throbber
R E V I E W
`
`doi: 10.1111/sji.12069
`
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`Inhibitors of BTK and ITK: State of the New Drugs for
`Cancer, Autoimmunity and Inflammatory Diseases
`
`L. Vargas*, A. Hamasy*†, B. F. Nore*‡ & C. I. E. Smith*
`
`Abstract
`
`*Department of Laboratory Medicine, Clinical
`Research Center, Karolinska Institutet,
`Karolinska University Hospital, Huddinge,
`Sweden; †Department of Clinical Biochemistry,
`College of Pharmacy, Hawler Medical University,
`Erbil, Kurdistan Region, Iraq (none of the work
`was carried out in Hawler Medical University,
`but the author receives salary from this
`university); and ‡Department of Biochemistry,
`School of Medicine, Faculty of Medical Sciences,
`University of Sulaimani, Sulaimani, Kurdistan
`Region, Iraq
`
`Received 18 April 2012; Accepted in revised
`form 5 May 2013
`
`Correspondence to: C. I. Edvard Smith, Karolinska
`Institutet, Clinical Research Center,
`level 5
`Novum, SE-141 86 Huddinge, Sweden. E-mail:
`edvard.smith@ki.se
`
`BTK and ITK are cytoplasmic tyrosine kinases of crucial importance for B and
`T cell development, with loss-of-function mutations causing X-linked agam-
`maglobulinemia and susceptibility to severe, frequently lethal, Epstein–Barr
`virus infection, respectively. Over the last few years, considerable efforts have
`been made in order to develop small-molecule inhibitors for these kinases to treat
`lymphocyte malignancies, autoimmunity or allergy/hypersensitivity. The ratio-
`nale is that even if complete lack of BTK or ITK during development causes
`severe immunodeficiency, inactivation after birth may result in a less severe
`phenotype. Moreover, therapy can be transient or only partially block the activity
`of BTK or ITK. Furthermore, a drug-induced B cell deficiency is treatable by
`gamma globulin substitution therapy. The newly developed BTK inhibitor
`PCI-32765, recently renamed Ibrutinib, has already entered several clinical trials
`for various forms of non-Hodgkin lymphoma as well as for multiple myeloma.
`Experimental animal studies have demonstrated highly promising treatment
`effects also in autoimmunity.
`ITK inhibitors are still under
`the early
`developmental phase, but it can be expected that such drugs will also become
`very useful. In this study, we present BTK and ITK with their signalling
`pathways and review the development of the corresponding inhibitors.
`
`Introduction
`
`BTK and ITK are TEC family kinases (TFKs) and loss-of-function
`mutations cause human disease
`
`Before reviewing the newly developed inhibitors of BTK
`and ITK, we provide a background to these tyrosine
`kinases. In the first section, we discuss their identification
`and the effect of inactivating mutations. In the following
`section, we describe the intracellular signalling pathway of
`BTK and ITK and summarize what is known about their
`regulation. This is followed by the description of the
`inhibitors.
`TFKs, consisting of BTK, BMX (ETK), ITK, TEC and
`TXK (RLK),
`form the second largest family of non-
`receptor kinases in humans, the largest being the SRC
`family. The TFK ancestor emerged already prior to the
`evolution of metazoans and shows evidence of differential
`evolutionary wiring [1–3]. BTK and ITK contain an
`N-terminal Pleckstrin homology (PH) domain, followed
`by a Tec homology (TH), Src homology (SH)-3, -2 and -1
`(catalytic) domains [4–6]. As depicted in Fig. 1, the TH
`domain consists of an N-terminal Zn2+-binding BTK
`motif, and one or two proline-rich motifs [4, 7–9].
`
`All the mammalian TFKs were identified in the 1990s.
`ITK [10, 11] and BTK [12, 13] were each cloned
`independently by two groups. This family of kinases soon
`received wide interest, owing to the fact
`that BTK
`mutations cause an X-linked form of B-lymphocyte
`deficiency (X-linked agammaglobulinemia, XLA) in man
`[13–16]. Today, more than 1000 patients with known
`mutations exist in the BTKbase registry [17]. In mice,
`mutations
`cause
`the phenotypically milder X-linked
`immunodeficiency (Xid) [18, 19]. In mice, it seems as if
`the TEC kinase has a unique compensatory role, because,
`while the TEC kinase is also expressed in human B
`lymphocytes, in mice, the double knockout of BTK and
`TEC causes an XLA-like phenotype. In contrast, TEC
`single-knockout mice do not have an overt phenotype [20].
`ITK deficiency in humans is much less common and was
`not reported until 16 years after the identification of
`mutations in BTK. Thus, ITK is one of the several genes in
`which loss-of-function mutations cause susceptibility to
`severe, often fatal, Epstein–Barr virus infections [21].
`BTK is constitutively expressed in myeloid and lym-
`phoid cells but absent in T cells and in mature plasma cells
`[22]. It is found during all stages of the B cell lineage up
`until the plasma-cell stage, where it is absent in the most
`
`130
`
`Ó 2013 John Wiley & Sons Ltd
`
`

`

`L. Vargas et al.
`
`BTK and ITK Inhibitors 131
`
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`Figure 1 Schematic representation of BTK and ITK showing membrane binding and regulatory tyrosine phosphorylation sites. BTK and ITK have
`similar domain organization, with the difference that BTK has two proline-rich repeats (PRR) in the Tec homology domain (TH). The highly conserved
`BTK motif binds a Zn2+ ion, which stabilizes the PH domain. The PH domain binds phosphatidylinoisitol-3,4,5-trisphosphate (PIP3), which is generated
`by PI3K (Fig. 2). In the PH domain of BTK, there is a 27 amino acid insertion not found in ITK (marked in blue). The SH3 domain binds to proline-rich
`regions, while the SH2 domain interacts with phosphorylated tyrosine residues forming reversible signalling complexes. The depicted trans-
`phosphorylated tyrosine residue in the catalytic domain has an activating function, whereas the role of the autophosphorylated tyrosine in the SH3 domain
`is less defined.
`
`mature form. Mutations cause a differentiation block at the
`stage of pre-B cells, with mature B cells being very few and
`non-responsive to foreign antigens, even if rare patients can
`have close to normal numbers [14, 23–25].
`for
`crucial
`ITK is
`less widely expressed and is
`T-lymphocyte development, as initially shown by knock-
`ing out the gene in mice [26]. More in-depth studies
`revealed that both ITK-deficient mice [5, 6, 27] and the
`few patients with ITK mutations analysed so far show
`almost complete absence of invariant natural killer T cells.
`While several different genetic defects show susceptibility
`to severe EBV infection, it was recently reported that ITK
`deficiency is clinically distinct
`from both signalling
`lymphocyte activation molecule (SLAM)-associated protein
`(SAP) and X-linked inhibitor of apoptosis protein (XIAP)
`deficiency [28]. ITK may also be a crucial host factor
`needed for the development of an HIV infection [29].
`Further studies have shown that ITK-deficient mice have
`drastically reduced lung inflammation, eosinophil infiltra-
`tion and mucous production in response to ovalbumin-
`induced induction of allergic asthma [30]. Therefore,
`inhibition of T cell activation has been one of the strategies
`for developing immunosuppressive agents to treat autoim-
`mune disorders and inflammation [31]. Suppression of host
`immune functions by blocking T cell activation is also a
`successful modality for preventing organ transplant rejec-
`tion [32].
`In contrast to BTK, ITK is not constitutively expressed.
`Thus, the corresponding transcript was initially identified
`from an IL-2-dependent mouse T cell line [11]. This means
`that even if BTK and ITK frequently are considered to be
`analogs,
`selectively
`expressed in T-
`and B cells,
`respectively, their differential expression with regard to
`
`the need for inducibility shows that this is not entirely
`true. This difference is also likely to influence the
`treatment effect of inhibitors.
`
`Signalling pathways of BTK and ITK and target diseases
`
`TFKs play central, but diverse, modulatory roles in various
`cellular processes. They participate in signal transduction
`in response to virtually all types of extracellular stimuli
`that are transmitted by growth factor receptors, cytokine
`receptors, G-protein-coupled receptors, antigen receptors
`and integrins [33, 34]. As illustrated in Fig. 2 (Step 1),
`following BCR, TCR stimulation, SRC family kinases are
`activated, leading to the phosphorylation of immunore-
`ceptor tyrosine activation motifs (ITAMs) of the CD79
`(Ig-a, Ig-b) and CD3 complex chains. Similarly, PI3K is
`activated to catalyse the conversion of membrane-associated
`PIP2 to PIP3 leading to BTK/ITK recruitment to the
`plasma membrane through the interaction of
`its PH
`domain with PIP3 [35]. Concomitantly, the phosphoryla-
`tion of Ig-a and Ig-b ITAMs leads to the recruitment of
`SYK/ZAP70 kinase via SYK SH2 domains. Following
`activation, BTK/ITK ignites multiple downstream signals
`generating pleiotropic effects (Fig. 2, step 2): (1) PLCc
`activation, generation of
`second messengers,
`such as
`inositol [1,4,5]-triphosphate (IP3), diacylglycerol (DAG)
`and calcium, (2) Cell proliferation, differentiation, apop-
`tosis and survival [16, 36]. However, the molecular basis of
`many of these pathways is not fully understood, and many
`interacting molecules remain to be isolated. Using affinity
`purification combined with tandem mass spectrometry, we
`have recently characterized the interaction of BTK with an
`ankyrin-repeat domain protein [37] and the protein 14-3-3,
`
`Ó 2013 John Wiley & Sons Ltd
`
`

`

`132 BTK and ITK Inhibitors
`
`L. Vargas et al.
`
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`Step 1
`
`TCR
`
`Step 2
`
`BCR
`
`~
`
`PIP2 ---+ PIP3
`
`I
`
`PLCy2/l
`
`IP3
`
`\
`i
`*
`
`~
`
`(lg-a) {lg-fi)
`
`.-8~
`.-8
`
`l
`
`r---- CypA
`
`PKC
`Pin- I -------,
`
`/
`
`DAG

`
`PKC - (Ca2+)
`
`Figure 2 BTK and ITK activation. B cell receptor (BCR) and BTK signalling components are depicted to the left and T cell receptor (TCR) to the right
`upon engagement of BCR and TCR, SRC family kinases including LYN and LCK are activated leading to phosphorylation of immunoreceptor tyrosine
`activation motifs (ITAMs) (Step 1). Activated PI3K converts PtdIns-4,5-bisphosphate (PIP2) into PIP3, which tethers BTK/ITK to the membrane
`(Fig. 1), where they phosphorylate regulatory tyrosine residues in PLCc2 and PLC, c1 respectively (Step 2). The increasing PLCc activity results in the
`production of the secondary messengers DAG and IP3 inducing activation of the transcription factors NF-jB (active mainly in B cells) and NF-AT (active
`mainly in T cells). Endogenous BTK/ITK inhibitors (PKC, Pin-1/CypA) regulate their activity affecting cellular responses such as cell survival, apoptosis,
`adhesion, migration and proliferation.
`
`which regulate nucleo-cytoplasmic shuttling [38] and
`attenuate signalling [39].
`To date, almost only B cell-derived tumours have been
`treated with the newly developed inhibitor for BTK,
`Ibrutinib (see below). The rationale is that tumours,
`similar to non-transformed B cells, may be dependent on
`BCR signalling for their survival. However, tumours with
`activating mutations downstream of BTK are likely to be
`resistant, and this also seems to be the case [40]. Although
`animal models of autoimmunity show very promising
`outcomes from Ibrutinib treatment, the drug has not yet
`been used in clinical studies in man. Allergies and other
`forms of hypersensitivity and inflammatory diseases with a
`strong B cell component could also become targets. Target
`diseases for ITK inhibitors are less defined and will await
`studies in relevant animal models. As always, potential side
`effects have to be balanced against the benefit of treatment.
`
`BTK inhibitors
`
`LFM-A13
`
`(alpha-
`The leflunomide metabolite analog (LFM-A13)
`cyano-beta-hydroxy-beta-methyl-N-(2,5-ibromophenyl)-pro-
`pen-amide) (Table 2) is one of the first rationally designed
`antileukemic agents targeting BTK [41, 42]. This small-
`molecule inhibitor binds non-covalently to the catalytic site
`
`of BTK in a reversible manner (half-maximal inhibitory
`concentration (IC50) = 17.2 lM for human BTK in vitro
`and IC50 = 2.5 lM for recombinant BTK). It does not
`affect the enzymatic activity of other protein tyrosine
`kinases, including EGFR, HCK, IRK JAK1, JAK3 and
`IRK at concentrations of 278 lM [41]. However, even if the
`molecule has been described as a highly specific inhibitor of
`BTK, it can also efficiently affect the activity of other
`kinases such as the erythropoietin receptor, JAK2 and
`downstream molecules [43].
`During the last decade, a plethora of in vitro and in vivo
`studies have suggested that LFM-A13 could act as a dual-
`function anticancer drug with apoptosis-promoting and
`antithrombotic properties [44–46]. In addition, LFM-A13
`also exhibits antiproliferative activity against Her2/Neu-
`overexpressing breast cancer cells [47]. Furthermore, LFM-
`A13 has been reported to prevent acute fatal graft-versus-
`host disease in a murine model of allogeneic bone marrow
`In addition, LFM-A13 has been
`transplantation [48].
`broadly used in vitro to inhibit BTK downstream signalling
`pathways (Fig. 2) and further to elucidate the role of SFKs
`[49–51]. On neutrophils, for example, it has been shown
`that LFM-A13 also negatively affects the translocation of
`Rac-2, RhoA, ADP ribosylation factor-1, TEC, BMX and
`BTK induced by fMet-Leu-Phe [52]. Moreover, LFM-A13
`could block the endogenous phosphorylation of Myd88
`adapter-like (Mal) on tyrosine in cells
`treated with
`
`Scandinavian Journal of Immunology, 2013, 78, 130–139
`
`

`

`L. Vargas et al.
`
`BTK and ITK Inhibitors 133
`
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`macrophage-activating lipopeptide-2 or LPS [53] and
`LFM-A13 inhibited Heme oxygenase (HO-1) induction
`by the classical TLR4 ligand LPS in cell cultures [54].
`Other reports show that LFM-A13 by inhibiting TFKs,
`rescues the suppression of TCR-induced CD25 expression
`in Jurkat cells [55]. In primary myeloma-bearing immu-
`nodeficient mice, LFM-A13 inhibited osteoclast activity,
`prevented myeloma-induced bone resorption and moder-
`ately suppressed myeloma growth [56]. Administration of
`LFM-A13 is not toxic to mice, rats or dogs at daily dose
`levels as high as 100 mg/kg [57]. However, as mentioned,
`rather high doses are needed for a pharmacological effect,
`and we are not aware of any ongoing, or planned, clinical
`studies.
`
`Dasatinib
`
`Dasatinib, BMS-354825 or Sprycel [N-(2-chloro-6-meth-
`ylphenyl)-2-(6-(4-(2-hydroxyethyl)-piperazin-1-yl)-2-meth-
`ylpyrimidin-4-ylamino)thiazole-5-carboxamide] (Table 2) is
`an orally available, dual ABL/SRC tyrosine kinase inhibitor
`(TKI), which was developed to treat patients with chronic
`myelogenous leukaemia (CML), who had failed, or were
`intolerant to, therapy with Imatinib BCR-ABL1 and SFK
`TKI [58, 59]. Other diseases in which bone metastases are
`frequent (e.g. breast or prostate tumours) could also benefit
`from the addition of Dasatinib to standard-of-care treat-
`ments [60, 61].
`Native targets of Dasatinib in CML cells have been
`identified using a chemical proteomics approach [62–64].
`Besides ABL and SRC kinases, BTK and TEC, but not
`ITK, were recognized as major binders
`inhibited by
`nanomolar concentrations. In addition, the gatekeeper
`residue as the critical determinant of Dasatinib suscepti-
`bility has been detected with the help of structure-based
`mutagenesis experiments. Mutation of Thr-474 in BTK to
`Ile and Thr-442 in TEC to Ile conferred resistance to
`Dasatinib, whereas mutation of the corresponding residue
`in ITK (Phe-435) to Thr sensitized the otherwise insen-
`sitive ITK [64]. Other studies have shown that Dasatinib
`induces apoptosis in primary chronic lymphocytic leukae-
`mia (CLL) cells blocking LYN kinase activity [65, 66].
`Moreover, Dasatinib decreased levels of the activated,
`phosphorylated forms of AKT, ERK1/2 and p38 and
`reduced the expression of the antiapoptotic proteins MCL-1
`and BCL-XL [67]. Thus, it seems that Dasatinib as a single
`agent has activity in relapsed and refractory CLL [66].
`In line with the role of TFKs in lymphoid and myeloid
`cells, Dasatinib inhibited the secretion of several immu-
`nomodulators [68, 69]. The observed inhibition of TFKs
`predicts immunosuppressive (side) effects of this drug and
`may offer therapeutic opportunities for inflammatory and
`immunological disorders [64, 70, 71]. However, further
`experiments are required to describe the exact mechanism
`of the above-mentioned hypothesis.
`
`Ó 2013 John Wiley & Sons Ltd
`
`In summary, this compound has been approved by the
`FDA for the treatment of patients with CML in all phases
`or Ph+-ALL, who were resistant, or intolerant, to therapy,
`with Imatinib. In Europe, it has been approved for therapy
`of patients with CML who are resistant, or intolerant, to
`Imatinib [59]. However, the drug has not been used to
`clinically interfere with TFKs.
`
`Ibrutinib (PCI-32765)
`
`(1-{(3R)-3-[4-amino-3-(4-phenoxyphenyl)-1H-
`Ibrutinib,
`pyrazolo[3,4-d]pyrimidin-1-yl]piperidin-1-yl}prop-2-en-1-
`one) (Table 2), is a selective and irreversible small-molecule
`BTK inhibitor that inhibits BCR signalling in human B
`cells. It was originally named PCI-32765 and re-named
`Ibrutinib by the World Health Organization (WHO) and
`the United States Adopted Name (USAN) Council. Orally
`administered Ibrutinib has demonstrated to be particularly
`active in different B cell malignancies including CLL,
`mantle
`cell
`lymphoma
`(MCL), diffuse
`large B cell
`lymphoma (DLBCL) and multiple myeloma (MM) [72–75].
`Ibrutinib inactivates BTK through covalent binding to
`the active site (Cys-481) in the ATP-binding domain of
`BTK with IC50 of 0.5 nmol/L [76]. Several TFKs with
`homology to BTK, including BMX and ITK, have similar
`cysteine residues that might also be irreversibly inhibited
`by Ibrutinib. Other kinases that can also be sensitive to
`Ibrutinib at nanomolar concentrations include BLK, TEC,
`EGFR, ERBB2, HER2, HER4 and JAK3 [72, 76, 77].
`
`Ibrutinib as a potential drug for B cell malignancies
`
`The use of Ibrutinib in preclinical and clinical trials
`appears to be a promising new strategy for treatment of B
`cell malignancies (Table 1). The in vivo effect of ibrutinib
`has been demonstrated in patients with CLL. Recent
`reports have shown that
`Ibrutinib inhibits CLL cell
`survival and proliferation as well as induces CLL apoptosis
`[77, 78]. In addition, treatment of CD40- or BCR-
`activated CLL cells with Ibrutinib results in inhibition of
`BTK tyrosine phosphorylation and also effectively abro-
`gates downstream survival pathways activated by this
`kinase, including ERK1/2, PI3K and NF-jB [74, 77].
`Ibrutinib also acts by modulating the interaction
`between CLL cells and their microenvironment. For
`example,
`it inhibits activation-induced proliferation of
`CLL cells and effectively blocks survival signals, which are
`provided externally to CLL cells from the microenviron-
`ment (CD40L, BAFF, IL-6, IL-4 and TNFa, fibronectin)
`engagement and stromal cell contact, as well as migration
`in response
`to tissue-homing chemokines
`(CXCL12,
`CXCL13) [79]. Moreover, the secretion of BCR-dependent
`cytokines such as CCL3 and CCL4 is effectively decreased
`both in vitro and in patients with CLL treated with
`Ibrutinib [78].
`
`

`

`134 BTK and ITK Inhibitors
`
`L. Vargas et al.
`
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`Table 1 Clinical trials of Ibrutinib in B cell malignancies.
`
`Disease
`
`Study description
`
`Drugs and doses
`
`Study phase
`
`CLL and
`SLL
`
`CLL, SLL
`
`Ibrutinib (420 mg)
`
`II
`
`ROR CLL,SLL
`with 17p deletion
`CLL, SLL in patients
`older than 65 or
`have 17p deletion
`
`Ibrutinib (420 mg)
`
`Ibrutinib (420 mg)
`
`CLL, SLL
`
`ROR
`CLL, SLL
`ROR
`CLL,SLL
`
`Ibrutinib (420 mg)
`VS chlorambucil
`Ibrutinib (420 mg)
`VS Ofatumumab
`Ibrutinib (420 mg) +
`Rituximab+
`Bendamustine
`Ibrutinib
`
`II
`
`II
`
`III
`
`III
`
`III
`
`II
`
`Estimated
`patients/age
`
`30
`≥18 years
`
`111
`≥18 years
`86
`≥65 years
`and ≥ 18 year
`for 17p deletion
`272
`≥65 years
`350
`≥18 years
`580
`≥18 years
`
`Objective
`
`Study duration
`
`Clinical trials.gov
`identifier
`
`Impact on
`leukemia cell
`trafficking
`and death
`ORR,PFS, OS
`
`2012–2015
`
`NCT01752426
`
`2013–2016
`
`NCT01744691
`
`ORR, OS, PFS
`
`2011-2015
`
`NCT01500733
`
`ORR, PFS
`
`2013–2016
`
`NCT01722487
`
`PFS, OS, ORR
`
`2012–2015
`
`NCT01578707
`
`PFS,OS, ORR
`
`2012–2018
`
`NCT01611090
`
`PFS, ORR,OS
`
`2012–2014
`
`NCT01589302
`
`NCT01325701
`
`CLL, SLL,
`B-PLL
`DLBCL
`
`FL
`
`ROR CLL,SLL,
`B-PLL
`ROR
`DLBCL
`Refractory FL
`
`MCL
`
`MCL
`
`ROR
`MCL
`ROR
`MCL
`MCL
`
`B cell
`neoplasm
`MM
`
`WM
`
`Recurrent mature B
`cell neoplasm
`Relapsed or relapsed
`and refractory MM
`WM
`
`Ibrutinib (560 mg)
`
`Ibrutinib
`(560 mg)
`Ibrutinib (560 mg)
`
`Ibruinib (560 mg)
`VS Temsirolimus
`Ibrutinib (560 mg)
`VS Temsirolimus
`Ibrutinib (560 mg) +
`Rituximab+
`Bendamustine
`Ibrutinib (420,
`560 mg)
`Ibrutinib (420,
`560, 840 mg)
`Ibrutinib
`
`II
`
`II
`
`II
`
`III
`
`III
`
`III
`
`I
`
`II
`
`II
`
`75
`≥18 years
`60
`≥18 years
`110
`≥18 years
`110
`≥18 years
`280
`≥18
`280
`≥18 years
`520
`≥65 years
`
`24
`≥20 years
`164
`≥18 years
`33
`≥18 years
`
`Efficacy and safety
`
`2011–2014
`
`ORR,OS, FPS
`
`2013–2016
`
`NCT01779791
`
`ORR, PFS, OS
`
`2012–2015
`
`NCT01599949
`
`PFS, OS
`
`2012
`
`PFS,ORR, OS
`
`2012–2017
`
`NC2012-
`000601-74
`NCT01646021
`
`PFS,OS, ORR
`
`2013–2019
`
`NCT01776840
`
`Safety and
`pharmacokinetic
`Efficacy and safety
`
`2012–2014
`
`NCT01704963
`
`2012–2016
`
`NCT01478581
`
`ORR, safety
`
`2012–2014
`
`NCT01614821
`
`Follicular lymphoma (FL), overall response rate (ORR), progression-free survival (PFS), overall survival (OS), chronic lymphocytic leukemia (CLL), small
`lymphocytic lymphoma (SLL), multiple myeloma (MM), mantle cell lymphoma (MCL), diffused large B cell lymphoma (DLBCL), B cell prolymphocytic
`leukemia (B-PLL), relapsed or refractory (ROR), Waldenstr€oms macroglobulinemia (WM).
`
`The initial phase I study of Ibrutinib enrolled patients
`with B cell lymphomas including CLL demonstrated that a
`dose of 420 mg is as efficient as 840 mg. In fact, the
`occupancy and inhibition of BTK were similar in both
`doses, so 420 mg was selected for further studies to
`minimize adverse effects [77]. Administration of Ibrutinib
`(420 mg/day)
`in patients with CLL induces a rapid
`shrinkage of enlarged lymph nodes and symptomatic
`improvement within the first few weeks of treatment [73].
`The expected pattern of initial rapid nodal response with
`sometimes marked lymphocytosis was also observed. This
`increase in lymphocyte count was transient and could be
`typically resolved after the first few months of therapy [72,
`In the 2012 American Society of Hematology
`77].
`meeting, it was reported that Ibrutinib induces an overall
`
`response rate (ORR) of 68% in previously untreated CLL
`patients, aged 65 or older, and an ORR of 71% in
`previously treated patients [72].
`It has been recently reported that BTK is highly
`expressed in malignant plasma cells from patients with
`MM [80, 81]. This is in contrast to the most mature
`normal plasma cells, where BTK is not expressed [22]. In
`MM models, Ibrutinib reduced osteoclast formation and
`bone resorption and also inhibited BTK-mediated osteo-
`clastogenesis induced by M-CSF and RANKL [80]. The
`chemokine and cytokine secretion from bone marrow
`stromal cells (BMSCs) and osteoclasts was significantly
`decreased by Ibrutinib, and it blocks SDF-1-induced
`adhesion and migration [81]. Furthermore, it also inhibited
`MM cell growth triggered by IL-6 or coculture with
`
`Scandinavian Journal of Immunology, 2013, 78, 130–139
`
`

`

`L. Vargas et al.
`
`BTK and ITK Inhibitors 135
`
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`Table 2 Chemical structure of BTK and ITK inhibitors.
`
`Structure
`
`BTK inhibitors
`
`BMSCs in vitro. In addition, Ibrutinib inhibits MM cell–
`induced osteolysis of implanted human bone chips in SCID
`mice [80].
`Ibrutinib also shows promising and encouraging results
`as a single-agent drug in MCL. Thus, Ibrutinib produced
`ORR of 66% in phase II study for patients receiving
`(560 mg/day) [82]. Co-administration of Ibrutinib with
`the proteasome inhibitor Bortezomib resulted in a syner-
`gistic effect and induced MCL cell-death [74].
`
`Ibrutinib as a therapeutic agent for autoimmune diseases
`
`Ibrutinib shows promising activity against experimental
`autoimmune diseases, particularly in rheumatoid arthritis
`(RA).
`In mice with collagen-induced arthritis
`(CIA),
`administration of 12.5 mg/kg effectively reduced arthritic
`symptoms after few days of treatment [72, 83]. These
`studies show that in CIA and collagen antibody-induced
`arthritis, Ibrutinib markedly reduced inflammation, bone
`resorption and cartilage destruction. Moreover, the infil-
`tration of
`inflammatory cells, cytokine and chemokine
`levels in synovial fluid are decreased [83, 84]. In the MRL-
`Fas (lpr) lupus model, treatment with Ibrutinib inhibits
`autoantibody production and renal impairment [76].
`
`Compound name
`
`Ibrutinib
`(PCI-32765)
`MW 440.5
`
`Dasatinib
`MW 488
`
`LFM-A13
`MW 360
`
`AVL-292
`MW 423.17
`
`Compound 3 (Sanofi)
`MW 297.38
`
`Compound 7 (Sanofi)
`MW 375.44
`
`Pharmacokinetics
`
`Ibrutinib is administered orally and rapidly absorbed.
`Between 1 and 2 hours after drug administration, it reaches
`the mean plasma concentration. It has a short half-life of 2-
`3 hours, but BTK inhibition will remain for 24 hours.
`Generally, the drug is well tolerated, with mild adverse
`effects like cough, fatigue, diarrhoea and infectious com-
`plications, which could be managed easily (Table S1) [85].
`
`ITK inhibitors
`
`..
`
`AVL-292
`
`AVL-292 is a new orally available potent compound that
`selectively inhibits BTK (Table 2). It also binds to Cys-481
`at the kinase domain, sustaining BTK inhibition for
`24 hours. Human clinical trials with AVL-292 have dem-
`onstrated that the drug is safe and like other BCR inhibitors,
`the drug caused lymphocytosis within few weeks of the
`treatment [73, 84]. AVL-292 also showed a promising effect
`in animal models with rheumatoid arthritis. Currently,
`according to ClinicalTrials.Gov, there are two studies
`evaluating AVL-292, one recruiting for non-Hodgkin
`lymphoma, CLL and Waldenstr€om’s macroglobulinemia
`(WM), and a second, active, but not recruiting, studying the
`safety and pharmacokinetic of this drug [84].
`
`BMS-509744
`MW 623.83
`
`?'.,
`
`-(_('yl/"'.)\,
`
`¥ (cid:173)
`
`0
`,,l
`""
`
`ITK inhibitors
`
`The crystal structure of the ITK kinase domain was
`elucidated for the phosphorylated and non-phosphorylated
`
`kinase domain bound to staurosporine, a potent broad-
`spectrum kinase inhibitor [86]. These structures are highly
`useful
`for the design of selective ITK inhibitors and
`provide insight into the influence of inhibitor binding and
`
`Ó 2013 John Wiley & Sons Ltd
`
`

`

`136 BTK and ITK Inhibitors
`
`L. Vargas et al.
`
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`ITK [86].
`conformation of
`phosphorylation on the
`Together, Met-398, Phe-403, Ala-407, Met-410 and
`Met-411 (from the a-C-helix), Phe-374 (from the Gly-
`rich loop), and Val-424, Leu-433 and Lys-391 define an
`extensive hydrophobic pocket lying adjacent to the ITK
`active site [86]. The size and shape of this pocket are
`different in SFKs as well as in Ser/Thr kinases such as
`CDK2. Although residues Met-398, Phe-403 and Met-410
`are conserved in ITK and BTK, Phe-435 is unique to ITK
`[86]. This residue is therefore identified as a gatekeeper (or
`blocker) of this extensive hydrophobic pocket. An inhibitor
`of the other TEC family members (BTK, TXK/RLK, TEC
`and BMX) may exploit the smaller threonine residue at
`this position, either via a hydrogen bonding interaction or
`by accessing the hydrophobic pocket
`[86]. A broad-
`spectrum inhibitor of all TFKs could be achieved using
`small lipophilic groups positioned close to residue Phe-435
`role in non-cytotoxic
`[86] and may have a general
`immunosuppression.
`A high-throughput screen campaign of a compound
`collection by Bristol-Myers Squibb and Boehringer Ingel-
`heim (Table 2) led to the identification of potent ITK
`inhibitors (Table S2) from a distinct scaffold series of
`2-aminothiazoles [87, 88] and benzimidazoles [89–91],
`respectively. Further development of benzimidazoles have
`demonstrated selective inhibition of ITK, improved the
`cellular and functional potency as well as the drug-like
`properties, with the 10n and 10o compounds as excellent
`agents for proof-of-concept studies [92].
`Novel ITK inhibitors based on (4- or 5-aryl)pyrazolyl-
`indole scaffolds were also found to be selective for ITK over
`other kinases
`[93]. Molecular modelling predictions
`showed that pyrazolyl-indoles are inhibitors of
`ITK.
`Docking models of various pyrazolyl-indole derivatives
`with ITK indicated a common binding mode, which
`involved three hydrogen bonds in the hinge region of ITK
`between: (i) N–H of the pyrazole ring with the carbonyl of
`Glu-436 of ITK, (ii) N of pyrazole and N–H of Met-438 of
`ITK and (iii) N–H of the indole ring with the carbonyl of
`Met-438 of ITK [93]. It was also apparent from docking,
`that among the three series examined, 5-benzylpyrazolyl-
`indoles (compound 34) show the best binding to ITK, and
`were followed in the decreasing order by 4-phenylpyraz-
`olyl-indoles (compound 13) and 5-phenylpyrazolyl-indoles
`(compound 24) (Table S2). On the other hand, compound
`44, which was developed by chemical optimization of an
`initial high-throughput screening hit,
`inhibited ITK’s
`activity with an IC50 in the nanomolar
`range [94].
`Compound 44 substantially reduced pro-inflammatory
`immune responses in vitro and in vivo after systemic
`administration in two acute
`contact hypersensitivity
`models [94].
`In another study, a series of ITK inhibitors, active in
`nanomolar concentrations, were synthesized based on
`indolylindazole libraries. The potential of this series of
`
`compounds was confirmed through in vivo tests in an
`anti-CD3-IL2 mouse model. The intravenous administra-
`tion of highly potent ITK inhibitor 11o (Table S2) resulted
`in dose-dependent, efficient suppression of IL-2 at 10 mg/
`kg [95].
`The discovery of a new series of potent and selective
`novel ITK inhibitors based on 3-aminopyride-2-ones has
`also been reported [96]. These inhibitors were identified
`by structure-based design,
`starting from a fragment
`generated de novo, the 3-aminopyrid-2-one motif. Among
`various derivatives, the compound 7v (Table S2) illustrates
`the fact that fragment-like de novo starting point can
`rapidly evolve into biologically inhibitors of ITK, with
`good potency and selectivity profile. Other derivatives 7w,
`7x and 7y also show potent inhibition of ITK (Table S2)
`[96].
`Another selective inhibitor, 7-benzyl-1-(3-(piperidin-1-
`yl)propyl)-2-(4-(pyridin-4-yl)phenyl)-1H-imidazo[4,5-g]qui-
`noxalin-6(5H)-one
`(CTA056), was developed through
`screening of 9600 compounds,
`followed by molecular
`modelling, and extensive structure–activity relationship
`studies [97]. CTA056 (Table S2) exhibits the highest
`inhibitory effects towards ITK,
`followed by BTK and
`BMX. Among the 41 cancer cell lines analysed, CTA056
`selectively targets acute lymphoblastic T cell leukaemia and
`cutaneous T cell lymphoma [97].
`Recently, novel and selective thienopyrazole inhibitors
`of ITK were generated by combining structure-based
`design and medicinal chemistry at Sanofi (Table 2) (Table
`S2). The most potent compounds crystallized with the
`target kinase ITK and also with SYK [98]. The mul

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket