throbber
Drugs (2020) 80:1901–1928
`https://doi.org/10.1007/s40265-020-01422-9
`
`REVIEW ARTICLE
`
`The Development of Cladribine Tablets for the Treatment of Multiple
`Sclerosis: A Comprehensive Review
`
`Kottil Rammohan1 · Patricia K. Coyle2 · Elke Sylvester3 · Andrew Galazka3 · Fernando Dangond4 · Megan Grosso5 ·
`Thomas P. Leist6
`
`Published online: 28 November 2020
`© The Author(s) 2020
`
`Abstract
`Cladribine is a purine nucleoside analog initially developed in the 1970s as a treatment for various blood cancers. Due to
`the molecule’s ability to preferentially reduce T and B lymphocytes, it has been developed into an oral formulation for the
`treatment of multiple sclerosis (MS). The unique proposed mechanism of action of cladribine allows for the therapy to be
`delivered orally over two treatment-week cycles per year, one cycle at the beginning of the first month and one cycle at the
`beginning of the second month of years 1 and 2, with the potential for no further cladribine treatment required in years 3
`and 4. This review summarizes the clinical development program for cladribine tablets in patients with MS, including the
`efficacy endpoints and results from the 2-year phase III CLARITY study in patients with relapsing–remitting MS (RRMS),
`the 2-year CLARITY EXTENSION study, and the phase III ORACLE-MS study in patients with a first clinical demyelinat-
`ing event at risk for developing MS. Efficacy results from the phase II ONWARD study, in which cladribine tablets were
`administered as an add-on to interferon-β therapy in patients with RRMS, are also summarized. A review of all safety data,
`including lymphopenia, infections, and malignancies, is provided based on data from all trials in patients with MS, includ-
`ing the initial parenteral formulation studies. Based on these data, cladribine tablets administered at 3.5 mg/kg over 2 years
`have been approved across the globe for various forms of relapsing MS.
`
`1 Introduction
`
`Multiple sclerosis (MS) is a chronic, inflammatory, immune-
`mediated demyelinating and neurodegenerative disease of
`the central nervous system (CNS). It is one of the most
`
`Enhanced Digital Features To view enhanced digital features for
`this article, go to https ://doi.org/10.1007/s4026 5-020-01422 -9
`
` * Kottil Rammohan
`krammohan@med.miami.edu
`
`1 Multiple Sclerosis Center, University of Miami, Miami, FL,
`USA
`
`2 Multiple Sclerosis Comprehensive Care Center, Stony Brook
`University, Stony Brook, NY, USA
`
`3 Merck KGaA, Darmstadt, Germany
`
`4
`
`5
`
`EMD Serono Research & Development Institute, Inc.,
`Billerica, MA, USA, an affiliate of Merck KGaA, Darmstadt,
`Germany
`
`EMD Serono, Inc., Rockland, MA, USA, an affiliate
`of Merck KGaA, Darmstadt, Germany
`
`6 Comprehensive MS Center, Jefferson University,
`Philadelphia, PA, USA
`
`common causes of serious neurological disability in young
`adults, and one of the most prevalent neurological disor-
`ders in the world [1]. In 2016, the estimated global preva-
`lence of MS was 2.2 million, an increase of 10.4% from the
`age-standardized MS prevalence in 1990 [2]. Among the
`more than 2 million people with MS, more than 900,000 are
`thought to reside in the United States (US) [3]. About 85%
`of patients with MS present with relapsing–remitting MS
`(RRMS), which is characterized by periodic acute exacerba-
`tions of disease activity (relapses) punctuated by periods of
`clinical stability. Relapses may be associated with partial or
`complete recovery [4].
`MS is associated with poor health-related quality of
`life and can have a profound effect on social functioning,
`employment status, and healthcare costs [5, 6]. The mean
`age at onset of symptoms is 30 years, and approximately
`75% of patients are female [7]. Therefore, many patients
`are women of childbearing age, and MS can impact fam-
`ily planning due to risks associated with treatments that
`may affect pregnancy [8]. While there are no adequately
`controlled studies of disease-modifying drugs (DMDs) and
`pregnancy, there is a significant body of evidence on the
`safety of DMDs during pregnancy, generated primarily
`
`Vol.:(0123456789)
`
`Merck 2023
`TWi v Merck
`IPR2023-00049
`
`

`

`
`
`1902
`
`Key Points
`
`Multiple sclerosis (MS) is a neurodegenerative disease
`that affects more than 2 million people globally and
`is associated with poor quality of life. MS involves an
`aberrant immune system attacking the central nervous
`system, and disease-modifying drugs are thought to act
`by suppressing or modulating the immune system.
`
`Cladribine tablets are the first oral therapy with a short-
`course, limited cycle dosing schedule approved for
`patients with relapsing forms of MS (RMS). In clinical
`studies, cladribine tablets have demonstrated efficacy in
`patients across the RMS spectrum.
`
`The safety profile of cladribine tablets monotherapy at
`the recommended dosage of 3.5 mg/kg includes data
`from > 3700 patient-years of cladribine exposure at this
`dose, and from > 2200 patient-years of placebo expo-
`sure in clinical trials. The most common adverse events
`reported with cladribine tablets include headaches and
`lymphopenia. Additional adverse events examined
`include malignancy and infections.
`
`from clinical experience and pregnancy registries. Large
`interferon (IFN)-β registry studies have found no evidence
`for differences in infant size [9], congenital anomalies, or
`miscarriages [10], and recently updated IFN-β Summaries
`of Product Characteristics recommend to continue treat-
`ment during pregnancy and while breastfeeding if clinically
`needed [11, 12]. Glatiramer acetate is also generally consid-
`ered to be safe for use during pregnancy [13, 14]. However,
`the labels for the majority of DMDs for patients with MS
`either state to use ‘only if clearly needed’ [15], to avoid these
`therapies unless the benefits outweigh the risks [16–19], or
`not to use at all, and a washout period is recommended prior
`to pregnancy [20–26]. Longitudinal studies indicate that life
`expectancy is shorter for patients with MS compared with
`the general population (approximately 7 years shorter in a
`Norwegian population studied over a 60-year period) [27],
`but rising prevalence without parallel changes in incidence,
`and a shift in peak age towards older age groups, suggest
`that survival is improving (based on Canadian data col-
`lected over 2 decades) [28]. An aging MS population may
`be associated with significant disability; in a self-reporting
`postal survey conducted in a Canadian cohort of MS patients
`aged > 55 years who had been living with MS for a mean of
`approximately 33 years, 28% either required a wheelchair or
`were bedridden [29].
`Lymphocytes play a central role in the pathogenesis of
`MS, with the actions of both B and T cells in the periphery
`and in the CNS implicated from an early stage in the disease
`
`K. Rammohan et al.
`
`process. Immune cells activated in the periphery enter the
`CNS, facilitated by chemokines [30, 31]. Autoreactive T
`cells are present in acute CNS lesions in early MS [30, 32];
`their reaction to myelin protein-derived antigens [32] con-
`tributes to direct cytotoxic effects and stimulation of mac-
`rophages [31, 33]. B cells have a role in the proliferation and
`reactivation of T cells, acting as antigen-presenting cells,
`and fully differentiated B cells further contribute to demyeli-
`nation via the production of antibodies to myelin by plasma
`cells [30, 31, 33, 34]. Thus, the combined actions of T cells,
`B cells, and macrophages contribute to demyelination and
`axonal damage, triggering neurodegenerative processes from
`disease onset [30, 31]. Later in the course of the disease,
`chronic CNS inflammation drives ongoing neurodegenera-
`tion, possibly via neurotoxic inflammatory mediators pro-
`duced by activated microglia and astrocytes [32].
`Treatment options for relapsing forms of MS (RMS) com-
`prise a number of immunosuppressive and immunomodu-
`latory agents. The first DMD, IFN-β-1b, was approved by
`the US Food and Drug Administration (FDA) for RMS in
`1993 [19] and by the European Medicines Agency (EMA) in
`1995 [35]. Alternative forms of IFN-β (subcutaneous [SC]
`and intramuscular [IM] IFN-β-1a) and glatiramer acetate
`entered the market between 1996 and 2002, becoming stand-
`ard treatment over the ensuing years [36]. The introduction
`of natalizumab, a recombinant monoclonal antibody against
`cell adhesion molecule α4-integrin [23] (initially approved
`in 2004, withdrawn from the market, then reintroduced in
`2006 [37]), marked a shift in the treatment paradigm for MS
`[38]. While this molecule was perceived to be more effective
`than the IFNs and glatiramer acetate [39], it is associated
`with greater risks, including progressive multifocal leukoen-
`cephalopathy (PML), which is rare but carries significant
`morbidity and a mortality rate of approximately 23% [40].
`After a span of nearly 20 years of injectable therapies, oral
`DMDs became available. While patients have generally pre-
`ferred oral DMDs over injections when given the choice
`[41], most approved oral DMDs, including fingolimod, terif-
`lunomide, dimethyl fumarate and siponimod, require regular
`and ongoing dosing once- or twice-daily [20–22].
`More recently, therapies with different hypothesized
`mechanisms of action (MOA) that require less frequent dos-
`ing have become available. Ocrelizumab is an anti-CD20
`antibody, administered via intravenous (IV) infusion every
`6 months (after the initial dose, which is split across two
`infusions 2 weeks apart) [25, 42]. Alemtuzumab is an anti-
`CD52 antibody with an infrequent dosing regimen, involv-
`ing infusions over a course of 5 days in year 1, followed
`by 3 days in year 2, with further 3-day courses adminis-
`tered as needed thereafter, separated by intervals of at least
`12 months [24, 43]. Both antibodies are thought to deplete
`lymphocytes via cytolysis and complement-mediated lysis,
`with different profiles based on expression of their respective
`
`

`

`Development of Cladribine Tablets
`
`1903
`
`target antigens on B cells, T cells, and other immune cells
`[24, 25, 44–46].
`Cladribine tablets are the first oral therapy with an infre-
`quent dosing schedule, administered in two yearly treatment
`courses, each divided into two treatment cycles compris-
`ing 4–5 days of treatment [12, 26]. Cladribine tablets are
`thought to exert their clinical effects via a transient reduction
`of selective lymphocyte subtypes, followed by a recovery
`period during which cell numbers return to the normal range
`[47, 48]. While immune function is restored as cell num-
`bers recover, cell subtype ratios are thought to be altered,
`resulting in a reduction in autoreactive lymphocytes [47–49].
`Thus, a short dosing period is thought to achieve sustained
`effects on lymphocytes that persist for an extended period
`after the treatment period has ended.
`Cladribine tablets have been approved worldwide by
`many different regulatory authorities, including the EMA
`in 2017 [50] and the FDA in 2019 [51]. In this review, we
`provide an overview of the development of cladribine tablets
`as a therapy for patients with MS, from initial discovery and
`development of the molecule to its putative MOA in MS. We
`also review the pertinent efficacy data from clinical studies,
`and an integrated safety analysis of > 3700 patient-years’
`clinical trial experience with cladribine tablets monotherapy
`at the recommended dosage.
`
`1.1 Discovery and Development
`
`Cladribine (2-chlorodeoxyadenosine [2-CdA]) was first
`synthesized nearly 50 years ago [52] (Fig. 1). It is a syn-
`thetic purine nucleoside analog of deoxyadenosine that was
`developed to selectively target lymphocytes in lymphopro-
`liferative diseases (e.g. hairy cell leukemia) and autoimmune
`disorders [53–55]. The design of cladribine was inspired
`by the consequences of adenosine deaminase (ADA) defi-
`ciency in children [55, 56]. The selective vulnerability of
`lymphocytes in this autosomal recessive genetic disorder
`was described by Carson and colleagues as resulting from
`the preferential accumulation of cytotoxic deoxyadenosine
`nucleotides in lymphocytes, causing lymphocytopenia [54,
`57]. Based on this understanding, several nucleoside analogs
`were synthesized, including cladribine in the early 1970s
`[54, 58, 59]. Following the efficacy observed in lymphoid
`leukemias and the ability of cladribine to selectively target
`lymphocyte populations, the drug was considered for poten-
`tial use in MS [53].
`
`1.2 Mechanism of Action
`
`Cladribine (2-CdA) is a small molecule (molecular mass
`285.7  g/mol [60]) prodrug that is taken up by cells via
`nucleoside transporter proteins and becomes active in cer-
`tain cells upon phosphorylation to 2-chlorodeoxyadenosine
`
`triphosphate (2-Cd-ATP). 2-CdA undergoes sequential intra-
`cellular phosphorylation, first to 2-chlorodeoxyadenosine
`monophosphate (2-Cd-AMP) mediated by deoxycytidine
`kinase (DCK), and subsequently to 2-chlorodeoxyadenosine
`diphosphate (2-Cd-ADP) and 2-Cd-ATP by other kinases.
`De-phosphorylation by 5(cid:397)-nucleotidases (5(cid:397)-NTases) pre-
`vents accumulation of 2-Cd-ATP in most cells [31]. Phos-
`phorylation preferentially occurs in B and T lymphocytes
`due to their unique constitutively high DCK and relatively
`low 5(cid:397)-NTase levels, compared with other cell types. DCK
`is a rate-limiting enzyme in the nucleoside salvage path-
`way that provides deoxyribonucleosides (dNTPs). The high
`DCK level is thought to be important for lymphocyte clonal
`expansion during development and immune reactions [61,
`62]. Lymphocytes are therefore susceptible to accumulation
`of 2-Cd-ATP (Fig. 2). In cells where 2-Cd-ATP accumulates,
`it incorporates into deoxyribonucleic acid (DNA) strands,
`disrupting DNA synthesis and cell cycle progression, and
`inhibits enzymes involved in DNA synthesis, leading to cell
`death in both proliferating and quiescent lymphocytes [31,
`59, 63].
`While the precise mechanisms by which cladribine exerts
`its therapeutic effects in patients with MS is not known, its
`effects on B and T lymphocytes are thought to play a central
`role. Cladribine preferentially reduces cells of the adaptive
`immune system, while leaving the innate immune system
`relatively spared. Absolute lymphocyte counts (ALC) and
`B- and T-cell subset counts rapidly reach nadir following
`administration of cladribine tablets, and gradual lymphocyte
`count recovery begins soon after treatment and continues
`for months afterwards [64]. In the phase III CLARITY and
`CLARITY Extension studies [65, 66], median ALC reached
`nadir, at the lower limit of normal (LLN) at week 13, and
`was followed by recovery of cell counts back to the nor-
`mal range (Fig. 3). In year 2, median ALC fell below the
`LLN but recovered into the normal range by week 84, or
`approximately 30 weeks postdose after completion of the
`second treatment cycle in year 2. Reduction of CD19+ B
`cells in year 1 also reached nadir at about week 13 (below
`LLN and approximately 80% change from baseline), fol-
`lowed by recovery towards baseline values (Fig. 4). CD19+
`B-cell counts also fell below LLN in year 2, and recovered
`to LLN by week 84. CD4+ T-cell median counts showed a
`lesser decline from baseline to nadir (approximately 50%
`change from baseline, also reached at week 13) in year 1, fol-
`lowed by a more gradual recovery towards baseline (Fig. 5).
`While CD4 + T cells did not reach the LLN in year 1, they
`fell below the LLN in year 2 and reached threshold values
`for recovery by week 96, approximately 43 weeks post last
`therapy dose. Median CD8 + T cells never dropped below
`LLN (Fig. 6) [64]. Analysis of CD4 + T-cell subtypes in
`ORACLE-MS showed that subpopulations displayed dif-
`ferent repopulation dynamics, leading to changes in the
`
`

`

`
`
`1904
`
`K. Rammohan et al.
`
`Clinical Development of Cladribine Tablets
`
`CLINICAL MILESTONES
`
`Discovery of
`ADA deficiency52
`
`2-CdA first synthesized
`by Christensen
`and colleagues122
`
`Global Phase 4
`program initiated
`
`Phase 3 CLARITY
`results published65,a
`
`Phase 3 ORACLE-MS
`results published94,b
`
`US Phase 4
`program initiated
`Integrated safety
`results published102,d
`
`Phase 3
`CLARITY-
`EXT results
`published66
`
`Phase 2 ONWARD
`results published100,c
`
`Completion of
`PREMIERE long-term
`safety registry study123
`
`1972
`
`1993
`
`2010
`
`2011
`
`2014
`
`2016
`
`2017
`
`2018
`
`2019
`
`IV formulation approved
`by FDA as an orphan
`drug for the treatment
`of hairy cell leukemia124
`
`First reviewed
`by EMA126
`FDA grants priority
`review for relapsing MS127
`
`REGULATORY MILESTONES
`
`Approved in
`Russia and
`Australia for
`RRMS but later
`withdrawn125
`
`CRL issued
`by the FDA
`for US
`approval128
`
`Marketing
`authorization
`application
`resubmitted to
`EMA with new
`clinical data from
`CLARITY-EXT,
`ORACLE-MS,
`and PREMIERE129
`
`EMA
`approval130
`
`NDA
`resubmitted to
`FDA with
`additional safety
`clinical data and
`analyses (no new
`efficacy data was
`submitted)131
`
`approval51
`
`Approved
`in more than
`75 countries
`as of July 2020
`
`Cladribine tablets were approved by the EMA in 2017 and the FDA in 2019
`
`Fig. 1 Cladribine tablets development milestones [51, 52, 65, 66, 94,
`100, 102, 122–131]. aNaive and treatment-experienced patients with
`RRMS. bTreatment-naive patients at high risk for developing MS.
`cPatients with active RRMS in combination with IFN-β. dIncluding
`patients from CLARITY, CLARITY–EXT, ORACLE-MS, and PRE-
`
`MIERE. ADA adenosine deaminase, 2-CdA 2-chlorodeoxyadenosine
`deaminase, CRL complete response letter, EMA European Medicines
`Agency, FDA Food and Drug Administration, IFN interferon, IV
`intravenous, MS multiple sclerosis, NDA new drug application, RRMS
`relapsing–remitting MS
`
`relative proportions of the CD4 +  T-cell subpopulations
`[67]. Median monocyte and neutrophil counts remained
`within the normal range throughout the CLARITY and
`CLARITY Extension studies (Figs. 7, 8), while the effects
`on natural killer (NK) cells were moderate and transient,
`with a 30–44% median decrease in NK cells followed by a
`recovery towards pretreatment levels by week 24 (Fig. 9).
`These findings support the view that the impact of cladribine
`on the innate immune system is relatively minor [67]. The
`preferential reduction of lymphocyte subpopulations, fol-
`lowed by the pattern of lymphocyte count recovery (termed
`immune reconstitution), may ‘reset’ the immune system to a
`less autoreactive state [64], a putative mechanism with con-
`siderable potential in the long-term treatment of MS [48].
`
`was required. Phase II and III studies that were ongoing at
`the time were completed, and a long-term safety registry
`was continued. The additional data served to support the
`thorough characterization of the safety profile of cladrib-
`ine tablets in MS. Evaluation of these additional data and
`analyses of the compound’s longer-term benefit–risk profile
`supported new submissions to regulatory authorities. Clad-
`ribine tablets received EMA approval in August 2017 [50]
`and FDA approval in March 2019 [26]. As of July 2020,
`cladribine tablets have gained marketing authorization in
`more than 75 countries for the treatment of patients with
`various forms of RMS.
`
`1.4 Dosing Schedule
`
`1.3 Approvals
`
`Cladribine tablets were developed as a potential treatment
`for MS in response to the efficacy seen in early studies of
`parenteral cladribine and the need for additional treatment
`options [53, 68, 69]. The pivotal CLARITY trial was com-
`pleted in 2009 and published in 2010; however, some regu-
`latory authorities indicated in 2011 that improved under-
`standing of safety risks and the overall benefit–risk profile
`
`Two dosages were investigated during the clinical develop-
`ment of cladribine tablets: 3.5 mg/kg and 5.25 mg/kg (cumu-
`lative doses over 2 years). In addition, in certain study arms,
`patients received retreatment in years 3 and 4 (cumulative
`doses over 4 years of 7.0 mg/kg and 8.75 mg/kg). The 3.5 mg/
`kg and 5.25 mg/kg doses appeared to be equally efficacious,
`but the 5.25 mg/kg dose was associated with an increased
`rate of higher-grade lymphopenia [65]. The 3.5 mg/kg dose
`was considered to have the most favorable benefit–risk profile.
`
`

`

`Development of Cladribine Tablets
`
`1905
`
`B and T lymphocytes
`
`HIGH
`kinase-to-phosphatase ratio
`
`Cladribine
`molecule
`
`Phosphatase
`Inactivation by
`5’NTase
`
`Kinase
`Activation
`by DCK
`
`High levels of
`activated cladribine
`molecule
`
`Other cells
`
`LOW
`kinase-to-phosphatase ratio
`
`Cladribine
`molecule
`
`Phosphatase
`Inactivation by
`5’NTase
`
`Kinase
`Activation
`by DCK
`
`Low levels of
`activated cladribine
`molecule
`
`CD4+ lymphocytes
`CD8+ lymphocytes
`Skin
`Heart
`Brain
`Ovary
`Liver
`Kidney
`Lung
`Testis germ cell
`Testis
`
`mRNA Expression of
`Inactivating Phosphatase
`
`mRNA Expression of
`Activating Kinase
`
`Fig. 2 Ratios of DCK to 5(cid:397)-NTase mRNA expression in T cells
`and various non-hematologic cells [58]. 5(cid:397)-NTase 5(cid:397)-nucleotidases,
`DCK deoxycytidine kinase, mRNA messenger RNA. *Calculated
`
`using data from the BioGPS website (available at https ://biogp
`s.org/#goto=welco me). Adapted with permission from Giovannoni
`[58]
`
`In countries where approved, the recommended cumu-
`lative dose of cladribine tablets is 3.5 mg/kg given over
`2 years (one treatment course is 1.75 mg/kg/year) [12, 26].
`Each course consists of two treatment weeks or cycles—one
`cycle at the beginning of the first month and one cycle at
`the beginning of the second month. Each treatment cycle
`lasts 4 or 5 consecutive days, depending on the patient’s
`weight, and patients receive one or two 10 mg tablets per
`
`day. Lymphocytes must be within normal limits prior to ini-
`tiating the first course and at least 800 cells/mm3 prior to
`initiating the second course. Modeling simulations suggest
`that 92% of patients would not require a delay in receiving
`the second treatment course and < 1% would be ineligible
`for treatment in year 2 due to a delay in recovery of more
`than 6 months [70]. No further treatment with cladribine
`tablets may be required in years 3 and 4; a patient cohort
`
`

`

`Weeks 1 & 5
`
`Weeks 48 & 52
`
`K. Rammohan et al.
`
`Cladribine tablets 3.5 mg/kg
`Placebo
`No additional active treatment
`No additional placebo
`Cladribine administration
`
`LLN 1.0 x 109/L
`
`Grade 1
`
`Grade 2
`
`Grade 3
`
`Grade 4
`
`0
`
`24
`
`48
`
`72
`
`96
`
`120
`
`144
`
`168
`
`192
`
`216
`
`240
`
`264
`
`288
`
`312
`
`Week
`
`3.0
`
`2.5
`
`2.0
`
`1.5
`
`1.0
`
`0.5
`
`0.0
`
`Median (Q1–Q3) lymphocyte counts
`
`(109/L)
`
`
`
`1906
`
`Fig. 3 Adaptive immune cell
`counts in patients with RMS
`receiving cladribine 3.5 mg/kg
`or placebo—absolute lympho-
`cyte counts [58]. LLN lower
`limit of normal, Q1–Q3 inter-
`quartile range, RMS relapsing
`multiple sclerosis. Pooled data
`from CLARITY, CLARITY
`EXT, and PREMIERE. Visits
`with a sample size ≥ 30 are
`displayed. Adapted with permis-
`sion from Giovannoni [58]
`
`Number of patients
`434
`
`683
`
`415
`
`645
`
`263
`
`437
`
`378
`
`624
`
`358
`
`574
`
`86
`
`298
`
`94
`
`265
`
`69
`
`167
`
`34
`
`147
`
`131
`
`116
`
`106
`
`66
`
`32
`
`LLN 0.1 x 109/L
`
`Weeks 1 & 5
`
`Weeks 48 & 52
`
`Cladribine tablets 3.5 mg/kg
`Placebo
`No additional active treatment
`Cladribine administration
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`
`Median (Q1–Q3) CD19+
`
`counts (109/L)
`
`Fig. 4 Adaptive immune cell
`counts in patients with RMS
`receiving cladribine 3.5 mg/
`kg or placebo—B (CD19+)
`cells [64]. LLN lower limit of
`normal, Q1–Q3 interquartile
`range, RMS relapsing multiple
`sclerosis. Pooled data from
`CLARITY, CLARITY EXT,
`and PREMIERE. Visits with a
`sample size ≥ 30 are displayed.
`Adapted with permission from
`Comi et al. [64]
`
`0.0
`
`0
`
`24
`
`48
`
`Number of patients
`79
`
`220
`
`81
`
`203
`
`61
`
`137
`
`72
`
`74
`
`191
`
`96
`Week
`
`78
`
`195
`
`120
`
`144
`
`168
`
`192
`
`38
`
`56
`
`68
`
`64
`
`that received additional treatments in years 3 and 4 had
`comparable clinical efficacy compared with a study cohort
`that received placebo in years 3 and 4 [66]. The safety and
`efficacy of additional treatment courses after year 4 have not
`been studied. There are no current definitions of treatment
`failure with cladribine tablets. Any definition would need to
`be on a case-by-case basis, most likely taking into account
`disease activity prior to commencing cladribine tablets [71].
`
`1.5 Clinical Pharmacology
`
`Following oral administration, cladribine is rapidly
`absorbed, reaching maximal plasma concentrations after
`approximately 0.5 h (in the fasted state). Food delays absorp-
`tion but does not affect overall exposure [72]. Oral bioa-
`vailability is approximately 40% compared with parenteral
`administration; reduced bioavailability is likely due to gas-
`trointestinal efflux mediated by the breast cancer resistance
`
`

`

`1907
`
`Cladribine tablets 3.5 mg/kg
`Placebo
`No additional active treatment
`Cladribine administration
`
`LLN 0.35 x 109/L
`
`72
`
`74
`
`191
`
`96
`Week
`
`78
`
`195
`
`120
`
`144
`
`168
`
`192
`
`38
`
`56
`
`68
`
`64
`
`Cladribine tablets 3.5 mg/kg
`Placebo
`No additional active treatment
`Cladribine administration
`
`LLN 0.2 x 109/L
`
`72
`
`74
`
`191
`
`96
`Week
`
`78
`
`195
`
`120
`
`144
`
`168
`
`192
`
`38
`
`56
`
`68
`
`64
`
`Weeks 1 & 5
`
`Weeks 48 & 52
`
`0
`
`24
`
`48
`
`1.2
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Median (Q1–Q3) CD4+
`
`counts (109/L)
`
`Development of Cladribine Tablets
`
`Fig. 5 Adaptive immune cell
`counts in patients with RMS
`receiving cladribine 3.5 mg/kg
`or placebo—T helper (CD4+)
`cells [64]. LLN lower limit of
`normal, Q1–Q3 interquartile
`range, RMS relapsing multiple
`sclerosis. Pooled data from
`CLARITY, CLARITY EXT,
`and PREMIERE. Visits with a
`sample size ≥ 30 are displayed.
`Adapted with permission from
`Comi et al. [64]
`
`Number of patients
`79
`
`220
`
`81
`
`203
`
`61
`
`137
`
`Weeks 1 & 5
`
`Weeks 48 & 52
`
`0
`
`24
`
`48
`
`0.6
`
`0.5
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`
`0.0
`
`Median (Q1–Q3) CD8+
`
`counts (109/L)
`
`Number of patients
`79
`
`220
`
`81
`
`203
`
`61
`
`137
`
`Fig. 6 Adaptive immune cell
`counts in patients with RMS
`receiving cladribine 3.5 mg/
`kg or placebo—T cytotoxic
`(CD8+) cells [64]. LLN lower
`limit of normal, Q1–Q3 inter-
`quartile range, RMS relapsing
`multiple sclerosis. Pooled data
`from CLARITY, CLARITY
`EXT, and PREMIERE. Visits
`with a sample size ≥ 30 are
`displayed. Adapted with permis-
`sion from Comi et al. [64]
`
`protein (BCRP), a transporter with affinity for cladribine
`[72]. Cladribine is rapidly taken up by lymphocytes, where
`it or its phosphorylated products accumulate, and reach
`intracellular concentrations that are approximately 30- to
`40-fold greater than in plasma within 1 h of administration
`[72]. No measurable accumulation of cladribine in plasma
`has been observed following repeated once-daily oral dosing
`[72]. The estimated terminal half-life is approximately 1 day
`[26]. About 50% of cladribine is cleared renally. Non-renal
`clearance occurs largely in lymphocytes; accumulated 2-Cd-
`AMP and 2-Cd-ATP are cleared via lymphocyte elimination
`
`pathways and their intracellular half-life is approximately
`10–15 h [72].
`Due to the risk of additive effects on the immune sys-
`tem, concomitant immunosuppressive or myelosuppressive
`therapy (e.g. cyclosporine, methotrexate) with cladribine is
`contraindicated. Previous immunomodulatory or immuno-
`suppressive therapy use, including washout periods, should
`be considered when initiating cladribine tablets [12, 26].
`Cladribine pharmacokinetics was not altered when used
`concomitantly with the proton pump inhibitor pantopra-
`zole or with IFN-β. However, the risk of lymphopenia may
`
`

`

`Weeks 1 & 5
`
`Weeks 48 & 52
`
`K. Rammohan et al.
`
`LLN 0.2 x 109/L
`
`Cladribine tablets 3.5 mg/kg
`Placebo
`No additional active treatment
`No additional placebo
`Cladribine administration
`
`0
`
`24
`
`48
`
`72
`
`96
`Week
`
`120
`
`144
`
`168
`
`192
`
`0.6
`
`0.5
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`
`0.0
`
`Median (Q1–Q3) monocyte
`
`counts (109/L)
`
`
`
`1908
`
`Fig. 7 Innate immune cell
`counts in patients with RMS
`receiving cladribine 3.5 mg/kg
`or placebo—monocytes [132].
`LLN lower limit of normal,
`Q1–Q3 interquartile range, RMS
`relapsing multiple sclerosis.
`Pooled data from CLARITY,
`CLARITY EXT, and PRE-
`MIERE. Visits with a sample
`size ≥ 30 are displayed
`
`Number of patients
`435
`
`683
`
`415
`
`645
`
`263
`
`437
`
`378
`
`624
`
`358
`
`574
`
`85
`
`293
`
`93
`
`249
`
`65
`
`148
`
`30
`
`116
`
`LLN 2.03 x 109/L
`
`Cladribine tablets 3.5 mg/kg
`Placebo
`No additional active treatment
`No additional placebo
`Cladribine administration
`
`72
`
`378
`
`624
`
`120
`
`144
`
`168
`
`192
`
`96
`Week
`
`358
`
`574
`
`85
`
`293
`
`93
`
`249
`
`65
`
`148
`
`30
`
`116
`
`Weeks 1 & 5
`
`Weeks 48 & 52
`
`0
`
`24
`
`48
`
`6.0
`
`5.0
`
`4.0
`
`3.0
`
`2.0
`
`1.0
`
`0.0
`
`Median (Q1–Q3) neutrophil
`
`counts (109/L)
`
`Fig. 8 Innate immune cell
`counts in patients with RMS
`receiving cladribine 3.5 mg/kg
`or placebo—neutrophils [132].
`LLN lower limit of normal,
`Q1–Q3 interquartile range, RMS
`relapsing multiple sclerosis.
`Pooled data from CLARITY,
`CLARITY EXT, and PRE-
`MIERE. Visits with a sample
`size ≥ 30 are displayed
`
`Number of patients
`435
`
`683
`
`415
`
`645
`
`263
`
`437
`
`increase with concomitant use of IFN-β. Cladribine may also
`result in hematological adverse events (AEs) when admin-
`istered with hematotoxic drugs (e.g. carbamazepine). As a
`substrate of BCRP, equilibrative nucleoside transporter 1
`(ENT1) and concentrative nucleoside transporter 3 (CNT3),
`coadministration with cladribine may interfere with cladrib-
`ine exposure. A decrease in cladribine exposure is possible
`when coadministered with potent inducers of BCRP (e.g.
`corticosteroids) or P-glycoprotein (e.g. rifampicin); however,
`
`acute short-term corticosteroid therapy can be concomitantly
`administered. Antivirals/antiretrovirals that require intracel-
`lular phosphorylation to become active (e.g. lamivudine,
`ribavirin) could potentially compete with cladribine over
`phosphorylation, affecting both cladribine and competing
`compound activity. The hydroxypropyl betadex component
`in cladribine may interact with active ingredients of other
`drugs to increase bioavailability, therefore a gap of at least
`3 h between administrations is recommended. Cladribine is
`
`

`

`1909
`
`Week 48
`
`LLN 0.09 x 109/L
`
`48
`
`83
`
`89
`
`116
`
`Weeks 1 & 5
`
`CLARITY cladribine tablets 3.5 mg/kg
`CLARITY EXT cladribine tablets 3.5 mg/kg*
`CLARITY placebo
`Cladribine administration
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`
`Median (Q1–Q3) NK
`
`counts (109/L)
`
`Development of Cladribine Tablets
`
`Fig. 9 Innate immune cell
`counts in patients with RMS
`receiving cladribine 3.5 mg/
`kg or placebo—natural killer
`(CD16 + /CD56 +) cells [67].
`LLN lower limit of normal,
`Q1–Q3 interquartile range, RMS
`relapsing multiple sclerosis.
`Visits with a sample size ≥ 30
`are displayed. *Patients who
`received placebo in CLARITY
`and cladribine tablets 3.5 mg/
`kg in CLARITY EXT. Adapted
`from Stuve et al. [67] under a
`Creative Commons CC-BY-NC
`license
`
`0.0
`
`0
`
`Number of patients
`77
`
`81
`
`122
`
`5
`
`84
`
`88
`
`110
`
`13
`
`83
`
`89
`
`116
`
`24
`Week
`
`81
`
`87
`
`124
`
`not a substrate of the cytochrome P450 (CYP) pathway and
`has no known inductive effect on CYP1A2, CYP2B6, and
`CYP3A4 enzymes [12, 26].
`It is not known whether cladribine can reduce the effec-
`tiveness of hormonal contraceptives; however, a clinical
`trial to examine this is ongoing (ClinicalTrials.gov identi-
`fier: NCT03745144). A barrier method is currently recom-
`mended during treatment with cladribine tablets and for at
`least 4 weeks after the last dose in each treatment course
`[26]. In humans, cladribine has a half-life of < 24 h and is
`rapidly eliminated [73]. However, in supratherapeutic doses,
`teratogenicity has been observed in mice and rabbits, and
`short-term effects have been observed in male mice germ
`cells [74]. Cladribine tablets should therefore not be admin-
`istered to pregnant women; pregnancy should be prevented
`using effective contraception during treatment and 6 months
`after the last dose in each treatment course in women and
`men of reproductive potential [26].
`As a small molecule, cladribine has been demonstrated
`to cross the blood–brain barrier (BBB) in animal [75] and
`human studies [76, 77]. In a study of parenteral cladribine
`in children with acute myeloid leukemia, cerebrospinal
`fluid (CSF) concentrations reached approximately 25%
`of those in plasma [76]. CNS penetration of cladribine
`may allow for a treatment effect on lymphocytes that have
`migrated into the CNS, possibly also affecting lymphoid
`follicles in the meninges of patients and reducing intrathe-
`cal immunoglobulin G (IgG) synthesis. In a study of 29
`patients with RRMS who had received parenteral cladrib-
`ine, 55% of patients tested negative for oligoclonal bands
`(OCBs) in CSF post-treatment, despite all patients testing
`
`positive for OCB and demonstrating raised Ig concentra-
`tions in CSF at baseline [78]. Furthermore, in a 10-year
`follow-up, Expanded Disability Status Scale (EDSS) pro-
`gression was significantly delayed in the OCB-negative
`patients despite both cohorts demonstrating equal charac-
`teristics at baseline.
`
`2 Clinical Studies in Relapsing Forms of 

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