1
|
Kaur C, Sharma B, Nepali K. Switch Pocket Kinase: An Emerging Therapeutic Target for the Design of Anticancer Agents. Anticancer Agents Med Chem 2022; 22:2662-2670. [PMID: 35379129 DOI: 10.2174/1871520622666220404081302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 11/22/2022]
Abstract
Protein kinases are amongst the most focused enzymes in current century to design, synthesize and formulate drugs ought to be effective in the treatment of various disordered and diseased states involving either overexpression or deficiency situations. The ATP pocket on the kinases is the binding active site for most of the kinase inhibitors. However, the kinase mutations prevent the binding of kinase inhibitors to ATP pocket. The switch pocket site on this enzyme when occupied by switch pocket inhibitors, the enzyme become inactive even in the mutated state. This review comprises the detailed information on various classical protein kinases and switch pocket kinase inhibitors with their mechanism of action so that new molecules can be designed to encounter mutations in the kinase enzyme.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Pharmaceutical Chemistry, Khalsa College of Pharmacy, Amritsar, Punjab, 143002
| | - Bhargavi Sharma
- Department of Pharmaceutical Chemistry, Khalsa College of Pharmacy, Amritsar, Punjab, 143002
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| |
Collapse
|
2
|
Multiparameter Evaluation of the Platelet-Inhibitory Effects of Tyrosine Kinase Inhibitors Used for Cancer Treatment. Int J Mol Sci 2021; 22:ijms222011199. [PMID: 34681859 PMCID: PMC8540269 DOI: 10.3390/ijms222011199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/01/2021] [Accepted: 10/13/2021] [Indexed: 12/30/2022] Open
Abstract
Current antiplatelet drugs for the treatment of arterial thrombosis often coincide with increased bleeding risk. Several tyrosine kinase inhibitors (TKIs) for cancer treatment inhibit platelet function, with minor reported bleeding symptoms. The aim of this study was to compare the antiplatelet properties of eight TKIs to explore their possible repurposing as antiplatelet drugs. Samples of whole blood, platelet-rich plasma (PRP), or isolated platelets from healthy donors were treated with TKI or the vehicle. Measurements of platelet aggregation, activation, intracellular calcium mobilization, and whole-blood thrombus formation under flow were performed. Dasatinib and sunitinib dose-dependently reduced collagen-induced aggregation in PRP and washed platelets; pazopanib, cabozantinib, and vatalanib inhibited this response in washed platelets only; and fostamatinib, axitinib, and lapatinib showed no/limited effects. Fostamatinib reduced thrombus formation by approximately 50% on collagen and other substrates. Pazopanib, sunitinib, dasatinib, axitinib, and vatalanib mildly reduced thrombus formation on collagen by 10–50%. Intracellular calcium responses in isolated platelets were inhibited by dasatinib (>90%), fostamatinib (57%), sunitinib (77%), and pazopanib (82%). Upon glycoprotein-VI receptor stimulation, fostamatinib, cabozantinib, and vatalanib decreased highly activated platelet populations by approximately 15%, while increasing resting populations by 39%. In conclusion, the TKIs with the highest affinities for platelet-expressed molecular targets most strongly inhibited platelet functions. Dasatinib, fostamatinib, sunitinib, and pazopanib interfered in early collagen receptor-induced molecular-signaling compared with cabozantinib and vatalanib. Fostamatinib, sunitinib, pazopanib, and vatalanib may be promising for future evaluation as antiplatelet drugs.
Collapse
|
3
|
Zheng TJ, Lofurno ER, Melrose AR, Lakshmanan HHS, Pang J, Phillips KG, Fallon ME, Kohs TCL, Ngo ATP, Shatzel JJ, Hinds MT, McCarty OJT, Aslan JE. Assessment of the effects of Syk and BTK inhibitors on GPVI-mediated platelet signaling and function. Am J Physiol Cell Physiol 2021; 320:C902-C915. [PMID: 33689480 PMCID: PMC8163578 DOI: 10.1152/ajpcell.00296.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/25/2022]
Abstract
Spleen tyrosine kinase (Syk) and Bruton's tyrosine kinase (BTK) play critical roles in platelet physiology, facilitating intracellular immunoreceptor tyrosine-based activation motif (ITAM)-mediated signaling downstream of platelet glycoprotein VI (GPVI) and GPIIb/IIIa receptors. Small molecule tyrosine kinase inhibitors (TKIs) targeting Syk and BTK have been developed as antineoplastic and anti-inflammatory therapeutics and have also gained interest as antiplatelet agents. Here, we investigate the effects of 12 different Syk and BTK inhibitors on GPVI-mediated platelet signaling and function. These inhibitors include four Syk inhibitors, Bay 61-3606, R406 (fostamatinib), entospletinib, TAK-659; four irreversible BTK inhibitors, ibrutinib, acalabrutinib, ONO-4059 (tirabrutinib), AVL-292 (spebrutinib); and four reversible BTK inhibitors, CG-806, BMS-935177, BMS-986195, and fenebrutinib. In vitro, TKIs targeting Syk or BTK reduced platelet adhesion to collagen, dense granule secretion, and alpha granule secretion in response to the GPVI agonist cross-linked collagen-related peptide (CRP-XL). Similarly, these TKIs reduced the percentage of activated integrin αIIbβ3 on the platelet surface in response to CRP-XL, as determined by PAC-1 binding. Although all TKIs tested inhibited phospholipase C γ2 (PLCγ2) phosphorylation following GPVI-mediated activation, other downstream signaling events proximal to phosphoinositide 3-kinase (PI3K) and PKC were differentially affected. In addition, reversible BTK inhibitors had less pronounced effects on GPIIb/IIIa-mediated platelet spreading on fibrinogen and differentially altered the organization of PI3K around microtubules during platelets spreading on fibrinogen. Select TKIs also inhibited platelet aggregate formation on collagen under physiological flow conditions. Together, our results suggest that TKIs targeting Syk or BTK inhibit central platelet functional responses but may differentially affect protein activities and organization in critical systems downstream of Syk and BTK in platelets.
Collapse
Affiliation(s)
- Tony J Zheng
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Elizabeth R Lofurno
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Alexander R Melrose
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | | | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | | | - Meghan E Fallon
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Tia C L Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Monica T Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
4
|
Kost-Alimova M, Sidhom EH, Satyam A, Chamberlain BT, Dvela-Levitt M, Melanson M, Alper SL, Santos J, Gutierrez J, Subramanian A, Byrne PJ, Grinkevich E, Reyes-Bricio E, Kim C, Clark AR, Watts AJ, Thompson R, Marshall J, Pablo JL, Coraor J, Roignot J, Vernon KA, Keller K, Campbell A, Emani M, Racette M, Bazua-Valenti S, Padovano V, Weins A, McAdoo SP, Tam FW, Ronco L, Wagner F, Tsokos GC, Shaw JL, Greka A. A High-Content Screen for Mucin-1-Reducing Compounds Identifies Fostamatinib as a Candidate for Rapid Repurposing for Acute Lung Injury. Cell Rep Med 2020; 1:100137. [PMID: 33294858 PMCID: PMC7691435 DOI: 10.1016/j.xcrm.2020.100137] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/23/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022]
Abstract
Drug repurposing has the advantage of identifying potential treatments on a shortened timescale. In response to the pandemic spread of SARS-CoV-2, we took advantage of a high-content screen of 3,713 compounds at different stages of clinical development to identify FDA-approved compounds that reduce mucin-1 (MUC1) protein abundance. Elevated MUC1 levels predict the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) and correlate with poor clinical outcomes. Our screen identifies fostamatinib (R788), an inhibitor of spleen tyrosine kinase (SYK) approved for the treatment of chronic immune thrombocytopenia, as a repurposing candidate for the treatment of ALI. In vivo, fostamatinib reduces MUC1 abundance in lung epithelial cells in a mouse model of ALI. In vitro, SYK inhibition by the active metabolite R406 promotes MUC1 removal from the cell surface. Our work suggests fostamatinib as a repurposing drug candidate for ALI.
Collapse
Affiliation(s)
| | - Eriene-Heidi Sidhom
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Abhigyan Satyam
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | - Moran Dvela-Levitt
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Seth L. Alper
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jean Santos
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Juan Gutierrez
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | - Choah Kim
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Abbe R. Clark
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrew J.B. Watts
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Jamie Marshall
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Juliana Coraor
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie Roignot
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Katherine A. Vernon
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Keith Keller
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Alissa Campbell
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Silvana Bazua-Valenti
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Astrid Weins
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephen P. McAdoo
- Department of Immunology and Inflammation, Imperial College, Hammersmith Hospital, London, UK
| | - Frederick W.K. Tam
- Department of Immunology and Inflammation, Imperial College, Hammersmith Hospital, London, UK
| | - Luciene Ronco
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - George C. Tsokos
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | - Anna Greka
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Barlaam B, Boiko S, Boyd S, Dry H, Gingipalli L, Ikeda T, Johnson T, Kawatkar S, Lorthioir O, Pike A, Pollard H, Read J, Su Q, Wang H, Wang H, Wang L, Wang P, Edmondson SD. Novel potent and selective pyrazolylpyrimidine-based SYK inhibitors. Bioorg Med Chem Lett 2020; 30:127523. [PMID: 32877741 DOI: 10.1016/j.bmcl.2020.127523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 10/23/2022]
Abstract
Hybridisation of amino-pyrimidine based SYK inhibitors (e.g. 1a) with previously reported diamine-based SYK inhibitors (e.g. TAK-659) led to the identification and optimisation of a novel pyrimidine-based series of potent and selective SYK inhibitors, where the original aminomethylene group was replaced by a 3,4-diaminotetrahydropyran group. The initial compound 5 achieved excellent SYK potency. However, it suffered from poor permeability and modest kinase selectivity. Further modifications of the 3,4-diaminotetrahydropyran group were identified and the interactions of those groups with Asp512 were characterised by protein X-ray crystallography. Further optimisation of this series saw mixed results where permeability and kinase selectivity were increased and oral bioavailability was achieved in the series, but at the expense of potent hERG inhibition.
Collapse
Affiliation(s)
| | - Scott Boiko
- R&D Oncology, AstraZeneca, Boston, MA, United States
| | - Scott Boyd
- R&D Oncology, AstraZeneca, Cambridge, United Kingdom
| | - Hannah Dry
- R&D Oncology, AstraZeneca, Boston, MA, United States
| | | | - Timothy Ikeda
- R&D Oncology, AstraZeneca, Boston, MA, United States
| | - Tony Johnson
- R&D Oncology, AstraZeneca, Cambridge, United Kingdom
| | | | | | - Andy Pike
- R&D Oncology, AstraZeneca, Cambridge, United Kingdom
| | - Hannah Pollard
- Discovery Sciences, AstraZeneca, Cambridge, United Kingdom
| | - Jon Read
- Discovery Sciences, AstraZeneca, Cambridge, United Kingdom
| | - Qibin Su
- R&D Oncology, AstraZeneca, Boston, MA, United States
| | - Haiyun Wang
- R&D Oncology, AstraZeneca, Boston, MA, United States
| | - Huimin Wang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, PR China
| | - Lianghe Wang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, PR China
| | - Peng Wang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, PR China
| | | |
Collapse
|
6
|
Alimova M, Sidhom EH, Satyam A, Dvela-Levitt M, Melanson M, Chamberlain BT, Alper SL, Santos J, Gutierrez J, Subramanian A, Grinkevich E, Bricio ER, Kim C, Clark A, Watts A, Thompson R, Marshall J, Pablo JL, Coraor J, Roignot J, Vernon KA, Keller K, Campbell A, Emani M, Racette M, Bazua-Valenti S, Padovano V, Weins A, McAdoo SP, Tam FW, Ronco L, Wagner F, Tsokos GC, Shaw JL, Greka A. A High Content Screen for Mucin-1-Reducing Compounds Identifies Fostamatinib as a Candidate for Rapid Repurposing for Acute Lung Injury during the COVID-19 pandemic. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.06.30.180380. [PMID: 32637960 PMCID: PMC7337390 DOI: 10.1101/2020.06.30.180380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Drug repurposing is the only method capable of delivering treatments on the shortened time-scale required for patients afflicted with lung disease arising from SARS-CoV-2 infection. Mucin-1 (MUC1), a membrane-bound molecule expressed on the apical surfaces of most mucosal epithelial cells, is a biochemical marker whose elevated levels predict the development of acute lung injury (ALI) and respiratory distress syndrome (ARDS), and correlate with poor clinical outcomes. In response to the pandemic spread of SARS-CoV-2, we took advantage of a high content screen of 3,713 compounds at different stages of clinical development to identify FDA-approved compounds that reduce MUC1 protein abundance. Our screen identified Fostamatinib (R788), an inhibitor of spleen tyrosine kinase (SYK) approved for the treatment of chronic immune thrombocytopenia, as a repurposing candidate for the treatment of ALI. In vivo , Fostamatinib reduced MUC1 abundance in lung epithelial cells in a mouse model of ALI. In vitro , SYK inhibition by Fostamatinib promoted MUC1 removal from the cell surface. Our work reveals Fostamatinib as a repurposing drug candidate for ALI and provides the rationale for rapidly standing up clinical trials to test Fostamatinib efficacy in patients with COVID-19 lung injury.
Collapse
Affiliation(s)
- Maria Alimova
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Eriene-Heidi Sidhom
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Abhigyan Satyam
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Moran Dvela-Levitt
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michelle Melanson
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Seth L. Alper
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Jean Santos
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Juan Gutierrez
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | | | | | - Choah Kim
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Abbe Clark
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Andrew Watts
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca Thompson
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jamie Marshall
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Juliana Coraor
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Julie Roignot
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Katherine A. Vernon
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keith Keller
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alissa Campbell
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Matthew Racette
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Silvana Bazua-Valenti
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Valeria Padovano
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Astrid Weins
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen P. McAdoo
- Department of Immunology and Inflammation, Imperial College, Hammersmith Hospital, London, UK
| | - Frederick W.K. Tam
- Department of Immunology and Inflammation, Imperial College, Hammersmith Hospital, London, UK
| | - Lucienne Ronco
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Florence Wagner
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - George C. Tsokos
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Jillian L. Shaw
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Anna Greka
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Fostamatinib for the treatment of chronic immune thrombocytopenia. Blood 2019; 133:2027-2030. [PMID: 30803989 DOI: 10.1182/blood-2018-11-852491] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/20/2019] [Indexed: 01/19/2023] Open
Abstract
Fostamatinib is a spleen tyrosine kinase inhibitor recently approved for the treatment of chronic immune thrombocytopenia (ITP) in patients without adequate response to at least 1 prior line of therapy. This article reviews fostmatinib's mechanism of action and its clinical safety and efficacy in 2 industry-sponsored multicenter phase 3 randomized controlled trials in North America, Australia, and Europe (FIT1 and FIT2). Cost comparisons are discussed as well as the role of fostamatinib in relation to other options for chronic ITP.
Collapse
|
8
|
Tullemans BME, Heemskerk JWM, Kuijpers MJE. Acquired platelet antagonism: off-target antiplatelet effects of malignancy treatment with tyrosine kinase inhibitors. J Thromb Haemost 2018; 16:1686-1699. [PMID: 29975003 DOI: 10.1111/jth.14225] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Indexed: 12/26/2022]
Abstract
Platelets can contribute to tumor progression and metastasis. Cancer patients are at increased risk of thrombosis, and advanced stages of cancer are associated with thrombocytosis or increased platelet reactivity. Tyrosine kinase inhibitors (TKIs) are widely used as a targeted strategy for cancer treatment, with the aim of prolonging progression-free survival of the patients. Because of their broad kinase target spectrum, most TKIs inevitably have off-target effects. Platelets rely on tyrosine kinase activity for their activation. Frequently observed side effects are lowering of platelet count and inhibition of platelet functions, whether or not accompanied by an increased bleeding risk. In this review, we aim to give insights into: (i) 38 TKIs that are currently used for the treatment of different types of cancer, either on the market or in clinical trials; (ii) how distinct TKIs can inhibit activation mechanisms in platelets; and (iii) the clinical consequences of the antiplatelet effects of TKI treatment. For several TKIs, the knowledge on affinity for their targets does not align with the published effects on platelets and reported bleeding events. This review should raise awareness of the potential antiplatelet effects of several TKIs, which will be enhanced in the presence of antithrombotic drugs.
Collapse
Affiliation(s)
- B M E Tullemans
- Cardiovascular Research Institute Maastricht, Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| | - J W M Heemskerk
- Cardiovascular Research Institute Maastricht, Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| | - M J E Kuijpers
- Cardiovascular Research Institute Maastricht, Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
9
|
Huang Y, Li Y, Dong G, Zhang W, Liu N, Sheng C. Identification of pyrazolopyridine derivatives as novel spleen tyrosine kinase inhibitors. Arch Pharm (Weinheim) 2018; 351. [PMID: 29952085 DOI: 10.1002/ardp.201800083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/03/2018] [Accepted: 06/08/2018] [Indexed: 11/12/2022]
Abstract
Inhibition of spleen tyrosine kinase (Syk) is a promising strategy for the treatment of various allergic and autoimmune disorders such as asthma, rheumatoid arthritis, and allergic rhinitis. Previously, a Syk inhibitor with novel indazole scaffold was discovered by structure-based virtual screening. Herein, the structure-activity relationship of the indazole Syk inhibitors was investigated. Several new inhibitors demonstrated potent activity against Syk. In particular, compound 18c showed good Syk inhibitory activity (IC50 = 1.2 µM), representing a good lead compound for further optimization.
Collapse
Affiliation(s)
- Yahui Huang
- School of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| | - Yu Li
- School of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| | - Guoqiang Dong
- School of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| | - Wannian Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| | - Na Liu
- School of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|
10
|
Bryan MC, Rajapaksa NS. Kinase Inhibitors for the Treatment of Immunological Disorders: Recent Advances. J Med Chem 2018; 61:9030-9058. [DOI: 10.1021/acs.jmedchem.8b00667] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Marian C. Bryan
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Naomi S. Rajapaksa
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
11
|
Van der Plas SE, Kelgtermans H, De Munck T, Martina SLX, Dropsit S, Quinton E, De Blieck A, Joannesse C, Tomaskovic L, Jans M, Christophe T, van der Aar E, Borgonovi M, Nelles L, Gees M, Stouten P, Van Der Schueren J, Mammoliti O, Conrath K, Andrews M. Discovery of N-(3-Carbamoyl-5,5,7,7-tetramethyl-5,7-dihydro-4H-thieno[2,3-c]pyran-2-yl)-lH-pyrazole-5-carboxamide (GLPG1837), a Novel Potentiator Which Can Open Class III Mutant Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Channels to a High Extent. J Med Chem 2018; 61:1425-1435. [DOI: 10.1021/acs.jmedchem.7b01288] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | - Hans Kelgtermans
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Tom De Munck
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | | | - Evelyne Quinton
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Ann De Blieck
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | - Linda Tomaskovic
- Fidelta Ltd., Prilaz Baruna Filipovića 29, Zagreb HR-10000, Croatia
| | - Mia Jans
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | | | - Monica Borgonovi
- Galapagos SASU, 102
Avenue Gaston Roussel, 93230 Romainville, France
| | - Luc Nelles
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Maarten Gees
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Pieter Stouten
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | - Oscar Mammoliti
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Katja Conrath
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Martin Andrews
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| |
Collapse
|
12
|
Alhazmi A, Choi J, Ulanova M. Syk inhibitor R406 downregulates inflammation in an in vitro model of Pseudomonas aeruginosa infection. Can J Physiol Pharmacol 2017; 96:182-190. [PMID: 29020462 DOI: 10.1139/cjpp-2017-0307] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As Pseudomonas aeruginosa infections are characterized by strong inflammation of infected tissues, anti-inflammatory therapies in combination with antibiotics have been considered for the treatment of associated diseases. Syk tyrosine kinase is an important regulator of inflammatory responses, and its specific inhibition was explored as a therapeutic option in several inflammatory conditions; however, this has not been studied in bacterial infections. We used a model of in vitro infection of human monocytic cell line THP-1 and lung epithelial cell line H292 with both wild-type and flagella-deficient mutant of P. aeruginosa strain K, as well as with clinical isolates from cystic fibrosis patients, to study the effect of a small molecule Syk inhibitor R406 on inflammatory responses induced by this pathogen. One-hour pretreatment of THP-1 cells with 10 μmol/L R406 resulted in a significant downregulation of the expression of the adhesion molecule ICAM-1, pro-inflammatory cytokines TNF-α and IL-1β, and phosphorylated signaling proteins ERK2, JNK, p-38, and IκBα, as well as significantly decreased TNF-α release by infected H292 cells. The results suggest that Syk is involved in the regulation of inflammatory responses to P. aeruginosa, and R406 may potentially be useful in dampening the damage caused by severe inflammation associated with this infection.
Collapse
Affiliation(s)
- Alaa Alhazmi
- a Department of Biology, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| | - Joshua Choi
- b Northern Ontario School of Medicine, Thunder Bay, ON P7B 5E1, Canada
| | - Marina Ulanova
- a Department of Biology, Lakehead University, Thunder Bay, ON P7B 5E1, Canada.,b Northern Ontario School of Medicine, Thunder Bay, ON P7B 5E1, Canada
| |
Collapse
|
13
|
Syk inhibitors interfere with erythrocyte membrane modification during P falciparum growth and suppress parasite egress. Blood 2017. [PMID: 28634183 DOI: 10.1182/blood-2016-11-748053] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Band 3 (also known as the anion exchanger, SLCA1, AE1) constitutes the major attachment site of the spectrin-based cytoskeleton to the erythrocyte's lipid bilayer and thereby contributes critically to the stability of the red cell membrane. During the intraerythrocytic stage of Plasmodium falciparum's lifecycle, band 3 becomes tyrosine phosphorylated in response to oxidative stress, leading to a decrease in its affinity for the spectrin/actin cytoskeleton and causing global membrane destabilization. Because this membrane weakening is hypothesized to facilitate parasite egress and the consequent dissemination of released merozoites throughout the bloodstream, we decided to explore which tyrosine kinase inhibitors might block the kinase-induced membrane destabilization. We demonstrate here that multiple Syk kinase inhibitors both prevent parasite-induced band 3 tyrosine phosphorylation and inhibit parasite-promoted membrane destabilization. We also show that the same Syk kinase inhibitors suppress merozoite egress near the end of the parasite's intraerythrocytic lifecycle. Because the entrapped merozoites die when prevented from escaping their host erythrocytes and because some Syk inhibitors have displayed long-term safety in human clinical trials, we suggest Syk kinase inhibitors constitute a promising class of antimalarial drugs that can suppress parasitemia by inhibiting a host target that cannot be mutated by the parasite to evolve drug resistance.
Collapse
|
14
|
Huang Y, Zhang Y, Fan K, Dong G, Li B, Zhang W, Li J, Sheng C. Discovery of new Syk inhibitors through structure-based virtual screening. Bioorg Med Chem Lett 2017; 27:1776-1779. [DOI: 10.1016/j.bmcl.2017.02.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/22/2017] [Accepted: 02/23/2017] [Indexed: 11/26/2022]
|
15
|
Bender AT, Gardberg A, Pereira A, Johnson T, Wu Y, Grenningloh R, Head J, Morandi F, Haselmayer P, Liu-Bujalski L. Ability of Bruton's Tyrosine Kinase Inhibitors to Sequester Y551 and Prevent Phosphorylation Determines Potency for Inhibition of Fc Receptor but not B-Cell Receptor Signaling. Mol Pharmacol 2017; 91:208-219. [PMID: 28062735 DOI: 10.1124/mol.116.107037] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 12/21/2016] [Indexed: 12/13/2022] Open
Abstract
Bruton's tyrosine kinase (Btk) is expressed in a variety of hematopoietic cells. Btk has been demonstrated to regulate signaling downstream of the B-cell receptor (BCR), Fc receptors (FcRs), and toll-like receptors. It has become an attractive drug target because its inhibition may provide significant efficacy by simultaneously blocking multiple disease mechanisms. Consequently, a large number of Btk inhibitors have been developed. These compounds have diverse binding modes, and both reversible and irreversible inhibitors have been developed. Reported herein, we have tested nine Btk inhibitors and characterized on a molecular level how their interactions with Btk define their ability to block different signaling pathways. By solving the crystal structures of Btk inhibitors bound to the enzyme, we discovered that the compounds can be classified by their ability to trigger sequestration of Btk residue Y551. In cells, we found that sequestration of Y551 renders it inaccessible for phosphorylation. The ability to sequester Y551 was an important determinant of potency against FcεR signaling as Y551 sequestering compounds were more potent for inhibiting basophils and mast cells. This result was true for the inhibition of FcγR signaling as well. In contrast, Y551 sequestration was less a factor in determining potency against BCR signaling. We also found that Btk activity is regulated differentially in basophils and B cells. These results elucidate important determinants for Btk inhibitor potency against different signaling pathways and provide insight for designing new compounds with a broader inhibitory profile that will likely result in greater efficacy.
Collapse
Affiliation(s)
- Andrew T Bender
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Anna Gardberg
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Albertina Pereira
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Theresa Johnson
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Yin Wu
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Roland Grenningloh
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Jared Head
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Federica Morandi
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Philipp Haselmayer
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| | - Lesley Liu-Bujalski
- TIP Immunology (A.T.B., A.P., Y.W., R.G.) and Discovery Technologies (A.G., T.J., J.H., F.M., L.L.-B.), EMD Serono Research and Development Institute, Billerica, Massachusetts; and TIP Immunology, Merck, Darmstadt, Germany (P.H.)
| |
Collapse
|
16
|
Fischer PM. Approved and Experimental Small-Molecule Oncology Kinase Inhibitor Drugs: A Mid-2016 Overview. Med Res Rev 2016; 37:314-367. [DOI: 10.1002/med.21409] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/04/2016] [Accepted: 08/09/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Peter M. Fischer
- School of Pharmacy and Centre for Biomolecular Sciences; University of Nottingham; Nottingham NG7 2RD UK
| |
Collapse
|
17
|
Lee SJ, Choi JS, Han BG, Kim HS, Song HJ, Lee J, Nam S, Goh SH, Kim JH, Koh JS, Lee BI. Crystal structures of spleen tyrosine kinase in complex with novel inhibitors: structural insights for design of anticancer drugs. FEBS J 2016; 283:3613-3625. [DOI: 10.1111/febs.13831] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/04/2016] [Accepted: 08/08/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Sang Jae Lee
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
- The Research Institute of Pharmaceutical Sciences; College of Pharmacy; Seoul National University; Korea
| | | | - Byeong-Gu Han
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
| | - Hyoun Sook Kim
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
| | | | | | - Seungyoon Nam
- Department of Life Sciences; College of BioNano Technology; Gachon University; Sungnam Korea
- Department of Genome Medicine and Science; Graduate School of Medicine; Gachon University; Incheon Korea
| | - Sung-Ho Goh
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
| | | | | | - Byung Il Lee
- Research Institute; National Cancer Center; Goyang Gyeonggi Korea
| |
Collapse
|
18
|
Bosques CJ, Manning AM. Fc-gamma receptors: Attractive targets for autoimmune drug discovery searching for intelligent therapeutic designs. Autoimmun Rev 2016; 15:1081-1088. [PMID: 27491569 DOI: 10.1016/j.autrev.2016.07.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/17/2016] [Indexed: 12/28/2022]
Abstract
Autoantibody immune complexes (ICs) mediate pathogenesis in multiple autoimmune diseases via direct interference with target function, complement fixation, and interaction with Fc-gamma receptors (FcγRs). Through high avidity interactions, ICs are able to crosslink low affinity FcγRs expressed on a wide variety of effector cells, leading to secretion of pro-inflammatory mediators and inducing cytotoxicity, ultimately resulting in tissue injury. Given their relevance in numerous autoimmune diseases, FcγRs have been considered as attractive therapeutic targets for the last three decades. However, a limited number of investigational drug candidates have been developed targeting FcγRs and only a few approved therapeutics have been associated with impacting FcγRs. This review provides a historical overview of the different therapeutic approaches used to target FcγRs for the treatment of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Carlos J Bosques
- Momenta Pharmaceuticals, 675 West Kendall Street, Cambridge, MA 02142, USA
| | - Anthony M Manning
- Momenta Pharmaceuticals, 675 West Kendall Street, Cambridge, MA 02142, USA.
| |
Collapse
|
19
|
Ma TKW, McAdoo SP, Tam FWK. Spleen Tyrosine Kinase: A Crucial Player and Potential Therapeutic Target in Renal Disease. Nephron Clin Pract 2016; 133:261-9. [PMID: 27476075 DOI: 10.1159/000446879] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/24/2016] [Indexed: 02/06/2023] Open
Abstract
Spleen tyrosine kinase (Syk), a 72 kDa cytoplasmic non-receptor protein-tyrosine kinase, plays an important role in signal transduction in a variety of cell types. Ever since its discovery in the early 1990s, there has been accumulating evidence to suggest a pathogenic role of Syk in various allergic disorders, autoimmune diseases and malignancies. Additionally, there is emerging data from both pre-clinical and clinical studies that Syk is implicated in the pathogenesis of proliferative glomerulonephritis (GN), including anti-glomerular basement membrane disease, anti-neutrophil cytoplasmic antibody-associated GN, lupus nephritis and immunoglobulin A nephropathy (IgAN). Moreover, recent animal studies have shed light on the importance of Syk in mediating acute renal allograft rejection, Epstein Barr virus-associated post-transplant lymphoproliferative disease and kidney fibrosis. Fostamatinib, an oral Syk inhibitor, has undergone clinical testing in rheumatoid arthritis, refractory immune thrombocytopenic purpura, leukemia and lymphoma. The recent STOP-IgAN trial showed that the addition of non-selective immunosuppressive therapy to intensive supportive care did not improve clinical outcomes in high-risk IgAN patients. A Syk-targeted approach may be beneficial and is currently being evaluated in a phase II randomized controlled trial. In this review, we will discuss the pathogenic role of Syk and potential use of Syk inhibitor in a variety of renal diseases.
Collapse
Affiliation(s)
- Terry King-Wing Ma
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | | | | |
Collapse
|
20
|
Allen JC, Talab F, Slupsky JR. Targeting B-cell receptor signaling in leukemia and lymphoma: how and why? Int J Hematol Oncol 2016; 5:37-53. [PMID: 30302202 DOI: 10.2217/ijh-2016-0003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/13/2016] [Indexed: 01/04/2023] Open
Abstract
B-lymphocytes are dependent on B-cell receptor (BCR) signaling for the constant maintenance of their physiological function, and in many B-cell malignancies this signaling pathway is prone to aberrant activation. This understanding has led to an ever-increasing interest in the signaling networks activated following ligation of the BCR in both normal and malignant cells, and has been critical in establishing an array of small molecule inhibitors targeting BCR-induced signaling. By dissecting how different malignancies signal through BCR, researchers are contributing to the design of more customized therapeutics which have greater efficacy and lower toxicity than previous therapies. This allows clinicians access to an array of approaches to best treat patients whose malignancies have BCR signaling as a driver of pathogenesis.
Collapse
Affiliation(s)
- John C Allen
- Department of Molecular & Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GE, UK.,Department of Molecular & Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GE, UK
| | - Fatima Talab
- Redx Oncology Plc, Duncan Building, Royal Liverpool University Hospital, Daulby Street, Liverpool, L69 3GA, UK.,Redx Oncology Plc, Duncan Building, Royal Liverpool University Hospital, Daulby Street, Liverpool, L69 3GA, UK
| | - Joseph R Slupsky
- Department of Molecular & Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK.,Department of Molecular & Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| |
Collapse
|
21
|
Yeo SC, Liew A, Barratt J. Emerging therapies in immunoglobulin A nephropathy. Nephrology (Carlton) 2016; 20:788-800. [PMID: 26032537 DOI: 10.1111/nep.12527] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2015] [Indexed: 01/29/2023]
Abstract
Despite advances in our understanding of immunoglobulin A nephropathy (IgAN) over the past decade, there are currently no specific therapies capable of targeting key pathways involved in the pathogenesis of the disease. Recent studies have, however, provided new insights into important molecular pathways that are likely to be amenable to therapeutic manipulation in the future. Specifically, a deeper understanding of the role of mucosal immunity, B-cell activation and mesangial cell activation in IgAN has provided the impetus for a number of exciting phase II/III clinical trials in IgAN. In this review, we examine some of these on-going studies, first examining studies that clarify the role of traditional immunosuppression in IgAN, then focusing on novel therapies in early clinical studies, looking closely at the rationale for these agents in relation to our current understanding of the pathogenesis of IgAN. Finally, we examine emerging pathways and therapeutic agents that have the potential to be developed as novel therapies in the coming years. It is hoped that as we continue to develop a greater understanding of IgAN, emerging therapies will soon become a reality in the day-to-day treatment of patients with IgAN.
Collapse
Affiliation(s)
- See Cheng Yeo
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore
| | - Adrian Liew
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore
| | - Jonathan Barratt
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK.,The John Walls Renal Unit, Leicester General Hospital, Leicester, UK
| |
Collapse
|
22
|
Martin P, Gillen M, Millson D, Oliver S, Brealey C, Surry D, Sweeny D, Lau D, Leese P. Effects of Fostamatinib on the Pharmacokinetics of the CYP2C8 Substrate Pioglitazone: Results From In Vitro and Phase 1 Clinical Studies. Clin Pharmacol Drug Dev 2016; 5:170-9. [PMID: 27163495 DOI: 10.1002/cpdd.243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 11/18/2016] [Accepted: 12/01/2016] [Indexed: 11/07/2022]
Abstract
Fostamatinib is a prodrug that undergoes gastrointestinal tract dephosphorylation to form the active metabolite, R406. Here we report its cytochrome P450-inducing potential. In vitro, R406 3 and 10 μM induced CYP2C8 to levels representing 53% and 75%, respectively, of the level achieved by the positive control, rifampicin. Induction of other enzymes was minor. The effect of fostamatinib (100 mg twice daily) on the pharmacokinetics of a single oral 30-mg dose of the CYP2C8 substrate pioglitazone and its metabolite, hydroxy pioglitazone, was then investigated (open-label, nonrandomized, 2-period phase I study [n = 15]). Coadministration of fostamatinib and pioglitazone (vs pioglitazone alone) was associated with lower mean maximum plasma concentration values for pioglitazone (geometric least-squares mean ratio, 82.8; 90% confidence interval, 64.2-106.8) and hydroxy pioglitazone (90.9; 78.6-105.1), an increase in pioglitazone AUC (117.8; 108.4-128.0), a decrease in hydroxy pioglitazone AUC(0-t) (89.7; 78.9-101.9), and an increase in pioglitazone geometric mean t1/2λz (9.4-12.8 hours). No tolerability concerns were identified upon coadministration. These data suggest that although clinical significance has not been formally evaluated, fostamatinib is unlikely to have a clinically significant effect on the pharmacokinetics of pioglitazone (which may be extrapolated to other CYP2C8 substrates). However, vigilance is advised should these agents be prescribed together.
Collapse
Affiliation(s)
| | | | | | | | | | | | - David Sweeny
- Rigel Pharmaceuticals, Inc, South San Francisco, CA, USA
| | - David Lau
- Rigel Pharmaceuticals, Inc, South San Francisco, CA, USA
| | | |
Collapse
|
23
|
Choi JS, Hwang HJ, Kim SW, Lee BI, Lee J, Song HJ, Koh JS, Kim JH, Lee PH. Highly potent and selective pyrazolylpyrimidines as Syk kinase inhibitors. Bioorg Med Chem Lett 2015; 25:4441-6. [PMID: 26384287 DOI: 10.1016/j.bmcl.2015.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 09/02/2015] [Accepted: 09/05/2015] [Indexed: 12/29/2022]
Abstract
A series of pyrazolylpyrimidine scaffold based Syk inhibitors were synthesized and evaluated for their biological activities and selectivity. Lead optimization efforts provided compounds with potent Syk inhibition in both enzymatic and TNF-α release assay.
Collapse
Affiliation(s)
- Jang-Sik Choi
- Department of Chemistry, Kangwon National University, Chuncheon 200-701, Republic of Korea; Oscotec Inc., 694-1 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | - Hae-Jun Hwang
- Oscotec Inc., 694-1 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | - Se-Won Kim
- Oscotec Inc., 694-1 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | - Byung Il Lee
- Biomolecular Function Research Branch, Division of Convergence Technology, Research Institute, National Cancer Center, Goyang, Gyeonggi 410-769, Republic of Korea
| | - Jaekyoo Lee
- Genosco, 767C Concord Avenue, 2nd Floor, Cambridge, MA 02138, USA
| | - Ho-Juhn Song
- Genosco, 767C Concord Avenue, 2nd Floor, Cambridge, MA 02138, USA
| | - Jong Sung Koh
- Genosco, 767C Concord Avenue, 2nd Floor, Cambridge, MA 02138, USA
| | - Jung-Ho Kim
- Oscotec Inc., 694-1 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea.
| | - Phil Ho Lee
- Department of Chemistry, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| |
Collapse
|
24
|
Salihoglu A, Ar MC, Soysal T. Novelties in the management of B-cell malignancies: B-cell receptor signaling inhibitors and lenalidomide. Expert Rev Hematol 2015; 8:765-83. [PMID: 26413907 DOI: 10.1586/17474086.2015.1091301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
B-cell lymphoproliferative disorders comprise 85% of Non-Hodgkin's lymphomas. Despite successful chemoimmunotherapy regimens, responses are not durable and the outcome is fatal in a considerable portion of patients. There is an inevitable need for less toxic and more potent therapeutic agents. Over the recent years, a plethora of agents including monoclonal antibodies, Bcl-2 antagonists, tyrosine kinase inhibitors, cyclin-dependent kinase inhibitors, mTOR inhibitors and immunomodulatory drugs have been developed in B-cell malignancies. The aim of this paper is to focus on B-cell receptor signaling inhibitors and lenalidomide as an immunomodulatory drug and to provide insight on how and when to incorporate these agents into the treatment algorithms.
Collapse
Affiliation(s)
- Ayse Salihoglu
- a Department of Internal Medicine, Division of Haematology, Istanbul University, Cerrahpasa Medical Faculty, Istanbul, Turkey
| | - Muhlis Cem Ar
- a Department of Internal Medicine, Division of Haematology, Istanbul University, Cerrahpasa Medical Faculty, Istanbul, Turkey
| | | |
Collapse
|
25
|
Liu C, Lin J, Hynes J, Wu H, Wrobleski ST, Lin S, Dhar TGM, Vrudhula VM, Sun JH, Chao S, Zhao R, Wang B, Chen BC, Everlof G, Gesenberg C, Zhang H, Marathe PH, McIntyre KW, Taylor TL, Gillooly K, Shuster DJ, McKinnon M, Dodd JH, Barrish JC, Schieven GL, Leftheris K. Discovery of ((4-(5-(Cyclopropylcarbamoyl)-2-methylphenylamino)-5-methylpyrrolo[1,2-f][1,2,4]triazine-6-carbonyl)(propyl)carbamoyloxy)methyl-2-(4-(phosphonooxy)phenyl)acetate (BMS-751324), a Clinical Prodrug of p38α MAP Kinase Inhibitor. J Med Chem 2015; 58:7775-84. [PMID: 26359680 DOI: 10.1021/acs.jmedchem.5b00839] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In search for prodrugs to address the issue of pH-dependent solubility and exposure associated with 1 (BMS-582949), a previously disclosed phase II clinical p38α MAP kinase inhibitor, a structurally novel clinical prodrug, 2 (BMS-751324), featuring a carbamoylmethylene linked promoiety containing hydroxyphenyl acetic acid (HPA) derived ester and phosphate functionalities, was identified. Prodrug 2 was not only stable but also water-soluble under both acidic and neutral conditions. It was effectively bioconverted into parent drug 1 in vivo by alkaline phosphatase and esterase in a stepwise manner, providing higher exposure of 1 compared to its direct administration, especially within higher dose ranges. In a rat LPS-induced TNFα pharmacodynamic model and a rat adjuvant arthritis model, 2 demonstrated similar efficacy to 1. Most importantly, it was shown in clinical studies that prodrug 2 was indeed effective in addressing the pH-dependent absorption issue associated with 1.
Collapse
Affiliation(s)
- Chunjian Liu
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - James Lin
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - John Hynes
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Hong Wu
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Stephen T Wrobleski
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Shuqun Lin
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - T G Murali Dhar
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Vivekananda M Vrudhula
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Jung-Hui Sun
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Sam Chao
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Rulin Zhao
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Bei Wang
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Bang-Chi Chen
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Gerry Everlof
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Christoph Gesenberg
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Hongjian Zhang
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Punit H Marathe
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Kim W McIntyre
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Tracy L Taylor
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Kathleen Gillooly
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - David J Shuster
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Murray McKinnon
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - John H Dodd
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Joel C Barrish
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Gary L Schieven
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| | - Katerina Leftheris
- Research and Development, Bristol-Myers Squibb , Princeton, New Jersey 08543, United States
| |
Collapse
|
26
|
Ryan J, Ma FY, Han Y, Ozols E, Kanellis J, Tesch GH, Nikolic-Paterson DJ. Myeloid cell-mediated renal injury in rapidly progressive glomerulonephritis depends upon spleen tyrosine kinase. J Pathol 2015; 238:10-20. [PMID: 26251216 DOI: 10.1002/path.4598] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/16/2015] [Accepted: 08/01/2015] [Indexed: 12/29/2022]
Abstract
Antibody-dependent activation of myeloid cells within the glomerulus plays a central role in rapidly progressive forms of glomerulonephritis. The spleen tyrosine kinase (Syk) is expressed by all leukocytes, except mature T cells, and is required for signalling via the B-cell receptor, Fc receptors, and some integrins. Syk has been proposed as a therapeutic target in glomerulonephritis. However, little is known of Syk activation in human kidney disease, while studies in experimental glomerulonephritis using non-selective Syk inhibitors require validation via conditional gene deletion. The current study addressed both of these important points. Syk activation (Tyr(525/526) phosphorylation) was examined in a cohort of 96 patients with different glomerulonephritides. Syk activation was evident in infiltrating leukocytes, mainly neutrophils and macrophages, in 36/40 cases of rapidly progressive glomerulonephritis. In contrast, non-proliferative diseases showed little or no Syk activation. Glomerular and interstitial cells exhibiting Syk activation correlated with renal function and systemic inflammation. Next, we examined mice with conditional Syk gene deletion in myeloid cells (Syk(My) ) versus Syk(f/f) littermate controls in nephrotoxic serum nephritis - a model of rapidly progressive glomerulonephritis. Control Syk(f/f) mice featured a transient neutrophil influx at 3 h and severe disease on day 9 of nephrotoxic serum nephritis, with crescent formation, macrophage infiltration, inflammation, kidney fibrosis, and renal dysfunction. In contrast, Syk(My) mice had significantly reduced neutrophil and macrophage infiltration despite equivalent glomerular deposition of humoral reactants. Syk(My) mice exhibited reduced crescent formation, inflammation, and fibrosis, with improved renal function on day 9 of nephrotoxic serum nephritis. In conclusion, Syk activation is prominent in infiltrating myeloid cells in human rapidly progressive glomerulonephritis, and functional studies demonstrate that Syk deletion in myeloid cells is protective in mouse nephrotoxic serum nephritis.
Collapse
Affiliation(s)
- Jessica Ryan
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria, Australia.,Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Frank Y Ma
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria, Australia.,Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Yingjie Han
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria, Australia.,Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Elyce Ozols
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria, Australia.,Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - John Kanellis
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria, Australia.,Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Greg H Tesch
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria, Australia.,Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria, Australia.,Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| |
Collapse
|
27
|
Witkowska M, Smolewski P. Emerging immunotherapy and strategies directly targeting B cells for the treatment of diffuse large B-cell lymphoma. Immunotherapy 2015; 7:37-46. [PMID: 25572478 DOI: 10.2217/imt.14.93] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
During the last decade, significant prolonged survival in diffusive large B-cell lymphoma (DLBCL) has been observed. The efficacy of initial treatment improved mostly due to addition of a chimeric anti-CD20 monoclonal antibody (rituximab) to standard chemotherapeutic regimens. Moreover, accurate understanding of DLBCL pathogenesis and remarkable progress in gene expression profiling have led to the development of a variety of tumor-specific regimens. Novel agents target directly the pathways involved in signal transduction, lead to apoptosis and cancer cells differentiation. In this article, we mainly focus on new treatment options, such as monoclonal antibodies, tyrosine kinase inhibitors and immunomodulatory drugs, currently investigated in aggressive B-cell lymphoma with particular attention to DLBCL type.
Collapse
Affiliation(s)
- Magdalena Witkowska
- Department of Experimental Hematology, Medical University of Lodz, Poland Copernicus Memorial Hospital, 93-510 Lodz, Ciołkowskiego 2, Poland
| | | |
Collapse
|
28
|
Loss of B-cell Receptor Expression Defines a Subset of Diffuse Large B-cell Lymphoma Characterized by Silent BCR/PI3K/AKT Signaling and a Germinal Center Phenotype Displaying Low-risk Clinicopathologic Features. Am J Surg Pathol 2015; 39:902-11. [DOI: 10.1097/pas.0000000000000396] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Zhang L, Liu W, Mao F, Zhu J, Dong G, Jiang H, Sheng C, Miao L, Huang L, Li J. Discovery of Benzylidene Derivatives as Potent Syk Inhibitors: Synthesis, SAR Analysis, and Biological Evaluation. Arch Pharm (Weinheim) 2015; 348:463-74. [PMID: 26032727 DOI: 10.1002/ardp.201500096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Lingling Zhang
- Shanghai Key Laboratory of New Drug Design; School of Pharmacy; East China University of Science and Technology; Shanghai China
| | - Wei Liu
- Department of Orthopaedic; The First Affiliated Hospital of Soochow University; Suzhou China
| | - Fei Mao
- Shanghai Key Laboratory of New Drug Design; School of Pharmacy; East China University of Science and Technology; Shanghai China
| | - Jin Zhu
- Shanghai Key Laboratory of New Drug Design; School of Pharmacy; East China University of Science and Technology; Shanghai China
| | - Guoqiang Dong
- School of Pharmacy; Second Military Medical University; Shanghai China
| | - Hualiang Jiang
- Shanghai Key Laboratory of New Drug Design; School of Pharmacy; East China University of Science and Technology; Shanghai China
| | - Chunquan Sheng
- School of Pharmacy; Second Military Medical University; Shanghai China
| | - Liyan Miao
- Department of Clinical Pharmacology Research Laboratory; The First Affiliated Hospital of Soochow University; Suzhou China
| | - Lixin Huang
- Department of Orthopaedic; The First Affiliated Hospital of Soochow University; Suzhou China
| | - Jian Li
- Shanghai Key Laboratory of New Drug Design; School of Pharmacy; East China University of Science and Technology; Shanghai China
| |
Collapse
|
30
|
Taylor PC, Genovese MC, Greenwood M, Ho M, Nasonov E, Oemar B, Stoilov R, Vencovsky J, Weinblatt M. OSKIRA-4: a phase IIb randomised, placebo-controlled study of the efficacy and safety of fostamatinib monotherapy. Ann Rheum Dis 2014; 74:2123-9. [DOI: 10.1136/annrheumdis-2014-205361] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 07/11/2014] [Indexed: 11/04/2022]
Abstract
ObjectivesOSKIRA-4 evaluated the efficacy of fostamatinib monotherapy versus placebo on the signs and symptoms of rheumatoid arthritis over 6 weeks by Disease Activity Score C reactive protein (DAS-28(CRP)) and assessed non-inferiority to adalimumab monotherapy at Week 24 by DAS-28(CRP).MethodsOverall, 279 patients not currently taking disease-modifying antirheumatic drugs were randomised to: (A) fostamatinib 100 mg twice daily for 24 weeks plus placebo injection every 2 weeks (PBOI); (B) fostamatinib 100 mg twice daily for 4 weeks, then 150 mg once daily up to Week 24, plus PBOI; (C) fostamatinib 100 mg twice daily for 4 weeks, then 100 mg once daily up to Week 24, plus PBOI; (D) adalimumab 40 mg every 2 weeks for 24 weeks, plus oral placebo twice daily; or (E) oral placebo twice daily for 6 weeks, plus PBOI, then a switch to arm A or B.ResultsFostamatinib demonstrated a significant improvement in DAS-28(CRP) score from baseline versus placebo at Week 6 for arms A and B, but not C. Fostamatinib was significantly less effective than adalimumab at Week 24 based on DAS-28(CRP). Adverse events observed with fostamatinib treatment were consistent with those reported in previous studies, including hypertension and diarrhoea.ConclusionsFostamatinib demonstrated efficacy as monotherapy, showing superior DAS-28(CRP) score changes between baseline and 6 weeks when compared with placebo in treatment arms A and B. However, all fostamatinib regimens demonstrated inferior responses compared with adalimumab at Week 24.Trial registration numberClinicaltrials.gov: NCT01264770.
Collapse
|
31
|
Currie KS, Kropf JE, Lee T, Blomgren P, Xu J, Zhao Z, Gallion S, Whitney JA, Maclin D, Lansdon EB, Maciejewski P, Rossi AM, Rong H, Macaluso J, Barbosa J, Di Paolo JA, Mitchell SA. Discovery of GS-9973, a selective and orally efficacious inhibitor of spleen tyrosine kinase. J Med Chem 2014; 57:3856-73. [PMID: 24779514 DOI: 10.1021/jm500228a] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Spleen tyrosine kinase (Syk) is an attractive drug target in autoimmune, inflammatory, and oncology disease indications. The most advanced Syk inhibitor, R406, 1 (or its prodrug form fostamatinib, 2), has shown efficacy in multiple therapeutic indications, but its clinical progress has been hampered by dose-limiting adverse effects that have been attributed, at least in part, to the off-target activities of 1. It is expected that a more selective Syk inhibitor would provide a greater therapeutic window. Herein we report the discovery and optimization of a novel series of imidazo[1,2-a]pyrazine Syk inhibitors. This work culminated in the identification of GS-9973, 68, a highly selective and orally efficacious Syk inhibitor which is currently undergoing clinical evaluation for autoimmune and oncology indications.
Collapse
Affiliation(s)
- Kevin S Currie
- Department of Chemistry, ‡Department of Biology, and §Department of Drug Metabolism, Gilead Sciences, Inc. , Branford, Connecticut 06405, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Accelerated therapeutic progress in diffuse large B cell lymphoma. Ann Hematol 2014; 93:541-56. [PMID: 24375125 DOI: 10.1007/s00277-013-1979-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/27/2013] [Indexed: 02/07/2023]
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma in the world. Clinically, biologically, and pathologically, DLBCL is a heterogeneous entity with a range of potential outcomes. Immunochemotherapy regimens, consisting of the chimeric monoclonal anti-CD20 antibody rituximab in combination with chemotherapy, have improved the outcomes. Relapsed DLBCL is generally treated with salvage immunochemotherapy followed by high-dose therapy and autologous stem cell transplantation; however, DLBCL is not yet curable in up to a third of patients. The real promise for cure lies in novel agents and their rational combinations. The improved understanding of DLBCL subtypes and gene expression profiling has led to the identification of targeted drugs that may allow for subtype specific therapy. We have summarized the existing data on the prognostic factors and the treatment of DLBCL, including the use of novel agents such as lenalidomide, carfilzomib, and ibrutinib. We also share our thoughts on the direction of future clinical trials.
Collapse
|
33
|
Lucas MC, Bhagirath N, Chiao E, Goldstein DM, Hermann JC, Hsu PY, Kirchner S, Kennedy-Smith JJ, Kuglstatter A, Lukacs C, Menke J, Niu L, Padilla F, Peng Y, Polonchuk L, Railkar A, Slade M, Soth M, Xu D, Yadava P, Yee C, Zhou M, Liao C. Using ovality to predict nonmutagenic, orally efficacious pyridazine amides as cell specific spleen tyrosine kinase inhibitors. J Med Chem 2014; 57:2683-91. [PMID: 24520947 DOI: 10.1021/jm401982j] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Inhibition of spleen tyrosine kinase has attracted much attention as a mechanism for the treatment of cancers and autoimmune diseases such as asthma, rheumatoid arthritis, and systemic lupus erythematous. We report the structure-guided optimization of pyridazine amide spleen tyrosine kinase inhibitors. Early representatives of this scaffold were highly potent and selective but mutagenic in an Ames assay. An approach that led to the successful identification of nonmutagenic examples, as well as further optimization to compounds with reduced cardiovascular liabilities is described. Select pharmacokinetic and in vivo efficacy data are presented.
Collapse
Affiliation(s)
- Matthew C Lucas
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, Small Molecule Research, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Norman P. Spleen tyrosine kinase inhibitors: a review of the patent literature 2010 - 2013. Expert Opin Ther Pat 2014; 24:573-95. [PMID: 24555683 DOI: 10.1517/13543776.2014.890184] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The non-receptor tyrosine kinase, spleen tyrosine kinase (Syk), is primarily expressed in haematopoietic cells and appears to be particularly important in B cells. Syk is involved in signal transduction processes and appears to regulate allergic, inflammatory and autoimmune responses. It also appears to play a significant role in the development of haematological malignancies. Inhibitors of Syk are potentially useful in treating asthma, rheumatoid arthritis, lupus, chronic lymphocytic leukaemia and lymphomas. AREAS COVERED This article reviews the increasing number of patent filings between 2010 and 2013 claiming Syk inhibitors and focuses on the multiple structural classes of Syk inhibitors disclosed. It also comments on recent developments with Syk inhibitors, both clinical results and licensing deals. EXPERT OPINION The increased interest in the identification of Syk inhibitors has seen a sharp increase in patent filings claiming such compounds. However, the number of these is well below that of filings relating to other pro-inflammatory kinases (p38, JAK). These filings have also claimed an increasingly diverse range of chemical classes moving away from the 2,4-diaminopyrimidine motif present in drugs such as fostamatinib and PRT-06207. Many of the claimed compounds are Syk inhibitors with potencies considerably better than fostamatinib. However, good kinase selectivity is also likely to be essential if a Syk inhibitor is to prove useful enough to emulate the JAK inhibitor tofacitinib in gaining marketing authorisation. Recent clinical failures with Syk inhibitors are expected to result in a decrease in the rate of patent filings claiming Syk inhibitors.
Collapse
Affiliation(s)
- Peter Norman
- Norman Consulting , 18 Pink Lane, Burnham, Bucks, SL1 8JW , UK
| |
Collapse
|
35
|
Padilla F, Bhagirath N, Chen S, Chiao E, Goldstein DM, Hermann JC, Hsu J, Kennedy-Smith JJ, Kuglstatter A, Liao C, Liu W, Lowrie LE, Luk KC, Lynch SM, Menke J, Niu L, Owens TD, O-Yang C, Railkar A, Schoenfeld RC, Slade M, Steiner S, Tan YC, Villaseñor AG, Wang C, Wanner J, Xie W, Xu D, Zhang X, Zhou M, Lucas MC. Pyrrolopyrazines as Selective Spleen Tyrosine Kinase Inhibitors. J Med Chem 2013; 56:1677-92. [DOI: 10.1021/jm301720p] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Fernando Padilla
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Niala Bhagirath
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Shaoqing Chen
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Eric Chiao
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - David M. Goldstein
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Johannes C. Hermann
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Jonathan Hsu
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Joshua J. Kennedy-Smith
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Andreas Kuglstatter
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Cheng Liao
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Wenjian Liu
- BioDuro Beijing Co. Ltd., Building E, No.
29, Life Science Park Road, Changping District, Beijing 102206, P.R.
China
| | - Lee E. Lowrie
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Kin Chun Luk
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Stephen M. Lynch
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - John Menke
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Linghao Niu
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Timothy D. Owens
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Counde O-Yang
- BioDuro Beijing Co. Ltd., Building E, No.
29, Life Science Park Road, Changping District, Beijing 102206, P.R.
China
| | - Aruna Railkar
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Ryan C. Schoenfeld
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Michelle Slade
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Sandra Steiner
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Yun-Chou Tan
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Armando G. Villaseñor
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Ce Wang
- BioDuro Beijing Co. Ltd., Building E, No.
29, Life Science Park Road, Changping District, Beijing 102206, P.R.
China
| | - Jutta Wanner
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Wenwei Xie
- BioDuro Beijing Co. Ltd., Building E, No.
29, Life Science Park Road, Changping District, Beijing 102206, P.R.
China
| | - Daigen Xu
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Xiaohu Zhang
- BioDuro Beijing Co. Ltd., Building E, No.
29, Life Science Park Road, Changping District, Beijing 102206, P.R.
China
| | - Mingyan Zhou
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Matthew C. Lucas
- Hoffmann-La Roche Inc., pRED, Pharma Research & Early Development, 340 Kingsland Street, Nutley, New Jersey 07110, United States
| |
Collapse
|
36
|
Lucas MC, Goldstein DM, Hermann JC, Kuglstatter A, Liu W, Luk KC, Padilla F, Slade M, Villaseñor AG, Wanner J, Xie W, Zhang X, Liao C. Rational design of highly selective spleen tyrosine kinase inhibitors. J Med Chem 2012; 55:10414-23. [PMID: 23151054 DOI: 10.1021/jm301367c] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A novel approach to design selective spleen tyrosine kinase (Syk) inhibitors is described. Inhibition of spleen tyrosine kinase has attracted much attention as a mechanism for the treatment of autoimmune diseases such as asthma, rheumatoid arthritis, and SLE. Fostamatinib, a Syk inhibitor that successfully completed phase II clinical trials, also exhibits some undesirable side effects. More selective Syk inhibitors could offer safer, alternative treatments. Through a systematic evaluation of the kinome, we identified Pro455 and Asn457 in the Syk ATP binding site as a rare combination among sequence aligned kinases and hypothesized that optimizing the interaction between them and a Syk inhibitor molecule would impart high selectivity for Syk over other kinases. We report the structure-guided identification of three series of selective spleen tyrosine kinase inhibitors that support our hypothesis and offer useful guidance to other researchers in the field.
Collapse
Affiliation(s)
- Matthew C Lucas
- Small Molecule Research, Discovery Chemistry, pRED, Pharma Research and Early Development, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kim MJ, McDaid JP, McAdoo SP, Barratt J, Molyneux K, Masuda ES, Pusey CD, Tam FWK. Spleen tyrosine kinase is important in the production of proinflammatory cytokines and cell proliferation in human mesangial cells following stimulation with IgA1 isolated from IgA nephropathy patients. THE JOURNAL OF IMMUNOLOGY 2012; 189:3751-8. [PMID: 22956578 DOI: 10.4049/jimmunol.1102603] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
IgA immune complexes are capable of inducing human mesangial cell (HMC) activation, resulting in release of proinflammatory and profibrogenic mediators. The subsequent inflammation, cellular proliferation, and synthesis of extracellular matrix lead to the progression of IgA nephropathy (IgAN). Spleen tyrosine kinase (SYK) is an intracellular protein tyrosine kinase involved in cell signaling downstream of immunoreceptors. In this study, we determined whether SYK is involved in the downstream signaling of IgA1 stimulation in HMC, leading to production of proinflammatory cytokines/chemokines and cell proliferation. Incubation of HMC with IgA1 purified from IgAN patients significantly increased the synthesis of MCP-1 in a dose-dependent manner. There was also significantly increased production of IL-6, IL-8, IFN-γ-inducible protein-10, RANTES, and platelet-derived growth factor-BB. Stimulation of HMC with heat-aggregated IgA1 purified from IgAN patients induced significantly increased HMC proliferation. Both pharmacological inhibition of SYK and knockdown of SYK by small interfering RNA significantly reduced the synthesis of these mediators and inhibited HMC proliferation. Moreover, positive immunostaining for total and phospho-SYK in glomeruli of kidney biopsies from IgAN patients strongly suggests the involvement of SYK in the pathogenesis of IgAN. To our knowledge, we demonstrate, for the first time, the involvement of SYK in the downstream signaling of IgA1 stimulation in HMC and in the pathogenesis of IgAN. Hence, SYK represents a potential therapeutic target for IgAN.
Collapse
Affiliation(s)
- Min Jeong Kim
- Imperial College Kidney and Transplant Institute, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|