151
|
Garnock-Jones KP. Nilotinib: in the first-line treatment of newly diagnosed Philadelphia chromosome-positive chronic myeloid leukaemia in chronic phase. Drugs 2011; 71:1579-90. [PMID: 21861543 DOI: 10.2165/11207770-000000000-00000] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Nilotinib is an effective first-line treatment for newly diagnosed Philadelphia chromosome-positive chronic myeloid leukaemia (CML) in chronic phase. It is an aminopyrimidine-based, high-affinity inhibitor of the tyrosine kinase activity of BCR-ABL. It thus decreases ABL-associated cell proliferation and kinase autophosphorylation. At 12 months, a significantly greater proportion of nilotinib 300 mg twice daily recipients experienced a major molecular response (primary endpoint) than those receiving imatinib 400 mg once daily, in the randomized, open-label, multicentre ENESTnd study in adults with newly diagnosed Philadelphia chromosome-positive CML in chronic phase. Moreover, a significantly greater proportion of nilotinib 300 mg twice daily than imatinib recipients had a complete molecular response at 12 months. Complete cytogenetic response rates were also significantly higher in the nilotinib 300 mg twice daily group than in the imatinib group at 12 months. Treatment differences in molecular response rates remained significant in an updated analysis, with data from a minimum follow-up of 24 months. Nilotinib 300 mg twice daily was generally well tolerated in the ENESTnd study. While nilotinib is associated with an increase in corrected QT interval (QTc), the incidence of cardiac-related adverse events in nilotinib recipients in the ENESTnd study was low.
Collapse
|
152
|
Brown RL. Tyrosine kinase inhibitor-induced hypothyroidism: incidence, etiology, and management. Target Oncol 2011; 6:217-26. [DOI: 10.1007/s11523-011-0197-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 10/19/2011] [Indexed: 11/29/2022]
|
153
|
Abstract
PURPOSE OF REVIEW To review recent developments in the molecular pathogenesis of tenosynovial giant cell tumor (TGCT) or pigmented villonodular synovitis (PVNS) and its therapeutic implications. RECENT FINDINGS TGCT or PVNS is a benign clonal neoplastic proliferation arising from the synovium characterized by a minor population of intratumoral cells that harbor a recurrent translocation. These cells overexpress CSF1, resulting in recruitment of CSF1R-bearing macrophages that are polyclonal and make up the bulk of the tumor. Inhibition of CSF1R using small molecule inhibitors such as imatinib, nilotinib or sunitinib can result in clinical, radiological and functional improvement in the affected joint. SUMMARY Currently, surgery remains the treatment of choice for patients with TGCT/PVNS. Localized TGCT/PVNS is managed by marginal excision. Recurrences occur in 8-20% of patients and are easily managed by re-excision. Diffuse TGCT/PVNS tends to recur more often (33-50%) and has a much more aggressive clinical course. Patients are often symptomatic and require multiple surgical procedures during their lifetime. For patients with unresectable disease or multiple recurrences, systemic therapy using CSF1R inhibitors may help delay or avoid surgical procedures and improve functional outcomes.
Collapse
|
154
|
Valent P, Gastl G, Geissler K, Greil R, Hantschel O, Lang A, Linkesch W, Lion T, Petzer AL, Pittermann E, Pleyer L, Thaler J, Wolf D. Nilotinib as frontline and second-line therapy in chronic myeloid leukemia: open questions. Crit Rev Oncol Hematol 2011; 82:370-7. [PMID: 21903413 DOI: 10.1016/j.critrevonc.2011.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 07/15/2011] [Accepted: 08/11/2011] [Indexed: 01/13/2023] Open
Abstract
Nilotinib is a second generation ABL tyrosine kinase inhibitor (TKI) that exerts major anti-leukemic effects in newly diagnosed patients with chronic myeloid leukemia (CML) as well as in most patients with imatinib-resistant CML. In freshly diagnosed patients, the anti-leukemic activity of nilotinib exceeds the efficacy of imatinib, and although long-term data for nilotinib are not available yet, the drug has recently been approved for firstline treatment of chronic phase CML in various countries. Still however, several questions concerning the optimal dose, follow-up parameters, long-term safety, and patient selection remain open. Likewise, it remains uncertain whether both Sokal low-risk and high-risk patients should receive nilotinib as frontline therapy in the future. Another question is whether nilotinib can completely eradicate CML in a subset of patients. Furthermore, it remains unclear whether and what comorbidity must be regarded as relative or absolute contra-indication for this TKI. To discuss these issues, the Austrian CML Working Group organized a series of meetings in 2010. In the current article, the outcomes from these discussions are summarized and presented together with recommendations for frontline use of TKIs in various groups of patients with CML. These recommendations should assist in daily practice as well as in the preparation and conduct of clinical trials.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Haematology & Haemostaseology, Medical University of Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Li YY, An J, Jones SJM. A computational approach to finding novel targets for existing drugs. PLoS Comput Biol 2011; 7:e1002139. [PMID: 21909252 PMCID: PMC3164726 DOI: 10.1371/journal.pcbi.1002139] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/14/2011] [Indexed: 01/08/2023] Open
Abstract
Repositioning existing drugs for new therapeutic uses is an efficient approach to drug discovery. We have developed a computational drug repositioning pipeline to perform large-scale molecular docking of small molecule drugs against protein drug targets, in order to map the drug-target interaction space and find novel interactions. Our method emphasizes removing false positive interaction predictions using criteria from known interaction docking, consensus scoring, and specificity. In all, our database contains 252 human protein drug targets that we classify as reliable-for-docking as well as 4621 approved and experimental small molecule drugs from DrugBank. These were cross-docked, then filtered through stringent scoring criteria to select top drug-target interactions. In particular, we used MAPK14 and the kinase inhibitor BIM-8 as examples where our stringent thresholds enriched the predicted drug-target interactions with known interactions up to 20 times compared to standard score thresholds. We validated nilotinib as a potent MAPK14 inhibitor in vitro (IC50 40 nM), suggesting a potential use for this drug in treating inflammatory diseases. The published literature indicated experimental evidence for 31 of the top predicted interactions, highlighting the promising nature of our approach. Novel interactions discovered may lead to the drug being repositioned as a therapeutic treatment for its off-target's associated disease, added insight into the drug's mechanism of action, and added insight into the drug's side effects.
Collapse
Affiliation(s)
- Yvonne Y Li
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | | | | |
Collapse
|
156
|
O'Sullivan S, Lin JM, Watson M, Callon K, Tong PC, Naot D, Horne A, Aati O, Porteous F, Gamble G, Cornish J, Browett P, Grey A. The skeletal effects of the tyrosine kinase inhibitor nilotinib. Bone 2011; 49:281-9. [PMID: 21550432 DOI: 10.1016/j.bone.2011.04.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 04/13/2011] [Accepted: 04/17/2011] [Indexed: 10/18/2022]
Abstract
Nilotinib is a tyrosine kinase inhibitor (TKI) developed to manage imatinib-resistance in patients with chronic myeloid leukemia (CML). It inhibits similar molecular targets to imatinib, but is a significantly more potent inhibitor of Bcr-Abl. Nilotinib exhibits off-target effects in other tissues, and of relevance to bone metabolism, hypophosphataemia has been reported in up to 30% of patients receiving nilotinib. We have assessed the effects of nilotinib on bone cells in vitro and on bone metabolism in patients receiving nilotinib for treatment of CML. We firstly investigated the effects of nilotinib on proliferating and differentiating osteoblastic cells, and on osteoclastogenesis in murine bone marrow cultures and RAW264.7 cells. Nilotinib potently inhibited osteoblast proliferation (0.01-1uM), through inhibition of the platelet-derived growth factor (PDGFR). There was a biphasic effect on osteoblast differentiation such that it was reduced by lower concentrations of nilotinib (0.1-0.5uM), with no effect at higher concentrations (1uM). Nilotinib also potently inhibited osteoclastogenesis, predominantly by stromal-cell dependent mechanisms. Thus, nilotinib decreased osteoclast development in murine bone marrow cultures, but did not affect osteoclastogenesis in RAW264.7 cells. Nilotinib treatment of osteoblastic cells increased expression and secretion of OPG and decreased expression of RANKL. In 10 patients receiving nilotinib, levels of bone turnover markers were in the low-normal range, despite secondary hyperparathyroidism, findings that are similar to those in patients treated with imatinib. Bone density tended to be higher than age and gender-matched normal values. These data suggest that nilotinib may have important effects on bone metabolism. Prospective studies should be conducted to determine the long-term effects of nilotinib on bone density and calcium metabolism.
Collapse
Affiliation(s)
- Susannah O'Sullivan
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Aichberger KJ, Herndlhofer S, Schernthaner GH, Schillinger M, Mitterbauer-Hohendanner G, Sillaber C, Valent P. Progressive peripheral arterial occlusive disease and other vascular events during nilotinib therapy in CML. Am J Hematol 2011; 86:533-9. [PMID: 21538470 DOI: 10.1002/ajh.22037] [Citation(s) in RCA: 201] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 03/17/2011] [Indexed: 12/16/2022]
Abstract
The second generation BCR/ABL kinase inhibitor nilotinib is increasingly used for the treatment of imatinib-resistant chronic myeloid leukemia (CML). So far, nilotinib is considered a well-tolerated drug with little if any side effects, although an increase in the fasting glucose level has been reported. We examined a series of 24 consecutive CML patients treated with nilotinib in our center for the development of non-hematologic adverse events. Three of these 24 CML patients developed a rapidly progressive peripheral arterial occlusive disease (PAOD) during treatment with nilotinib. In all three cases, PAOD required repeated angioplasty and/or multiple surgeries within a few months. No PAOD was known before nilotinib-therapy in these patients, although all three had received imatinib. In two patients, pre-existing risk factors predisposing for PAOD were known, and one of them had developed diabetes mellitus during nilotinib. In the other 21 patients treated with nilotinib in our center, one less severe PAOD, one myocardial infarction, one spinal infarction, one subdural hematoma, and one sudden death of unknown etiology were recorded. In summary, treatment with nilotinib may be associated with an increased risk of vascular adverse events, including PAOD development. In a subgroup of patients, these events are severe or even life-threatening. Although the exact mechanisms remain unknown, we recommend screening for pre-existing PAOD and for vascular risk factors such as diabetes mellitus in all patients before starting nilotinib and in the follow up during nilotinib-therapy.
Collapse
MESH Headings
- Adult
- Aged
- Benzamides
- Blood Glucose/metabolism
- Cohort Studies
- Constriction, Pathologic/blood
- Constriction, Pathologic/chemically induced
- Constriction, Pathologic/surgery
- Drug Resistance, Neoplasm/drug effects
- Fasting/blood
- Female
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Male
- Middle Aged
- Peripheral Arterial Disease/blood
- Peripheral Arterial Disease/chemically induced
- Peripheral Arterial Disease/surgery
- Piperazines/administration & dosage
- Piperazines/adverse effects
- Protein Kinase Inhibitors/administration & dosage
- Protein Kinase Inhibitors/adverse effects
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Pyrimidines/administration & dosage
- Pyrimidines/adverse effects
Collapse
Affiliation(s)
- Karl J Aichberger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
158
|
Hammerman PS, Sos ML, Ramos AH, Xu C, Dutt A, Zhou W, Brace LE, Woods BA, Lin W, Zhang J, Deng X, Lim SM, Heynck S, Peifer M, Simard JR, Lawrence MS, Onofrio RC, Salvesen HB, Seidel D, Zander T, Heuckmann JM, Soltermann A, Moch H, Koker M, Leenders F, Gabler F, Querings S, Ansén S, Brambilla E, Brambilla C, Lorimier P, Brustugun OT, Helland Å, Petersen I, Clement JH, Groen H, Timens W, Sietsma H, Stoelben E, Wolf J, Beer DG, Tsao MS, Hanna M, Hatton C, Eck MJ, Janne PA, Johnson BE, Winckler W, Greulich H, Bass AJ, Cho J, Rauh D, Gray NS, Wong KK, Haura EB, Thomas RK, Meyerson M. Mutations in the DDR2 kinase gene identify a novel therapeutic target in squamous cell lung cancer. Cancer Discov 2011; 1:78-89. [PMID: 22328973 PMCID: PMC3274752 DOI: 10.1158/2159-8274.cd-11-0005] [Citation(s) in RCA: 369] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
UNLABELLED While genomically targeted therapies have improved outcomes for patients with lung adenocarcinoma, little is known about the genomic alterations which drive squamous cell lung cancer. Sanger sequencing of the tyrosine kinome identified mutations in the DDR2 kinase gene in 3.8% of squamous cell lung cancers and cell lines. Squamous lung cancer cell lines harboring DDR2 mutations were selectively killed by knock-down of DDR2 by RNAi or by treatment with the multi-targeted kinase inhibitor dasatinib. Tumors established from a DDR2 mutant cell line were sensitive to dasatinib in xenograft models. Expression of mutated DDR2 led to cellular transformation which was blocked by dasatinib. A squamous cell lung cancer patient with a response to dasatinib and erlotinib treatment harbored a DDR2 kinase domain mutation. These data suggest that gain-of-function mutations in DDR2 are important oncogenic events and are amenable to therapy with dasatinib. As dasatinib is already approved for use, these findings could be rapidly translated into clinical trials. SIGNIFICANCE DDR2 mutations are present in 4% of lung SCCs, and DDR2 mutations are associated with sensitivity to dasatinib. These findings provide a rationale for designing clinical trials with the FDA-approved drug dasatinib in patients with lung SCCs.
Collapse
Affiliation(s)
- Peter S Hammerman
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Martin L Sos
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
- Department I of Internal Medicine and Laboratory of Translational Cancer Genomics, Center of Integrated Oncology Köln – Bonn, University of Köln, Köln, Germany
| | | | - Chunxiao Xu
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Amit Dutt
- Broad Institute, Cambridge, Massachusetts, USA
| | - Wenjun Zhou
- Department of Biological Chemistry and Molecular Pharmacology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Lear E Brace
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Brittany A Woods
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Wenchu Lin
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jianming Zhang
- Department of Biological Chemistry and Molecular Pharmacology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Xianming Deng
- Department of Biological Chemistry and Molecular Pharmacology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sang Min Lim
- Department of Biological Chemistry and Molecular Pharmacology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Stefanie Heynck
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
| | - Martin Peifer
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
| | - Jeffrey R Simard
- Chemical Genomics Centre of the Max Planck Society, Otto-Hahn-Strasse 15, D-44227 Dortmund, Germany
| | | | | | - Helga B Salvesen
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021 Bergen, Norway
- Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway
| | - Danila Seidel
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
| | - Thomas Zander
- Department I of Internal Medicine and Laboratory of Translational Cancer Genomics, Center of Integrated Oncology Köln – Bonn, University of Köln, Köln, Germany
- Department I for Internal Medicine, Center for Integrated Oncology Köln-Bonn, University Hospital of Cologne, Germany
| | - Johannes M Heuckmann
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
| | | | | | - Mirjam Koker
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
| | - Frauke Leenders
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
| | - Franziska Gabler
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
| | - Silvia Querings
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
| | - Sascha Ansén
- Department I for Internal Medicine, Center for Integrated Oncology Köln-Bonn, University Hospital of Cologne, Germany
| | - Elisabeth Brambilla
- Institut Albert Bonniot INSERM U823; Université Joseph Fourier Grenoble France
| | - Christian Brambilla
- Institut Albert Bonniot INSERM U823; Université Joseph Fourier Grenoble France
| | - Philippe Lorimier
- Institut Albert Bonniot INSERM U823; Université Joseph Fourier Grenoble France
| | - Odd Terje Brustugun
- Division of Surgery and Cancer, Oslo University Hospital Radiumhospitalet, Montebello 0301, Oslo, Norway
| | - Åslaug Helland
- Division of Surgery and Cancer, Oslo University Hospital Radiumhospitalet, Montebello 0301, Oslo, Norway
| | - Iver Petersen
- Jena University Hospital, Department Hematology/Oncology, Jena, Germany
| | - Joachim H Clement
- Jena University Hospital, Department Hematology/Oncology, Jena, Germany
| | - Harry Groen
- University Medical Center Groningen and University of Groningen, Pulmonology and Pathology, Groningen, Netherlands
| | - Wim Timens
- University Medical Center Groningen and University of Groningen, Pulmonology and Pathology, Groningen, Netherlands
| | - Hannie Sietsma
- University Medical Center Groningen and University of Groningen, Pulmonology and Pathology, Groningen, Netherlands
| | | | - Jürgen Wolf
- Department I of Internal Medicine and Laboratory of Translational Cancer Genomics, Center of Integrated Oncology Köln – Bonn, University of Köln, Köln, Germany
- Department I for Internal Medicine, Center for Integrated Oncology Köln-Bonn, University Hospital of Cologne, Germany
| | - David G Beer
- Section of Thoracic Surgery, Department of Surgery, Ann Arbor, Michigan, USA
| | - Ming Sound Tsao
- Ontario Cancer Institute and Princess Margaret Hospital, Toronto, Canada
| | - Megan Hanna
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
- Center for Cancer Genome Discovery, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Charles Hatton
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
- Center for Cancer Genome Discovery, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael J Eck
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Pasi A Janne
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Bruce E Johnson
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Heidi Greulich
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Adam J Bass
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jeonghee Cho
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Daniel Rauh
- Chemical Genomics Centre of the Max Planck Society, Otto-Hahn-Strasse 15, D-44227 Dortmund, Germany
- Technical University Dortmund, Otto-Hahn-Strasse 6, D-44221 Dortmund, Germany
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kwok-Kin Wong
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Eric B Haura
- Departments of Thoracic Oncology and Experimental Therapeutics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Roman K Thomas
- Max Planck Institute for Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society and the Medical Faculty of the University of Köln, Köln, Germany
- Department I of Internal Medicine and Laboratory of Translational Cancer Genomics, Center of Integrated Oncology Köln – Bonn, University of Köln, Köln, Germany
- Chemical Genomics Center of the Max Planck Society, Dortmund, Germany
| | - Matthew Meyerson
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
- Center for Cancer Genome Discovery, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
159
|
Minimal cross-intolerance with nilotinib in patients with chronic myeloid leukemia in chronic or accelerated phase who are intolerant to imatinib. Blood 2011; 117:5600-6. [PMID: 21467546 PMCID: PMC4186645 DOI: 10.1182/blood-2010-11-318949] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Nilotinib has significant efficacy in patients with newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP) and in patients with CML-CP or CML in accelerated phase (CML-AP) after imatinib failure. We investigated the occurrence of cross-intolerance to nilotinib in imatinib-intolerant patients with CML. Only 1/75 (1%) patients with nonhematologic imatinib intolerance experienced a similar grade 3/4 adverse event (AE), and 3/75 (4%) experienced a similar persistent grade 2 nonhematologic AE on nilotinib. Only 7/40 (18%) patients with hematologic imatinib intolerance discontinued nilotinib, all because of grade 3/4 thrombocytopenia. Ninety percent of imatinib-intolerant patients with CML-CP who did not have complete hematologic response (CHR) at baseline (n = 52) achieved CHR on nilotinib. Nilotinib induced a major cytogenetic response in 66% and 41% of patients with imatinib-intolerant CML-CP and CML-AP (complete cytogenetic response in 51% and 30%), respectively. Minimal cross-intolerance was confirmed in patients with imatinib-intolerant CML. The favorable tolerability of nilotinib in patients with imatinib intolerance leads to alleviation of AE-related symptoms and significant and durable responses. In addition to its established clinical benefit in patients with newly diagnosed CML and those resistant to imatinib, nilotinib is effective and well-tolerated for long-term use in patients with imatinib intolerance. This study is registered at http://www.clinicaltrials.gov as NCT00471497.
Collapse
|
160
|
El-Agamy DS. Nilotinib ameliorates lipopolysaccharide-induced acute lung injury in rats. Toxicol Appl Pharmacol 2011; 253:153-60. [PMID: 21473879 DOI: 10.1016/j.taap.2011.03.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/05/2011] [Accepted: 03/24/2011] [Indexed: 12/20/2022]
Abstract
The present study aimed to investigate the effect of the new tyrosine kinase inhibitor, nilotinib on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in rats and explore its possible mechanisms. Male Sprague-Dawley rats were given nilotinib (10mg/kg) by oral gavage twice daily for 1week prior to exposure to aerosolized LPS. At 24h after LPS exposure, bronchoalveolar lavage fluid (BALF) samples and lung tissue were collected. The lung wet/dry weight (W/D) ratio, protein level and the number of inflammatory cells in the BALF were determined. Optical microscopy was performed to examine the pathological changes in lungs. Malondialdehyde (MDA) content, superoxidase dismutase (SOD) and reduced glutathione (GSH) activities as well as nitrite/nitrate (NO(2)(-)/NO(3)(-)) levels were measured in lung tissues. The expression of inflammatory cytokines, tumor necrosis factor-α (TNF-α), transforming growth factor-β(1) (TGF-β(1)) and inducible nitric oxide synthase (iNOS) were determined in lung tissues. Treatment with nilotinib prior to LPS exposure significantly attenuated the LPS-induced pulmonary edema, as it significantly decreased lung W/D ratio, protein concentration and the accumulation of the inflammatory cells in the BALF. This was supported by the histopathological examination which revealed marked attenuation of LPS-induced ALI in nilotinib treated rats. In addition, nilotinib significantly increased SOD and GSH activities with significant decrease in MDA content in the lung. Nilotinib also reduced LPS mediated overproduction of pulmonary NO(2)(-)/NO(3)(-) levels. Importantly, nilotinib caused down-regulation of the inflammatory cytokines TNF-α, TGF-β(1) and iNOS levels in the lung. Taken together, these results demonstrate the protective effects of nilotinib against the LPS-induced ALI. This effect can be attributed to nilotinib ability to counteract the inflammatory cells infiltration and hence ROS generation and regulate cytokine effects.
Collapse
Affiliation(s)
- Dina S El-Agamy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
161
|
Abstract
The development of tyrosine kinase inhibitors (TKIs) for the treatment of chronic myelogenous leukemia (CML) was based on the discovery that CML stem and progenitor cells overexpress the abnormal fusion protein kinase BCR-ABL. The prototype TKI, imatinib, selectively inhibits BCR-ABL, as well as several other kinases, including stem cell factor receptor (KIT), discoidin domain receptor (DDR), platelet-derived growth factor receptor (PDGFR), and colony-stimulating factor receptor-1 (CSF-1R). Although the management of CML improved dramatically with the introduction of imatinib, not all patients benefit from treatment because of resistance or intolerance. Consequently, research efforts have focused on developing more potent TKIs with the ability to circumvent imatinib resistance. Nilotinib, a second-generation oral TKI, was rationally designed based on the crystal structure of imatinib to be highly active against a wide range of imatinib-resistant BCR-ABL mutants and is approved for the treatment of newly diagnosed or imatinib-resistant or -intolerant CML, and has shown superiority over imatinib in first-line treatment for newly diagnosed CML. Furthermore, the activity of nilotinib against KIT and PDGFRα has led to its evaluation in advanced gastrointestinal stromal tumors (GIST). The purpose of this review is to describe the development of nilotinib, providing a structural explanation for the differential activity of nilotinib and imatinib in GIST. Activity of nilotinib against KIT and PDGFR and emerging evidence of differences in cellular uptake between nilotinib and imatinib are discussed.
Collapse
Affiliation(s)
- Jean-Yves Blay
- University Claude Bernard Lyon, Centre Léon Bérard, Department of Medicine, Lyon, France
| | | |
Collapse
|
162
|
Reichardt P, Montemurro M. Clinical Experience to Date With Nilotinib in Gastrointestinal Stromal Tumors. Semin Oncol 2011; 38 Suppl 1:S20-7. [DOI: 10.1053/j.seminoncol.2011.01.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
163
|
DeRemer DL, Katsanevas K, Ustun C. Critical appraisal of nilotinib in frontline treatment of chronic myeloid leukemia. Cancer Manag Res 2011; 3:65-78. [PMID: 21556318 PMCID: PMC3085241 DOI: 10.2147/cmr.s11948] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Indexed: 12/13/2022] Open
Abstract
The development of imatinib has revolutionized the treatment of chronic myeloid leukemia. Follow-up analysis of IRIS trial participants continues to demonstrate durable responses for imatinib at 400 mg/day. However, 10%-15% of patients with chronic myeloid leukemia will become imatinib-resistant or intolerant of adverse events. Phase II studies have shown that most of these patients will respond to second-generation tyrosine kinase inhibitors, such as nilotinib, dasatinib, and bosutinib. Both nilotinib and dasatinib have recently demonstrated clinical efficacy as frontline therapy in Phase III studies. In the ENESTnd trial, nilotinib 600-800 mg/day produced significantly higher major molecular rates and complete cytogenetic response rates in comparison with imatinib at 12 months. Recently, 18-month follow-up analysis of this trial continues to demonstrate superiority for nilotinib. It is unknown whether this will ultimately translate into improved long-term outcomes, such as event-free survival or overall survival. Nilotinib continues to be generally well tolerated and tends to produce less Grade 3/4 toxicity in frontline therapy when compared with its use following imatinib failure. With three tyrosine kinase inhibitors for potential frontline therapy and an active drug discovery pipeline, treatment for chronic myeloid leukemia is still subject to change with time as clinical algorithms continue to evolve.
Collapse
Affiliation(s)
- David L DeRemer
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA, USA
- Medical College of Georgia Health Inc, Augusta, GA, USA
| | - Katerina Katsanevas
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA, USA
- Medical College of Georgia Health Inc, Augusta, GA, USA
| | - Celalettin Ustun
- Department of Medicine, Section of Hematology/Oncology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
164
|
Fedorov O, Huber K, Eisenreich A, Filippakopoulos P, King O, Bullock AN, Szklarczyk D, Jensen LJ, Fabbro D, Trappe J, Rauch U, Bracher F, Knapp S. Specific CLK inhibitors from a novel chemotype for regulation of alternative splicing. CHEMISTRY & BIOLOGY 2011; 18:67-76. [PMID: 21276940 PMCID: PMC3145970 DOI: 10.1016/j.chembiol.2010.11.009] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 11/08/2010] [Accepted: 11/10/2010] [Indexed: 12/02/2022]
Abstract
There is a growing recognition of the importance of protein kinases in the control of alternative splicing. To define the underlying regulatory mechanisms, highly selective inhibitors are needed. Here, we report the discovery and characterization of the dichloroindolyl enaminonitrile KH-CB19, a potent and highly specific inhibitor of the CDC2-like kinase isoforms 1 and 4 (CLK1/CLK4). Cocrystal structures of KH-CB19 with CLK1 and CLK3 revealed a non-ATP mimetic binding mode, conformational changes in helix αC and the phosphate binding loop and halogen bonding to the kinase hinge region. KH-CB19 effectively suppressed phosphorylation of SR (serine/arginine) proteins in cells, consistent with its expected mechanism of action. Chemical inhibition of CLK1/CLK4 generated a unique pattern of splicing factor dephosphorylation and had at low nM concentration a profound effect on splicing of the two tissue factor isoforms flTF (full-length TF) and asHTF (alternatively spliced human TF).
Collapse
Affiliation(s)
- Oleg Fedorov
- University of Oxford, Nuffield Department of Clinical Medicine, Structural Genomics Consortium, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Kilian Huber
- Ludwig-Maximilians Universität, Department of Pharmacy-Center for Drug Research, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Andreas Eisenreich
- Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Centrum für Herz-und Kreislaufmedizin, Berlin, Germany
| | - Panagis Filippakopoulos
- University of Oxford, Nuffield Department of Clinical Medicine, Structural Genomics Consortium, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Oliver King
- University of Oxford, Nuffield Department of Clinical Medicine, Structural Genomics Consortium, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Alex N. Bullock
- University of Oxford, Nuffield Department of Clinical Medicine, Structural Genomics Consortium, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Damian Szklarczyk
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Lars J. Jensen
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Doriano Fabbro
- Novartis Pharma AG, Klybeckstrasse 141, CH-4002 Basel, Switzerland
| | - Jörg Trappe
- Novartis Pharma AG, Klybeckstrasse 141, CH-4002 Basel, Switzerland
| | - Ursula Rauch
- Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Centrum für Herz-und Kreislaufmedizin, Berlin, Germany
| | - Franz Bracher
- Ludwig-Maximilians Universität, Department of Pharmacy-Center for Drug Research, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Stefan Knapp
- University of Oxford, Nuffield Department of Clinical Medicine, Structural Genomics Consortium, Old Road Campus Research Building, Oxford OX3 7DQ, UK
- University of Oxford, Department of Clinical Pharmacology, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| |
Collapse
|
165
|
Liniger M, Neuhaus C, Hofmann T, Fransioli-Ignazio L, Jordi M, Drueckes P, Trappe J, Fabbro D, Altmann KH. Kinase inhibition by deoxy analogues of the resorcylic lactone L-783277. ACS Med Chem Lett 2011; 2:22-7. [PMID: 24900250 DOI: 10.1021/ml1001807] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 09/23/2010] [Indexed: 12/22/2022] Open
Abstract
The natural product L-783277 is a resorcylic lactone type covalent kinase inhibitor. We have prepared the 5'-deoxy analogue of L-783277 (1) in a stereoselective fashion. Remarkably, this analogue retains almost the full kinase inhibitory potential of natural L-783277, with low nanomolar IC50 values against the most sensitive kinases, and it exhibits essentially the same selectivity profile (within the panel of 39 kinases investigated). In contrast, removal of both the 4'- and the 5'-hydroxyl groups leads to a more significant reduction in kinase inhibitory activity and so does a change in the geometry of the C7'-C8' double bond in 1 from Z to E. These findings offer new perspectives for the design of second generation resorcylic lactone-based kinase inhibitors.
Collapse
Affiliation(s)
- Marc Liniger
- Swiss Federal Institute of Technology (ETH) Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, 8093 Zurich, Switzerland
| | - Christian Neuhaus
- Swiss Federal Institute of Technology (ETH) Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, 8093 Zurich, Switzerland
| | - Tatjana Hofmann
- Swiss Federal Institute of Technology (ETH) Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, 8093 Zurich, Switzerland
| | - Luca Fransioli-Ignazio
- Swiss Federal Institute of Technology (ETH) Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, 8093 Zurich, Switzerland
| | - Michel Jordi
- Swiss Federal Institute of Technology (ETH) Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, 8093 Zurich, Switzerland
| | - Peter Drueckes
- Novartis Institute for Biomedical Research, Center for Proteomic Chemistry, Expertise Platform Kinases, 4002 Basel, Switzerland
| | - Jörg Trappe
- Novartis Institute for Biomedical Research, Center for Proteomic Chemistry, Expertise Platform Kinases, 4002 Basel, Switzerland
| | - Doriano Fabbro
- Novartis Institute for Biomedical Research, Center for Proteomic Chemistry, Expertise Platform Kinases, 4002 Basel, Switzerland
| | - Karl-Heinz Altmann
- Swiss Federal Institute of Technology (ETH) Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, 8093 Zurich, Switzerland
| |
Collapse
|
166
|
Comparative suppressive effects of tyrosine kinase inhibitors imatinib and nilotinib in models of autoimmune arthritis. Mod Rheumatol 2010; 21:267-75. [PMID: 21188449 DOI: 10.1007/s10165-010-0392-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 11/16/2010] [Indexed: 11/25/2022]
Abstract
Imatinib and nilotinib are inhibitors that selectively target a set of protein tyrosine kinases, including abelson kinase (Abl), together with the chimeric oncoprotein, breakpoint cluster region-abelson kinase (Bcr-Abl), as well as stem cell factor receptor (KIT), platelet-derived growth factor receptor (PDGFR), discoidin domain receptor (DDR), and colony stimulating factor-1 receptor (CSF-1R). The aim of the present study was to investigate whether imatinib or nilotinib was effective against arthritis in the glucose-6-phosphate isomerase (GPI)-induced arthritis mouse model. Imatinib or nilotinib was administered orally to the arthritic mice at different time points. Efficacy was evaluated by visual scoring and by determining the production of anti-GPI antibody. Splenocytes from the arthritic mice were cultured with GPI in the presence of imatinib or nilotinib in vitro, and cytokine levels in the culture supernatants were analyzed. To investigate the effects of imatinib and nilotinib on T-cell proliferation, lymph node cells from the arthritic mice were cultured with GPI in the presence of imatinib or nilotinib in vitro. Interleukin (IL)-17 mRNA expression in the arthritic ankle joints from the onset of arthritis was analyzed by real-time polymerase chain reaction (PCR). The administration of imatinib from day 0 showed suppression of arthritis (P < 0.05), the administration of nilotinib from day 0 resulted in pronounced suppression of arthritis (P < 0.01), and that from day 7 showed significant inhibition of the progression of arthritis (P < 0.05). A reduction in anti-GPI antibodies was correlated with the therapeutic efficacy of imatinib, but not with that of nilotinib. Imatinib dose-dependently inhibited tumor necrosis factor (TNF)-α, IL-6, interferon (IFN)-γ, and IL-17 production by splenocytes in vitro, while nilotinib inhibited only IL-17 and IFN-γ production in a dose-dependent fashion. Imatinib at 3 μM exerted a mild antiproliferative effect on CD4+ T cells (P < 0.05), whereas imatinib at 10 μM and nilotinib at 3 and 10 μM demonstrated a marked antiproliferative effect (P < 0.01). The IL17 gene expression level on day 7 tended to be higher than that on day 14. These findings suggest that imatinib and nilotinib could prevent autoimmune arthritis, essentially via distinct mechanisms, in that imatinib inhibits both inflammatory and T-cell-derived cytokine production, whereas nilotinib suppresses T-cell-derived cytokine production. Imatinib and nilotinib could have therapeutic potential for rheumatoid arthritis (RA) and other inflammatory diseases.
Collapse
|
167
|
Posy SL, Hermsmeier MA, Vaccaro W, Ott KH, Todderud G, Lippy JS, Trainor GL, Loughney DA, Johnson SR. Trends in Kinase Selectivity: Insights for Target Class-Focused Library Screening. J Med Chem 2010; 54:54-66. [DOI: 10.1021/jm101195a] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Shana L. Posy
- Computer-Assisted Drug Design, Applied Biotechnology, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Mark A. Hermsmeier
- Chemistry Informatics, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Wayne Vaccaro
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Karl-Heinz Ott
- Bioinformatics, Applied Biotechnology, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Gordon Todderud
- Lead Evaluation, Applied Biotechnology, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Jonathan S. Lippy
- Lead Evaluation, Applied Biotechnology, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - George L. Trainor
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Deborah A. Loughney
- Computer-Assisted Drug Design, Applied Biotechnology, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Stephen R. Johnson
- Computer-Assisted Drug Design, Applied Biotechnology, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| |
Collapse
|
168
|
Nilotinib is effective in patients with chronic myeloid leukemia in chronic phase after imatinib resistance or intolerance: 24-month follow-up results. Blood 2010; 117:1141-5. [PMID: 21098399 DOI: 10.1182/blood-2010-03-277152] [Citation(s) in RCA: 264] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nilotinib is a potent selective inhibitor of the BCR-ABL tyrosine kinase approved for use in patients with newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP), and in CML-CP and CML-accelerated phase after imatinib failure. Nilotinib (400 mg twice daily) was approved on the basis of the initial results of this phase 2 open-label study. The primary study endpoint was the proportion of patients achieving major cytogenetic response (CyR). All patients were followed for ≥ 24 months or discontinued early. Of 321 patients, 124 (39%) continue on nilotinib treatment. Overall, 59% of patients achieved major CyR; this was complete CyR (CCyR) in 44%. Of patients achieving CCyR, 56% achieved major molecular response. CyRs were durable, with 84% of patients who achieved CCyR maintaining response at 24 months. The overall survival at 24 months was 87%. Adverse events were mostly mild to moderate, generally transient, and easily managed. This study indicates that nilotinib is effective, with a manageable safety profile, and can provide favorable long-term benefits for patients with CML-CP after imatinib failure.
Collapse
|
169
|
Kim TD, Schwarz M, Nogai H, Grille P, Westermann J, Plöckinger U, Braun D, Schweizer U, Arnold R, Dörken B, le Coutre P. Thyroid dysfunction caused by second-generation tyrosine kinase inhibitors in Philadelphia chromosome-positive chronic myeloid leukemia. Thyroid 2010; 20:1209-14. [PMID: 20929406 DOI: 10.1089/thy.2010.0251] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Thyroid dysfunction is a well-known adverse effect of first-generation tyrosine kinase inhibitors (TKIs), like sunitinib. The aim of this study was to investigate the effect of second-generation TKIs on thyroid function. METHODS We retrospectively assessed the effect of the first-generation TKI imatinib and the second-generation TKI nilotinib and dasatinib on thyroid function tests in 73 Philadelphia chromosome-positive (Ph-positive) chronic myeloid leukemia patients. RESULTS Overall, 33 of 73 (45%) had one or more thyroid function test abnormalities during follow-up. Hypothyroidism or hyperthyroidism were found in 18 of 73 (25%) and 21 of 73 (29%) cases after a median of 6 and 22 weeks, respectively. In most patients (29 of 39, 74%) thyroid dysfunction was transient without clinical symptoms. Therapy of hypo-/hyperthyroidism was required in three patients. Thyroid dysfunction never resulted in the discontinuation of TKI therapy. Under treatment with imatinib, nilotinib, and dasatinib, thyroid abnormalities were detected in 25%, 55%, and 70%, respectively. Four of 55 patients (7%) treated with nilotinib had evidence for an autoimmune thyroiditis (antibody positive in 3 of 4 patients) with an episode of hyperthyroidism preceding hypothyroidism. CONCLUSIONS Thyroid dysfunction is a common adverse event with second-generation TKI therapy in patients with Ph-positive chronic myeloid leukemia. Although the mechanism is still unclear, the high frequency of thyroid abnormalities, including autoimmune thyroiditis, warrants regular and long-term monitoring of thyroid function in these patients.
Collapse
Affiliation(s)
- Theo D Kim
- Clinic for Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Nilotinib in patients with GIST who failed imatinib and sunitinib: importance of prior surgery on drug bioavailability. Cancer Chemother Pharmacol 2010; 68:285-91. [PMID: 20957481 DOI: 10.1007/s00280-010-1479-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 09/24/2010] [Indexed: 10/18/2022]
Abstract
PURPOSE To evaluate the efficacy of nilotinib in patients with advanced gastrointestinal stromal tumors (GISTs) resistant or intolerant to both imatinib and sunitinib and to explore the potential relationship between nilotinib pharmacokinetics and clinical outcomes. PATIENTS AND METHODS We analyzed the efficacy, tolerability and pharmacokinetic parameters of nilotinib (400 mg twice daily) in 17 GIST patients with histories of prior gastrointestinal surgery. RESULTS Median patient age was 59 years (range, 35-71 years), 14 of 17 patients (82.4%) were male, and mean body weight was 59.4 kg. Of the 17 patients, 2 (11.8%) had partial responses (PR), 10 (58.8%) had stable disease (SD), and 5 (29.4%) had progressive disease (PD), with a clinical benefit rate (CR + PR + SD) at 24 weeks of 47.0%. Median progression-free survival (PFS) and overall survival (OS) were 23.6 weeks (95% confidence interval [CI] 0.0-50.6 weeks) and 74.0 weeks (95% CI 27.4-120.6 weeks), respectively. Most observed adverse events were mild (grade 1, 41.2%; grade 2, 52.9%), with no grade 3/4 events. Pharmacokinetic parameters of nilotinib were as follows: C (max) of 1,754 ± 970 μg/L, T(1/2) of 13.4 ± 8.94 h and AUC (0-12 h) of 14,190 ± 6,853 h μg/L. The AUC (0-12 h) of nilotinib was significantly lower in the 4 patients with prior major (total or subtotal) gastrectomy than in the other 13 patients (8,526 ± 7,869 h μg/L vs. 15,930 ± 5,759 h μg/L, P = 0.014). Of the 4 gastrectomized patients, two (50%) showed markedly decreased nilotinib exposure (AUC (0-12 h) of 1,914 and 3,194 h μg/L) and rapid disease progression (PFS of 4.6 and 7.1 weeks). CONCLUSION Nilotinib was active and safe in patients with advanced GIST resistant to both imatinib and sunitinib. Major gastrectomy decreased the bioavailability of nilotinib and, in some patients, lowered its clinical activity.
Collapse
|
171
|
Schlemmer M, Schinwald N, Bruns C, Berger F, Reichardt P. Response to nilotinib as a first-line treatment for metastatic gastrointestinal stromal tumors. J Gastrointest Cancer 2010; 43:385-7. [PMID: 20922581 DOI: 10.1007/s12029-010-9208-2] [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: 11/26/2022]
Abstract
PURPOSE Effective and safe treatment options are needed for patients with advanced gastrointestinal stromal tumors (GIST) who are initially unresponsive to the tyrosine kinase inhibitor (TKI) imatinib, or develop acquired secondary imatinib resistance. CASE REPORT We report a 39-year-old woman with primary rectal GIST who underwent abdominoperineal resection in December 2004, achieving R0 margins. In August 2009, the patient was referred to our clinic, and we detected metastatic GIST of the liver, as well as peritoneal and gluteal lesions. The patient was treated with imatinib 400 mg/day for 3 weeks and subsequently switched to nilotinib (400 mg bid) after enrolling in a clinical trial. After 8 weeks of nilotinib treatment, a response was observed in the liver metastasis, and metabolic activity was no longer detected. Also, the gluteal and peritoneal lesions were no longer detected. After 16 weeks of nilotinib treatment, a cystic mass was identified in the liver metastasis. Tumor rupture was considered a strong possibility, prompting resection of the liver metastasis. Greater than 80% of the resected tumor mass was necrotic, consistent with the lack of metabolism observed 8 weeks prior. The patient resumed nilotinib treatment (400 mg bid) shortly after surgery and continues treatment while remaining disease-free for more than 9 months with normal liver function. CONCLUSION This is the first report demonstrating the feasibility of nilotinib (400 mg bid) for the first-line treatment of metastatic GIST. Furthermore, these results underscore that responses to TKIs may be underestimated by Response Evaluation Criteria in Solid Tumors.
Collapse
Affiliation(s)
- Marcus Schlemmer
- Department of Internal Medicine III-Hematology/Oncology, University Hospital Munich-Campus Grosshadern, Munich, Germany.
| | | | | | | | | |
Collapse
|
172
|
The small molecule specific EphB4 kinase inhibitor NVP-BHG712 inhibits VEGF driven angiogenesis. Angiogenesis 2010; 13:259-67. [PMID: 20803239 PMCID: PMC2941628 DOI: 10.1007/s10456-010-9183-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 08/05/2010] [Indexed: 01/02/2023]
Abstract
EphB4 and its cognitive ligand ephrinB2 play an important role in embryonic vessel development and vascular remodeling. In addition, several reports suggest that this receptor ligand pair is also involved in pathologic vessel formation in adults including tumor angiogenesis. Eph/ephrin signaling is a complex phenomena characterized by receptor forward signaling through the tyrosine kinase of the receptor and ephrin reverse signaling through various protein–protein interaction domains and phosphorylation motifs of the ephrin ligands. Therefore, interfering with EphR/ephrin signaling by the means of targeted gene ablation, soluble receptors, dominant negative mutants or antisense molecules often does not allow to discriminate between inhibition of Eph/ephrin forward and reverse signaling. We developed a specific small molecular weight kinase inhibitor of the EphB4 kinase, NVP-BHG712, which inhibits EphB4 kinase activity in the low nanomolar range in cellular assays showed high selectivity for targeting the EphB4 kinase when profiled against other kinases in biochemical as well as in cell based assays. Furthermore, NVP-BHG712 shows excellent pharmacokinetic properties and potently inhibits EphB4 autophosphorylation in tissues after oral administration. In vivo, NVP-BHG712 inhibits VEGF driven vessel formation, while it has only little effects on VEGF receptor (VEGFR) activity in vitro or in cellular assays. The data shown here suggest a close cross talk between the VEGFR and EphR signaling during vessel formation. In addition to its established function in vascular remodeling and endothelial arterio-venous differentiation, EphB4 forward signaling appears to be an important mediator of VEGF induced angiogenesis since inhibition of EphB4 forward signaling is sufficient to inhibit VEGF induced angiogenesis.
Collapse
|
173
|
Manley PW, Stiefl N, Cowan-Jacob SW, Kaufman S, Mestan J, Wartmann M, Wiesmann M, Woodman R, Gallagher N. Structural resemblances and comparisons of the relative pharmacological properties of imatinib and nilotinib. Bioorg Med Chem 2010; 18:6977-86. [PMID: 20817538 DOI: 10.1016/j.bmc.2010.08.026] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 07/08/2010] [Accepted: 08/11/2010] [Indexed: 02/06/2023]
Abstract
Although orphan drug applications required by the EMEA must include assessments of similarity to pre-existing products, these can be difficult to quantify. Here we illustrate a paradigm in comparing nilotinib to the prototype kinase inhibitor imatinib, and equate the degree of structural similarity to differences in properties. Nilotinib was discovered following re-engineering of imatinib, employing structural biology and medicinal chemistry strategies to optimise cellular potency and selectivity towards BCR-ABL1. Through evolving only to conserve these properties, this resulted in significant structural differences between nilotinib and imatinib, quantified by a Daylight-fingerprint-Tanimoto similarity coefficient of 0.6, with the meaning of this absolute measure being supported by an analysis of similarity distributions of similar drug-like molecules. This dissimilarity is reflected in the drugs having substantially different preclinical pharmacology and a lack of cross-intolerance in CML patients, which translates into nilotinib being an efficacious treatment for CML, with a favourable side-effect profile.
Collapse
Affiliation(s)
- Paul W Manley
- Novartis Institutes for BioMedical Research, Basel, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Sauter KAD, Magun EA, Iordanov MS, Magun BE. ZAK is required for doxorubicin, a novel ribotoxic stressor, to induce SAPK activation and apoptosis in HaCaT cells. Cancer Biol Ther 2010; 10:258-66. [PMID: 20559024 DOI: 10.4161/cbt.10.3.12367] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Doxorubicin is an anthracycline drug that is one of the most effective and widely used anticancer agents for the treatment of both hematologic and solid tumors. The stress-activated protein kinases (SAPKs) are frequently activated by a number of cancer chemotherapeutics. When phosphorylated, the SAPKs initiate a cascade that leads to the production of proinflammatory cytokines. Some inhibitors of protein synthesis, known as ribotoxic stressors, coordinately activate SAPKs and lead to apoptotic cell death. We demonstrate that doxorubicin effectively inhibits protein synthesis, activates SAPKs, and causes apoptosis. Ribotoxic stressors share a common mechanism in that they require ZAK, an upstream MAP3K, to activate the pro-apoptotic and proinflammatory signaling pathways that lie downstream of SAPKs. By employing siRNA mediated knockdown of ZAK or administration of sorafenib and nilotinib, kinase inhibitors that have a high affinity for ZAK, we provide evidence that ZAK is required for doxorubicin-induced proinflammatory and apoptotic responses in HaCaT cells, a pseudo-normal keratinocyte cell line, but not in HeLa cells, a cancerous cell line. ZAK has two different isoforms, ZAK-α (91 kDa) and ZAK-β (51 kDa). HaCaT or HeLa cells treated with doxorubicin and immunoblotted for ZAK displayed a progressive decrease in the ZAK-α band and the appearance of ZAK-β bands of larger size. Abrogation of these changes after exposure of cells to sorafenib and nilotinib suggests that these alterations occur following stimulation of ZAK. We suggest that ZAK inhibitors such as sorafenib or nilotinib may be effective when combined with doxorubicin to treat cancer patients.
Collapse
Affiliation(s)
- Kristin A D Sauter
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR, USA
| | | | | | | |
Collapse
|
175
|
Abstract
IMPORTANCE OF THE FIELD Inflammatory diseases are one of the major health issues and have become a major focus in the pharmaceutical and biotech industries. To date, drugs prescribed for treatment of these diseases target enzymes that are not specific to the immune system resulting in adverse effects. The main challenge of this research field is, therefore, identifying targets that act specifically on the diseased tissue. AREAS COVERED IN THIS REVIEW This review summarizes drug discovery efforts on kinases that have been identified as key players mediating inflammation and autoimmune disorders. In particular, we discuss recent developments on well-established targets such as mammalian target of rapamycin, JAK3, spleen tyrosine kinase, p38α and lymphocyte specific kinase but provide also a perspective on emerging targets. WHAT THE READER WILL GAIN The reader will obtain an overview of drug discovery efforts on kinases in inflammation, recent clinical and preclinical data and developed inhibitor scaffolds. In addition, the reader will be updated on issues in target validation of current drug targets and the potential of selected novel kinase targets in this important disease area. TAKE HOME MESSAGE Cellular signaling networks that regulate inflammatory response are still poorly understood making rational selection of targets challenging. Recent data suggest that kinase targets that are specific to the immune system and mediate signals immediately downstream of surface receptors are most efficacious in the clinic.
Collapse
Affiliation(s)
- Susanne Müller
- University of Oxford, Structural Genomics Consortium (SGC), Department of Clinical Medicine, Old Road Campus Research Building, Oxford OX3 7DQ, UK + 44 1865 617584 ; + 44 1865 617575 ;
| | | |
Collapse
|
176
|
Abstract
Ricin exhibits well characterized ribotoxic actions that lead to the inhibition of protein synthesis and the phosphorylation of stress activated protein kinases (SAPKs). Proinflammatory effects of ricin are thought to be caused by upregulation of genes encoding proinflammatory transcripts as a result of the activation of c-Jun N-terminal kinase (JNK) and p38 MAPK. We reported previously that macrophages and interleukin-1β (IL-1β) signaling are required for murine host immune responses to ricin delivered to the lungs. Here we report that ricin-mediated IL-1β release from bone-marrow derived macrophages is dependent on the NALP3 inflammasome, a scaffolding complex that mediates pro-IL-1β cleavage to active IL-1β by caspase-1. Release of IL-1β from macrophages was suppressed by the reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC) and high extracellular K(+), which are two agents known to inhibit NALP3/cryopyrin/CIAS1 inflammasome formation. By employing inhibitors of p38 MAPK and JNK, we demonstrated that ricin-mediated release of IL-1β was enhanced, rather than suppressed, by inhibition of SAPK phosphorylation. In contrast, proteasomal inhibitors bortezomib and MG-132 completely suppressed ricin-induced IL-1β release from macrophages. These data suggest that ricin-mediated translational inhibition itself, by fostering the disappearance of labile protein(s) that normally suppress inflammasome formation, may constitute the mechanism underlying IL-1-dependent inflammatory signaling by ricin.
Collapse
Affiliation(s)
| | | | - Bruce Magun
- Author to whom correspondence should be addressed; ; Tel.: +00-1-503-494-5824. Fax: +00-1-503-494-4253
| |
Collapse
|
177
|
Discovery of a small-molecule type II inhibitor of wild-type and gatekeeper mutants of BCR-ABL, PDGFRalpha, Kit, and Src kinases: novel type II inhibitor of gatekeeper mutants. Blood 2010; 115:4206-16. [PMID: 20299508 DOI: 10.1182/blood-2009-11-251751] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Many clinically validated kinases, such as BCR-ABL, c-Kit, PDGFR, and EGFR, become resistant to adenosine triphosphate-competitive inhibitors through mutation of the so-called gatekeeper amino acid from a threonine to a large hydrophobic amino acid, such as an isoleucine or methionine. We have developed a new class of adenosine triphosphate competitive inhibitors, exemplified by HG-7-85-01, which is capable of inhibiting T315I- BCR-ABL (clinically observed in chronic myeloid leukemia), T670I-c-Kit (clinically observed in gastrointestinal stromal tumors), and T674I/M-PDGFRalpha (clinically observed in hypereosinophilic syndrome). HG-7-85-01 is unique among all currently reported kinase inhibitors in having the ability to accommodate either a gatekeeper threonine, present in the wild-type forms of these kinases, or a large hydrophobic amino acid without becoming a promiscuous kinase inhibitor. The distinctive ability of HG-7-85-01 to simultaneously inhibit both wild-type and mutant forms of several kinases of clinical relevance is an important step in the development of the next generation of tyrosine kinase inhibitors.
Collapse
|