1
|
Berdel AF, Koch R, Gerss J, Hentrich M, Peceny R, Bartscht T, Steffen B, Bischoff M, Spiekermann K, Angenendt L, Mikesch JH, Kewitz T, Butterfass-Bahloul T, Serve H, Lenz G, Berdel WE, Krug U, Schliemann C. A randomized phase 2 trial of nintedanib and low-dose cytarabine in elderly patients with acute myeloid leukemia ineligible for intensive chemotherapy. Ann Hematol 2023; 102:63-72. [PMID: 36399194 PMCID: PMC9807538 DOI: 10.1007/s00277-022-05025-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
We investigated the safety and efficacy of nintedanib added to low-dose cytarabine (LDAC) in a phase 1/2 study in patients 60 years or older with newly diagnosed or relapsed/refractory (r/r) AML ineligible for intensive chemotherapy. The results of the dose-finding phase 1 part have been previously published. Patients were randomized 1:1 to LDAC plus nintedanib or LDAC plus placebo stratified by AML status (newly diagnosed vs r/r). LDAC was applied subcutaneously at 20 mg twice daily on days 1 to 10. Nintedanib/placebo was orally administered twice daily on days 1 to 28 in 28-day cycles. The primary endpoint was overall survival (OS). Between 05/2017 and 09/2019, 31 patients were randomized and 30 were treated, before the study was terminated prematurely due to slow recruitment. Median (range) age of patients was 76 (60-84) years. Twenty-two patients (73%) had r/r AML. Median OS in patients treated with LDAC and nintedanib was 3.4 months, compared with 3.6 months in those treated in the placebo arm, with a HR adjusted for AML status of 1.19 (corresponding confirmatory adjusted 95% CI, 0.55-2.56; univariate log-rank P = 0.96). In the 22 patients with r/r AML, median OS was 3.0 months in the nintedanib and 3.6 months in the placebo arm (P = 0.36). One patient in the nintedanib and two patients in the placebo arm achieved a CR and entered maintenance treatment. Nintedanib showed no superior therapeutic activity over placebo when added to LDAC in elderly AML patients considered unfit for intensive chemotherapy. The trial was registered at clinicaltrials.gov NCT01488344.
Collapse
Affiliation(s)
- Andrew F Berdel
- Department of Medicine A, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Raphael Koch
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Joachim Gerss
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Marcus Hentrich
- Department of Hematology and Oncology, Red Cross Hospital, Munich, Germany
| | - Rudolf Peceny
- Department of Oncology, Hematology and Stem Cell Transplantation, Klinikum Osnabrück, Osnabrück, Germany
| | - Tobias Bartscht
- Department of Medicine I, University Hospital Lübeck, Lübeck, Germany
| | - Björn Steffen
- Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Marina Bischoff
- Department of Hematology and Oncology, Klinikum Idar-Oberstein, Idar-Oberstein, Germany
| | - Karsten Spiekermann
- Department of Medicine III, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Linus Angenendt
- Department of Medicine A, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Jan-Henrik Mikesch
- Department of Medicine A, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Tobias Kewitz
- Centre for Clinical Trials, University of Münster, Münster, Germany
| | | | - Hubert Serve
- Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Georg Lenz
- Department of Medicine A, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Utz Krug
- Department of Medicine A, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
- Department of Medicine, III, Hospital Leverkusen, Leverkusen, Germany
| | - Christoph Schliemann
- Department of Medicine A, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
2
|
Yamashita M, Dellorusso PV, Olson OC, Passegué E. Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis. Nat Rev Cancer 2020; 20:365-382. [PMID: 32415283 PMCID: PMC7658795 DOI: 10.1038/s41568-020-0260-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2020] [Indexed: 12/17/2022]
Abstract
Haematopoiesis is governed by haematopoietic stem cells (HSCs) that produce all lineages of blood and immune cells. The maintenance of blood homeostasis requires a dynamic response of HSCs to stress, and dysregulation of these adaptive-response mechanisms underlies the development of myeloid leukaemia. Leukaemogenesis often occurs in a stepwise manner, with genetic and epigenetic changes accumulating in pre-leukaemic HSCs prior to the emergence of leukaemic stem cells (LSCs) and the development of acute myeloid leukaemia. Clinical data have revealed the existence of age-related clonal haematopoiesis, or the asymptomatic clonal expansion of mutated blood cells in the elderly, and this phenomenon is connected to susceptibility to leukaemic transformation. Here we describe how selection for specific mutations that increase HSC competitive fitness, in conjunction with additional endogenous and environmental changes, drives leukaemic transformation. We review the ways in which LSCs take advantage of normal HSC properties to promote survival and expansion, thus underlying disease recurrence and resistance to conventional therapies, and we detail our current understanding of leukaemic 'stemness' regulation. Overall, we link the cellular and molecular mechanisms regulating HSC behaviour with the functional dysregulation of these mechanisms in myeloid leukaemia and discuss opportunities for targeting LSC-specific mechanisms for the prevention or cure of malignant diseases.
Collapse
Affiliation(s)
- Masayuki Yamashita
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Paul V Dellorusso
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Oakley C Olson
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
3
|
Brattås MK, Reikvam H, Tvedt THA, Bruserud Ø. Precision medicine for TP53-mutated acute myeloid leukemia. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1644164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | - Håkon Reikvam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Øystein Bruserud
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
4
|
Kessler T, Koschmieder S, Schliemann C, Crysandt M, Mikesch JH, von Stillfried S, Stelljes M, Pohlen M, Lenz G, Kirsch A, Vehring K, Wardelmann E, Hartmann W, Bormann E, Gerss J, Brümmendorf TH, Müller-Tidow C, Berdel WE. Phase II clinical trial of pazopanib in patients with acute myeloid leukemia (AML), relapsed or refractory or at initial diagnosis without an intensive treatment option (PazoAML). Ann Hematol 2019; 98:1393-1401. [DOI: 10.1007/s00277-019-03651-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/02/2019] [Indexed: 12/01/2022]
|
5
|
Lunardi F, Pezzuto F, Vuljan SE, Calabrese F. Idiopathic Pulmonary Fibrosis and Antifibrotic Treatments: Focus on Experimental Studies. Arch Pathol Lab Med 2018; 142:1090-1097. [DOI: 10.5858/arpa.2018-0080-ra] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
Idiopathic pulmonary fibrosis (IPF) is a progressive fatal disease that up to now has been associated with a poor outcome. Some advances have been made in understanding the multiple interrelated pathogenic pathways underlying IPF. The disease is now believed to result from complex interactions among genetic, epigenetic, transcriptional, posttranscriptional, metabolic, and environmental factors. The discovery and validation of theranostic biomarkers are necessary to enable a more precise and earlier diagnosis of IPF and to improve the prediction of future disease behavior. Two drugs recently approved by the US Food and Drug Administration, pirfenidone and nintedanib, have shown the ability to reduce the progression of the disease, although survival benefits are only minimal and neither drug prevents or reverses the disease.
Objective.—
To provide a critical overview of the main experimental studies carried out for testing the principal effects of pirfenidone and nintedanib on IPF.
Data Sources.—
Experimental (animal and in vitro) studies concerning both drugs were used.
Conclusions.—
Pirfenidone has a longer history of preclinical experimental studies than nintedanib. Many studies have been reported more recently (after 2014) and some of them evaluated the association of both drugs, thus suggesting their combination in future therapeutic approaches. Future investigations focusing on targets at molecular, cellular, and tissue levels are necessary to have a better in-depth knowledge of the properties of these drugs and to explore the potential efficacy of both or other drug combinations.
Collapse
Affiliation(s)
| | | | | | - Fiorella Calabrese
- From the Department of Cardiac, Thoracic and Vascular Sciences, University of Padova Medical School, Padova, Italy
| |
Collapse
|
6
|
Zhang J, Liu T, Chen M, Liu F, Liu X, Zhang J, Lin J, Jin Y. Synthesis and Biological Evaluation of Indole-2-carbohydrazide Derivatives as Anticancer Agents with Anti-angiogenic and Antiproliferative Activities. ChemMedChem 2018; 13:1181-1192. [DOI: 10.1002/cmdc.201800033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/02/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Jianqiang Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resources; Ministry of Education and Yunnan Province; School of Chemical Science and Technology; Yunnan University; No. 2 Cuihu North Road Kunming 650091 P.R. China
- Key Laboratory of Subtropical Medicinal Edible Resources Development and Utilization in Yunnan Province; College of Biology and Chemistry; Puer University; No. 6 Xueyuan's Road Puer 665000 P.R. China
| | - Tongyang Liu
- Laboratory of Molecular Genetics of Aging and Tumors; Medical School; Kunming University of Science and Technology; No. 68 Wenchang Road, 121 Avenue Kunming 650500 P.R. China
| | - Mei Chen
- Key Laboratory of Subtropical Medicinal Edible Resources Development and Utilization in Yunnan Province; College of Biology and Chemistry; Puer University; No. 6 Xueyuan's Road Puer 665000 P.R. China
| | - Feifei Liu
- Laboratory of Molecular Genetics of Aging and Tumors; Medical School; Kunming University of Science and Technology; No. 68 Wenchang Road, 121 Avenue Kunming 650500 P.R. China
| | - Xingyuan Liu
- Key Laboratory of Subtropical Medicinal Edible Resources Development and Utilization in Yunnan Province; College of Biology and Chemistry; Puer University; No. 6 Xueyuan's Road Puer 665000 P.R. China
| | - Jihong Zhang
- Laboratory of Molecular Genetics of Aging and Tumors; Medical School; Kunming University of Science and Technology; No. 68 Wenchang Road, 121 Avenue Kunming 650500 P.R. China
| | - Jun Lin
- Key Laboratory of Medicinal Chemistry for Natural Resources; Ministry of Education and Yunnan Province; School of Chemical Science and Technology; Yunnan University; No. 2 Cuihu North Road Kunming 650091 P.R. China
| | - Yi Jin
- Key Laboratory of Medicinal Chemistry for Natural Resources; Ministry of Education and Yunnan Province; School of Chemical Science and Technology; Yunnan University; No. 2 Cuihu North Road Kunming 650091 P.R. China
| |
Collapse
|
7
|
Hilberg F, Tontsch-Grunt U, Baum A, Le AT, Doebele RC, Lieb S, Gianni D, Voss T, Garin-Chesa P, Haslinger C, Kraut N. Triple Angiokinase Inhibitor Nintedanib Directly Inhibits Tumor Cell Growth and Induces Tumor Shrinkage via Blocking Oncogenic Receptor Tyrosine Kinases. J Pharmacol Exp Ther 2017; 364:494-503. [PMID: 29263244 DOI: 10.1124/jpet.117.244129] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/11/2017] [Indexed: 12/11/2022] Open
Abstract
The triple-angiokinase inhibitor nintedanib is an orally available, potent, and selective inhibitor of tumor angiogenesis by blocking the tyrosine kinase activities of vascular endothelial growth factor receptor (VEGFR) 1-3, platelet-derived growth factor receptor (PDGFR)-α and -β, and fibroblast growth factor receptor (FGFR) 1-3. Nintedanib has received regulatory approval as second-line treatment of adenocarcinoma non-small cell lung cancer (NSCLC), in combination with docetaxel. In addition, nintedanib has been approved for the treatment of idiopathic lung fibrosis. Here we report the results from a broad kinase screen that identified additional kinases as targets for nintedanib in the low nanomolar range. Several of these kinases are known to be mutated or overexpressed and are involved in tumor development (discoidin domain receptor family, member 1 and 2, tropomyosin receptor kinase A (TRKA) and C, rearranged during transfection proto-oncogene [RET proto oncogene]), as well as in fibrotic diseases (e.g., DDRs). In tumor cell lines displaying molecular alterations in potential nintedanib targets, the inhibitor demonstrates direct antiproliferative effects: in the NSCLC cell line NCI-H1703 carrying a PDGFRα amplification (ampl.); the gastric cancer cell line KatoIII and the breast cancer cell line MFM223, both driven by a FGFR2 amplification; AN3CA (endometrial carcinoma) bearing a mutated FGFR2; the acute myeloid leukemia cell lines MOLM-13 and MV-4-11-B with FLT3 mutations; and the NSCLC adenocarcinoma LC-2/ad harboring a CCDC6-RET fusion. Potent kinase inhibition does not, however, strictly translate into antiproliferative activity, as demonstrated in the TRKA-dependent cell lines CUTO-3 and KM-12. Importantly, nintedanib treatment of NCI-H1703 tumor xenografts triggered effective tumor shrinkage, indicating a direct effect on the tumor cells in addition to the antiangiogenic effect on the tumor stroma. These findings will be instructive in guiding future genome-based clinical trials of nintedanib.
Collapse
Affiliation(s)
- Frank Hilberg
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Ulrike Tontsch-Grunt
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Anke Baum
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Anh T Le
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Robert C Doebele
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Simone Lieb
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Davide Gianni
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Tilman Voss
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Pilar Garin-Chesa
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Christian Haslinger
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| |
Collapse
|
8
|
Farag AK, Elkamhawy A, Londhe AM, Lee KT, Pae AN, Roh EJ. Novel LCK/FMS inhibitors based on phenoxypyrimidine scaffold as potential treatment for inflammatory disorders. Eur J Med Chem 2017; 141:657-675. [PMID: 29107425 DOI: 10.1016/j.ejmech.2017.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/19/2017] [Accepted: 10/02/2017] [Indexed: 01/24/2023]
Abstract
Tyrosine kinases including LCK and FMS are involved in inflammatory disorders as well as many types of cancer. Our team has designed and synthesized thirty novel pyrimidine based inhibitors targeting LCK, classified into four different series (amides, ureas, imines (Schiff base) and benzylamines). Twelve of them showed nanomolar IC50 values. Compound 7g showed excellent selectivity profile and was selectively potent over FMS kinase (IC50 value of 4.6 nM). Molecular docking study was performed to help us rationalize the obtained results and predict the possible binding mode for our compounds in both LCK and FMS. Based on the obtained biological assay data and modelling results, a detailed SAR study was discussed. As a further testing regarding the anti-inflammatory effect of the new compounds, in vitro cellular assay over RAW 264.7 macrophages was performed. Compound 7g exhibited excellent anti-inflammatory effect. Therefore, we report the design of novel phenoxypyrimidine derivatives as potent and selective LCK inhibitors and the discovery of 7g as potent and selective FMS/LCK dual inhibitor for the potential application in inflammatory disorders including rheumatoid arthritis (RA).
Collapse
Affiliation(s)
- Ahmed Karam Farag
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ahmed Elkamhawy
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ashwini M Londhe
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Kyung-Tae Lee
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Ae Nim Pae
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
9
|
Schliemann C, Gerss J, Wiebe S, Mikesch JH, Knoblauch N, Sauer T, Angenendt L, Kewitz T, Urban M, Butterfass-Bahloul T, Edemir S, Vehring K, Müller-Tidow C, Berdel WE, Krug U. A Phase I Dose Escalation Study of the Triple Angiokinase Inhibitor Nintedanib Combined with Low-Dose Cytarabine in Elderly Patients with Acute Myeloid Leukemia. PLoS One 2016; 11:e0164499. [PMID: 27716819 PMCID: PMC5055288 DOI: 10.1371/journal.pone.0164499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/21/2016] [Indexed: 11/18/2022] Open
Abstract
Nintedanib (BIBF 1120), a potent multikinase inhibitor of VEGFR-1/-2/-3, FGFR-1/-2/-3 and PDGFR-α/-β, exerts growth inhibitory and pro-apoptotic effects in myeloid leukemic cells, especially when used in combination with cytarabine. This phase I study evaluated nintedanib in combination with low-dose cytarabine (LDAC) in elderly patients with untreated or relapsed/refractory acute myeloid leukemia (AML) ineligible for intensive chemotherapy in a 3+3 design. Nintedanib (dose levels 100, 150, and 200 mg orally twice daily) and LDAC (20 mg subcutaneous injection twice daily for 10 days) were administered in 28-day cycles. Dose-limiting toxicity (DLT) was defined as non-hematological severe adverse reaction CTC grade ≥ 4 with possible or definite relationship to nintedanib. Between April 2012 and October 2013, 13 patients (median age 73 [range: 62-86] years) were enrolled. One patient did not receive study medication and was replaced. Nine (69%) patients had relapsed or refractory disease and 6 (46%) patients had unfavorable cytogenetics. The most frequently reported treatment-related adverse events (AE) were gastrointestinal events. Twelve SAEs irrespective of relatedness were reported. Two SUSARs were observed, one fatal hypercalcemia and one fatal gastrointestinal infection. Two patients (17%) with relapsed AML achieved a complete remission (one CR, one CRi) and bone marrow blast reductions without fulfilling PR criteria were observed in 3 patients (25%). One-year overall survival was 33%. Nintedanib combined with LDAC shows an adequate safety profile and survival data are promising in a difficult-to-treat patient population. Continuation of this trial with a phase II recommended dose of 2 x 200 mg nintedanib in a randomized, placebo-controlled phase II study is planned. The trial is registered to EudraCT as 2011-001086-41. TRIAL REGISTRATION ClinicalTrials.gov NCT01488344.
Collapse
Affiliation(s)
| | - Joachim Gerss
- Institute of Biostatistics and Clinical Research, University Hospital Muenster, Muenster, Germany
| | - Stefanie Wiebe
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Jan-Henrik Mikesch
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Nicola Knoblauch
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Tim Sauer
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Linus Angenendt
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Tobias Kewitz
- Centre for Clinical Trials, University Hospital Muenster, Muenster, Germany
| | - Marc Urban
- Centre for Clinical Trials, University Hospital Muenster, Muenster, Germany
| | | | - Sabine Edemir
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Kerstin Vehring
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | | | - Wolfgang E. Berdel
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Utz Krug
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
- * E-mail:
| |
Collapse
|
10
|
Dai SP, Xie C, Ding N, Zhang YJ, Han L, Han YW. Targeted inhibition of genome-wide DNA methylation analysis in epigenetically modulated phenotypes in lung cancer. Med Oncol 2015; 32:615. [PMID: 25926333 DOI: 10.1007/s12032-015-0615-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/06/2015] [Indexed: 11/28/2022]
Abstract
DNA methylation analysis, an epigenetic specification, has been explored for partial determination of cancer cell phenotypes. The development of metastasis in cancerogenesis has led its feasible association with the epigenetic modulations. We generated highly aggressive non-small cell lung cancer cell lines (HTB56 and A549) by using in vivo selection approach. These were, then, subjected to DNA methylation analysis (genome-wide). We also explored the therapeutic effects of azacytidine, an epigenetic agent, on DNA methylation patterns as well as the in vivo phenotypes. During the development of highly aggressive cell lines, we observed widespread modulations in DNA methylation. Reduced representation bisulfite sequencing was used and compared with the less aggressive parental cell lines to identify the differential methylation, which was achieved up to 2.7 % of CpG-rich region. Azacytidine inhibited DNA methyltransferase and reversed the prometastatic phenotype. We found its high association with the preferential loss of DNA methylation from hypermethylated sites. After persisted exposure of azacytidine, we observed that DNA methylation affected the polycomb-binding sites. We found close association of DNA methylome modifications with metastatic capability of non-small cell lung cancer. We also concluded that epigenetic modulation could be used as a potential therapeutic approach to prevent metastasis formation as prometastatic phenotype was reversed due to inhibition of DNA methyltransferase.
Collapse
Affiliation(s)
- Shou-Ping Dai
- Department of Medical Imaging, Linyi People's Hospital, Linyi, Shandong, China
| | | | | | | | | | | |
Collapse
|
11
|
Myllärniemi M, Kaarteenaho R. Pharmacological treatment of idiopathic pulmonary fibrosis - preclinical and clinical studies of pirfenidone, nintedanib, and N-acetylcysteine. Eur Clin Respir J 2015; 2:26385. [PMID: 26557253 PMCID: PMC4629756 DOI: 10.3402/ecrj.v2.26385] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/13/2015] [Indexed: 01/01/2023] Open
Abstract
Three recent clinical trials on the pharmacologic treatment of idiopathic pulmonary fibrosis (IPF) mark a new chapter in the management of patients suffering from this very severe fibrotic lung disease. This review article summarizes the published investigations on the preclinical studies of three novel IPF drugs, namely pirfenidone, nintedanib, and N-acetylcysteine (NAC). In addition, the study protocols, differences, and the main findings in the recent clinical trials of these pharmacological treatments are reviewed. The strategy for drug development and the timeline from the discovery to the clinical use have been very different in these regimens. Pirfenidone was discovered in 1976 but only recently received approval in most countries, and even now its exact mechanism of action is unknown. On the contrary, nintedanib (BIBF1120) was identified in large drug screening tests as a very specific inhibitor of certain tyrosine kinases, but no published data on preclinical tests existed until 2014. NAC, a mucolytic drug with an antioxidant mechanism of action was claimed to possess distinct antifibrotic properties in several experimental models but proved to be ineffective in a recent randomized placebo-controlled trial. At present, no curative treatment is available for IPF. A better understanding of the molecular mechanisms of IPF as well as relevant preclinical tests including animal models and in vitro experiments on human lung cells are needed to promote the development of therapeutic drugs.
Collapse
Affiliation(s)
- Marjukka Myllärniemi
- Department of Pulmonary Medicine, Helsinki University Central Hospital, Heart and Lung Center and the University of Helsinki, Helsinki, Finland
| | - Riitta Kaarteenaho
- Department of Internal Medicine, Respiratory Diseases, Institute of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Respiratory Research Unit, Oulu University Hospital, Oulu, Finland
- Unit of Medicine and Clinical Research, Pulmonary Division, University of Eastern Finland, Kuopio, Finland
- Division of Respiratory Medicine, Center for Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
12
|
Hascher A, Haase AK, Hebestreit K, Rohde C, Klein HU, Rius M, Jungen D, Witten A, Stoll M, Schulze I, Ogawa S, Wiewrodt R, Tickenbrock L, Berdel WE, Dugas M, Thoennissen NH, Müller-Tidow C. DNA Methyltransferase Inhibition Reverses Epigenetically Embedded Phenotypes in Lung Cancer Preferentially Affecting Polycomb Target Genes. Clin Cancer Res 2013; 20:814-26. [DOI: 10.1158/1078-0432.ccr-13-1483] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Hoang VT, Zepeda-Moreno A, Ho AD. Identification of leukemia stem cells in acute myeloid leukemia and their clinical relevance. Biotechnol J 2012; 7:779-88. [PMID: 22588704 DOI: 10.1002/biot.201100350] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 12/13/2022]
Abstract
Acute myeloid leukemia (AML) is considered to be a disease of stem cells. A rare defective stem cell population is purported to drive tumor growth. Similarly to their normal counterparts, leukemic stem cells (LSC) divide extreme slowly. This may explain the ineffectiveness of conventional chemotherapy in combatting this disease. Novel treatment strategies aimed at disrupting the binding of LSC to stem cell niches within the bone marrow might render the LSC vulnerable to chemotherapy and thus improving treatment outcome. This review focuses on the detection of LSC, our current knowledge about their cellular and molecular biology, and LSC interaction with the niche. Finally, we discuss the clinical relevance of LSC and prospective targeted treatment strategies for patients with AML.
Collapse
Affiliation(s)
- Van Thanh Hoang
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | |
Collapse
|
14
|
Kudo K, Arao T, Tanaka K, Nagai T, Furuta K, Sakai K, Kaneda H, Matsumoto K, Tamura D, Aomatsu K, De Velasco MA, Fujita Y, Saijo N, Kudo M, Nishio K. Antitumor activity of BIBF 1120, a triple angiokinase inhibitor, and use of VEGFR2+pTyr+ peripheral blood leukocytes as a pharmacodynamic biomarker in vivo. Clin Cancer Res 2010; 17:1373-81. [PMID: 21131553 DOI: 10.1158/1078-0432.ccr-09-2755] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE BIBF 1120 is a potent, orally available triple angiokinase inhibitor that inhibits VEGF receptors (VEGFR) 1, 2, and 3, fibroblast growth factor receptors, and platelet-derived growth factor receptors. This study examined the antitumor effects of BIBF 1120 on hepatocellular carcinoma (HCC) and attempted to identify a pharmacodynamic biomarker for use in early clinical trials. EXPERIMENTAL DESIGN We evaluated the antitumor and antiangiogenic effects of BIBF 1120 against HCC cell line both in vitro and in vivo. For the pharmacodynamic study, the phosphorylation levels of VEGFR2 in VEGF-stimulated peripheral blood leukocytes (PBL) were evaluated in mice inoculated with HCC cells and treated with BIBF 1120. RESULTS BIBF 1120 (0.01 μmol/L) clearly inhibited the VEGFR2 signaling in vitro. The direct growth inhibitory effects of BIBF 1120 on four HCC cell lines were relatively mild in vitro (IC(50) values: 2-5 μmol/L); however, the oral administration of BIBF 1120 (50 or 100 mg/kg/d) significantly inhibited the tumor growth and angiogenesis in a HepG2 xenograft model. A flow cytometric analysis revealed that BIBF 1120 significantly decreased the phosphotyrosine (pTyr) levels of VEGFR2(+)CD45(dim) PBLs and the percentage of VEGFR2(+)pTyr(+) PBLs in vivo; the latter parameter seemed to be a more feasible pharmacodynamic biomarker. CONCLUSIONS We found that BIBF 1120 exhibited potent antitumor and antiangiogenic activity against HCC and identified VEGFR2(+)pTyr(+) PBLs as a feasible and noninvasive pharmacodynamic biomarker in vivo.
Collapse
Affiliation(s)
- Kanae Kudo
- Department of Genome Biology and Gastroenterology, Kinki University School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Guerrouahen BS, Futami M, Vaklavas C, Kanerva J, Whichard ZL, Nwawka K, Blanchard EG, Lee FY, Robinson LJ, Arceci R, Kornblau SM, Wieder E, Cayre YE, Corey SJ. Dasatinib inhibits the growth of molecularly heterogeneous myeloid leukemias. Clin Cancer Res 2010; 16:1149-58. [PMID: 20145167 DOI: 10.1158/1078-0432.ccr-09-2416] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Dasatinib is a dual Src/Abl inhibitor recently approved for Bcr-Abl+ leukemias with resistance or intolerance to prior therapy. Because Src kinases contribute to multiple blood cell functions by triggering a variety of signaling pathways, we hypothesized that their molecular targeting might lead to growth inhibition in acute myeloid leukemia (AML). EXPERIMENTAL DESIGN We studied growth factor-dependent and growth factor-independent leukemic cell lines, including three cell lines expressing mutants of receptor tyrosine kinases (Flt3 or c-Kit) as well as primary AML blasts for responsiveness to dasatinib. RESULTS Dasatinib resulted in the inhibition of Src family kinases in all cell lines and blast cells at approximately 1 x 10(-9) mol/L. It also inhibited mutant Flt3 or Kit tyrosine phosphorylation at approximately 1 x 10(-6) mol/L. Mo7e cells expressing the activating mutation (codon 816) of c-Kit were most sensitive to growth inhibition with a GI(50) of 5 x 10(-9) mol/L. Primary AML blast cells exhibited a growth inhibition of <1 x 10(-6) mol/L. Cell lines that showed growth inhibition at approximately 1 x 10(-6) mol/L showed a G(1) cell cycle arrest and correlated with accumulation of p21 and p27 protein. The addition of rapamycin or cytotoxic agents enhanced growth inhibition. Dasatinib also caused the apoptosis of Mo7e cells expressing oncogenic Kit. CONCLUSIONS Although all of the precise targets for dasatinib are not known, this multikinase inhibitor causes either growth arrest or apoptosis in molecularly heterogeneous AML. The addition of cytotoxic or targeted agents can enhance its effects.
Collapse
Affiliation(s)
- Bella S Guerrouahen
- Division of Pediatrics, Departments of Leukemia and Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas 60611, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Roth GJ, Heckel A, Colbatzky F, Handschuh S, Kley J, Lehmann-Lintz T, Lotz R, Tontsch-Grunt U, Walter R, Hilberg F. Design, Synthesis, and Evaluation of Indolinones as Triple Angiokinase Inhibitors and the Discovery of a Highly Specific 6-Methoxycarbonyl-Substituted Indolinone (BIBF 1120). J Med Chem 2009; 52:4466-80. [DOI: 10.1021/jm900431g] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
| | | | | | | | | | | | - Ralf Lotz
- Department of Drug Discovery Support
| | | | | | - Frank Hilberg
- Boehringer Ingelheim RCV GmbH & Co. KG, A-1121 Vienna, Austria
| |
Collapse
|
17
|
Tatake RJ, O’Neill MM, Kennedy CA, Wayne AL, Jakes S, Wu D, Kugler SZ, Kashem MA, Kaplita P, Snow RJ. Identification of pharmacological inhibitors of the MEK5/ERK5 pathway. Biochem Biophys Res Commun 2008; 377:120-5. [DOI: 10.1016/j.bbrc.2008.09.087] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Accepted: 09/19/2008] [Indexed: 11/27/2022]
|
18
|
Hilberg F, Roth GJ, Krssak M, Kautschitsch S, Sommergruber W, Tontsch-Grunt U, Garin-Chesa P, Bader G, Zoephel A, Quant J, Heckel A, Rettig WJ. BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 2008; 68:4774-82. [PMID: 18559524 DOI: 10.1158/0008-5472.can-07-6307] [Citation(s) in RCA: 815] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Inhibition of tumor angiogenesis through blockade of the vascular endothelial growth factor (VEGF) signaling pathway is a novel treatment modality in oncology. Preclinical findings suggest that long-term clinical outcomes may improve with blockade of additional proangiogenic receptor tyrosine kinases: platelet-derived growth factor receptors (PDGFR) and fibroblast growth factor receptors (FGFR). BIBF 1120 is an indolinone derivative potently blocking VEGF receptor (VEGFR), PDGFR and FGFR kinase activity in enzymatic assays (IC(50), 20-100 nmol/L). BIBF 1120 inhibits mitogen-activated protein kinase and Akt signaling pathways in three cell types contributing to angiogenesis, endothelial cells, pericytes, and smooth muscle cells, resulting in inhibition of cell proliferation (EC(50), 10-80 nmol/L) and apoptosis. In all tumor models tested thus far, including human tumor xenografts growing in nude mice and a syngeneic rat tumor model, BIBF 1120 is highly active at well-tolerated doses (25-100 mg/kg daily p.o.), as measured by magnetic resonance imaging of tumor perfusion after 3 days, reducing vessel density and vessel integrity after 5 days, and inducing profound growth inhibition. A distinct pharmacodynamic feature of BIBF 1120 in cell culture is sustained pathway inhibition (up to 32 hours after 1-hour treatment), suggesting slow receptor off-kinetics. Although BIBF 1120 is rapidly metabolized in vivo by methylester cleavage, resulting in a short mean residence time, once daily oral dosing is fully efficacious in xenograft models. These distinctive pharmacokinetic and pharmacodynamic properties may help explain clinical observations with BIBF 1120, currently entering phase III clinical development.
Collapse
Affiliation(s)
- Frank Hilberg
- Boehringer Ingelheim Austria GmbH, Vienna, Austria and Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zhou J, Khng J, Jasinghe VJ, Bi C, Neo CHS, Pan M, Poon LF, Xie Z, Yu H, Yeoh AEJ, Lu Y, Glaser KB, Albert DH, Davidsen SK, Chen CS. In vivo activity of ABT-869, a multi-target kinase inhibitor, against acute myeloid leukemia with wild-type FLT3 receptor. Leuk Res 2007; 32:1091-100. [PMID: 18160102 DOI: 10.1016/j.leukres.2007.11.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 11/11/2007] [Accepted: 11/13/2007] [Indexed: 11/26/2022]
Abstract
Neoangiogenesis plays an important role in leukemogenesis. We investigated the in vivo anti-leukemic effect of ABT-869 against AML with wild-type FLT3 using RFP transfected HL60 cells with in vivo imaging technology on both the subcutaneous and systemic leukemia xenograft models. ABT-869 showed a five-fold inhibition of tumor growth in comparison with vehicle control. IHC analysis revealed that ABT-869 decreased p-VEGFR1, Ki-67 labeling index, VEGF and remarkably increased apoptotic cells in the xenograft models. ABT-869 also reduced the leukemia burden and prolonged survival. Our study supports the rationale for clinically testing an anti-angiogenesis agent in AML with wild-type FLT3.
Collapse
Affiliation(s)
- Jianbiao Zhou
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Samlowski WE, Vogelzang NJ. Emerging drugs for the treatment of metastatic renal cancer. Expert Opin Emerg Drugs 2007; 12:605-18. [PMID: 17979602 DOI: 10.1517/14728214.12.4.605] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
For decades, options for the treatment for metastatic renal cancer have been limited and mostly ineffective. During this time, immunotherapy agents, such as IFN-alpha and IL-2, have represented the major treatment options. Over the last 3 years, advances in cancer biology have characterized important signaling pathways that regulate blood vessel growth and cell proliferation. These studies have identified a number of novel 'druggable' targets. Since 2004, this has resulted in regulatory approval of four additional agents that are active against renal cancer (bevacizumab, sorafenib, sunitinib and temsirolimus). A large number of additional candidate molecules that block the vascular endothelial growth factor and mTOR pathways have subsequently been identified. These agents are rapidly progressing through clinical testing in renal cancer and in other malignancies. This paper overviews the status of these investigational agents and anticipates areas of future research and development.
Collapse
Affiliation(s)
- Wolfram E Samlowski
- Section of Melanoma, Renal Cancer and Immunotherapy Nevada Cancer Institute, One Breakthrough Way, Las Vegas, NV 89135, USA.
| | | |
Collapse
|