throbber
review
`
`Breaking good: the inexorable rise of BTK inhibitors in the
`treatment of chronic lymphocytic leukaemia
`
`Claire V. Hutchinson1,2 and Martin J. S. Dyer1,2,3
`
`1Department of of Cancer Studies and Molecular Medicine, University of Leicester, 2The Ernest and Helen Scott Haematological
`Research Institute, University of Leicester, and 3Department of Biochemistry, University of Leicester, Leicester, UK
`
`Summary
`
`lineages and
`Although expressed in several haematological
`involved in multiple different signalling pathways, Bruton
`tyrosine kinase (BTK) plays an indispensible role in B cells in
`signalling from the B cell receptor (BCR) for antigen. Many
`B cell malignancies remain dependent on constitutive BCR
`signalling, making BTK a functional therapeutic target. Sev-
`eral BTK inhibitors (BTKi) with different kinomes and
`modes of action are being assessed clinically. This review
`documents the efficacy and toxicity of BTKi in chronic lym-
`phocytic leukaemia (CLL). Clinically, the furthest in develop-
`ment is ibrutinib (trade name, Imbruvica), an irreversible
`BTKi, which has shown spectacular preliminary efficacy, with
`rapid reductions in lymph nodes accompanied by peripheral
`blood lymphocytosis. The lymphocytosis resolves slowly and
`most patients do not enter a complete remission. Neverthe-
`less, it is possible to maintain many CLL patients, even those
`with adverse cytogenetic features, on drug for many months
`with minimal
`toxicities,
`thus potentially transforming the
`therapeutic paradigms for CLL. The efficacy, lack of toxicity
`and oral administration of BTKi will ensure their adoption
`in a wide range of B cell malignancies. An outstanding chal-
`lenge is to incorporate BTKi with other precision medicines
`in a mechanism-based manner in order to dispense with
`conventional chemotherapy.
`
`Keywords: chronic lymphocytic leukaemia, Bruton tyrosine
`kinase, ibrutinib, B cell receptor signalling.
`
`If the multi-billion dollar industry that has developed since
`the ‘war on cancer’ was announced by Richard Nixon in
`1971 (Sporn, 1997) is eventually to succeed in ‘eliminating
`the suffering and death from cancer’ (von Eschenbach,
`2006), then it must provide tools that allow either eradica-
`tion or long-term control of disease with minimal toxicity.
`In terms of the malignancies of mature B cells, significant
`
`Correspondence: Professor Martin J. S. Dyer, Room 3/57, Henry
`
`Wellcome Building, Lancaster Road, Leicester LE1 9HN, UK.
`
`E-mail: mjsd1@le.ac.uk
`
`progress has been made and there is now a plethora of
`‘precision medicines’
`including both engineered antibodies
`and targeted small molecules that are having great therapeu-
`tic impact even in early phase clinical trials (Dyer et al,
`2013).
`One class of small molecule inhibitor currently arousing
`intense interest comprises compounds targeting Bruton tyro-
`sine kinase (BTK). Importantly, BTK is not a genetically-
`defined therapeutic target; there are no mutations in the
`BTK gene or gene fusions that result in constitutive activity.
`Rather, BTK is defined functionally as a therapeutic target,
`playing a key role in several pathways that maintain B cell
`survival. The efficacy of BTK inhibitors (BTKi) in several
`forms of mature B cell malignancy has been spectacular,
`resulting in the first-in-class BTKi (Ibrutinib; Pharmacyclics,
`Sunnyvale, CA, USA) being awarded ‘Breakthrough Drug’
`status by the US Food and Drug Administration (FDA) for
`mantle cell lymphoma (MCL), CLL and Waldenstrom mac-
`roglobulinaemia (Dolgin, 2013); breakthrough drug status is
`given if preliminary clinical evidence indicates the drug may
`offer a substantial improvement over available therapies for
`patients with serious or life-threatening diseases. A product
`licence for ibrutinib, for the treatment of relapsed MCL, was
`awarded by the US FDA on 13 November 2013 and for
`relapsed CLL on 11 February 2014; at the time of writing,
`similar licences are anticipated imminently in Europe.
`This review addresses the rationale for targeting BTK in
`CLL, the nature and mode of action of BTKi and the data
`emerging from clinical trials in CLL, alone and in combina-
`tion with other agents. Despite their huge promise and huge
`enthusiasm for their use from both patients and clinicians
`alike, there remain several barriers to the incorporation of
`these novel agents into CLL therapy.
`
`An overview of BTK structure and functions
`
`BTK takes its name from Colonel Ogden C. Bruton (1908–
`2003), an American Army paediatrician based at the Walter
`Reed Army Hospital
`in Washington DC, who,
`in 1952,
`described a condition now referred to as X-linked agamma-
`globulinaemia,
`following the study of an 8-year-old boy,
`Joseph S. Holtoner, Jr., who had recurrent pneumonia and
`
`First published online 18 April 2014
`doi: 10.1111/bjh.12895
`
`ª 2014 John Wiley & Sons Ltd
`British Journal of Haematology, 2014, 166, 12–22
`
` 13652141, 2014, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.12895, Wiley Online Library on [29/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1019, p. 1 of 11
`
`

`

`absence of gamma globulin in his serum (Bruton, 1952). Dr
`Bruton and his discovery were featured in ‘TIME’ magazine
`18 May 1953. It was noted that all patients were boys; disease
`identification followed the development of successful antibi-
`otic therapy for recurrent bacterial
`infections. Forty years
`later,
`following extensive mapping experiments,
`the gene
`responsible for this condition (BTK), was cloned and charac-
`terized (Tsukada et al, 1993; Vetrie et al, 1993). Mapping to
`chromosome Xq21.3-q22, BTK encodes a kinase of the TEC
`subfamily (Yu & Smith, 2011; and other articles in that issue).
`Collectively, these kinases (BTK, ITK, BMX, TXK and TEC)
`are characterized by five structural domains including PH
`(pleckstrin homology) mediating phosphatidylinositol (3,4,5)-
`trisphosphate (PIP3) binding, TEC homology (BH), SH3 (Src
`homology 3), SH2 (Src homology 2) and kinase domains
`(Fig 1). The crystal structures of the major BTK domains
`have been solved (Mao et al, 2001; Marcotte et al, 2010).
`These structures have been of fundamental
`importance in
`terms of the design of targeted inhibitors, especially the irre-
`versible inhibitors, such as ibrutinib, AVL-292/CC-292 and
`ONO-4059 (Kuglstatter et al, 2011). BTK is localized pre-
`dominantly in the cytoplasm (but interestingly, also in the
`nucleus) with translocation to the plasma membrane via the
`PH domain following PIP3 binding,
`for phosphorylation
`and activation. Within BTK there are two major tyrosine
`
`Review
`
`phosphorylation sites. Phosphorylation of tyrosine (Tyr)551
`within the kinase domain regulates the transition between
`active (open) and inactive (closed) states. Phosphorylation of
`Tyr551 occurs on localization to the membrane by SRC fam-
`ily kinases (LYN/FYN/BLK) or SYK kinase. Conformational
`change then enables auto-phosphorylation at Tyr223. Once
`activated, BTK can then phosphorylate phospholipase Cc2,
`leading to calcium mobilization (King & Freedman, 2009)
`and subsequent activation of downstream signalling pathways,
`cytoskeletal
`rearrangements and transcriptional pathways
`(including NF-jB, NFAT and ARID3A) involved in prolifera-
`tion, cell survival and migration (Fig 1). Over 600 unique
`inherited BTK mutations have been described that lead to
`profound defects in B cell development (failure of develop-
`ment of pro to pre B cells) and immunoglobulin production
`(Valiaho
`et al,
`2006;
`http://structure.bmc.lu.se/idbase/
`BTKbase/). BTK mutations fall within all domains with the
`exception of the SH3 domain and many affect crucial residues
`involved in either ATP binding or in the catalytic site; others
`affect substrate recognition. Affected boys have normal levels
`of pre-B cells in their bone marrow but virtually no circulat-
`ing mature B lymphocytes, with a resultant lack of immuno-
`globulins of all classes and recurrent bacterial infections in
`the first few years of life. Interestingly, mice with identical Btk
`mutations have a much milder phenotype,
`indicating a
`
`pPLCγ2
`
`RAS
`
`PKCβ
`
`pERK
`
`IKK
`
`B-cell activation /
`Proliferation
`
`CD79B
`
`CD79A
`
`CD19
`
`PI3K
`
`PIP3
`
`pSYK
`
`pLYN
`
`BTK
`
`pTyr223
`
`Cys481
`
`pTyr551
`
`1
`
`PH
`
`BH PPR SH3
`
`SH2
`
`TK
`
`659
`
`Fig 1. BTK and its involvement in BCR signalling. Bruton tyrosine kinase (BTK) comprises several domains including: (i) pleckstrin homology
`(PH) domain. Mediates binding to inositol polyphosphate and phosphoinositides, leading to targeting of BTK to the plasma membrane. Plasma
`membrane localization is a critical step in the activation of BTK. (ii) BTK homology (BH) domain. Contains a highly conserved zinc finger motif
`that mediates Zn2+ ion binding necessary for protein stability. (iii) Polyproline (PPR) domain. (iv) Src homology (SH3 and SH2) domains. The
`SH2 domain of BTK binds to the B cell adapter protein BLNK, which is required for full BTK activation. (v) Tyrosine kinase (TK) domain.
`Unphosphorylated BTK is catalytically inactive. As shown in the figure, full activation of BTK requires both phosphorylation and binding of
`PIP3. Following BCR engagement and activation, and following PIP3 binding, BTK translocates to the plasma membrane, where it is phosphory-
`lated at Tyr551 by LYN and SYK, leading to autophosphorylation of Tyr223. Once bound to BLNK, BTK phosphorylates PLCG2 at several sites,
`leading to calcium mobilization and activation of the protein kinase C (PKC) family members and other effectors including RAS and lead to
`NFjB activation and phosphorylation of ERK. The BTK inhibitors ibrutinib, CC-292 and ONO-4059 all bind irreversibly to Cys481 within the
`ATP binding site of the BTK kinase domain, thus preventing BTK activation. In contrast, the highly specific BTKi, CGI-1746, binds to the ATP
`binding pocket of BTK and stabilizes the non-phosphorylated form of the protein by internalizing Tyr551. Apart from its role in BCR signalling,
`BTK is also implicated in cytokine and Toll receptor signalling pathways (not shown).
`
`ª 2014 John Wiley & Sons Ltd
`British Journal of Haematology, 2014, 166, 12–22
`
`13
`
` 13652141, 2014, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.12895, Wiley Online Library on [29/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1019, p. 2 of 11
`
`

`

`Review
`
`species-specific functional redundancy; combined inactivation
`of both BTK and TEC is necessary to produce a similar phe-
`notype in mice.
`Although recent focus has been on the role of BTK in
`terms of modulating signalling from the BCR as outlined
`below, BTK is, in fact, activated in response to a very broad
`range of growth/differentiation stimuli in haemopoietic tis-
`sues, including growth factor receptors, cytokine receptors,
`G-protein coupled receptors and integrins (Qiu & Kung,
`2000). BTK is implicated in the control of several pathways
`of central importance to mature B cells. Moreover, BTK is
`expressed in all bone marrow-derived cell lineages, with the
`exception of T lymphocytes and plasma cells (de Weers et al,
`1993; Genevier et al, 1994; Smith et al, 1994). In terms of
`these other lineages the functions of BTK are relatively unex-
`plored, but recent data indicate key functions in myeloid
`cells (Honda et al, 2012). Recent exciting in vitro data indi-
`cate that BTK may be a therapeutic target in acute myeloid
`leukaemias (Rushworth et al, 2014) as well as B cell malig-
`nancies. In macrophages, BTK functions downstream of sev-
`eral toll-like receptors and regulates apoptotic cell uptake
`(Byrne et al, 2013). These and other observations may also
`have important clinical implications in terms of design of
`BTKi combination studies,
`for example, with therapeutic
`monoclonal antibodies dependent on macrophages for their
`clinical efficacy. BTKi may also interfere with platelet activa-
`tion (Byrd et al, 2013a; Hsu et al, 2013; Rushworth et al,
`2013). Rushworth et al
`(2013)
`showed that
`ibrutinib
`impaired the platelet activation in response to collagen and
`ADP; however, the clinical significance of these observations
`is not clear (Farooqui et al, 2012). Finally, BTK is highly
`homologous to the interleukin-2 inducible T cell kinase
`(ITK) expressed in T cells, a homology that may be of thera-
`peutic relevance (Dubovsky et al, 2013). Inherited mutations
`in ITK predispose to viral infections, including Epstein–Barr
`virus (EBV) infection as well as EBV-driven lymphoprolifera-
`tive conditions (Huck et al, 2009; Stepensky et al, 2011).
`Collectively, these data do not appear to augur well for
`targeting BTK therapeutically. The expression of BTK in
`multiple haemopoietic lineages, the involvement of BTK in
`multiple different pathways, and the complete block of nor-
`mal B cell development in individuals with constitutional
`BTK mutations suggest that BTK inhibition might be cata-
`strophic! However, all of the above appear to be trumped by
`functional data concerning the central role of BTK activity in
`the maintenance of several forms of mature B cell malig-
`nancy.
`
`Functions of BTK in malignant B cells; BTK as a
`potential therapeutic target
`
`In mouse models, BCR signalling is vital for the survival of
`normal mature B cells (Kraus et al, 2004): interestingly, in
`mice this defect is rescued by phosphatidylinositide 3-kinase
`(PI3K) rather than BTK signalling (Srinivasan et al, 2009). In
`
`14
`
`human B cell malignancies, maintained BCR signalling has
`been shown to play an essential role in the survival of the
`activated B cell subtype of diffuse large B cell
`lymphoma
`(DLBCL) and in CLL (please see two recent reviews, Burger
`& Chiorazzi, 2013; Young & Staudt, 2013). In activated B cell
`(ABC)-DLBCL, constitutive BCR signalling is largely driven
`by somatic mutations in key domains of signalling molecules
`including CD79A/B and CARD11 (Davis et al, 2010). In CLL
`however these mutations are not present, but a fascinating
`recent finding indicates that the configuration of the BCR
`itself may drive antigen-independent cell-autonomous signal-
`ling (Duhren-von Minden et al, 2012). Inhibition of BTK
`may therefore be effective therapy in all cases of CLL rather
`than just with maintained signalling capacity following BCR
`cross-linking in vitro (Apollonio et al, 2013).
`However,
`the above scenario is complicated by the
`involvement of BTK in other signalling pathways controlling
`B cell migration and motility. Mature lymphocytes are not
`static; they are constantly moving between blood and tissues
`(Gowans & Knight, 1964). For normal lymphocytes this pro-
`cess enables immune surveillance; for malignant lymphocytes
`it offers a protective niche in which to receive survival and
`proliferation signals from the microenvironmental and solu-
`ble factors within the lymph node. Lymphocyte movement
`is not random and depends on a complex process of signal-
`ling pathways involving adhesion molecules and chemokines,
`which ultimately control the cytoskeleton. Two such path-
`ways of particular importance are the chemokine receptor-
`ligand pathways CXCR4-CXCL12 and CXCR5-CXCL13.
`BTK is known to be important for B cell migration and
`homing (Ortolano et al, 2006; de Gorter et al, 2007), and is
`activated upon chemokine binding to CXCR4 and CXCR5
`through direct interaction with the chemokine receptor G
`protein subunits (Jiang et al, 1998; Lowry & Huang, 2002).
`Thus, BTKi may have a significant effect on lymphocyte
`homing and recirculation; this has been observed clinically
`in CLL patients receiving BTKi, as discussed below. The rel-
`ative importance of BTKi-mediated blockade of the various
`different pathways will presumably vary from patient
`to
`patient, and moreover in CLL cells at different anatomical
`locations.
`
`Pharmacological development of BTKi
`
`As is so often the case in drug development, there are several
`BTKi currently in clinical trial or in late preclinical develop-
`ment. Some of the BTKi being assessed are listed in Table I;
`Ibrutinib (Pharmacyclics) is furthest along in terms of clini-
`cal development but there are a currently a number of others
`undergoing clinical trials including AVL-292/CC-292 (Cel-
`gene, Summit, NJ, USA) and ONO-4059 (ONO Pharmaceu-
`ticals, Osaka, Japan). Whilst this plethora of new molecules
`is of considerable scientific and clinical interest and reflects
`the activities of unfettered market forces, one must question
`the logic of commercially developing so many molecules
`
`ª 2014 John Wiley & Sons Ltd
`British Journal of Haematology, 2014, 166, 12–22
`
` 13652141, 2014, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.12895, Wiley Online Library on [29/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1019, p. 3 of 11
`
`

`

`Table I. BTKi currently in clinical trial or preclinical development.
`
`Molecule
`
`Company
`
`Irreversible
`inhibitor?
`
`Ibrutinib
`
`Pharmacyclics/Jansen Yes
`
`AVL-292/CC-292 Avila/Celgene
`
`Yes
`
`BTK
`BTK IC50
`specific?
`0 5 nmol/l No – broad
`kinome
`<0 5 nmol/l Yes
`
`ONO-4059
`
`Ono Pharma
`
`Yes
`
`2 2 nmol/l Yes
`
`HM71224
`
`Hanmi Pharma
`
`GDC-0834
`
`CGI-1746
`
`CGI Pharma
`Genentech
`CGI Pharma
`
`NK
`
`No
`
`No
`
`NK
`Yes
`5 9 nmol/l NK
`1 9 nmol/l Yes
`
`Review
`
`Clinical
`development
`
`Clinical trials
`
`References
`
`Multiple Phase
`II and Phase III
`clinical trials
`Phase II clinical trial NCT01766583
`With Lenalidomide
`in ABC-DLBCL
`NCT01659255
`Dose escalation
`in B cell
`malignancies
`NCT01765478
`
`Phase I clinical trial
`
`Phase I in normal
`individuals
`Phase I normal
`individuals
`Preclinical
`
`See text
`
`See text
`
`Salles et al
`(2013) and
`Rule et al
`(2013)
`NK
`
`Liu et al
`(2011)
`Di Paolo
`et al (2011)
`
`BTK, Bruton tyrosine kinase; BTKi, Bruton tyrosine kinase inhibitor; IC50, 50% inhibitory concentration; ABC-DLBCL, activated B cell-diffuse
`large B cell; NK, not known.
`
`against the same target. Which might be best clinically is
`quite unclear, and given the clinical success of ibrutinib may
`well be impossible to assess. The chemical structures of the
`three irreversible BTKi are very similar and yet the kinomes
`of each molecule are very different.
`The development of ibrutinib is of interest for at least two
`reasons. Firstly, and most
`importantly,
`ibrutinib is an
`irreversible BTKi that binds covalently to BTK (Pan et al,
`2007). A series of highly selective and irreversible BTKi were
`developed at Celera using a bioinformatics approach using
`the crystal structures of BTK. These molecules inactivate BTK
`by covalent binding to Cys481 near the ATP binding domain
`of BTK and thus inhibited phosphorylation at Tyr223. The
`lead molecule (PCI-32765) was tested in in vivo models of
`both rheumatoid arthritis and lymphoma (using a spontane-
`ously occurring canine lymphoma model) and showed con-
`siderable activity (Honigberg et al, 2010). Historically, the
`pharmaceutical industry has been wary of irreversible kinase
`inhibitors because of concerns over possible toxicity, particu-
`larly so in kinases with long protein half-lives. (Interestingly,
`BTK is a long-lived protein with a half-life in B cells of over
`8 h). However, there are now an increasing number of irre-
`versible kinase inhibitors entering the market, including afati-
`nib an irreversible inhibitor of EGFR (Sanderson, 2013). The
`second interesting aspect about ibrutinib is that its kinome is
`very broad and includes several other kinases with pivotal
`roles in normal and malignant B cell signalling,
`including
`numerous SRC kinase family members including LYN (Pan
`et al, 2007). Not unsurprisingly, given the close homology of
`the two proteins, ibrutinib irreversibly inactivates ITK as well
`as BTK (Dubovsky et al, 2013). One of the implications of
`this finding might be a role for ibrutinib in T cell malignan-
`cies dependent on ITK signalling as well as those with ITK
`
`ª 2014 John Wiley & Sons Ltd
`British Journal of Haematology, 2014, 166, 12–22
`
`fusions (Streubel et al, 2006; Pechloff et al, 2010). Less
`positively, it is difficult to reconcile the lack of observed tox-
`icity seen in patients taking ibrutinib for many months with
`this broad kinome, especially perhaps in patients immuno-
`compromised by their disease and prior therapies.
`A variety of more specific BTKi have been developed
`and three of these are currently undergoing clinical trials in
`B cell malignancies. Some of these are shown in Table I.
`AVL-292/CC-292 and ONO-4059 are both irreversible
`inhibitors, which are closely related to ibrutinib structurally
`but are considerably more specific in terms of other kinases
`inhibited; both are being assessed in patients with B cell
`malignancies. ONO-4059 is in early phase I development
`but
`initial results presented at
`the American Society of
`Hematology (ASH) annual meeting in 2013 indicate compa-
`rable activity to ibrutinib in CLL with possibly less toxicity;
`significant efficacy was also observed in MCL (Salles et al,
`2013; Rule et al, 2013). Data with AVL-292/CC-292 in CLL
`is given below and with twice daily dosing suggests
`improved nodal responses (Harb et al, 2013). The combina-
`tion of AVL-292/CC-292 and lenalidomide in ABC-DLBCL
`is of
`interest, given that both molecules have significant
`activity against this disease subtype (Yang et al, 2012; Vose
`et al, 2013; NCT01766583). This combination is also being
`assessed in CLL (NCT01732861); it is a mechanistically and
`financially logical combination, given that both lenalidomide
`and CC-292 are now owned by Celgene. Another BTKi,
`CGI-1746 [CGI Pharmaceuticals (Branford, CT, USA) (now
`part of Gilead)]
`is highly BTK-specific and may have
`unique and potentially advantageous properties. CGI-1746
`binds to the ATP-binding pocket of BTK and stabilizes the
`non-phosphorylated form of
`the protein by internalizing
`the crucial residue Tyr551, which has to be phosphorylated
`
`15
`
` 13652141, 2014, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.12895, Wiley Online Library on [29/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1019, p. 4 of 11
`
`

`

`Review
`
`for BTK activation. CGI-1746 therefore has exquisite sensi-
`tivity for BTK (Di Paolo et al, 2011). Whether this mole-
`cule is now being developed for clinical use is not clear.
`There are insufficient data to compare the BTKi even indi-
`rectly at the current time. Paradoxically, given the data from
`the clinical studies to date, the ‘dirtier’ inhibitor, ibrutinib
`might possibly have significant advantages.
`
`Clinical studies with BTKi
`
`Ibrutinib as single agent has been used both in elderly patients
`(≥65 years) with treatment-na€ıve CLL and those with relapsed
`and refractory disease. A phase Ib-II trial was recently reported
`for relapsed and refractory CLL (Byrd et al, 2013b). Eighty-five
`patients considered to be ‘high risk’ were treated with either
`420 mg (51 patients) or 840 mg (34 patients). The overall
`response rate (ORR) was 71% in both cohorts [two complete
`responses (CR) and 34 partial responses (PR)] with a further
`20% (420 mg cohort) and 15% (840 mg cohort) achieving a
`PR with lymphocytosis. The response was independent of the
`number of previous therapies or high-risk features, including
`17p 13 deletion. At 26 months, progression-free survival (PFS)
`was estimated at 75% and OS at 83%. A lymphocytosis was
`observed in 78% of patients. Ibrutinib was very well tolerated;
`however, the higher dose was associated with more adverse
`events leading to discontinuation of treatment (two patients
`on 420 mg and four patients on the 840 mg dose). Perhaps
`not surprizingly in this patient group, pneumonia was the
`most common grade 3 or higher adverse event, seen in 10
`patients, and infections were seen more commonly early in
`treatment. Interestingly, despite the low levels of immunoglob-
`ulins seen in patients with the genetic mutation of BTK,
`immunoglobulin levels remained relatively stable during treat-
`ment. Indeed, IgA and IgM serum levels increase on treatment,
`signifying recovery of humoral
`immunity (Farooqui et al,
`2013). The treatment-na€ıve arm of this trial in patients older
`than 65 years (n = 31) was updated at the International Work-
`shop
`on Chronic
`Lymphocytic
`Leukaemia
`in
`2013
`(iwCLL2013): with a median follow up of 22 1 months, the
`ORR was 71% (CR 13%) with a 96% PFS (O’Brien et al,
`2014).
`An unanticipated finding was that ibruitinib induced a
`prompt lymphocytosis in the peripheral blood. A similar
`effect was observed in patients receiving PI3 delta kinase
`inhibitors (reviewed in Macias-Perez & Flinn, 2013) as well
`as CXCR4 antagonists. In ibrutinib-treated CLL patients the
`lymphocytosis is usually seen by 7 d, peaking within 4 weeks
`and then slowly decreasing with time, which is generally
`slower than that seen with treatment in MCL. The lympho-
`cytosis is not restricted to subtypes of CLL but patients with
`unmutated immunoglobulin variable-region heavy chain
`(IGHV) gene segments had an earlier resolution of lympho-
`cytosis compared to patients with mutated IGHV rearrange-
`ments (median 6 4 vs. 14 8 months) (Byrd et al, 2013a). It is
`not yet clear whether the rate of fall in lymphocyte count
`
`16
`
`will perhaps become a prognostic factor as important as
`lymphocyte doubling time.
`
`What are the practicalities?
`
`Ibrutinib is given orally once a day. In CLL patients, the
`optimal dose, determined from the above trials and being
`taken forward in future trials, is 420 mg. Given the broad
`expression of BTK in haematological cells and the ibrutinib
`kinome, it is surprising that the drug is so well tolerated.
`Toxicities are very manageable on an out-patient basis with
`the most common treatment-related adverse event being
`mild to moderate diarrhoea, fatigue and nausea, with pneu-
`monia more commonly seen in CLL patients. Haematological
`grade 3 or higher neutropenia, thrombocytopenia and anae-
`mia are seen in 10–20% of patients with previously treated
`CLL and these improve with treatment. Bleeding is seen in a
`subset of patients and current trials exclude the concurrent
`use of warfarin but not other anticoagulant therapies.
`
`How will ibrutinib compare to other single agents?
`
`Two randomized, multicentre phase III trials are currently
`ongoing to address this question. RESONATE-2 is comparing
`chlorambucil against
`ibrutinib in a 1:1 randomization in
`treatment–na€ıve CLL/SLL patients older
`than 65 years
`(NCT01722487), whilst RESONATE compared the novel
`CD20 monoclonal antibody ofatumumab against ibrutinib in
`relapsed/refractory CLL/SLL patients (NCT01578707). The
`latter study was stopped early by the data monitoring com-
`mittee after an an interim analysis showed that patients on
`ibrutinib had a statistically significant improvement not only
`in PFS (the primary study end point), but also in OS (a sec-
`ondary end point), when compared with ofatumumab
`(http://www.pmlive.com/pharma_news/j_and_js_imbruvica_
`tops_arzerra_in_phase_iii_trial_531853 and please see also
`http://www.btktrials.com.)
`
`Will remission be achieved using single agent ibrutinib?
`
`The present indication is that there are very few complete
`responses with ibrutinib (2% in previously treated CLL, 13%
`in treatment na€ıve CLL) and progression is rapidly seen when
`treatment is interrupted. However, most patients continue to
`have an ongoing response with continued treatment and the
`role of combination therapy to improve responses, with the
`aim of attaining a minimal residual disease (MRD) negative
`response without significant toxicity is keenly debated.
`
`What about the chance of developing resistance to BTK
`inhibition?
`
`In CLL patients who had developed resistance to ibrutinib,
`comparative DNA sequence analysis of
`the genome was
`performed in samples at baseline and at disease progression.
`
`ª 2014 John Wiley & Sons Ltd
`British Journal of Haematology, 2014, 166, 12–22
`
` 13652141, 2014, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/bjh.12895, Wiley Online Library on [29/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1019, p. 5 of 11
`
`

`

`In five patients analysed, four patients developed a mutation
`resulting in the substitution of cysteine for serine at position
`481 with a further mutation seen in the major downstream
`BTK target, PLCc2. All five patients had high risk cytogenet-
`ics. It is therefore likely that mutations will develop with
`prolonged therapy, highlighting the necessity for the develop-
`ment of mechanism-based combinations.
`
`What are the mechanisms of action of BTKi in vivo?
`
`Despite the clinical success of ibrutinib, there remain some
`fundamental questions
`concerning the mechanism(s) of
`action of this drug and other BTKi
`in vivo. Specifically,
`how much of the tumour cell death induced by BTK inhi-
`bition is due to a direct cytotoxic effect on the malignant B
`cells and how much is due to death by deprivation of pro-
`survival signalling? The relative importance of
`these two
`mechanisms may vary according to disease type. Firstly, it
`is clear that BTKi can have significant activity against some
`cases of chemotherapy-refractory non-germinal center B
`cell-like (GCB) DLBCL (deVos et al, 2013; Yoshizawa et al,
`2013). Given that these malignancies are rapidly prolifera-
`tive,
`this suggests that BTK inhibition directly results in
`DLBCL cell death without significant
`lymphocytosis. The
`precise mechanism by which this occurs remains unclear;
`only one ABC-DLBCL cell line (TMD8) is sensitive to BTKi
`in vitro (Kozaki et al, 2013). Similar remarks may apply to
`nodal MCL. However,
`this
`simple mechanism is con-
`founded in CLL where the malignant lymphocytes initially
`increase significantly and persist in the peripheral blood for
`many months during therapy and decline only slowly with
`time. These clinical data suggest ‘death by deprivation’. In
`contrast however,
`lymph node size diminishes rapidly in
`CLL, with the majority of effects seen in the first 2 months
`of therapy (Byrd et al, 2013b). The egress of CLL cells into
`the peripheral blood is not sufficient to account for the
`observed decreases in lymph node size (Garg et al, 2013;
`CV Hutchinson and MJS Dyer, unpublished observations).
`BTK inhibition clearly has significant activity across all sub-
`types of CLL including patients with poor risk disease indi-
`cating a TP53 independent mechanism of action. However,
`precise mechanisms of action of BTKi in vivo need to be
`determined in order to design rational combination thera-
`pies.
`
`What about combination therapy?
`
`In CLL, the combination with rituximab would seem logical
`and potentially synergistic;
`‘mobilize’ the cells with BTKi
`and then target them in the circulation with a monoclonal
`antibody. Burger et al (2012) reported on a single centre
`study of 40 patients with high risk CLL [del17p or TP53
`mutation (treated or untreated), PFS <36 months after
`frontline
`chemoimmunotherapy or
`relapsed CLL with
`del11q]. Patients received daily ibrutinib at 420 mg plus
`
`ª 2014 John Wiley & Sons Ltd
`British Journal of Haematology, 2014, 166, 12–22
`
`Review
`
`rituximab 325 mg/m2 weekly for
`then
`the first month,
`monthly for a total 6 months and then continued on daily
`ibrutinib. The results were recently updated at
`the ASH
`annual meeting in 2013 (Keating et al, 2013), at a median
`follow up of 14 months, 32 of 40 patients continued on
`therapy without disease progression (16 out of 20 with
`del17p or TP53 mutation). Of the 39 patients evaluable,
`there was an ORR of 95% (PR 87%, CR 8%). The regimen
`was generally well
`tolerated although infections were the
`most common complication (six cases of pneumonia, three
`cases of upper respiratory infections). Interestingly, the lym-
`phocytosis peaked earlier
`(within first month) and the
`duration was
`shorter
`(back
`to at
`least baseline
`at
`3 months). Whether this will equate to better responses is
`keenly anticipated. The combination of
`ibrutinib and of-
`atumumab, the fully humanized CD20 antibody, also has
`shown favourable responses in patients with relapsed/refrac-
`tory CLL/small lymphocytic lymphoma/prolymphocytic leu-
`kaemia, with 100% ORR (24/24 patients) within six cycles
`with minimal toxicity and rapid response rate (Jaglowski
`et al, 2012).
`Going one step further, will improved responses be seen
`with BTKi in combination with chemoimmunotherapy? The
`final result

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