ, Porgador A, Prendergast GC, Rabinovich GA, Restifo NP, Rizvi N, Sautès-Fridman C, Schreiber H, Seliger B, Shiku H, Silva-Santos B, Smyth MJ, Speiser DE, Spisek R, Srivastava PK, Talmadge JE, Tartour E, Van Der Burg SH, Van Den Eynde BJ, Vile R, Wagner H, Weber JS, Whiteside TL, Wolchok JD, Zitvogel L, Zou W, Kroemer G. Classification of current anticancer immunotherapies. Oncotarget 2014; 5:12472-508. [PMID: 25537519 PMCID: PMC4350348 DOI: 10.18632/oncotarget.2998] [Citation(s) in RCA: 319] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 12/15/2014] [Indexed: 11/25/2022] Open
Abstract
During the past decades, anticancer immunotherapy has evolved from a promising therapeutic option to a robust clinical reality. Many immunotherapeutic regimens are now approved by the US Food and Drug Administration and the European Medicines Agency for use in cancer patients, and many others are being investigated as standalone therapeutic interventions or combined with conventional treatments in clinical studies. Immunotherapies may be subdivided into "passive" and "active" based on their ability to engage the host immune system against cancer. Since the anticancer activity of most passive immunotherapeutics (including tumor-targeting monoclonal antibodies) also relies on the host immune system, this classification does not properly reflect the complexity of the drug-host-tumor interaction. Alternatively, anticancer immunotherapeutics can be classified according to their antigen specificity. While some immunotherapies specifically target one (or a few) defined tumor-associated antigen(s), others operate in a relatively non-specific manner and boost natural or therapy-elicited anticancer immune responses of unknown and often broad specificity. Here, we propose a critical, integrated classification of anticancer immunotherapies and discuss the clinical relevance of these approaches.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Erika Vacchelli
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - José-Manuel Bravo-San Pedro
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Aitziber Buqué
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Laura Senovilla
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Elisa Elena Baracco
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Norma Bloy
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Francesca Castoldi
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Medicine, Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
- Sotio a.c., Prague, Czech Republic
| | - Jean-Pierre Abastado
- Pole d'innovation thérapeutique en oncologie, Institut de Recherches Internationales Servier, Suresnes, France
| | - Patrizia Agostinis
- Cell Death Research and Therapy (CDRT) Laboratory, Dept. of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Ron N. Apte
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Fernando Aranda
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maha Ayyoub
- INSERM, U1102, Saint Herblain, France
- Institut de Cancérologie de l'Ouest, Saint Herblain, France
| | - Philipp Beckhove
- Translational Immunology Division, German Cancer Research Center, Heidelberg, Germany
| | - Jean-Yves Blay
- Equipe 11, Centre Léon Bérard (CLR), Lyon, France
- Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
| | - Laura Bracci
- Dept. of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anne Caignard
- INSERM, U1160, Paris, France
- Groupe Hospitalier Saint Louis-Lariboisière - F. Vidal, Paris, France
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumors, Dept. of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center, Dept. of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Estaban Celis
- Cancer Immunology, Inflammation and Tolerance Program, Georgia Regents University Cancer Center, Augusta, GA, USA
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Aled Clayton
- Institute of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, UK
- Velindre Cancer Centre, Cardiff, UK
| | - Mario P. Colombo
- Unit of Immunotherapy of Human Tumors, Dept. of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | - Lisa Coussens
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Madhav V. Dhodapkar
- Sect. of Hematology and Immunobiology, Yale Cancer Center, Yale University, New Haven, CT, USA
| | | | | | - Wolf H. Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | - Jitka Fučíková
- Sotio a.c., Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Dmitry I. Gabrilovich
- Dept. of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jérôme Galon
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers, Paris, France
| | - Abhishek Garg
- Cell Death Research and Therapy (CDRT) Laboratory, Dept. of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - François Ghiringhelli
- INSERM, UMR866, Dijon, France
- Centre Georges François Leclerc, Dijon, France
- Université de Bourgogne, Dijon, France
| | - Giuseppe Giaccone
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Eli Gilboa
- Dept. of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Sacha Gnjatic
- Sect. of Hematology/Oncology, Immunology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Axel Hoos
- Glaxo Smith Kline, Cancer Immunotherapy Consortium, Collegeville, PA, USA
| | - Anne Hosmalin
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- INSERM, U1016, Paris, France
- CNRS, UMR8104, Paris, France
- Hôpital Cochin, AP-HP, Paris, France
| | - Dirk Jäger
- National Center for Tumor Diseases, University Medical Center Heidelberg, Heidelberg, Germany
| | - Pawel Kalinski
- Dept. of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
- Dept. of Immunology and Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Klas Kärre
- Dept. of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Oliver Kepp
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Rolf Kiessling
- Dept. of Oncology, Karolinska Institute Hospital, Stockholm, Sweden
| | - John M. Kirkwood
- University of Pittsburgh Cancer Institute Laboratory, Pittsburgh, PA, USA
| | - Eva Klein
- Dept. of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Alexander Knuth
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Claire E. Lewis
- Academic Unit of Inflammation and Tumour Targeting, Dept. of Oncology, University of Sheffield Medical School, Sheffield, UK
| | - Roland Liblau
- INSERM, UMR1043, Toulouse, France
- CNRS, UMR5282, Toulouse, France
- Laboratoire d'Immunologie, CHU Toulouse, Université Toulouse II, Toulouse, France
| | - Michael T. Lotze
- Dept. of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Jean-Pierre Mach
- Dept. of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Fabrizio Mattei
- Dept. of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Institute, Rozzano, Italy
- Dept. of Medical Biotechnologies and Translational Medicine, University of Milan, Rozzano, Italy
| | - Ignacio Melero
- Dept. of Immunology, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- Dept. of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Cornelis J. Melief
- ISA Therapeutics, Leiden, The Netherlands
- Dept. of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Elizabeth A. Mittendorf
- Research Dept. of Surgical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Adekunke Odunsi
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Hideho Okada
- Dept. of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Marcus E. Peter
- Div. of Hematology/Oncology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Kenneth J. Pienta
- The James Buchanan Brady Urological Institute, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Angel Porgador
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
- Dept. of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Philadelphia, PA, USA
- Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gabriel A. Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Buenos Aires, Argentina
| | - Nicholas P. Restifo
- National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Naiyer Rizvi
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Catherine Sautès-Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | - Hans Schreiber
- Dept. of Pathology, The Cancer Research Center, The University of Chicago, Chicago, IL, USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Hiroshi Shiku
- Dept. of Immuno-GeneTherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Daniel E. Speiser
- Dept. of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Pramod K. Srivastava
- Dept. of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
- Carole and Ray Neag Comprehensive Cancer Center, Farmington, CT, USA
| | - James E. Talmadge
- Laboratory of Transplantation Immunology, Dept. of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric Tartour
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- INSERM, U970, Paris, France
- Paris-Cardiovascular Research Center (PARCC), Paris, France
- Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou (HEGP), AP-HP, Paris, France
| | | | - Benoît J. Van Den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Brussels, Belgium
- Université Catholique de Louvain, Brussels, Belgium
| | - Richard Vile
- Dept. of Molecular Medicine and Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Hermann Wagner
- Institute of Medical Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
| | - Jeffrey S. Weber
- Donald A. Adam Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, FL, USA
| | - Theresa L. Whiteside
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jedd D. Wolchok
- Dept. of Medicine and Ludwig Center, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, U1015, Villejuif, France
- Centre d'Investigation Clinique Biothérapie 507 (CICBT507), Gustave Roussy Cancer Campus, Villejuif, France
| | - Weiping Zou
- University of Michigan, School of Medicine, Ann Arbor, MI, USA
| | - Guido Kroemer
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou (HEGP), AP-HP, Paris, France
| |
Collapse
|
26
|
Bianchi G, Anderson KC. Understanding biology to tackle the disease: Multiple myeloma from bench to bedside, and back. CA Cancer J Clin 2014; 64:422-44. [PMID: 25266555 DOI: 10.3322/caac.21252] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/21/2014] [Accepted: 08/21/2014] [Indexed: 02/01/2023] Open
Abstract
Multiple myeloma (MM) is a cancer of antibody-producing plasma cells. The pathognomonic laboratory finding is a monoclonal immunoglobulin or free light chain in the serum and/or urine in association with bone marrow infiltration by malignant plasma cells. MM develops from a premalignant condition, monoclonal gammopathy of undetermined significance (MGUS), often via an intermediate stage termed smoldering multiple myeloma (SMM), which differs from active myeloma by the absence of disease-related end-organ damage. Unlike MGUS and SMM, active MM requires therapy. Over the past 6 decades, major advancements in the care of MM patients have occurred, in particular, the introduction of novel agents (ie, proteasome inhibitors, immunomodulatory agents) and the implementation of hematopoietic stem cell transplantation in suitable candidates. The effectiveness and good tolerability of novel agents allowed for their combined use in induction, consolidation, and maintenance therapy, resulting in deeper and more sustained clinical response and extended progression-free and overall survival. Previously a rapidly lethal cancer with few therapeutic options, MM is the hematologic cancer with the most novel US Food and Drug Administration-approved drugs in the past 15 years. These advances have resulted in more frequent long-term remissions, transforming MM into a chronic illness for many patients.
Collapse
Affiliation(s)
- Giada Bianchi
- Hematology Oncology Fellow, Jerome Lipper Multiple Myeloma Center and LeBow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
27
|
Novel thalidomide analogs: Anti-angiogenic and apoptotic effects on Hep-G2 and MCF-7 cancer cell lines. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.biomag.2014.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
28
|
Zahran MAH, Abdin YG, Osman AMA, Gamal-Eldeen AM, Talaat RM, Pedersen EB. Synthesis and Evaluation of Thalidomide and Phthalimide Esters as Antitumor Agents. Arch Pharm (Weinheim) 2014; 347:642-9. [DOI: 10.1002/ardp.201400073] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/15/2014] [Accepted: 04/17/2014] [Indexed: 11/07/2022]
Affiliation(s)
- Magdy A. H. Zahran
- Faculty of Science, Department of Chemistry; Menoufia University; Shebin El-Koam Egypt
| | - Yasmin G. Abdin
- Faculty of Science, Department of Chemistry; Menoufia University; Shebin El-Koam Egypt
| | - Amany M. A. Osman
- Faculty of Science, Department of Chemistry; Menoufia University; Shebin El-Koam Egypt
- Department of Physics, Chemistry and Pharmacy, Nucleic Acid Center; University of Southern Denmark; Odense Denmark
| | - Amira M. Gamal-Eldeen
- Cancer Biology Laboratory; Center of Excellence for Advanced Sciences, Biochemistry National Research Center; Dokki Cairo Egypt
| | - Roba M. Talaat
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute (GEBRI); Menoufiya University; Egypt
| | - Erik B. Pedersen
- Department of Physics, Chemistry and Pharmacy, Nucleic Acid Center; University of Southern Denmark; Odense Denmark
| |
Collapse
|
29
|
Abstract
Multiple myeloma continues to be an incurable disease. The understanding of the disease's pathophysiology has significantly improved over the past few years, partly due to the discovery of the role of immunomodulatory agents and the study of their mechanism of action. Thalidomide, the first of the immunomodulatory family to be used in the management of multiple myeloma, proved not only to be effective in the treatment of multiple myeloma, but also instigated a wide range of in vitro and in vivo studies to define the pathophysiology of the plasma cell dyscrasia. The attention thalidomide has received in the past and recent history has not been without a price. The drug has a side-effect profile that, if managed appropriately, provides the most unique active molecule in the management of the disease, where it maintains the same response rate in newly diagnosed patients as in advanced relapsed/refractory multiple myeloma patients.
Collapse
Affiliation(s)
- Mohamad A Hussein
- Cleveland Clinic Multidisciplinary Multiple Myeloma Research Program, Cleveland, OH 44195, USA.
| |
Collapse
|
30
|
Raje N, Hideshima T, Anderson KC. Therapeutic use of immunomodulatory drugs in the treatment of multiple myeloma. Expert Rev Anticancer Ther 2014; 6:1239-47. [PMID: 17020458 DOI: 10.1586/14737140.6.9.1239] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Immunomodulatory drugs, such as thalidomide, lenalidomide (Revlimid, CC-5013) and actimid (CC-4047), have a broad spectrum of activity and have shown remarkable responses in patients with multiple myeloma and related hematological diseases, such as myelodysplastic syndrome. They are currently being tested in other cancer types. This review will focus on the preclinical and clinical activity of thalidomide and its more potent immunomodulatory derivatives that are used to treat multiple myeloma. They represent a new class of antitumor agents that not only target the tumor cell directly, but also have significant activity within the bone marrow milieu. These agents have shown high responses in all phases of multiple myeloma, including the upfront setting, relapsed refractory stage and also as maintenance therapy for the disease. They have been used in combination with dexamethasone, chemotherapy and, more recently, with other novel agents, such as proteasome inhibitors. Thalidomide and lenalidomide in combination with dexamethasone have recently been approved by the US FDA for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Noopur Raje
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| | | | | |
Collapse
|
31
|
Laine AM, Westover KD, Choy H. Radiation therapy as a backbone of treatment of locally advanced non-small cell lung cancer. Semin Oncol 2013; 41:57-68. [PMID: 24565581 DOI: 10.1053/j.seminoncol.2013.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Locally advanced non-small cell lung cancer (LA-NSCLC) is a heterogeneous disease, encompassing stage IIIA, for which surgery in combination with chemotherapy and/or radiation therapy (RT) represents a potential treatment approach for select patients, and stage IIIB, for which chemoradiation represents the standard of care. Recent advances in systemic cytotoxic and molecularly targeted therapies coupled with technologic innovations in radiotherapy have the potential to improve outcomes for this patient population. Many ongoing clinical trials use specific genetic mutations or histologic status to determine the combination of targeted therapies and RT, as well as to determine the optimal chemoradiotherapy platforms. Additionally, use of modern RT techniques has improved outcomes for some patients with limited metastatic disease, thereby prompting further studies on how to best integrate aggressive management of oligometastases using RT with chemotherapeutic regimens.
Collapse
Affiliation(s)
- Aaron M Laine
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Kenneth D Westover
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Hak Choy
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
32
|
Ruchelman AL, Man HW, Zhang W, Chen R, Capone L, Kang J, Parton A, Corral L, Schafer PH, Babusis D, Moghaddam MF, Tang Y, Shirley MA, Muller GW. Isosteric analogs of lenalidomide and pomalidomide: synthesis and biological activity. Bioorg Med Chem Lett 2012; 23:360-5. [PMID: 23168019 DOI: 10.1016/j.bmcl.2012.10.071] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/12/2012] [Accepted: 10/15/2012] [Indexed: 11/26/2022]
Abstract
A series of analogs of the immunomodulary drugs lenalidomide (1) and pomalidomide (2), in which the amino group is replaced with various isosteres, was prepared and assayed for immunomodulatory activity and activity against cancer cell lines. The 4-methyl and 4-chloro analogs 4 and 15, respectively, displayed potent inhibition of tumor necrosis factor-α (TNF-α) in LPS-stimulated hPBMC, potent stimulation of IL-2 in a human T cell co-stimulation assay, and anti-proliferative activity against the Namalwa lymphoma cell line. Both of these analogs displayed oral bioavailability in rat.
Collapse
Affiliation(s)
- Alexander L Ruchelman
- Drug Discovery Department, Celgene Corporation, 86 Morris Avenue, Summit, NJ 07901, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Paesler J, Gehrke I, Poll-Wolbeck SJ, Kreuzer KA. Targeting the vascular endothelial growth factor in hematologic malignancies. Eur J Haematol 2012; 89:373-84. [DOI: 10.1111/ejh.12009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2012] [Indexed: 12/16/2022]
Affiliation(s)
- Julian Paesler
- Department I of Internal Medicine I; University at Cologne; Cologne; Germany
| | - Iris Gehrke
- Department I of Internal Medicine I; University at Cologne; Cologne; Germany
| | | | - Karl-Anton Kreuzer
- Department I of Internal Medicine I; University at Cologne; Cologne; Germany
| |
Collapse
|
34
|
Bianchi G, Ghobrial IM. Molecular mechanisms of effectiveness of novel therapies in multiple myeloma. Leuk Lymphoma 2012; 54:229-41. [DOI: 10.3109/10428194.2012.706287] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
35
|
Kim DN, Nam TK, Choe KS, Choy H. Personalized Combined Modality Therapy for Locally Advanced Non-small Cell Lung Cancer. Cancer Res Treat 2012; 44:74-84. [PMID: 22802745 PMCID: PMC3394867 DOI: 10.4143/crt.2012.44.2.74] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 12/20/2011] [Indexed: 11/21/2022] Open
Abstract
Locally advanced non-small cell lung cancer (NSCLC) is a heterogeneous disease, and we have embarked on an era where patients will benefit from individualized therapeutic strategies based on identifiable molecular characteristics of the tumor. The landmark studies demonstrating the importance of molecular characterization of tumors for NSCLC patients, the promising molecular pathways, and the potential molecular targets/agents for treatment of this disease will be reviewed. Understanding these issues will aid in the development of rationally designed clinical trials, so as to determine best means of appropriately incorporating these molecular strategies, to the current standard of radiation and chemotherapy regimens, for the treatment of locally advanced NSCLC.
Collapse
Affiliation(s)
- D Nathan Kim
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | |
Collapse
|
36
|
Induction therapy and stem cell mobilization in patients with newly diagnosed multiple myeloma. Stem Cells Int 2012; 2012:607260. [PMID: 22701493 PMCID: PMC3369512 DOI: 10.1155/2012/607260] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 04/06/2012] [Indexed: 12/31/2022] Open
Abstract
Autologous stem cell transplantation (ASCT) is considered the standard therapy for younger patients with newly diagnosed symptomatic multiple myeloma (MM). The introduction into clinical practice of novel agents, such as the proteasome inhibitor bortezomib and the immunomodulatory derivatives (IMiDs) thalidomide and lenalidomide, has significantly contributed to major advances in MM therapy and prognosis. These novel agents are incorporated into induction regimens to enhance the depth of response before ASCT and further improve post-ASCT outcomes. Between January 2000 and November 2011, 65 patients with MM were transplanted in the Department of Biomedical Science and Clinical Oncology at the University of Bari. According to Durie-Salmon, 60 patients had stage III of disease and 5 stage II. Only 7 patients were in stage B (renal failure). Induction regimens that were administered in two or more cycles were VAD (vincristine, adriamycin, and dexamethasone), Thal-Dex (thalidomide, dexamethasone), Len-Dex (lenalidomide, dexamethasone), Vel-Dex (bortezomib, dexamethasone), VTD (bortezomib, thalidomide, and dexamethasone), and PAD (bortezomib, pegylated liposomal doxorubicin, and dexamethasone). In mobilization procedure, the patients received cyclophosphamide and granulocyte colony-stimulating factor (G-CSF). The number of cells collected through two or more leukapheresess, response after induction, and toxicity were evaluated to define the more adequate up-front induction regimen in transplantation-eligible MM patients.
Collapse
|
37
|
Bugaev IM, Dmitriev AI, Prosenko AE. Direct phthalimidomethylation of 2,6-dialkylphenols. Reactions of N-(4-hydroxybenzyl)phthalimides with electrophiles and nucleophiles. Russ Chem Bull 2012. [DOI: 10.1007/s11172-012-0041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
38
|
Takada K, Munemasa Y, Kuribayashi J, Fujino H, Kitaoka Y. Protective effect of thalidomide against N-methyl-D-aspartate-induced retinal neurotoxicity. J Neurosci Res 2011; 89:1596-604. [PMID: 21702058 DOI: 10.1002/jnr.22698] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 12/23/2022]
Abstract
Thalidomide, an inhibitor of tumor necrosis factor-α (TNF-α) production, has been indicated to be useful for many inflammatory and oncogenic diseases. In the present study, we examined whether thalidomide (50 mg/kg/day, p.o.) has a protective effect against N-methyl-D-aspartate (NMDA)-induced retinal neurotoxicity in rats. A morphometric analysis showed that systemic administration of thalidomide protects neural cells in the ganglion cell layer (GCL) in a dose-dependent manner and significantly decreases the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells in GCL and in the inner nuclear layer (INL). ELISA showed that thalidomide significantly suppressed the elevation of TNF-α 6 and 24 hr after an NMDA injection. Western blot analysis revealed a significant increase in nuclear factor-κB (NF-κB) p65 level in the retinas treated with NMDA at 24 hr after the injection, but not at 6 or 72 hr. Furthermore, an increase in p-JNK and p-p38 levels was also observed in the retina after NMDA injection. Thalidomide suppressed the increased expressions of NF-κB p65, p-JNK, and p-p38 after NMDA injection. Immunohistochemical analysis showed that thalidomide attenuated NF-κB p65 immunoreactivity in the GCL induced by NMDA treatment. In the NMDA-treated group, translocation of NF-κB p65 from the cytoplasm to the nucleus was detected in TUNEL-positive cells exposed to NMDA treatment. These results suggest new indications for thalidomide against neurodegenerative diseases.
Collapse
Affiliation(s)
- Kazuhide Takada
- 5th Year, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | | | | | | | | |
Collapse
|
39
|
Kang MH, Ju JH, Kim HG, Kang JH, Jeon KN, Kim HC, Lee GW. Thalidomide induced nonspecific interstitial pneumonia in patient with relapsed multiple myeloma. Korean J Intern Med 2010; 25:447-9. [PMID: 21179284 PMCID: PMC2997975 DOI: 10.3904/kjim.2010.25.4.447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2007] [Revised: 02/14/2008] [Accepted: 03/28/2008] [Indexed: 11/27/2022] Open
Abstract
A 63-year-old female diagnosed with relapsed multiple myeloma visited our hospital complaining of a persistent cough. Since July 2006, she had been taking 100 mg thalidomide daily and gradually developed shortness of breath and a persistent dry cough. A chest X-ray and computed tomography showed ground glass opacities in both lungs. An open lung biopsy of the right middle lobe under general anesthesia revealed chronic peribronchial inflammation, mild interstitial fibrosis, and intra-alveolar macrophage infiltration, with some hemosiderin features, compatible with non-specific interstitial pneumonia (NSIP). After discontinuing the thalidomide, the patient's symptoms did not deteriorate, although the radiographs did not improve. The patient is alive and well with regular outpatient follow-up without progression of the NSIP.
Collapse
Affiliation(s)
- Myung-Hee Kang
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Ji-Hyun Ju
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Hoon-Gu Kim
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Jung Hun Kang
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Kyung-Nyeo Jeon
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Korea
- Department of Diagnostic Radiology, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Ho-Cheol Kim
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Gyeong-Won Lee
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
- Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Korea
| |
Collapse
|
40
|
PI3K/Akt signaling pathway is required for neuroprotection of thalidomide on hypoxic–ischemic cortical neurons in vitro. Brain Res 2010; 1357:157-65. [DOI: 10.1016/j.brainres.2010.08.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 07/31/2010] [Accepted: 08/04/2010] [Indexed: 01/06/2023]
|
41
|
Kim DH, Kim YJ, Chang SA, Lee HW, Kim HN, Kim HK, Chang HJ, Sohn DW, Park YB. The protective effect of thalidomide on left ventricular function in a rat model of diabetic cardiomyopathy. Eur J Heart Fail 2010; 12:1051-60. [PMID: 20601373 DOI: 10.1093/eurjhf/hfq103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS To evaluate the protective effect of thalidomide, a potent anti-inflammatory drug, on the development of diabetic cardiomyopathy (DMCMP). METHODS AND RESULTS We induced type 1 diabetes using streptozocin in 8-week-old Sprague-Dawley rats, divided them into two groups-a thalidomide treatment group (DM-T, n = 15) and a non-treatment group (DM-N, n = 15)-and compared them with a normal control (n = 10). Ten weeks after diabetes induction, heart and lung mass indices were higher in the DM-N group compared with the control group. In the DM-T group, increases in heart and lung mass indices were attenuated compared with the DM-N group. On echocardiographic examination, systolic and diastolic mitral annulus velocities were impaired in the DM-N group, but they remained normal in the DM-T group. On haemodynamic analyses, left ventricular (LV) systolic function, represented by end-systolic elastance (0.35 ± 0.14 vs. 0.18 ± 0.07 mmHg/μl, P < 0.001) and preload-recruitable stroke work (90.5 ± 24.3 vs. 51.8 ± 22.0 mmHg, P < 0.001), was preserved in the DM-T group compared with the DM-N group. Likewise, deterioration of LV diastolic function was attenuated in the DM-T group. Increases in serum levels of TNF-α were attenuated in the DM-T group compared with the DM-N group. On histological analysis, thalidomide treatment lowered total myocardial collagen content and the expression of TNF-α, IL-1β, ICAM-1, and VCAM-1. CONCLUSION In an animal model of DMCMP, deterioration of LV systolic and diastolic function was partially prevented by thalidomide treatment.
Collapse
Affiliation(s)
- Dae-Hee Kim
- Department of Internal Medicine, Cardiovascular Center, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Brown RD, Spencer A, Joy Ho P, Kennedy N, Kabani K, Yang S, Sze DM, Aklilu E, Gibson J, Joshua DE. Prognostically significant cytotoxic T cell clones are stimulated after thalidomide therapy in patients with multiple myeloma. Leuk Lymphoma 2009; 50:1860-4. [DOI: 10.3109/10428190903216804] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
43
|
Quintás-Cardama A, Kantarjian HM, Manshouri T, Thomas D, Cortes J, Ravandi F, Garcia-Manero G, Ferrajoli A, Bueso-Ramos C, Verstovsek S. Lenalidomide plus prednisone results in durable clinical, histopathologic, and molecular responses in patients with myelofibrosis. J Clin Oncol 2009; 27:4760-6. [PMID: 19720904 PMCID: PMC4879697 DOI: 10.1200/jco.2009.22.6548] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE To investigate the safety and efficacy of the combination of lenalidomide and prednisone in patients with myelofibrosis (MF). PATIENTS AND METHODS Forty patients with MF were treated. Therapy consisted of lenalidomide 10 mg/d (5 mg/d if baseline platelet count < 100 x 10(9)/L) on days 1 through 21 of a 28-day cycle for six cycles, in combination with prednisone 30 mg/d orally during cycle 1, 15 mg/d during cycle 2, and 15 mg/d every other day during cycle 3. Lenalidomide therapy was continued indefinitely in patients exhibiting clinical benefit. RESULTS The median follow-up was 22 months (range, 6 to 27). Responses were recorded in 12 patients (30%) and are ongoing in 10 (25%). The median time to response was 12 weeks (range, 2 to 32). According to the International Working Group for Myelofibrosis Research and Treatment consensus criteria, three patients (7.5%) had partial response and nine patients (22.5%) had clinical improvement durable for a median of 18 months (range, 3.5 to 24+). Overall response rates were 30% for anemia and 42% for splenomegaly. Moreover, 10 of 11 assessable responders who started therapy with reticulin fibrosis grade 4 experienced reductions to at least a score of 2. All eight JAK2(V617F)-positive responders experienced a reduction of the baseline mutant allele burden, which was greater than 50% in four, including one of whom the mutation became undetectable. Grade 3 to 4 hematologic adverse events included neutropenia (58%), anemia (42%), and thrombocytopenia (13%). CONCLUSION The combination of lenalidomide and prednisone induces durable clinical, molecular, and pathologic responses in MF.
Collapse
Affiliation(s)
- Alfonso Quintás-Cardama
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Hagop M. Kantarjian
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Taghi Manshouri
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Deborah Thomas
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Jorge Cortes
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Guillermo Garcia-Manero
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Alessandra Ferrajoli
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Carlos Bueso-Ramos
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX
| | - Srdan Verstovsek
- From the Departments of Leukemia and Hematopathology, M.D. Anderson Cancer Center, Houston, TX.,Corresponding author: Srdan Verstovsek, MD, PhD, University of Texas M. D. Anderson Cancer Center, Department of Leukemia, Unit 428, 1515 Holcombe Blvd, Houston, TX 77030; e-mail:
| |
Collapse
|
44
|
Koskela K, Pelliniemi TT, Pulkki K, Remes K. Treatment of Multiple Myeloma with All-Trans Retinoic Acid Alone and in Combination with Chemotherapy: a Phase I/II Trial. Leuk Lymphoma 2009; 45:749-54. [PMID: 15160951 DOI: 10.1080/10428190310001628158] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
All-trans retinoic acid (ATRA) is a derivative of vitamin A. ATRA inhibits the growth of human myeloma cell lines and freshly isolated myeloma cells in vitro mainly by down-regulating interleukin-6 receptor. Clinically, however, ATRA alone has not been efficacious and adverse events, notably hypercalcemia, have been common. In the present study 10 patients with stable multiple myeloma after conventional chemotherapy received ATRA alone for 2 months, followed by a combination of ATRA and the chemotherapy regimen during which no further reduction of the paraprotein had occurred. The purpose of the combination therapy was to sensitize the myeloma cells with ATRA to chemotherapy by blocking the growth-promoting effect of IL-6. Although ATRA was well tolerated, ATRA alone lacked clinical efficacy. The combination therapy resulted minimal responses in 4 patients and relatively long progression-free survival in 4 patients was achieved. In 3 of these responding patients serum concentrations of interleukin-6 and/or soluble interleukin-6 receptor were elevated prior to the study. The bone marrow cells of responding patients were sensitive to ATRA in vitro. These results show that ATRA alone is not effective to treat multiple myeloma. There may be some beneficial effect of ATRA in combination chemotherapy in selected patients who have activated IL-6 signaling.
Collapse
Affiliation(s)
- Kari Koskela
- Department of Medicine, Turku University Central Hospital, Turku, Finland.
| | | | | | | |
Collapse
|
45
|
Kees M, Dimou G, Sillaber C, Drach J, Ackermann J, Lechner K, Gisslinger H. Low Dose Thalidomide in Patients with Relapsed or Refractory Multiple Myeloma. Leuk Lymphoma 2009; 44:1943-6. [PMID: 14738147 DOI: 10.1080/1042819031000123492] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Remarkable results of the treatment of refractory multiple myeloma with thalidomide have been reported. In most preceding studies, the given thalidomide dose was escalated to a maximum tolerated dose of up to 800 mg/d. The frequency of adverse effects correlates with dose intensity. Since a significant gain of therapeutic effects could not be observed as thalidomide dosage was escalated, the optimal dose of thalidomide remains to be determined. We report the results of a study with low dose thalidomide (median administered dose 100 mg/d, range 50-400 mg/d). Twenty-four relapsed (n = 19) or resistant (n = 5) multiple myeloma patients were included in the study. Twelve patients (50%) received thalidomide as monotherapy, 8 patients (33%) received a combination of thalidomide and dexamethasone (every 4 weeks 40 mg/day for 4 days) and 4 patients (17%) who were resistant to vincristine, doxorubicin, dexamethasone (VAD) received VAD combined with thalidomide. Overall, a response was observed in 12 patients (50%). Of the 12 patients treated with low dose thalidomide alone 5 (42%) responded, of the 8 patients who received a combination of thalidomide and dexamethasone 5 (63%) responded and of the 4 patients who had thalidomide in addition to VAD 2 patients (50%) responded. In 3 patients, thalidomide treatment had to be discontinued because of side effects and 1 patient died before response could be assessed. We conclude that low dose thalidomide is an effective and safe rescue therapy in relapsing or refractory multiple myeloma. Response to thalidomide might be dependent on prognostic parameters and tumor burden. To answer these questions larger prospective studies are necessary.
Collapse
Affiliation(s)
- M Kees
- Division of Haematology and Blood Coagulation, Department of Internal Medicine I, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
46
|
Garderet L, Mazurier C, Pellat-Deceunynck C, Karim A, Baudin B, Funck-Brentano C, Bouchet S, Geffroy A, Bataille R, Gorin NC, Lopez M. Poorex vivoinduction of T-cell responses to idiotype or tumor cell lysate-pulsed autologous dendritic cells in advanced pre-treated multiple myeloma. Leuk Lymphoma 2009; 47:1340-7. [PMID: 16923566 DOI: 10.1080/10428190500465242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This study evaluated the feasibility of using dendritic cells (DCs) to generate, ex vivo, anti-tumor cytotoxic T lymphocytes (CTL) in patients with stage III multiple myeloma (MM). Nucleated cells from eight patients who had received chemotherapy (three of whom had undergone autologous hemopoeitic stem cell transplantation) were collected by apheresis. Their monocytes were enriched using counter-current centrifugation, differentiated into DCs which were further co-cultured with autologous CD8 lymphocytes to induce CTL. The DCs were pulsed either with the idiotypic paraprotein (regarded as a tumor-specific antigen) or with autologous MM cell lysate before co-culture. Specific T-cell responses were measured in IFNgamma enzyme-linked immunospot and chromium release assays of autologous plasmocyte targets. A slight increase in IFNgamma secretion by T-cells was observed for two patients (DCs pulsed with idiotypic paraprotein for one, MM cell lysate for the other). No or weak specific lysis of plasmocyte targets was observed in the chromium release assays. In conclusion, the T-cell response to pulsed DCs was very weak or absent. There are clinical and technical reasons that could explain, in part, this lack of response.
Collapse
Affiliation(s)
- Laurent Garderet
- Inserm and EA1638, Faculté de Médecine Saint Antoine, Université Pierre et Marie Curie (Paris 6), Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kodama T, Abe M, Iida S, Ozaki S, Sakai A, Sawamura M, Shimazaki C, Miyata A, Wakayama T, Murakami H. A Pharmacokinetic Study Evaluating the Relationship Between Treatment Efficacy and Incidence of Adverse Events with Thalidomide Plasma Concentrations in Patients with Refractory Multiple Myeloma. ACTA ACUST UNITED AC 2009; 9:154-9. [DOI: 10.3816/clm.2009.n.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Preclinical activity of P276-00, a novel small-molecule cyclin-dependent kinase inhibitor in the therapy of multiple myeloma. Leukemia 2009; 23:961-70. [PMID: 19151776 DOI: 10.1038/leu.2008.378] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cyclin D dysregulation and overexpression is noted in the majority of multiple myeloma (MM) patients, suggesting its critical role in MM pathogenesis. Here, we sought to identify the effects of targeting cyclin D in MM. We first confirmed cyclin D mRNA overexpression in 42 of 64 (65%) patient plasma cells. Silencing cyclin D1 resulted in >50% apoptotic cell death suggesting its validity as a potential therapeutic target. We next evaluated P276-00, a clinical-grade small-molecule cyclin-dependent kinase inhibitor as a way to target the cyclins. P276-00 resulted in dose-dependent cytotoxicity in MM cells. Cell-cycle analysis confirmed either growth arrest or caspase-dependent apoptosis; this was preceded by inhibition of Rb-1 phosphorylation with associated downregulation of a range of cyclins suggesting a regulatory role of P276-00 in cell-cycle progression through broad activity. Proliferative stimuli such as interleukin-6, insulin-like growth factor-1 and bone-marrow stromal cell adherence induced cyclins; P276-00 overcame these growth, survival and drug resistance signals. Because the cyclins are substrates of proteasome degradation, combination studies with bortezomib resulted in synergism. Finally, in vivo efficacy of P276-00 was confirmed in an MM xenograft model. These studies form the basis of an ongoing phase I study in the treatment of relapsed/refractory MM.
Collapse
|
49
|
Husain S, Yates PW, Crosson CE. Latanoprost-induced changes in rat intraocular pressure: direct or indirect? J Ocul Pharmacol Ther 2009; 24:367-72. [PMID: 18665807 DOI: 10.1089/jop.2008.0042] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION The topical application of prostaglandin F(2 ) (FP)-receptor agonists has been shown to significantly lower intraocular pressure (IOP) in humans and is now considered the first-line treatment for ocular hypertension. Despite the prominent role FP-receptor agonists play in the treatment of glaucoma, our understanding of how these agents lower IOP remains incomplete. The present study was designed to evaluate the role of matrix metalloproteinase (MMP) activation and the cytokine, tumor necrosis factor alpha (TNF-alpha), in latanoprost-induced changes in IOP. METHODS Changes in IOP following an acute topical administration of latanoprost (60 ng) in normotensive Brown Norway rats were evaluated by means of a commercially available rebound tonometer. To examine the role of MMPs and TNF-alpha in this response, the rats were pretreated with a broad-spectrum MMP inhibitor, GM-6001 (100 microg), or the TNF-alpha inhibitor, thalidomide (25 microg). RESULTS The topical administration of latanoprost (60 ng) alone produced a biphasic change in ipsilateral IOP: an initial hypertension (4.21 +/- 0.52), followed by a prolonged hypotension (-4.79 +/- 0.65). In rats, pretreatment with GM-6001 blocked the latanoprost-induced reduction in IOP but did not prevent the initial rise in IOP. Pretreatment with thalidomide also blocked the ocular hypotension induced by latanoprost; however, thalidomide pretreatment enhanced the duration of the initial hypertension. CONCLUSIONS These results provide evidence that the secretion and activation of MMPs and the release of TNF-alpha play a central role in the ocular hypotension induced by FP-agonists. The administration of FP-agonists appears to lower IOP directly by inducing the activation of MMPs within the ciliary body, leading to improved uveoscleral outflow and indirectly through the release of TNF within the ciliary body. Secreted TNF-alpha may then activate TNF-receptors in the uvea and trabecular meshwork, increasing both uveoscleral and conventional outflow.
Collapse
Affiliation(s)
- Shahid Husain
- Hewitt Laboratory of the Ola B. Williams Glaucoma Center, Department of Ophthalmology, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | |
Collapse
|
50
|
Vallet S, Palumbo A, Raje N, Boccadoro M, Anderson KC. Thalidomide and lenalidomide: Mechanism-based potential drug combinations. Leuk Lymphoma 2008; 49:1238-45. [PMID: 18452080 DOI: 10.1080/10428190802005191] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Thalidomide and its analogue lenalidomide are potent anti-inflammatory, anti-angiogenic and immunomodulatory drugs, successfully used for the treatment of hematological cancers, in particular multiple myeloma (MM). Both drugs reveal a dual mechanism of action: they target tumour cells by direct cytotoxicity and, indirectly, by interfering with several components of the bone marrow microenvironment. Lenalidomide and thalidomide are versatile drugs with a broad range of activities that potentiate the anti-MM effects of conventional and novel agents. Here, we review the mechanism of action of these drugs, providing a rationale for combination studies in order to improve patient outcome and reduce side effects.
Collapse
Affiliation(s)
- Sonia Vallet
- Division of Hematology and Oncology, Cancer Center, Massachusetts General Hospital, Boston, MA, USA.
| | | | | | | | | |
Collapse
|