Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Allard Kaptein is active.

Publication


Featured researches published by Allard Kaptein.


The New England Journal of Medicine | 2016

Acalabrutinib (ACP-196) in Relapsed Chronic Lymphocytic Leukemia

John C. Byrd; Bonnie K. Harrington; Susan O'Brien; Jeffrey A. Jones; Anna Schuh; S Devereux; Jorge Chaves; William G. Wierda; Farrukh T. Awan; Jennifer R. Brown; Peter Hillmen; Deborah M. Stephens; Paolo Ghia; Jacqueline C. Barrientos; John M. Pagel; Jennifer A. Woyach; Dave Johnson; Jane Huang; Xiaolin Wang; Allard Kaptein; Brian Lannutti; Todd Covey; Maria Fardis; Jesse McGreivy; Ahmed Hamdy; Wayne Rothbaum; Raquel Izumi; Thomas G. Diacovo; Amy J. Johnson; Richard R. Furman

BACKGROUND Irreversible inhibition of Brutons tyrosine kinase (BTK) by ibrutinib represents an important therapeutic advance for the treatment of chronic lymphocytic leukemia (CLL). However, ibrutinib also irreversibly inhibits alternative kinase targets, which potentially compromises its therapeutic index. Acalabrutinib (ACP-196) is a more selective, irreversible BTK inhibitor that is specifically designed to improve on the safety and efficacy of first-generation BTK inhibitors. METHODS In this uncontrolled, phase 1-2, multicenter study, we administered oral acalabrutinib to 61 patients who had relapsed CLL to assess the safety, efficacy, pharmacokinetics, and pharmacodynamics of acalabrutinib. Patients were treated with acalabrutinib at a dose of 100 to 400 mg once daily in the dose-escalation (phase 1) portion of the study and 100 mg twice daily in the expansion (phase 2) portion. RESULTS The median age of the patients was 62 years, and patients had received a median of three previous therapies for CLL; 31% had chromosome 17p13.1 deletion, and 75% had unmutated immunoglobulin heavy-chain variable genes. No dose-limiting toxic effects occurred during the dose-escalation portion of the study. The most common adverse events observed were headache (in 43% of the patients), diarrhea (in 39%), and increased weight (in 26%). Most adverse events were of grade 1 or 2. At a median follow-up of 14.3 months, the overall response rate was 95%, including 85% with a partial response and 10% with a partial response with lymphocytosis; the remaining 5% of patients had stable disease. Among patients with chromosome 17p13.1 deletion, the overall response rate was 100%. No cases of Richters transformation (CLL that has evolved into large-cell lymphoma) and only one case of CLL progression have occurred. CONCLUSIONS In this study, the selective BTK inhibitor acalabrutinib had promising safety and efficacy profiles in patients with relapsed CLL, including those with chromosome 17p13.1 deletion. (Funded by the Acerta Pharma and others; ClinicalTrials.gov number, NCT02029443.).


Journal of Medicinal Chemistry | 2012

Irreversible protein kinase inhibitors: balancing the benefits and risks.

Tjeerd Barf; Allard Kaptein

■ INTRODUCTION In the relatively young but expanding field of irreversible kinase inhibitor drug discovery, there are two main developments that are of central importance. First, the patients have watched the first wave of low molecular weight protein kinase inhibitors becoming available to them over the past decade. These kinase inhibitors have been approved by the U.S. Food and Drug Administration (FDA) for therapeutic use in oncology indications and constitute an important addition to the arsenal of drugs to combat cancer (Table 1). Without exception, these marketed protein kinase inhibitors have been identified and developed using conventional approaches, i.e., reversible inhibitors that (partly) occlude the ATP pocket in the catalytic domain of the kinase. Although protein kinases are regarded as an attractive drug target family, it took (and still takes) huge efforts to master the human kinome, which comprises more than 500 protein kinases. The search for clinically applicable kinase inhibitors that target the highly conserved ATP pocket has been thwarted by a couple of well-known hurdles. Selectivity, cellular potency, and an increasingly crowded intellectual property arena are major points of attention. The second important development is the renewed interest in covalent binding drugs (reviewed by Potashman and Duggan and by Singh et al). This recent revival results from a better understanding of the benefits of the covalent binding principle and the approval of effective and safe covalent drugs. Historically, drug discoverers have been taught to stay away from small molecular entities that harbor reactive electrophilic groups because these used to be equivalent to promiscuity. Promiscuous hits that relied on reactive groups were traditionally hard to optimize toward leads, since these were more than often interfering with the biochemical assay rather than truly modifying the activity of the target of interest. Even if the target modulation was real, indiscriminant reactivity was believed to trigger insurmountable toxic events that may surface in late stage clinical trials when larger patient populations are involved. As a consequence, “suicide inhibitors”, “warheads”, and covalent irreversible inhibitors developed a negative flavor over time and became almost synonymous with toxicity in some organizations. The skepticism toward irreversible drugs may evaporate as more examples of irreversible drugs progress clinically that demonstrate good efficacy and safety margins. In a nutshell, the therapeutic applicability or the success of irreversible binding kinase inhibitors is dependent on whether or not the covalent bond can be confined solely to the protein kinase of interest. So this approach is in essence a story about two T’s: treatment and toxicity. When relying on the covalent binding principle, it is important to discern adduct-based toxicity and adduct-based treatment, since the adducting molecular entities in question obey overlapping fundamental rules in terms of reactivity. The only key difference is the nature and the function of the proteins that are covalently modified. Covalent kinase inhibitors with well-balanced recognition and reactivity should provide efficacy, selectivity, and ultimately the safety margins that are required for regulatory approval. If we strike the right balance, a third “T” will give enough comfort: therapeutic window. This Perspective aims to give a comprehensive account from a medicinal chemists point of view on the progress of irreversible kinase inhibitor drug discovery. The “state of the art” is reviewed by means of reported irreversible kinase inhibitors profiles and their chemical structures. The potential upsides and pitfalls that are associated with this concept are highlighted to provide a general understanding of the differences with respect to conventional drug discovery, as well as the future potential of this approach.


PLOS ONE | 2016

Preclinical Evaluation of the Novel BTK Inhibitor Acalabrutinib in Canine Models of B-Cell Non-Hodgkin Lymphoma.

Bonnie K. Harrington; Heather L. Gardner; Raquel Izumi; Ahmed Hamdy; Wayne Rothbaum; Kevin R. Coombes; Todd Covey; Allard Kaptein; Michael Gulrajani; Bart Van Lith; Cecile M. Krejsa; Christopher C. Coss; Duncan S. Russell; Xiaoli Zhang; Bridget K. Urie; Cheryl A. London; John C. Byrd; Amy J. Johnson; William C. Kisseberth

Acalabrutinib (ACP-196) is a second-generation inhibitor of Bruton agammaglobulinemia tyrosine kinase (BTK) with increased target selectivity and potency compared to ibrutinib. In this study, we evaluated acalabrutinib in spontaneously occurring canine lymphoma, a model of B-cell malignancy similar to human diffuse large B-cell lymphoma (DLBCL). First, we demonstrated that acalabrutinib potently inhibited BTK activity and downstream effectors in CLBL1, a canine B-cell lymphoma cell line, and primary canine lymphoma cells. Acalabrutinib also inhibited proliferation in CLBL1 cells. Twenty dogs were enrolled in the clinical trial and treated with acalabrutinib at dosages of 2.5 to 20mg/kg every 12 or 24 hours. Acalabrutinib was generally well tolerated, with adverse events consisting primarily of grade 1 or 2 anorexia, weight loss, vomiting, diarrhea and lethargy. Overall response rate (ORR) was 25% (5/20) with a median progression free survival (PFS) of 22.5 days. Clinical benefit was observed in 30% (6/20) of dogs. These findings suggest that acalabrutinib is safe and exhibits activity in canine B-cell lymphoma patients and support the use of canine lymphoma as a relevant model for human non-Hodgkin lymphoma (NHL).


Clinical Cancer Research | 2017

The Bruton's tyrosine kinase (BTK) inhibitor acalabrutinib demonstrates potent on-target effects and efficacy in two mouse models of chronic lymphocytic leukemia

Sarah E.M. Herman; Arnau Montraveta; Carsten U. Niemann; Helena Mora-Jensen; Michael Gulrajani; Fanny Krantz; Rose Mantel; Lisa L. Smith; Fabienne McClanahan; Bonnie K. Harrington; Dolors Colomer; Todd Covey; John C. Byrd; Raquel Izumi; Allard Kaptein; Roger Ulrich; Amy J. Johnson; Brian Lannutti; Adrian Wiestner; Jennifer A. Woyach

Purpose: Acalabrutinib (ACP-196) is a novel, potent, and highly selective Bruton tyrosine kinase (BTK) inhibitor, which binds covalently to Cys481 in the ATP-binding pocket of BTK. We sought to evaluate the antitumor effects of acalabrutinib treatment in two established mouse models of chronic lymphocytic leukemia (CLL). Experimental Design: Two distinct mouse models were used, the TCL1 adoptive transfer model where leukemic cells from Eμ-TCL1 transgenic mice are transplanted into C57BL/6 mice, and the human NSG primary CLL xenograft model. Mice received either vehicle or acalabrutinib formulated into the drinking water. Results: Utilizing biochemical assays, we demonstrate that acalabrutinib is a highly selective BTK inhibitor as compared with ibrutinib. In the human CLL NSG xenograft model, treatment with acalabrutinib demonstrated on-target effects, including decreased phosphorylation of PLCγ2, ERK, and significant inhibition of CLL cell proliferation. Furthermore, tumor burden in the spleen of the mice treated with acalabrutinib was significantly decreased compared with vehicle-treated mice. Similarly, in the TCL1 adoptive transfer model, decreased phosphorylation of BTK, PLCγ2, and S6 was observed. Most notably, treatment with acalabrutinib resulted in a significant increase in survival compared with mice receiving vehicle. Conclusions: Treatment with acalabrutinib potently inhibits BTK in vivo, leading to on-target decreases in the activation of key signaling molecules (including BTK, PLCγ2, S6, and ERK). In two complementary mouse models of CLL, acalabrutinib significantly reduced tumor burden and increased survival compared with vehicle treatment. Overall, acalabrutinib showed increased BTK selectivity compared with ibrutinib while demonstrating significant antitumor efficacy in vivo on par with ibrutinib. Clin Cancer Res; 23(11); 2831–41. ©2016 AACR.


Bioorganic & Medicinal Chemistry Letters | 2011

Structure-based lead identification of ATP-competitive MK2 inhibitors.

Tjeerd Barf; Allard Kaptein; Sander de Wilde; Ruud van der Heijden; Richard van Someren; Dennis Demont; Carsten Schultz-Fademrecht; Judith Versteegh; Mario van Zeeland; Nicole Seegers; Bert Kazemier; Bas van de Kar; Maaike van Hoek; Jeroen de Roos; Henri Klop; Rl Smeets; Claudia Hofstra; Jorrit Hornberg; Arthur Oubrie

MK2 kinase is a promising drug discovery target for the treatment of inflammatory diseases. Here, we describe the discovery of novel MK2 inhibitors using X-ray crystallography and structure-based drug design. The lead has in vivo efficacy in a short-term preclinical model.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of selective and orally available spiro-3-piperidyl ATP-competitive MK2 inhibitors

Allard Kaptein; Arthur Oubrie; Edwin de Zwart; Niels Hoogenboom; Joeri J.P. de Wit; Bas van de Kar; Maaike van Hoek; Gerard Vogel; Vera de Kimpe; Carsten Schultz-Fademrecht; Judith Borsboom; Mario van Zeeland; Judith Versteegh; Bert Kazemier; Jeroen de Roos; Frank Wijnands; John Dulos; Martin Jaeger; Paula Leandro-Garcia; Tjeerd Barf

The identification of a potent, selective, and orally available MK2 inhibitor series is described. The initial absence of oral bioavailability was successfully tackled by moving the basic nitrogen of the spiro-4-piperidyl moiety towards the electron-deficient pyrrolepyridinedione core, thereby reducing the pK(a) and improving Caco-2 permeability. The resulting racemic spiro-3-piperidyl analogues were separated by chiral preparative HPLC, and the activity towards MK2 inhibition was shown to reside mostly in the first eluting stereoisomer. This led to the identification of new MK2 inhibitors, such as (S)-23, with low nanomolar biochemical inhibition (EC(50) 7.4 nM) and submicromolar cellular target engagement activity (EC(50) 0.5 μM).


Bioorganic & Medicinal Chemistry Letters | 2010

6-Phenyl-1H-imidazo[4,5-c]pyridine-4-carbonitrile as cathepsin S inhibitors

Jiaqiang Cai; Mark Baugh; Darcey Black; Clive Long; D. Jonathan Bennett; Maureen Dempster; Xavier Fradera; Jonathan Gillespie; Fiona Elizabeth Andrews; Sylviane Boucharens; John Bruin; Kenneth S. Cameron; Iain Cumming; William Hamilton; Philip Jones; Allard Kaptein; Emma Kinghorn; Maurice Maidment; Iain Martin; Ann Mitchell; Zoran Rankovic; John E. Robinson; Paul Scullion; Joost C.M. Uitdehaag; Paul Vink; Paul Westwood; Mario van Zeeland; Leon van Berkom; Martijn Bastiani; Tommi Meulemans

6-Phenyl-1H-imidazo[4,5-c]pyridine-4-carbonitrile analogues were identified as potent and selective cathepsin S inhibitor against both purified enzyme and in human JY cell based cellular assays. This core has a very stable thio-trapping nitrile war-head in comparison with the well reported pyrimidine-2-carbonitrile cysteine cathepsin inhibitors. Compound 47 is also very potent in in vivo mouse spleenic Lip10 accumulation assays.


Cancer Research | 2015

Abstract 2596: ACP-196: a novel covalent Bruton's tyrosine kinase (Btk) inhibitor with improved selectivity and in vivo target coverage in chronic lymphocytic leukemia (CLL) patients

Todd Covey; Tjeerd Barf; Michael Gulrajani; Fanny Krantz; Bart Van Lith; Elena Bibikova; Bas van de Kar; Edwin de Zwart; Ahmed Hamdy; Raquel Izumi; Allard Kaptein

Ibrutinib, a first generation Btk inhibitor, is approved for the treatment of CLL and mantle cell lymphoma; known toxicities include atrial fibrillation, diarrhea, rash, arthralgia and bleeding events (1). Recent reports show ibrutinib9s off target effects may negatively impact its potential for combined therapy with anti-CD20 antibodies (2,3). Here we describe the pharmacologic characterization of ACP-196 a potent, novel second generation Btk inhibitor, which binds covalently to Cys481 with improved selectivity and in vivo target coverage. Compared to ibrutinib and CC-292, ACP-196 demonstrated higher selectivity for Btk when profiled against a panel of 395 non-mutant kinases (1 μM) in a competitive binding assay. IC50 determinations on 9 kinases with a Cys in the same position as Btk showed ACP-196 to be the most selective. The improved selectivity is related to the reduced intrinsic reactivity of ACP-1969s electrophile. Importantly, unlike ibrutinib, ACP-196 did not inhibit EGFR, Itk or Txk. Phosphoflow assays on EGFR expressing cell lines confirmed ibrutinib9s EGFR inhibition (EC50: 47-66 nM) with no inhibition observed for ACP-196 at 10 μM. These data may explain the ibrutinib-related incidence of diarrhea and rash. Ibrutinib9s potency on Itk and Txk may explain why it interferes with cell-mediated anti-tumor activities of therapeutic CD20 antibodies and immune-mediated killing in the tumor microenvironment (2,3). In human whole blood, ACP-196 and ibrutinib showed robust and equipotent inhibitory activity on B-cell receptor induced responses in the low nM range, whereas CC-292 was 10-20 fold less potent. In vivo, oral administration of ACP-196 in mice resulted in dose-dependent inhibition of anti-IgM-induced CD86 expression in CD19+ splenocytes with an ED50 of 0.34 mg/kg compared to 0.91 mg/kg for ibrutinib. A similar model was used to compare the duration of Btk inhibition after a single oral dose of 25 mg/kg. ACP-196 and ibrutinib inhibited CD86 expression >90% at 3h and ∼50% at 24h postdose. In contrast, CC-292 inhibited ∼50% at 3h and ∼20% at 24h postdose. An ELISA based Btk target occupancy assay was developed to measure target coverage in preclinical and clinical studies. In healthy volunteers, ACP-196 at an oral dose of 100 mg QD showed >90% target coverage over a 24h period. Btk occupancy and regulation of the PD markers (CD69 and CD86) correlated with PK parameters for exposure. In CLL patients, after 7 days of dosing with ACP-196 at 200 mg QD, 94% Btk target occupancy was observed compared with ∼80% reported for ibrutinib at 420 mg QD (4). In conclusion, ACP-196 is a novel Btk inhibitor with key pharmacologic differentiators versus ibrutinib and CC-292. ACP-196 is currently being evaluated in clinical trials. 1. IMBRUVICA package insert 2014 2. Rajasekaran Blood 2014 Abstr # 3118 3. Da Roit Haematologica 2014 4. Byrd NEJM 2013 Citation Format: Todd Covey, Tjeerd Barf, Michael Gulrajani, Fanny Krantz, Bart van Lith, Elena Bibikova, Bas van de Kar, Edwin de Zwart, Ahmed Hamdy, Raquel Izumi, Allard Kaptein. ACP-196: a novel covalent Bruton9s tyrosine kinase (Btk) inhibitor with improved selectivity and in vivo target coverage in chronic lymphocytic leukemia (CLL) patients. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2596. doi:10.1158/1538-7445.AM2015-2596


Journal of Pharmacology and Experimental Therapeutics | 2017

Acalabrutinib (ACP-196): A Covalent Bruton Tyrosine Kinase Inhibitor with a Differentiated Selectivity and In Vivo Potency Profile

Tjeerd Barf; Todd Covey; Raquel Izumi; Bas van de Kar; Michael Gulrajani; Bart Van Lith; Maaike van Hoek; Edwin de Zwart; Diana Mittag; Dennis Demont; Saskia Verkaik; Fanny Krantz; Paul G. Pearson; Roger Ulrich; Allard Kaptein

Several small-molecule Bruton tyrosine kinase (BTK) inhibitors are in development for B cell malignancies and autoimmune disorders, each characterized by distinct potency and selectivity patterns. Herein we describe the pharmacologic characterization of BTK inhibitor acalabrutinib [compound 1, ACP-196 (4-[8-amino-3-[(2S)-1-but-2-ynoylpyrrolidin-2-yl]imidazo[1,5-a]pyrazin-1-yl]-N-(2-pyridyl)benzamide)]. Acalabrutinib possesses a reactive butynamide group that binds covalently to Cys481 in BTK. Relative to the other BTK inhibitors described here, the reduced intrinsic reactivity of acalabrutinib helps to limit inhibition of off-target kinases having cysteine-mediated covalent binding potential. Acalabrutinib demonstrated higher biochemical and cellular selectivity than ibrutinib and spebrutinib (compounds 2 and 3, respectively). Importantly, off-target kinases, such as epidermal growth factor receptor (EGFR) and interleukin 2-inducible T cell kinase (ITK), were not inhibited. Determination of the inhibitory potential of anti-immunoglobulin M–induced CD69 expression in human peripheral blood mononuclear cells and whole blood demonstrated that acalabrutinib is a potent functional BTK inhibitor. In vivo evaluation in mice revealed that acalabrutinib is more potent than ibrutinib and spebrutinib. Preclinical and clinical studies showed that the level and duration of BTK occupancy correlates with in vivo efficacy. Evaluation of the pharmacokinetic properties of acalabrutinib in healthy adult volunteers demonstrated rapid absorption and fast elimination. In these healthy individuals, a single oral dose of 100 mg showed approximately 99% median target coverage at 3 and 12 hours and around 90% at 24 hours in peripheral B cells. In conclusion, acalabrutinib is a BTK inhibitor with key pharmacologic differentiators versus ibrutinib and spebrutinib and is currently being evaluated in clinical trials.


Bioorganic & Medicinal Chemistry Letters | 2012

Novel ATP competitive MK2 inhibitors with potent biochemical and cell-based activity throughout the series.

Arthur Oubrie; Allard Kaptein; Edwin de Zwart; Niels Hoogenboom; Rianne Goorden; Bas van de Kar; Maaike van Hoek; Vera de Kimpe; Ruud van der Heijden; Judith Borsboom; Bert Kazemier; Jeroen de Roos; Michiel Scheffers; Jos Lommerse; Carsten Schultz-Fademrecht; Tjeerd Barf

Optimization of our previously described pyrrolopiperidone series led to the identification of a new benzamide sub-series, which exhibits consistently high potency in biochemical and cell-based assays throughout the series. Strong inhibition of LPS-induced production of the cytokine TNFα is coupled to the regulation of HSP27 phosphorylation, indicating that the observed cellular effects result from the inhibition of MK2. X-ray crystallographic and computational analyses provide a rationale for the high potency of the series.

Collaboration


Dive into the Allard Kaptein's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmed Hamdy

Hamad Medical Corporation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John C. Byrd

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge