1
|
Papadopoulos KP, McKean M, Goldoni S, Genvresse I, Garrido MF, Li R, Wilkinson G, Kneip C, Yap TA. First-in-Human Dose-Escalation Study of the First-in-Class PDE3A-SLFN12 Complex Inducer BAY 2666605 in Patients with Advanced Solid Tumors Coexpressing SLFN12 and PDE3A. Clin Cancer Res 2024; 30:5568-5576. [PMID: 39437010 DOI: 10.1158/1078-0432.ccr-24-2713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE The study aims to evaluate the safety, tolerability, and pharmacokinetics of BAY 2666605, a velcrin that induces complex formation between the phosphodiesterase PDE3A and the protein Schlafen 12 (SLFN12), leading to a cytotoxic response in cancer cells. PATIENTS AND METHODS This was a first-in-human phase I study of BAY 2666605 (NCT04809805), an oral, potent first-in-class PDE3A-SLFN12 complex inducer, with reduced PDE3A inhibition. Adults with advanced solid tumors that coexpress SLFN12 and PDE3A received BAY 2666605 at escalating doses starting at 5 mg once daily in 28-day cycles. Forty-seven patients were prescreened for SLFN12 and PDE3A overexpression, and five biomarker-positive patients received ≥1 BAY 2666605 dose. RESULTS The most common adverse event was grade 3 to 4 thrombocytopenia in three of the five patients treated. The long half-life (>360 hours) and associated accumulation of BAY 2666605 led to the selection of an alternative schedule consisting of a loading dose with a once-daily maintenance dose. The maximum tolerated dose was not established as the highest doses of both schedules were intolerable. No objective responses were observed. Due to the high expression of PDE3A in platelets compared with tumor tissues, the ex vivo dose-dependent inhibitory effect of BAY 2666605 on megakaryocytes, and the pharmacokinetic profile of the compound, alternative schedules were not predicted to ameliorate the mechanism-based thrombocytopenia. CONCLUSIONS Despite the decreased PDE3A enzymatic inhibition profile of BAY 2666605, the occurrence of thrombocytopenia in treated patients, an on-target effect of the compound, precluded the achievement of a therapeutic window, consequently leading to trial termination.
Collapse
Affiliation(s)
| | | | - Silvia Goldoni
- Research and Early Development Oncology, Bayer Pharma, Cambridge, Massachusetts
| | | | | | - Rui Li
- Clinical Statistics, Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | | | | | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
2
|
Meanwell NA. Anagrelide: A Clinically Effective cAMP Phosphodiesterase 3A Inhibitor with Molecular Glue Properties. ACS Med Chem Lett 2023; 14:350-361. [PMID: 37077378 PMCID: PMC10108399 DOI: 10.1021/acsmedchemlett.3c00092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
The mode of action by which the orphan drug anagrelide (1), a potent cAMP phosphodiesterase 3A inhibitor, reduces blood platelet count in humans is not well understood. Recent studies indicate that 1 stabilizes a complex between PDE3A and Schlafen 12, protecting it from degradation while activating its RNase activity.
Collapse
Affiliation(s)
- Nicholas A. Meanwell
- The Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| |
Collapse
|
3
|
Kellner A, Dombi P, Illes A, Demeter J, Homor L, Ercsei I, Simon Z, Karadi E, Herczeg J, Gy Korom V, Gasztonyi Z, Szerafin L, Udvardy M, Egyed M. Anagrelide influences thrombotic risk, and prolongs progression-free and overall survival in essential thrombocythaemia vs hydroxyurea plus aspirin. Eur J Haematol 2020; 105:408-418. [PMID: 32557810 DOI: 10.1111/ejh.13459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE We report an extension study of patients with essential thrombocythaemia (ET) in the Hungarian Myeloproliferative Neoplasm (HUMYPRON) Registry, which demonstrated that over 6 years anagrelide significantly decreased the number of patients experiencing minor arterial and minor venous thrombotic events (TEs) vs hydroxyurea+aspirin. METHODS Data on patients with ET were collected through completion of a questionnaire developed according to 2008 WHO diagnostic criteria and with regard to Landolfi, Tefferi and IPSET criteria for thrombotic risk. Data were entered into the registry from 14 haematological centres. TEs, secondary malignancies, disease progression and survival were compared between patients with ET treated with anagrelide (n = 116) and with hydroxyurea+aspirin (n = 121). RESULTS Patients were followed for (median) 10 years. A between-group difference in the number of patients with TEs was observed (25.9% anagrelide vs 38.0% hydroxyurea+aspirin; P = .052). Minor arterial events were more frequently reported in the hydroxyurea+aspirin group (P < .001); there were marginally more reports of major arterial events in the anagrelide group (P = .049). TE prior to diagnosis was found to significantly influence TE incidence (P > .001). Progression-free survival (P = .004) and survival (P = .001) were significantly increased for the anagrelide group vs hydroxyurea+aspirin. CONCLUSIONS Anagrelide reduced TEs, and increased progression-free and overall survival vs hydroxyurea+aspirin over (median) 10 years.
Collapse
Affiliation(s)
- Adam Kellner
- Department of Haematology, Somogy County Kaposi Mor Teaching Hospital, Kaposvár, Hungary
| | - Peter Dombi
- Szent Borbala County Hospital, Tatabánya, Hungary
| | - Arpad Illes
- Department of Haematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Demeter
- First Department of Internal Medicine, Division of Haematology, Semmelweis University of Budapest, Budapest, Hungary
| | - Lajos Homor
- Faculty of Humanities and Social Sciences, Pazmany Peter Catholic University, Budapest, Hungary
| | - Ibolya Ercsei
- Department of Haematology, Somogy County Kaposi Mor Teaching Hospital, Kaposvár, Hungary
| | - Zsofia Simon
- Department of Haematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eva Karadi
- Department of Haematology, Somogy County Kaposi Mor Teaching Hospital, Kaposvár, Hungary
| | - Jozsef Herczeg
- Department of Haematology, Somogy County Kaposi Mor Teaching Hospital, Kaposvár, Hungary
| | - Viktoria Gy Korom
- Department of Haematology, Somogy County Kaposi Mor Teaching Hospital, Kaposvár, Hungary
| | - Zoltan Gasztonyi
- Karolina General Hospital Mosonmagyarovar, Mosonmagyarovar, Hungary
| | | | - Miklos Udvardy
- Department of Haematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklos Egyed
- Department of Haematology, Somogy County Kaposi Mor Teaching Hospital, Kaposvár, Hungary
| |
Collapse
|
4
|
Takaishi K, Takeuchi M, Tsukamoto S, Takayama N, Oshima M, Kimura K, Isshiki Y, Kayamori K, Hino Y, Oshima-Hasegawa N, Mitsukawa S, Takeda Y, Mimura N, Ohwada C, Iseki T, Nakamura S, Eto K, Iwama A, Yokote K, Nakaseko C, Sakaida E. Suppressive effects of anagrelide on cell cycle progression and the maturation of megakaryocyte progenitor cell lines in human induced pluripotent stem cells. Haematologica 2019; 105:e216-e220. [PMID: 31488559 DOI: 10.3324/haematol.2018.214841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Koji Takaishi
- Department of Hematology, Chiba University Hospital, Chiba
| | | | | | - Naoya Takayama
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba
| | - Motohiko Oshima
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo
| | - Kenji Kimura
- Department of Hematology, Chiba University Hospital, Chiba
| | - Yusuke Isshiki
- Department of Hematology, Chiba University Hospital, Chiba
| | | | - Yutaro Hino
- Department of Hematology, Chiba University Hospital, Chiba
| | | | - Shio Mitsukawa
- Department of Hematology, Chiba University Hospital, Chiba.,Department of Transfusion Medicine and Cell Therapy, Chiba University Hospital, Chiba
| | - Yusuke Takeda
- Department of Hematology, Chiba University Hospital, Chiba
| | - Naoya Mimura
- Department of Hematology, Chiba University Hospital, Chiba.,Department of Transfusion Medicine and Cell Therapy, Chiba University Hospital, Chiba
| | - Chikako Ohwada
- Department of Hematology, Chiba University Hospital, Chiba
| | - Tohru Iseki
- Department of Hematology, Chiba University Hospital, Chiba.,Department of Transfusion Medicine and Cell Therapy, Chiba University Hospital, Chiba
| | - Sou Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto
| | - Koji Eto
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba.,Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo
| | - Koutaro Yokote
- Department of Clinical Biology and Medicine, Chiba University Graduate School of Medicine, Chiba
| | | | - Emiko Sakaida
- Department of Hematology, Chiba University Hospital, Chiba
| |
Collapse
|
5
|
Stasik S, Middeke JM, Kramer M, Röllig C, Krämer A, Scholl S, Hochhaus A, Crysandt M, Brümmendorf TH, Naumann R, Steffen B, Kunzmann V, Einsele H, Schaich M, Burchert A, Neubauer A, Schäfer-Eckart K, Schliemann C, Krause S, Herbst R, Hänel M, Frickhofen N, Noppeney R, Kaiser U, Baldus CD, Kaufmann M, Rácil Z, Platzbecker U, Berdel WE, Mayer J, Serve H, Müller-Tidow C, Ehninger G, Bornhäuser M, Schetelig J, Thiede C. EZH2 mutations and impact on clinical outcome: an analysis in 1,604 patients with newly diagnosed acute myeloid leukemia. Haematologica 2019; 105:e228-e231. [PMID: 31413097 DOI: 10.3324/haematol.2019.222323] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Sebastian Stasik
- Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Jan M Middeke
- Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Michael Kramer
- Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Christoph Röllig
- Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Alwin Krämer
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Heidelberg, Germany
| | - Sebastian Scholl
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Jena, Germany
| | - Andreas Hochhaus
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Jena, Germany
| | - Martina Crysandt
- Uniklinik RWTH Aachen, Klinik für Hämatologie, Onkologie, Hämostasiologie und Stammzelltransplantation, Aachen, Germany
| | - Tim H Brümmendorf
- Uniklinik RWTH Aachen, Klinik für Hämatologie, Onkologie, Hämostasiologie und Stammzelltransplantation, Aachen, Germany
| | - Ralph Naumann
- St. Marien-Krankenhaus Siegen, Medizinische Klinik III, Siegen, Germany
| | - Björn Steffen
- Universitätsklinikum Frankfurt, Medizinische Klinik II, Frankfurt am Main, Germany
| | - Volker Kunzmann
- Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany
| | - Hermann Einsele
- Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany
| | - Markus Schaich
- Rems-Murr-Klinikum Winnenden, Klinik für Hämatologie, Onkologie und Palliativmedizin, Winnenden, Germany
| | - Andreas Burchert
- Philipps Universität Marburg, Klinik für Hämatologie, Onkologie, Immunologie, Marburg, Germany
| | - Andreas Neubauer
- Philipps Universität Marburg, Klinik für Hämatologie, Onkologie, Immunologie, Marburg, Germany
| | | | | | - Stefan Krause
- Universitätsklinikum Erlangen, Medizinische Klinik V, Erlangen, Germany
| | - Regina Herbst
- Klinikum Chemnitz, Medizinische Klinik III, Chemnitz, Germany
| | - Mathias Hänel
- Klinikum Chemnitz, Medizinische Klinik III, Chemnitz, Germany
| | | | - Richard Noppeney
- Universitätsklinikum Essen, Klinik für Hämatologie, Essen, Germany
| | - Ulrich Kaiser
- St. Bernward Krankenhaus, Medizinische Klinik II, Hildesheim, Germany
| | - Claudia D Baldus
- Charité-Universitätsmedizin Berlin, Hämatologie und Onkologie, Berlin, Germany
| | - Martin Kaufmann
- Robert-Bosch-Krankenhaus, Abteilung für Hämatologie, Onkologie und Palliativmedizin, Stuttgart, Germany
| | - Zdenek Rácil
- Masaryk University and University Hospital, Department of Internal Medicine, Hematology and Oncology, Brno, Czech Republic
| | - Uwe Platzbecker
- Universitätsklinikum Leipzig, Medizinische Klinik und Poliklinik I, Hämatologie und Zelltherapie, Leipzig, Germany
| | - Wolfgang E Berdel
- Universitätsklinikum Münster, Medizinische Klinik A, Münster, Germany
| | - Jiri Mayer
- Masaryk University and University Hospital, Department of Internal Medicine, Hematology and Oncology, Brno, Czech Republic
| | - Hubert Serve
- Universitätsklinikum Frankfurt, Medizinische Klinik II, Frankfurt am Main, Germany
| | | | - Gerhard Ehninger
- Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Martin Bornhäuser
- Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Johannes Schetelig
- Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany.,DKMS Clinical Trials Unit, Dresden, Germany
| | - Christian Thiede
- Universitätsklinikum Carl Gustav Carus, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | | |
Collapse
|
6
|
Pulkka OP, Gebreyohannes YK, Wozniak A, Mpindi JP, Tynninen O, Icay K, Cervera A, Keskitalo S, Murumägi A, Kulesskiy E, Laaksonen M, Wennerberg K, Varjosalo M, Laakkonen P, Lehtonen R, Hautaniemi S, Kallioniemi O, Schöffski P, Sihto H, Joensuu H. Anagrelide for Gastrointestinal Stromal Tumor. Clin Cancer Res 2018; 25:1676-1687. [DOI: 10.1158/1078-0432.ccr-18-0815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/23/2018] [Accepted: 12/04/2018] [Indexed: 11/16/2022]
|
7
|
The Use of Anagrelide in Myeloproliferative Neoplasms, with Focus on Essential Thrombocythemia. Curr Hematol Malig Rep 2017; 11:348-55. [PMID: 27497846 PMCID: PMC5031713 DOI: 10.1007/s11899-016-0335-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Anagrelide (ANA) is a drug with specific platelet-lowering activity, used primarily in ET, registered as a second-line drug in essential thrombocythemia (ET) in Europe and in some countries as first-line therapy, in USA licensed by FDA for thrombocythemia in myeloproliferative neoplasms (MPN). The platelet-lowering efficacy is similar to that of hydroxycarbamide (HC), around 70 % complete response and 90 % partial response. Side effects are common, especially headache and tachycardia, but usually subside or disappear within a few weeks. Around 20 % of patients stop ANA therapy due to side effects or insufficient response. Studies of treatment patterns in Europe show that ANA is preferentially given to younger patients, probably because of the concern for a possible leukemogenic effect of the common first-line drug, HC. Only two randomized studies have compared the efficacy of ANA and HC in preventing thrombosis and haemorrhage, the larger of them showing a slightly better efficacy of HC, the other showing non-inferiority of ANA to HC. A recent observational 5-year study of 3600 patients shows a low and basically similar efficacy of ANA and other cytoreductive therapies in ET. ANA does not appear to inhibit fibrosis development, and probably due to its anticoagulation properties, the combination of ASA and ANA produces an increased rate of haemorrhage. Combination of ANA with HC or interferon (IFN) is feasible and effective in patients with insufficient platelet response to mono-therapy.
Collapse
|
8
|
Ahluwalia M, Butcher L, Donovan H, Killick-Cole C, Jones PM, Erusalimsky JD. The gene expression signature of anagrelide provides an insight into its mechanism of action and uncovers new regulators of megakaryopoiesis. J Thromb Haemost 2015; 13:1103-12. [PMID: 25851510 DOI: 10.1111/jth.12959] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/02/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND Anagrelide is a cytoreductive agent used to lower platelet counts in essential thrombocythemia. Although the drug has been known to selectively inhibit megakaryopoiesis for many years, the molecular mechanism accounting for this activity is still unclear. OBJECTIVES AND METHODS To address this issue we have compared the global gene expression profiles of human hematopoietic cells treated ex-vivo with and without anagrelide while growing under megakaryocyte differentiation conditions, using high-density oligonucleotide microarrays. Gene expression data were validated by the quantitative polymerase chain reaction and mined to identify functional subsets and regulatory pathways. RESULTS We identified 328 annotated genes differentially regulated by anagrelide, including many genes associated with platelet functions and with the control of gene transcription. Prominent among the latter was TRIB3, whose expression increased in the presence of anagrelide. Pathway analysis revealed that anagrelide up-regulated genes that are under the control of the transcription factor ATF4, a known TRIB3 inducer. Notably, immunoblot analysis demonstrated that anagrelide induced the phosphorylation of eIF2α, which is an upstream regulator of ATF4, and increased ATF4 protein levels. Furthermore, salubrinal, an inhibitor of eIF2α dephosphorylation, increased the expression of ATF4-regulated genes and blocked megakaryocyte growth. CONCLUSIONS These findings link signaling through eIF2α/ATF4 to the anti-megakaryopoietic activity of anagrelide and identify new potential modulators of megakaryopoiesis.
Collapse
Affiliation(s)
- M Ahluwalia
- School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - L Butcher
- School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - H Donovan
- School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - C Killick-Cole
- School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - P M Jones
- School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - J D Erusalimsky
- School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| |
Collapse
|
9
|
Espasandin YR, Glembotsky AC, Grodzielski M, Lev PR, Goette NP, Molinas FC, Marta RF, Heller PG. Anagrelide platelet-lowering effect is due to inhibition of both megakaryocyte maturation and proplatelet formation: insight into potential mechanisms. J Thromb Haemost 2015; 13:631-42. [PMID: 25604267 DOI: 10.1111/jth.12850] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 01/04/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Anagrelide represents a treatment option for essential thrombocythemia patients. It lowers platelet counts through inhibition of megakaryocyte maturation and polyploidization, although the basis for this effect remains unclear. Based on its rapid onset of action, we assessed whether, besides blocking megakaryopoiesis, anagrelide represses proplatelet formation (PPF) and aimed to clarify the underlying mechanisms. METHODS AND RESULTS Exposure of cord blood-derived megakaryocytes to anagrelide during late stages of culture led to a dose- and time-dependent inhibition of PPF and reduced proplatelet complexity, which were independent of the anagrelide-induced effect on megakaryocyte maturation. Whereas anagrelide was shown to phosphorylate cAMP-substrate VASP, two pharmacologic inhibitors of the cAMP pathway were completely unable to revert anagrelide-induced repression in megakaryopoiesis and PPF, suggesting these effects are unrelated to its ability to inhibit phosphodiesterase (PDE) 3. The reduction in thrombopoiesis was not the result of down-regulation of transcription factors which coordinate PPF, while the myosin pathway was identified as a candidate target, as anagrelide was shown to phosphorylate the myosin light chain and the PPF phenotype was partially rescued after inhibition of myosin activity with blebbistatin. CONCLUSIONS The platelet-lowering effect of anagrelide results from impaired megakaryocyte maturation and reduced PPF, both of which are deregulated in essential thrombocythemia. These effects seem unrelated to PDE3 inhibition, which is responsible for anagrelide's cardiovascular side-effects and antiplatelet activity. Further work in this field may lead to the potential development of drugs to treat thrombocytosis in myeloproliferative disorders with an improved pharmacologic profile.
Collapse
Affiliation(s)
- Y R Espasandin
- Departamento de Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Troy S, Parks V, Purkayastha J, Gossart S, Goodman DB, Achenbach H, Armstrong M, Martin PT. Effect of anagrelide on cardiac repolarization in healthy volunteers: a randomized, double-blind, placebo- and positive-controlled, thorough QT study. Pharmacol Res Perspect 2014. [DOI: 10.1002/prp2.80] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Steven Troy
- Global Clinical Pharmacology and Pharmacokinetics; Shire Pharmaceuticals; Wayne Pennsylvania
| | - Virginia Parks
- Global Clinical Pharmacology and Pharmacokinetics; Shire Pharmaceuticals; Paris France
| | - Jaideep Purkayastha
- Clinical Development Operations and Biometrics; Shire Pharmaceuticals; Wayne Pennsylvania
| | - Sophie Gossart
- Global Clinical Pharmacology and Pharmacokinetics; Shire Pharmaceuticals; Paris France
| | | | | | | | - Patrick T. Martin
- Global Clinical Pharmacology and Pharmacokinetics; Shire Pharmaceuticals; Wayne Pennsylvania
| |
Collapse
|
11
|
Abstract
Platelets respond to a wide variety of exogenous agonists that bind to distinct receptors on the platelet surface resulting in the intracellular generation of second messengers or the opening of ion channels, setting off a cascade of events leading to both physical and functional changes in the platelet. The cyclic nucleotides, cyclic adenosine 3'5'-monophosphate (CAMP) and cyclic guanosine 3'5'-monophosphate (cGMP) initiate a sequence of intracellular events that modulate many of these reactions in the platelet.
Collapse
Affiliation(s)
- S B Sheth
- Sol Sherry Thrombosis Research Center, 3400 North Broad Street, Philadelphia, PA, 19140, USA, (215) 707-4684, (215) 707-2783
| | | |
Collapse
|
12
|
Abstract
Headache is frequently reported as one of the neurological manifestations of essential thrombocythaemia (ET) and other myeloproliferative neoplasms. It is associated with considerable morbidity; yet, it is a frequently under-recognised symptom. In patients with ET, headaches may be attributable to the disease, to the prescribed ET treatment, or unrelated to ET. The majority of headaches in ET are self-limiting and can be managed with standard headache therapies such as paracetamol, but it is vital that the clinician managing these conditions is able to recognise the headaches with a more sinister pathology. In this article, we will review the incidence and management of headaches in ET, whether they are primarily related to the disease or a result of its treatment. Identification of specific headache types in patients with ET may enable physicians to employ the most effective headache medication. This would enhance the patient-physician relationship, increasing patient compliance and thus reducing the risk of adverse outcomes.
Collapse
Affiliation(s)
- R Frewin
- Pathology Department, Gloucester Royal Hospital, Gloucester, UK.
| | | |
Collapse
|
13
|
Dingli D, Tefferi A. A critical review of anagrelide therapy in essential thrombocythemia and related disorders. Leuk Lymphoma 2009; 46:641-50. [PMID: 16019501 DOI: 10.1080/10428190400029817] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Thrombocytosis is a common clinical problem and it represents either a primary myeloid disorder (a clonal process) or a reactive phenomenon. While reactive thrombocytosis is often inconsequential, clonal thrombocytosis may require cytoreductive therapy to prevent thrombohemorrhagic complications. In this regard, a controlled clinical trial has previously demonstrated the efficacy of hydroxyurea in reducing the risk of thrombosis in high-risk patients with essential thrombocythemia (ET). Despite the absence of similar evidence for clinical benefit, the platelet-lowering agent anagrelide has been widely used in both ET and polycythemia vera (PV) and recent reports of serious side-effects suggest that such practice might be detrimental to patients. In the current review we provide basic drug information as well as a critical assessment of anagrelide treatment in ET and related disorders.
Collapse
Affiliation(s)
- David Dingli
- Division of Hematology, Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | |
Collapse
|
14
|
Petrides PE, Gisslinger H, Steurer M, Linkesch W, Krumpl G, Schüller A, Widmann R. Pharmacokinetics, bioequivalence, tolerability, and effects on platelet counts of two formulations of anagrelide in healthy volunteers and patients with thrombocythemia associated with chronic myeloproliferation. Clin Ther 2009; 31:386-98. [PMID: 19302911 DOI: 10.1016/j.clinthera.2009.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Anagrelide hydrochloride is an anti-thrombotic agent indicated for the treatment of essential thrombocythemia (ET). In various previously published clinical trials of 2 branded formulations of anagrelide in patients with ET at high risk for thrombohemorrhagic events, the rates of adverse events and discontinuation were strikingly divergent between brands. Because the formulations and manufacturers differed, the differences in tolerability, as well as platelet counts, might have been related to differences in pharmacokinetic properties between the 2 formulations. OBJECTIVES The present series of investigations (1) determined the pharmacokinetic profile of anagrelide and its metabolites; (2) compared the pharmacokinetic profiles of the test and reference formulations of anagrelide; (3) investigated the in vitro release of anagrelide as a marker of intragastric anagrelide release of the test and reference formulations; and (4) compared the platelet-reducing effects of the test and reference formulations in patients with thrombocythemia in 2 longitudinal studies over 4 weeks. METHODS A series of 4 in vivo studies and 1 in vitro study were conducted. In a pilot, prospective, singledose study in healthy volunteers, the pharmacokinetic properties (C(max), T(max), and AUC(0-infinity)) of a test formulation of anagrelide were assessed using high-performance liquid chromatography analysis of plasma samples. Based on the results from that study, a single-dose, randomized, double-blind, 2-period crossover study in healthy volunteers was conducted to determine bioequivalence of 2 formulations of anagrelide 2 mg/d (taken as 4 capsules). In vitro dissolution properties of the test or reference formulation containing 0.5 mg anagrelide as the active ingredient were studied in an assay mimicking gastrointestinal release. To test for effects on platelet counts of switching from the reference formulation (previous treatment on stable dose for 3 months) to the test formulation, two 4-week longitudinal trials were conducted: one in patients with ET (in Germany), and one in patients with thrombocythemia associated with chronic myeloproliferative disorders (CMPDs) (in Austria). RESULTS The pilot pharmacokinetic study of the test formulation in 16 volunteers (10 women, 6 men; mean [SD] age, 20.5 [1.5] years; weight, 69.0 [10.0 kg) suggested that anagrelide was metabolized to 3-hydroxyanagrelide (AUC(0-infinity) 50% compared with anagrelide) and the inactive metabolite 2-amino-5,6-dichloro-,4-dihydroquinazolone. The subsequent bioequivalence study in 24 volunteers (14 women, 10 men; mean [SD] age, 23 [4] years; white, 100%; weight, 67.5 [10.2] kg) found that the test formulation was associated with a significantly lower C(max) (point estimation [PE], 66%; 90% CI, 58%-76%; P < 0.001) and AUC(0-infinity) (PE, 77%; 90% CI, 68%-86%; P = 0.001). T(max) values for anagrelide and 3-hydroxyanagrelide were 1 hour longer with the test formulation compared with the reference formulation. The total number of adverse events with the reference formulation was 46; the test formulation, 29 (P = 0.05). In vitro, anagrelide from the reference formulation was immediately released (89.1% at 5 minutes), whereas there was a delayed release (93.6% at 30 minutes) from the test formulation (P < 0.05). In the last 2 studies, 2 cohorts of white patients (cohort 1, 15 patients with ET; 10 women, 5 men; mean [SD] age, 49.0 [10.7] years [range, 31-66 years]; weight, 73.2 [12.6] kg; cohort 2, 19 patients with thrombocythemia associated with CMPD; 12 women, 7 men; age, 62.6 [12.4] years [range, 38-80 years]; weight, 66.1 [13.3] kg) who had received treatment for > or =3 months with the reference formulation were switched to the same dose of the test formulation and maintained on this dose for 4 weeks. Platelet counts did not change significantly from baseline over 4 weeks and stayed within a predefined margin of 150 x 10(3) cells/microL. CONCLUSIONS The pharmacokinetic properties, adverse event rates, and in vitro dissolution profile differed between the test and reference anagrelide formulations in these healthy volunteers. In patients with ET or thrombocythemia associated with CMPD, platelet counts did not differ significantly from baseline at 4 weeks when subjects were switched from the reference to the test anagrelide formulation.
Collapse
Affiliation(s)
- Petro E Petrides
- Hematology Oncology Center Munich and Ludwig Maximilians University of Munich Medical School, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Stoclet JC, Keravis T, Komas N, Lugnier C. Section Review: Cardiovascular & Renal: Cyclic nucleotide phosphodiesterases as therapeutic targets in cardiovascular diseases. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.4.11.1081] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
16
|
Hong Y, Wang G, Del Arroyo AG, Hernandez J, Skene C, Erusalimsky JD. Comparison between anagrelide and hydroxycarbamide in their activities against haematopoietic progenitor cell growth and differentiation: selectivity of anagrelide for the megakaryocytic lineage. Leukemia 2006; 20:1117-22. [PMID: 16557242 DOI: 10.1038/sj.leu.2404180] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Anagrelide (ANA) and hydroxycarbamide (HC) are two distinct pharmacological agents used to treat thrombocythaemia associated with myeloproliferative disorders. Although both drugs have been in clinical use for a number of years, comparative studies of their selectivity and mode of action are still lacking. Here, we have evaluated the activities of ANA and HC on the growth and differentiation of human haematopoietic progenitor cells in liquid culture. Both drugs inhibited thrombopoietin-induced megakaryocytopoiesis in a dose-dependent manner, but with strikingly different potencies (IC(50)=26 nM for ANA and 30 muM for HC) and modes of action. Whereas HC inhibited cell proliferation, ANA acted primarily on the differentiation process. At doses that abrogated megakaryocytopoiesis, HC also inhibited the expansion of CD34(+) cells stimulated by stem cell factor, interleukin-3 and Flt-3 ligand and also induced apoptosis. Furthermore, HC inhibited erythroid and myelomonocytic cell growth, induced by erythropoietin or granulocyte-macrophage colony-stimulating factor, respectively. In contrast, ANA showed none of these additional effects. Taken together, these results demonstrate that ANA is a potent and selective inhibitor of megakaryocytopoiesis, having no significant activity against haematopoietic progenitor cell expansion or differentiation into other lineages. In contrast, the anti-megakaryocytopoietic activity of HC cannot be dissociated from its more general cytoreductive and cytotoxic actions.
Collapse
Affiliation(s)
- Y Hong
- The Wolfson Institute for Biomedical Research, University College London, London, UK
| | | | | | | | | | | |
Collapse
|
17
|
Wang G, Franklin R, Hong Y, Erusalimsky JD. Comparison of the biological activities of anagrelide and its major metabolites in haematopoietic cell cultures. Br J Pharmacol 2006; 146:324-32. [PMID: 16041400 PMCID: PMC1576287 DOI: 10.1038/sj.bjp.0706341] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The platelet-lowering drug anagrelide inhibits bone marrow megakaryocytopoiesis by an unknown mechanism. Recently, it was found that anagrelide is bio-transformed in humans into two major metabolites (6,7-dichloro-3-hydroxy-1,5 dihydro-imidazo[2,1-b]quinazolin-2-one (BCH24426) and 2-amino-5,6-dichloro-3,4,-dihydroquinazoline (RL603). Whether these metabolites have biological activities that may underlie the mode of action of the parent drug is presently unclear. To clarify this question here we have compared the activities of anagrelide, BCH24426 and RL603 on the growth and differentiation of CD34(+) haematopoietic progenitor cells in liquid culture and on the migration of differentiated megakaryocytes. Incubation with either anagrelide, BCH24426 or RL603 did not affect the early expansion of CD34(+) cells stimulated by thrombopoietin. In contrast, both anagrelide and BCH24426 potently inhibited the development of megakaryocytes (IC(50) +/- s.e.m. = 26 +/- 4 and 44 +/- 6 nM, respectively), whereas RL603 showed no significant effect. Anagrelide and BCH24426 did not affect erythroid or myelomonocytic differentiation stimulated by erythropoietin or granulocyte-macrophage colony-stimulating factor, demonstrating the selectivity of these compounds against the megakaryocytic lineage. Neither anagrelide nor its metabolites showed a significant effect on the migratory response of megakaryocytes towards stromal cell-derived factor-1alpha. Although BCH24426 was shown to be considerably more potent than anagrelide as an inhibitor of phosphodiesterase type III (PDEIII) (IC(50) = 0.9 vs 36 nM) this activity did not correlate with the potency of inhibition of megakaryocyte development. Furthermore, other PDEIII inhibitors of widely differing potency were shown to have negligible effects on megakaryocytopoiesis. Taken together our results demonstrate that anagrelide and BCH24426 target a cellular event involved specifically in the megakaryocyte differentiation programme, which is independent of PDEIII inhibition.
Collapse
Affiliation(s)
- Guosu Wang
- The Wolfson Institute for Biomedical Research, University College London, Cruciform Building, Gower Street, London WC1E 6BT
| | - Richard Franklin
- Shire Pharmaceutical Development, Hampshire International Business Park, Chineham, Basingstoke, Hampshire RG24 8EP
| | - Ying Hong
- The Wolfson Institute for Biomedical Research, University College London, Cruciform Building, Gower Street, London WC1E 6BT
| | - Jorge D Erusalimsky
- The Wolfson Institute for Biomedical Research, University College London, Cruciform Building, Gower Street, London WC1E 6BT
- Author for correspondence:
| |
Collapse
|
18
|
Petrides PE. Anagrelide: a decade of clinical experience with its use for the treatment of primary thrombocythaemia. Expert Opin Pharmacother 2005; 5:1781-98. [PMID: 15264993 DOI: 10.1517/14656566.5.8.1781] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Primary thrombocythaemia (PT) is the most frequent among the rare chronic myeloproliferative disorders. Life expectancy is determined by thromboembolic and haemorrhagic complications, which can be prevented by cytoreductive therapy. For a long time, hydroxyurea has been considered as the therapeutic gold standard. However, hydroxyurea treatment is not lineage-specific, may not be tolerated because of adverse effects (skin, gastrointestinal tract) and is leukaemogenic when sequentially used with other DNA-targeting drugs. Hence, anagrelide was welcomed in 1988 when it was first described as being efficient at normalising elevated platelet counts, specific for megakaryocytes and non-mutagenic. Since then, anagrelide has been approved in the US and Canada (Agrylin), Shire Pharmaceuticals) as well as in Austria and other countries of the EU (Thromboreductin), AOP Orphan Pharmaceuticals). Clinical Phase III trials (PT1 and ANAHYDRET) are underway to directly compare the efficacy and safety of anagrelide and hydroxyurea.
Collapse
Affiliation(s)
- Petro E Petrides
- Hematology Oncology Center, Zweibruckenstr. 2, 80331 Munich, Germany.
| |
Collapse
|
19
|
Leoncini G, Signorello MG, Bruzzese D, Di Braccio M, Grossi GC, Roma G. Mechanisms involved in the antiplatelet activity of 8-methyl-4-(1-piperazinyl)-7-(3-pyridinylmethoxy)-2H-1-benzopyran-2-one (RC414). Biochem Pharmacol 2004; 67:911-8. [PMID: 15104244 DOI: 10.1016/j.bcp.2003.10.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The effect on human platelets of 8-methyl-4-(1-piperazinyl)-7-(3-pyridinylmethoxy)-2H-1-benzopyran-2-one (RC414) was tested in vitro by measuring aggregation induced by several agonists, cAMP and cGMP levels, cAMP phosphodiesterase and PKC activities and [Ca2+]i. The RC414 effect on nitric oxide production was also evaluated. RC414 in a dose-dependent manner inhibited aggregation both in platelet rich plasma and in washed platelets. It was particularly effective in platelets challenged by collagen, ADP and thrombin: IC50 values are 0.51 +/- 0.12 microM, 0.98 +/- 0.36 microM and 1.00 +/- 0.15 microM, respectively. RC414 increased cAMP levels, through the specific inhibition of the cAMP high affinity phosphodiesterase (IC50 = 1.73 +/- 0.35 microM). RC414 reduced [Ca2+]i transients and PKC activation induced by thrombin. In addition RC414 was able to increase nitric oxide formation involving the stimulation of constitutive nitric oxide synthase enzyme. In conclusion, RC414 exerts its powerful anti-platelet activity by increasing cAMP intracellular levels and nitric oxide formation.
Collapse
Affiliation(s)
- Giuliana Leoncini
- Department of Experimental Medicine, Biochemistry Section, University of Genoa, Viale Benedetto XV 1, 16132 Genova, Italy.
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
Platelets are intimately involved in the pathogenesis of thromboembolic disorders, especially arterial forms of thrombosis. Although most arterial thromboses develop on the basis of endothelial injuries, some do not. In these instances "hyperactive" platelets could be the cause. Hyperaggregable platelets have been described in association with a number of acquired disease entities whereby the cause-and-effect relationship is unclear. In contrast, the sticky platelet syndrome is a congenital, autosomal dominant disorder, characterized by hyperaggregable platelets in response to ADP, epinephrine, or both. Patients usually seek treatment for transient or permanent arterial vascular occlusions. These are often precipitated by stressful events. Treatment with low-dose aspirin (81 mg/day) reverses clinical symptoms and hyperaggregability in the laboratory.
Collapse
Affiliation(s)
- Eugene P Frenkel
- Department of Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8852, USA.
| | | |
Collapse
|
21
|
Abstract
Over a century has elapsed since the first description of polycythemia vera (PV), and current treatment recommendations are primarily based on the results of clinical trials that were performed in the late 1960s and early 1970s. Continued identification and appropriate utilization of PV-specific biologic parameters may allow substantial modification of early diagnostic criteria. New cytoreductive treatment agents are increasingly being used without any evidence of superiority over conventional therapy. The role of aspirin is being readdressed by an ongoing controlled study. Transformation of PV into either myelofibrosis with myeloid metaplasia or acute leukemia remains a major complication that may not be influenced by current therapy.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Lane WJ, Hattori K, Dias S, Peerschke EI, Moore MA, Blanset DL, Lang PC, Petrone M, Rafii S. Anagrelide metabolite induces thrombocytopenia in mice by inhibiting megakaryocyte maturation without inducing platelet aggregation. Exp Hematol 2001; 29:1417-24. [PMID: 11750100 DOI: 10.1016/s0301-472x(01)00742-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The mechanism for anagrelide's potent platelet lowering activity in human subjects is not well defined. Studies related to anagrelide function have been hampered by its lack of activity in nonhuman primates and water insolubility. In an effort to define the mechanism whereby anagrelide exerts its therapeutic effect, we identified a water-soluble metabolite (anagrelide.met). The availability of anagrelide.met allowed, for the first time, parallel in vitro and in vivo animal studies centered on the mechanisms by which anagrelide lowers platelet levels. MATERIALS AND METHODS The effects of anagrelide.met on proliferation and maturation of mega-karyocytes (MKs) as well as platelet production were studied both in vitro and in vivo. RESULTS Anagrelide.met is capable of blocking in vitro MK migration by 20% to 40%. At 100 ng/mL, anagrelide.met selectively blocked in vitro MK maturation, resulting in a 50% decrease in the total number of CD41a(+) MKs, corresponding with a 30% decrease in MK ploidy by day 10 and a 60% decrease by day 20. Daily intraperitoneal injections of anagrelide.met 100 microg into BALB/c mice was sufficient to significantly decrease platelet counts within 24 to 48 hours, stabilizing to 40 to 50% of normal levels by day 5. This was associated with a 45% decrease in the number of developing MKs and an increase in thrombopoietin levels. Anagrelide.met did not alter WBC counts, hematocrit, or bleeding time, or lead to any apparent signs of toxicity. Furthermore, unlike the parent anagrelide compound, anagrelide.met did not inhibit ADP-induced platelet aggregation even at high concentrations (10 microg/mL). CONCLUSIONS We describe a cross-species reactive anagrelide metabolite that selectively inhibits MK maturation and migration, lowering platelet levels without influencing platelet aggregation.
Collapse
Affiliation(s)
- W J Lane
- Division of Hematology-Oncology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Chezal JM, Moreau E, Delmas G, Gueiffier A, Blache Y, Grassy G, Lartigue C, Chavignon O, Teulade JC. Heterocyclization of functionalized vinylic derivatives of imidazo. J Org Chem 2001; 66:6576-84. [PMID: 11578207 DOI: 10.1021/jo015582x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Heterocyclization of functionalized vinylic derivatives of imidazo[1,2-a]pyridines was explored experimentally and theoretically using semiempirical AM1 and ab initio methods. A range of functionalized vinylic derivatives (azido, amino, and carbodiimide groups) were prepared for conversion into pyrroloazaindoles 19-22, imidazo[1,x]-, (x = 5, 6, 7, 8), [2,6]-, and [2,7]naphthyridines 28-30, 35-38 by thermal reaction. In the case of vinylic groups in the 5 position, peri annulation also was observed. The experimental and theoretical data are compared and discussed.
Collapse
Affiliation(s)
- J M Chezal
- UMR INSERM 484, Université d'Auvergne, Faculté de Pharmacie, 28 Pl. H. Dunant, B.P. 38, 63001 Clermont-Ferrand CEDEX 1, France
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Myeloproliferative diseases (MPD) include polycythemia vera (PV), essential thrombocythemia, agnogenic myeloid metaplasia, and chronic myelogenous leukemia. The focus of this report is on PV, which is characterized by an increase in red blood cells, granulocytes, and platelets. Complications associated with PV are an increased risk of thrombosis and abnormal bleeding. Phlebotomy to a hematocrit less than 45% is the mainstay of treatment for erythrocythemia, but may further increase the platelet count, necessitating the use of a platelet-lowering agent in conjunction with phlebotomy. Other treatment strategies include low-dose aspirin or other antithrombotic therapy and cytoreduction. Mounting evidence of the leukemogenicity and mutagenicity of radioactive phosphorus and alkylating agents, as administered using "conventional" regimens, has restricted the liberal use of these treatments. Three drugs have emerged as useful because of their efficacy in reducing the elevated platelet count: anagrelide, hydroxyurea (HU), and interferon alfa (IFN). It is clear that no single agent satisfies all the needs for cytoreduction that arise during the course of PV. Future protocols should be designed that draw on the large body of experience already gained with these drugs to transcend the limitations of single-agent therapy and to improve quality of life as well as survival. Semin Hematol 38(suppl 2):25-28.
Collapse
Affiliation(s)
- H S Gilbert
- Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
25
|
Laguna MS, Kornblihtt LI, Marta RF, Michiels JJ, Molinas FC. Effectiveness of anagrelide in the treatment of symptomatic patients with essential thrombocythemia. Clin Appl Thromb Hemost 2000; 6:157-61. [PMID: 10898276 DOI: 10.1177/107602960000600307] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We prospectively evaluated the effect of anagrelide on platelet counts and the clinical manifestations of microvascular circulation disturbances in 17 newly diagnosed patients with essential thrombocythemia. Ten patients had symptoms related to thrombocythemia, eight at the time of starting anagrelide treatment. The platelet counts before anagrelide treatment and during maintained remission of essential thrombocythemia by anagrelide were 980 (range, 610-2030) and 378 (range, 212-546) x 10(9)/L, respectively. Spontaneous platelet aggregation was found in 6 patients (35%), which disappeared on remission of essential thrombocythemia in five cases (P = 0.02). Essential thrombocythemia-related microvascular thrombotic and hemorrhagic symptoms disappeared with the normalization of platelet count in all cases during maintained remission of essential thrombocythemia by long term continuous anagrelide treatment with a follow-up period of between 2 and 6 years. However, ET-related symptoms reappeared in three patients, coinciding with increased platelet count up to 600 x 10(9)/L caused by anagrelide dose reduction. We conclude that reduction of increased platelet to normal (< 400 x 10(9)/L) in symptomatic patients with essential thrombocythemia through use of maintained anagrelide treatment is associated with the disappearance of spontaneous platelet aggregation and the complete relief of thrombotic and hemorrhagic manifestations.
Collapse
Affiliation(s)
- M S Laguna
- Sección Hematología Investigación, Instituto de Investigaciones Médices Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
26
|
Abstract
The activity of phosphodiesterase (PDE)3A requires divalent cations. Putative metal-binding sites are expected at 2 highly conserved metal-binding motifs, HXXXH(X)25E. A functional truncated recombinant PDE3A containing the catalytic domain (PDE3A▵1) and mutant proteins were expressed in a baculovirus/Sf9 cell system. All the mutant proteins had decreased catalytic efficiency (kcat/Km). Mutants H752A, H756A, and E825A had kcat of less than 0.0008 s−1 to 0.0475 s−1 compared to PDE3A▵1, with 1.86 second−1, with unchanged Km. Although E866A had a kcat of 0.235 s−1, the Kmfor cyclic adenosine monophosphate (cAMP) was increased 11-fold and the Ki for cyclic guanosine monophosphate (cGMP) was 27-fold higher than PDE3A▵1. The Ki of H836A for cGMP was 177-fold higher than that of PDE3A▵1. The Km for E971A was 5-fold higher than PDE3A▵1. These results suggest that the cAMP and cGMP binding sites are overlapping, but not identical, involving both common and different amino acids. Mutants E825A, H836A, and E866A showed low activity in a metal ion-free assay; however, their enzymatic activities were increased 4- to 10-fold in buffers containing Mn2+, Mg2+, or Co2+. This observation indicates that conserved amino acids in the second metal-binding motif might not be involved in binding divalent cations but may serve other functions such as substrate or inhibitor binding in PDE3A.
Collapse
|
27
|
Conserved amino acids in metal-binding motifs of PDE3A are involved in substrate and inhibitor binding. Blood 2000. [DOI: 10.1182/blood.v95.11.3380] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe activity of phosphodiesterase (PDE)3A requires divalent cations. Putative metal-binding sites are expected at 2 highly conserved metal-binding motifs, HXXXH(X)25E. A functional truncated recombinant PDE3A containing the catalytic domain (PDE3A▵1) and mutant proteins were expressed in a baculovirus/Sf9 cell system. All the mutant proteins had decreased catalytic efficiency (kcat/Km). Mutants H752A, H756A, and E825A had kcat of less than 0.0008 s−1 to 0.0475 s−1 compared to PDE3A▵1, with 1.86 second−1, with unchanged Km. Although E866A had a kcat of 0.235 s−1, the Kmfor cyclic adenosine monophosphate (cAMP) was increased 11-fold and the Ki for cyclic guanosine monophosphate (cGMP) was 27-fold higher than PDE3A▵1. The Ki of H836A for cGMP was 177-fold higher than that of PDE3A▵1. The Km for E971A was 5-fold higher than PDE3A▵1. These results suggest that the cAMP and cGMP binding sites are overlapping, but not identical, involving both common and different amino acids. Mutants E825A, H836A, and E866A showed low activity in a metal ion-free assay; however, their enzymatic activities were increased 4- to 10-fold in buffers containing Mn2+, Mg2+, or Co2+. This observation indicates that conserved amino acids in the second metal-binding motif might not be involved in binding divalent cations but may serve other functions such as substrate or inhibitor binding in PDE3A.
Collapse
|
28
|
Lackner H, Urban C, Beham-Schmid C, Benesch M, Kerbl R, Schwinger W. Treatment of children with anagrelide for thrombocythemia. J Pediatr Hematol Oncol 1998; 20:469-73. [PMID: 9787322 DOI: 10.1097/00043426-199809000-00012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE Essential thrombocythemia (ET) is a rare myeloproliferative disease characterized by hyperproliferation of megakaryocytes and persistent elevation of platelets. Major complications are thrombosis, bleeding, and microvascular occlusive symptoms, sometimes resulting in serious morbidity. In patients with platelet counts > 1000 x 10(9)/l, cytoreduction is important to reduce the risk for thrombotic or hemorrhagic events. However, the frequent side effects and the carcinogenic potential of many cytoreductive agents limit their use, especially in younger patients. Anagrelide, a noncarcinogenic agent with moderate toxicity, has been successfully used in adult patients with ET, but experience with anagrelide in pediatric patients with ET is limited. PATIENTS AND METHODS Three children who received anagrelide (Induction dose 1 mg/day, maintenance dose 1 to 2.5 mg/day) as therapy for ET are described. RESULTS All children had a rapid and marked decline of their platelet counts, and only one child developed mild and transient abdominal side effects. CONCLUSIONS Treatment with anagrelide seems to be a promising approach for children with ET. It should be considered as first-line therapy because of its selective activity against platelet production, tolerable toxicity, suspected negligible cancerigenic effect, and the possibility of oral administration.
Collapse
Affiliation(s)
- H Lackner
- Department of Pediatrics, University of Graz, Austria
| | | | | | | | | | | |
Collapse
|
29
|
Leoncini G, Signorello MG, Roma G, Di Braccio M. Effect of 2-(1-piperazinyl)-4H-pyrido[1,2-a]pyrimidin-4-one (AP155) on human platelets in vitro. Biochem Pharmacol 1997; 53:1667-72. [PMID: 9264319 DOI: 10.1016/s0006-2952(97)00019-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The effect on human platelets of 2-(1-piperazinyl)-4H-pyrido[1,2-a]pyrimi din-4-one (AP155) was tested in vitro by measuring cyclic adenosine monophosphate (cAMP) level, cytosolic Ca++, [(125I)]fibrinogen binding as well as aggregation induced by several agonists. AP155 dose-dependently inhibited aggregation both in platelet rich plasma (PRP) and in washed platelets (WP), exerting its maximal power in the presence of collagen, ADP and platelet activating factor (PAF). It specifically inhibited the activity of cAMP high affinity phosphodiesterase (PDE), resulting in a sufficient increase in cAMP levels to activate cAMP-dependent protein kinase. AP155 was able to inhibit aggregation, the increase in cytosolic Ca++ induced by thrombin, and fibrinogen binding to ADP or thrombin-stimulated platelets. Thus, this new pyridopyrimidine derivative exerts its antiplatelet activity by increasing cAMP intracellular concentration.
Collapse
Affiliation(s)
- G Leoncini
- Istituto di Chimica Biologica, Università di Genova, Italy
| | | | | | | |
Collapse
|
30
|
Tefferi A, Elliott MA, Solberg LA, Silverstein MN. New drugs in essential thrombocythemia and polycythemia vera. Blood Rev 1997; 11:1-7. [PMID: 9218101 DOI: 10.1016/s0268-960x(97)90001-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Among the chronic myeloproliferative disorders, polycythemia vera and essential thrombocythemia are unique because of their association with thrombohemorrhagic manifestations and their relatively indolent clinical course. Patients with essential thrombocythemia may not have a significant shortening of life-expectancy and most may not require specific therapy. However, patients with polycythemia vera have a significant risk of transformation of polycythemia vera into acute leukemia or postpolycythemic myelofibrosis (or both). 'High-risk-for-thrombosis' patients with either polycythemia vera or essential thrombocythemia require specific therapy with a platelet-lowering agent to prevent thrombotic complications. Currently, the standard agent used for this is hydroxyurea. However, its tetratogenic and leukemogenic potential has been of concern. As a result, new platelet-lowering agents are being evaluated in the treatment of polycythemia vera and essential thrombocythemia. Anagrelide and interferon alfa are two such agents and have been shown to be effective in reducing platelet counts in patients with chronic myeloproliferative disorders. The putative mechanism of action of these drugs, their specific activity in polycythemia vera and essential thrombocythemia, side-effect profile, and current indications are discussed herein.
Collapse
Affiliation(s)
- A Tefferi
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
31
|
Trinka P, Sl�gel P, Reiter J. A convenient synthesis of ethyl (2-amino-5,6-dichlorobenzyl)glycinate. ACTA ACUST UNITED AC 1996. [DOI: 10.1002/prac.199633801129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Handa RK, Dillingham EO, Gollamudi R. Antithrombotic nipecotamide increases cyclic AMP levels and inhibits protein phosphorylation in human platelets. Life Sci 1995; 57:983-8. [PMID: 7643723 DOI: 10.1016/0024-3205(95)02033-f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
alpha,alpha'-bis[3-(N,N-diethylcarbamoyl)piperidino]-p-xylene dihydrobromide (A-1), a typical antithrombotic nipecotamide, elevated the levels of cyclic adenosine monophosphate (cAMP) in human platelets in vitro, without inhibiting cAMP-phosphodiesterase (PDE). The compound elevated the basal cAMP levels, enhanced the prostaglandin (PG)E1-stimulated platelet adenylyl cyclase (AC) activity, and prevented the ADP-induced decline of the latter. Collagen-induced phosphorylation of 20 and 47 kDa proteins was inhibited by IC50 and 0.5 x IC50 concentrations. In light of the known actions of A-1, it is suggested that stimulation of AC and inhibition of agonist-induced rise in cytosolic ionized calcium ([Ca2+]i) may constitute an aspect of its mechanism of action.
Collapse
Affiliation(s)
- R K Handa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee, Memphis 38163, USA
| | | | | |
Collapse
|
33
|
Affiliation(s)
- S Garattini
- Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
34
|
Seiler S, Gillespie E, Arnold AJ, Brassard CL, Meanwell NA, Fleming JS. Imidazoquinoline derivatives: potent inhibitors of platelet cAMP phosphodiesterase which elevate cAMP levels and activate protein kinase in platelets. Thromb Res 1991; 62:31-42. [PMID: 1649498 DOI: 10.1016/0049-3848(91)90666-k] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Compounds containing the imidazoquinoline nucleus are a new class of potent, broad-spectrum inhibitors of platelet aggregation. This report describes studies with a simply-substituted imidazoquinoline (BMY 20844) and several new ether-linked side chain derivatives (BMY 21638 and BMY 43351). These compounds are potent inhibitors of platelet cAMP phosphodiesterase (IC50 values: BMY 20844, 1.3 X 10(-8); BMY 21638, 2 X 10(-10); and BMY 43351, 1 X 10(-10) M, measured using 0.15 microM cAMP) but have little effect on platelet homogenate cGMP phosphodiesterase (IC50 greater than 10(-5) M). Inhibition of different cAMP phosphodiesterase isozymes was tested to determine if the compounds inhibited similar isozymes in other tissues. Rabbit heart cAMP phosphodiesterase isozymes were resolved by ion-exchange chromatography and three peaks of activity were obtained. BMY 20844 inhibited only fraction III (a "cGMP-inhibitable, low Km" cAMP-specific phosphodiesterase) with an IC50 value of 5 X 10(-8) M. These compounds also inhibited canine cardiac sarcoplasmic reticulum membrane-bound "cGMP-inhibitable, low Km" cAMP-specific phosphodiesterase with virtually the same potency as inhibition of cAMP phosphodiesterase in platelet homogenate. In washed platelets these compounds elevated cAMP levels and activated the platelet cAMP dependent protein kinase. Activation of cAMP-dependent protein kinase was determined by cAMP-dependent protein kinase ratio measurements and phosphorylation of intracellular proteins. These studies suggest that this potent new class of agents inhibits platelet phosphodiesterase activity in intact platelets causing an elevation in cAMP levels sufficient to activate the cAMP-dependent protein kinase and stimulate protein phosphorylation. This mechanism is, at least in part, responsible for the ability of these compounds to prevent platelet aggregation and thrombosis in experimental animal models.
Collapse
Affiliation(s)
- S Seiler
- Department of Cardiovascular Biochemistry, Bristol-Myers Squibb Company, Wallingford, Connecticut 06492
| | | | | | | | | | | |
Collapse
|