throbber
The safety of etanercept for the treatment
`of plaque psoriasis
`
`R E V I E W S
`
`Kim A Papp
`University of Western Ontario, and
`K Papp Clinical Research, Waterloo,
`ON, Canada
`
`Correspondence: Kim A Papp
`University of Western Ontario, and K
`Papp Clinical Research, 135 Union Street
`East, Waterloo, ON, Canada N2J 1C4
`Tel +1 519 579 9535
`Fax +1 519 579 8312
`Email kapapp@probitymedical.com
`
`Abstract: Effective treatment with etanercept results from a congregation of immunological
`signaling and modulating roles played by tumor necrosis factor-alpha (TNF-alpha), a pervasive
`member of the TNF super-family of cytokines participating in numerous immunologic and
`metabolic functions. Macrophages, lymphocytes and other cells produce TNF as part of the
`deregulated immune response resulting in psoriasis or other chronic infl ammatory disorders.
`Tumor necrosis factor is also produced by macrophages and lymphocytes responding to foreign
`antigens as a primary response to potential infection. Interference with cytokine signaling by
`etanercept yields therapeutic response. At the same time, interference with cytokine signaling by
`etanercept exposes patients to potential adverse events. While the effi cacy of etanercept for the
`treatment of psoriasis is evident, the risks of treatment continue to be defi ned. Of the potential
`serious adverse events, response to infection is the best characterized in terms of physiology,
`incidence, and management. Rare but serious events: activation of latent tuberculosis, multiple
`sclerosis, lymphoma, and others, have been observed but have questionable or yet to be defi ned
`association with therapeutic uses of etanercept. The safe use of etanercept for the treatment of
`psoriasis requires an appreciation of potential adverse events as well as screening and monitoring
`strategies designed to manage patient risk
`Keywords: etanercept, psoriasis, demyelination, tumor necrosis factor, lymphoma, tuberculosis,
`infection, safety
`
`Characterizing the safety of a drug is rarely simple and never complete. Both short
`and long-term drug safety profi les require episodic, critical reviews of available
`information. Episodic reviews are necessary to survey case reports and put previous
`summaries into perspective. Critical evaluation is important to determine relevance,
`veracity, and adequacy of available information. Etanercept is no exception.
`The short-term safety of etanercept is well established by rigorous clinical trials in
`rheumatoid arthritis, psoriatic arthritis, and psoriasis (Leonardi et al 2003; Papp 2004;
`Keystone 2005; Kavanaugh et al 2006). Registries, now abundant in the rheumatology
`arena, are resources for assessing long-term risk and harm (Sokka 2004). Psoriasis
`registries should provide useful data over the next few years. Nonetheless, information
`extracted from registries must be put into context. The underlying disease may have
`epidemiologic characteristics different from the target disease. And, by their nature,
`registries are not as restrictive or as selective as controlled trials (Krishnan and Fries
`2004). There is a treatment bias: treatment tends to be given to a sicker population.
`There may be a confounding indication: not every enrollee fulfi lls appropriate
`diagnostic criteria. Biased patient selection, good or bad, may exaggerate effectiveness
`or safety. In addition, patients enrolled in clinical registries have few if any restriction on
`concurrent therapy thus confounding drug-drug interactions and attribution of effi cacy
`or adverse effects. On the other hand, registries are thought to be more refl ective of
`
`Therapeutics and Clinical Risk Management 2007:3(2) 245–258
`© 2007 Dove Medical Press Limited. All rights reserved
`
`245
`
`Biogen Exhibit 2119
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 1 of 14
`
`

`

`Papp
`
`real world experience. In addition, registries often provide
`larger number of patients, long periods of observation and
`data collection compared with registration trials.
`Case reports will identify unanticipated adverse events
`attributed to etanercept but are limited by the potential for
`inappropriate association of cause and effect. Etanercept is an
`effective therapy for psoriasis: effectiveness is advantageous
`to its adoption as a new therapy. This advantage is potentially
`offset by heightened scrutiny, off-label use, and sub-standard
`post marketing reports of adverse events.
`In this review, every effort is made to provide a balanced
`appraisal of risk. Pathophysiology and likelihood of
`association are considered as complements to incident reports
`when evaluating safety (Mulrow et al 1997; Ioannidis et al
`2006). Where assessment of risk is hampered by insuffi cient
`epidemiologic data, provisional estimates or cautionary
`comments are inserted.
`
`Background
`TNF-alpha, often referred to as tumor necrosis factor (TNF),
`is one member of the TNF superfamily of cytokines (Zhou
`et al 2002). TNF was initially described as a hemorrhagic
`necrosis factor produced by lipopolysaccharide-stimulated
`tumours. We now appreciated that TNF is a ubiquitous
`cytokine expressed by many cell types and having activity
`in innate and adaptive immune pathways (Zhou et al 2002).
`As a consequence of its important immunological function,
`TNF plays a central role in acute and chronic infl ammation
`(Liz-Grana and Carnota 2001; Pfeffer, 2003). The variety
`of immunological and metabolic processes affected by
`TNF-alpha, the functions of soluble and membrane bound
`TNF-alpha, and interactions between members of the TNF
`superfamily are impacted by the molecular activity of TNF-
`inhibitors such as etanercept. Both unanticipated risks and
`unanticipated benefi ts may arise through long-term high
`frequency exposure to a TNF-inhibitor. Given the unique
`molecular characteristics of each TNF-inhibitor, we expect
`common risk-benefi t profi les and differences.
`Etanercept is a dimeric fusion protein produced using
`recombinant genetic programming of Chinese hamster ovary
`cells. The protein has a molecular weight of 150 kDa and
`consists of two 75 kDa TNF-alpha receptors linked to the Fc
`portion of human immunoglobulin G1 (Dembic et al 1990;
`Mohler et al 1993).
`Clinical study reports are suffi ciently detailed to provide
`short term safety data, but none are powered to identify rare
`events. The National Data Bank for Rheumatic Diseases
`
`and the publicly available BIOBADASER are examples of
`registries that provide excellent longitudinal information
`on patients with rheumatologic diseases treated with TNF-
`antagonists.
`The structure of this review is as follows: Broad
`categories of adverse events are identifi ed. Within each
`category there may be specifi c, noteworthy concerns. Inciting
`observations and scientifi c rationale preface each general and
`specifi c category. Data relevant to the category are presented
`accompanied by brief commentary. Comparative data for
`TNF-antagonists as a group are avoided where possible to
`limit the scope of the review.
`
`Mechanisms of action
`Tumor necrosis factor-alpha engages in many aspect of
`immunological function. By its activity on TNF-alpha,
`etanercept will impact immunological and infl ammatory
`processes ranging from innate and extrinsic immunological
`response, cellular traffi cking, acute and chronic infl ammation,
`fever, and neuroendocrine regulation (Gruss and Dower
`1995). TNF interacts with glucocorticoids to regulate Toll-
`like receptor 2 gene expression (Hermoso et al 2004).
`The precise mechanisms of action of TNF-antagonists
`are not known. Certainly etanercept binds to free, soluble,
`or non-membrane-bound TNF-alpha but etanercept also
`has activity against the p55 receptor TNF-beta, also called
`lymphotoxin (Williams and Griffi ths 2002; Keystone and
`Dinarello 2005). Contrary to the effects on TNF-alpha, the
`activity of etanercept against p55 may stimulate immuno-
`reactivity (Han et al 2005) in addition to having effects on B-,
`T-, NK-cells and lymphoid architecture (Spahn et al 2005).
`Moreover, there is evidence that both etanercept and infl iximab
`induce apoptosis in macrophages, but not lymphocytes within
`rheumatoid arthritis (RA) joints (Catrina et al 2005). In general,
`the effects of etanercept are mediated by its binding of soluble
`TNF-alpha, but other TNF-antagonist activities are recognized.
`The implications of accessory TNF-antagonist activities are
`not known. Etanercept has peak absorption at 51 hours and a
`mean half-life of 68 hours (Korth-Bradley et al 2000).
`Whether or not TNF is an intrinsic pyrogen remains
`controversial (Stefferl et al 1996; Luheshi et al 1997;
`Dinarello 2005). In the mouse model, TNF does not appear
`to have pyrogen activity (Dinarello 2005). Nonetheless,
`resolving the question for humans is signifi cant as fever is a
`common, early, ubiquitous sign of infection and infections
`remain the most prominent safety concern during treatment
`with TNF-antagonists.
`
`246
`
`Therapeutics and Cl nical Risk Management 2007:3(2)
`
`Biogen Exhibit 2119
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 2 of 14
`
`

`

`A more complex role is refl ected in the effects TNF may have
`on neuroendocrine response. Pituitary and hypothalamic response
`are potentiated when TNF is present in high levels (Turnbull
`and Rivier 1999). The neuroendocrine effects of TNF may be
`refl ected in the psychological state of patients experiencing
`chronic infl ammatory disease (Tyring et al 2006).
`
`Adverse events
`Injection site reactions
`Mechanical processes such as poor injection technique,
`irritation, or immunologically mediated inflammatory
`processes associated with either drug or excipients cause
`injection site reactions. Foreign proteins may cause direct
`or indirect infl ammatory response (Shepherd 2003). It is not
`surprising that injection site reactions are by far the most
`common side effect associated with etanercept.
`Studies evaluating etanercept for the treatment of RA
`report a high incidence of injection site reactions with
`34%–37% of etanercept-treated patients compared with
`7%–10% of controls reporting reactions (Lebwohl 2002;
`Fleischmann and Yocum 2004). The high incidence in the
`RA population contrasts with a much lower incidence seen
`in psoriasis studies: 14%–20% (Leonardi et al 2003; Papp
`2004; Papp et al 2005). Why there are stark differences in
`the incidence of injection site reactions between RA and
`psoriasis populations is not known.
`Injection site reactions with 25 mg doses of etanercept
`are mild, well tolerated, self-limiting, and tend to occur early
`in the course of therapy (Zeltser et al 2001; Papp 2004).
`Irritation during and briefl y following injection is very
`common with the 50 mg single dose compared with the 25
`mg dosing formulation. Occasionally, persistent reactions
`of moderate severity are noted. Persistent reactions are
`characterized by erythematous, indurated, and urticarial like
`plaques (Edwards et al 2003). The histology of etanercept
`injection site reactions is consistent with a delayed-type
`hypersensitivity reaction (Werth and Levinson 2001; Zeltser
`et al 2001). Delayed and recall injection site reactions are
`infrequent but tend to be somewhat more severe than typical
`etanercept-associated injection site reactions (Zeltser et al
`2001; Rajakulendran and Deighton 2004). Signifi cant
`and severe injection site reactions are rare with etanercept
`regardless of dose (Papp 2004).
`
`Infection
`Clinical trials and post-marketing experience suggest that
`infection is the most common signifi cant category of adverse
`
`Safety of etanercept for the treatment of plaque psoriasis
`
`events experienced by patients treated with etanercept. Less
`common infections including tuberculosis and opportunistic
`infections, specifi cally histoplasmosis and listeriosis, are
`considered separately.
`TNF is involved in the immune response to bacterial
`and viral infections (Imanishi 2000; Herbein and O’Brien
`2000). More specifi cally, TNF plays an essential role in
`host response to intracellular pathogens (Choy and Panayi
`2001). The important role of TNF in immune response to
`intracellular organisms is further supported by TNF-defi cient
`animal models (Marino et al 1997). For Gram-positive and
`Gram-negative infections, clearance of organisms may
`be impeded by TNF-suppression (Takashima et al 1997;
`O’Brien et al 1999; Rijneveld et al 2003; Moore et al 2003,
`2005). Clinical studies, registries, and case reports confi rm
`that host response to infection is the most common signifi cant
`safety concern in patients treated with etanercept.
`Serious infection is defi ned within studies and for safety
`monitoring as one requiring intravenous antibiotics or
`hospitalization (Keystone 2004). The incidence of serious
`infections in patients treated with etanercept varies according
`to the population treated, severity of disease, concomitant
`medication, and adherence to a defi nition of serious infection.
`Some report serious infections as those requiring systemic
`therapy.
`During the clinical trial development of etanercept, the
`observed serious infection rate in the RA population was
`0.03–0.04 serious infectious events per patient-year (SIE/
`pt-yr), equal to rates seen in placebo controls (Cush 2004a
`2004b). Post-marketing surveillance across all indicated
`diseases has shown the rates of SIE with etanercept and
`infl iximab to be 0.007 SIE/pt-yr (confi dence interval [CI]
`0.03–0.09/pt-yr). Signifi cant under-reporting with post-
`marketing surveillance is expected but confounding effects
`include a less well defi ned treated population, inclusion of
`indications other than those reported in the clinical studies,
`and thus these rates may be substantially more common in
`the RA population (Cush 2004a, 2004b).
`Within the RA population, an increased risk of
`serious infection is seen in patients having extra-articular
`manifestations of RA, presence of comorbid diseases, and
`immunosuppressive therapy (Doran et al 2002a). These
`infections tend to be upper respiratory, skin, and urinary
`tract infections.
`Tumor necrosis factor-antagonist therapy may increase
`the risk of infection in the RA population. Reports of
`the number of infection-related adverse events per 100
`
`Therapeutics and Clinical Risk Management 2007:3(2)
`
`247
`
`Biogen Exhibit 2119
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 3 of 14
`
`

`

`Papp
`
`patient-years during an 18 month period show that etanercept
`had a rate of 22.6 (18.7–27.2) events per hundred years
`compared with controls (those receiving disease modifying
`and remitting drugs [DMARDs]) with a rate of 6.8 (5.0–9.4)
`per hundred patient years (Listing et al 2005). The rates
`for serious infections were 6.4 (4.5–9.1) and 2.3 (1.3–3.9)
`for etanercept and control groups respectively. Adjusting for
`case-patient mix, the rates of serious infection were similar
`for etanercept and infl iximab. These results suggest there
`is an increased risk of infection in those treated with TNF-
`antagonists. This study is limited by small numbers, a short
`observation period, and bias in populations: those on anti-
`TNF cannot be the same population as those on DMARDs
`since general patients with more severe disease are treated
`with TNF-antagonists, which introduces potential bias in the
`study populations.
`It is certain that TNF-antagonists exacerbate septicemia
`with increase in mortality among septic patients on etanercept
`(Fisher et al 1996; Baghai et al 2001).
`Patients developing new infections while on etanercept
`should be closely monitored and discontinued in those with
`serious infections or sepsis. Etanercept should not be initiated
`in patients with active infections including chronic or local-
`ized infections. Caution should be exercised when initiating
`etanercept in patients with a history of or predisposition to
`frequent recurrent infections.
`
`Mycobacterium tuberculosis
`Susceptibility to mycobacterium tuberculosis (TB) impacted
`by multiple factors including age, environment, immune
`status, and microbial virulence (Mitsos et al 2003) and
`genetic susceptibility (Abel and Casanova 2000; Casanova
`and Abel 2002). Latent tuberculosis remains a signifi cant
`global health concern with nearly 30% of the world
`population infected (Jasmer et al 2002). TNF is necessary
`for cell recruitment, granuloma formation, and clearance of
`mycobacterial infection (Roach et al 2002). Recent clinical
`results demonstrating reactivation of latent TB in patients
`receiving anti-TNF monoclonal antibody therapy solidifi es
`the importance of TNF in host response to TB (Keane et al
`2001; Keane 2004, 2005).
`The role of TNF in initial host response and subsequent
`confi nement of TB organisms is complex and not completely
`elucidated. TNF regulates chemokine induction that in turn
`orchestrates cell recruitment, granuloma formation, and
`clearance of mycobacterial infection (Roach et al 2002;
`Stenger 2005). Mice lacking TNF mount delayed chemokine
`
`response and cellular infi ltrate. Subsequent high chemokine
`production produced disorganized T-cell and macrophage
`responses capable of producing high levels of interferon-
`gamma but unable to protect against fatal TB infections 28
`days post inoculation. Wild mouse strains survived 16 weeks
`or longer. The response of TNF-defi cient mice exposed to
`mycobacteria anticipates the prominent role TNF plays in the
`initial response to infection and subsequent maintenance of
`granulomas. In large part, mortality results from unchecked
`type-1 infl ammatory response producing tissue necrosis
`(Zganiacz et al 2004). Animal models also suggest there
`are differences in the activity of membrane-bound and -free
`TNF in acute and chronic response to TB (Olleros et al 2005;
`Saunders et al 2005). TNF is also required to maintain latency
`of TB (Botha and Ryffel 2003). Thus, the role of TNF in
`immune response to TB is poly-modal: initiate infl ammatory
`response, regulate and suppress the infl ammatory response,
`and maintain chronic immunological response.
`RA patients on etanercept showed a linear incidence
`of TB infection (Wallis et al 2004a, 2004b) suggesting
`that acquisition of TB was related to exposure and not
`re-activation of latent infection. Infl iximab-treated patients
`demonstrated an accelerated incidence of TB infection in
`keeping with activation of latent disease (Wallis et al 2004a,
`2004b). While the incidence of TB in patients on etanercept
`may not be signifi cantly greater than the background rate,
`treatment with etanercept does alter the clinical presentation
`of TB (Arend et al 2003; Gardam et al 2003). Approximately
`half of patients treated with etanercept who develop clinical
`TB present with extra-pulmonary manifestations including
`disseminated TB. The expected rate of extra-pulmonary
`TB in immunocompetent hosts is less than 15% (Dye et al
`1999).
`In summary, there is no scientifi c data in support of
`screening for latent TB prior to initiating therapy with
`etanercept. However, patients treated with etanercept who
`acquire infection with TB are more likely to have atypical
`presenting signs and symptoms. Etanercept-treated patients
`developing TB may be at increased risk of severe and
`potentially fatal infection. Screening for active TB with a
`chest X-ray is medically prudent. A more cautious approach
`is to screen for latent and active TB by chest X-ray (CXR)
`and tuberculin (PPD) testing prior to initiating etanercept
`in patients at high risk. Based upon minimal data, but
`highlighting the need for extreme safety, some suggest a
`CXR and PPD prior to introducing any immunosuppressive
`treatment (CDC 2004; Keane 2005).
`
`248
`
`Therapeutics and Cl nical Risk Management 2007:3(2)
`
`Biogen Exhibit 2119
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 4 of 14
`
`

`

`Opportunistic infections
`Opportunistic infections occur in patients on TNF-blocking
`agents (Jarvis and Faulds 1999; Garrison and McDonnell
`1999; Mease et al 2000; Doran et al 2002a, 2002b; Netea
`et al 2003; Elkayam et al 2004), but these are rare (Keystone
`2004).
`Histoplasmosis is an uncommon opportunistic infection
`endemic to many regions of the world (Cano and Hajjeh
`2001). Normal host defense to infection is dependent upon
`TNF expression (Smith et al 1990; Zhou et al 1998). Infection
`with histoplasmosis may be exacerbated in patients on therapy
`with TNF-antagonists, but lack of control comparators and
`cases occurring in histoplasmosis-endemic regions of the
`US make any conclusions tentative (Lee et al 2002). Less
`certain is the question of risk of reactivation of latent infection
`with histoplasmosis. This uncertainty is highlighted by
`reports of disseminated histoplasmosis occurring in patients
`on low-dose methotrexate (Berry 1969; Witty et al 1992;
`LeMense and Sahn 1994; Voloshin et al 1995; Roy and
`Hammerschmidt 2000; Arunkumar et al 2004).
`Listeria monocytogenes is an uncommon but ubiquitous,
`opportunistic, intracellular pathogen causing gastroenteritis,
`meningitis, encephalitis, and septicemia (Hamon et al
`2006). TNF is essential to effect normal host response to
`listeria (Havell, 1989, Rothe et al 1993; Kanaly et al 1999;
`Dinarello 2003; Torres et al 2005) and treatment with
`etanercept may predispose patients to infection (Schett et al
`2005). Infection with listeria is reported in patients treated
`with TNF-antagonists and particularly etanercept (Slifman
`et al 2003; Ehlers 2005; La Montagna and Valentini 2005;
`Nadarajah and Pritchard 2005; Rachapalli and O’Daunt 2005;
`Schett et al 2005). What is not evident is whether there is a
`real increased risk of infection or a modifi cation of clinical
`presentation and host response (Pagliano et al 2004). Given
`that etanercept does affect lymphotoxin (Williams and
`Griffi ths 2002) and that lymphotoxin is essential in providing
`normal immune response to listeria (Ehlers et al 2003), the
`possible increased susceptibility to infection with listeria
`must be considered.
`Rare cases of disseminated sporotrichosis further stress
`the importance of TNF in maintaining normal host response
`to infections (Gottlieb et al 2003).
`
`Vaccination
`TNF plays a signifi cant role in immune response to pathogens
`(Herbein and O’Brien 2000) and may therefore modulate
`host response to vaccination. A number of studies have
`
`Safety of etanercept for the treatment of plaque psoriasis
`
`evaluated response to infl uenza vaccine in RA patients treated
`with etanercept. In general, response to infl uenza vaccine is
`blunted but not completely suppressed (Fomin et al 2006).
`The addition of methotrexate further suppresses the response
`to vaccination (Kapetanovic et al 2006).
`
`Malignancy exclusive of lymphoma
`The role of TNF in carcinogenicity and tumor surveillance
`has not been established. Early cell culture studies indicated
`that TNF is cytotoxic for certain tumor cell lines (Old 1985;
`Creasey et al 1986; Palladino, Patton, et al 1987; Palladino,
`Srivastava, et al 1987). Subsequent studies revealed that, for
`certain types of malignancies, TNF may act as a growth factor
`(Freedman et al 1992; Warzocha et al 1995; Filella et al 1996;
`Warzocha and Salles 1998; Warzocha, Bienvenu, et al 1998;
`Warzocha, Ribero, et al 1998; Renard et al 1999; Moore
`et al 1999) and may even enhance the metastatic potential of
`certain tumors (Balkwill et al 1990; Malik et al 1990).
`Review of the clinical studies and registries for TNF-
`antagonists shows no increase in the incidence of solid tumors
`in the RA population (Keystone 2003, 2005). Likewise, in
`the clinical studies evaluating etanercept for the treatment of
`psoriasis, there is no evidence of increased risk of malignancy
`(Leonardi et al 2003; Papp 2004; Papp et al 2005). The
`potential for increased risk of solid tumors in patients
`receiving concurrent etanercept and alkylating agents must
`be considered (Mukhtyar and Luqmani 2005; WGET 2005;
`Hellmich et al 2006; Stone et al 2006). There are cases of
`rapidly developing squamous cell carcinomas in RA patients
`initiating therapy with etanercept (Smith and Skelton 2001).
`To the contrary, TNF-alpha defi cient mice are resistant to
`cutaneous carcinogenesis (Arnott et al 2002).
`
`Lymphoma
`There is a strong association of non-Hodgkin’s lymphoma with
`Epstein-Barr virus (EBV) infection and immunosuppression
`(Liebowitz 1998; Meyer et al 2004; Poppema 2005).
`Approximately 95% of adults are infected with EBV. Many
`develop subclinical reactivation (Rickinson and Kieff 1996).
`Members of the TNF superfamily of receptors play a role
`in pathogenesis of EBV-positive lymphomas arising in
`immunosupressed patients, but the role of TNF-alpha is not
`established (Liebowitz 1998; Herbein and O’Brien 2000).
`Chronic infl ammation produces elevated TNF levels, which
`in turn produce indirect alterations in immunological function
`(Khan 2006; Weyand et al 2006) and modulatory effects
`of TNF on T-cell surveillance (Baran-Marszak et al 2006).
`
`Therapeutics and Clinical Risk Management 2007:3(2)
`
`249
`
`Biogen Exhibit 2119
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 5 of 14
`
`

`

`Papp
`
`Confounding the role of TNF in the develoment of lymphoma
`is the association of chronic infl ammatory processes and
`lymphoma (Kato et al 2003; Chang et al 2005). The potential
`association of TNF, TNF-antagonism, and the development
`of lymphomas is confounded by epidemiologic surveys
`showing a strong trend in risk in patients with psoriasis
`(Hannuksela-Svahn et al 2000; Gelfand et al 2003, 2006)
`though these fi ndings are not substantiated by larger surveys
`(Smedby et al 2006). The association of lymphoma and
`rheumatoid arthritis appear more certain (Baecklund et al
`1998, 2003, 2004, 2006; Ekstrom et al 2003).
`There are case reports of lymphoma developing in
`patients receiving TNF-antagonists (Brown et al 2002). Of
`the 26 cases reported, 18 developed in patients on etanercept.
`The mean time to onset of lesions after commencing therapy
`was 8 weeks. Data from an RA registry suggests an increased
`risk of lymphoma in patients treated with TNF-antagonists
`or methotrexate compared with those who are not (Wolfe
`and Michaud 2004b). There is; however, a strong selection
`bias in that those receiving anti-TNF therapy tend to be the
`most severely affected patients, a cohort already known to
`have a greater risk of lymphoma (Baecklund et al 1998)
`and that the strongest association is not with therapy but the
`underlying RA itself (Baecklund et al 2006). In addition, two
`cases of cutaneous and systemic T-cell lymphoma progressed
`rapidly after initiating TNF-blockade: one with etanercept
`and one with infl iximab (Adams et al 2004). Both cases
`were described as rapid in onset with fulminate courses:
`extensive cutaneous, and systemic involvement resulting in
`death within months of diagnosis.
`While there are numerous case reports of lymphoma
`developing in patients treated with etanercept, the relative
`risk of lymphoma remains constant for RA patients regardless
`of therapy with TNF-antagonists (Keystone 2005). The high
`incidence of lymphoma in RA patients makes risk assessment
`complex. The lower incidence of lymphoma in the psoriasis
`population may be instrumental in the assessment of risk
`associated with long-term treatment with etanercept.
`Central nervous system
`demyelination events
`We have some understanding of the incidence of multiple
`sclerosis (MS) in the general population: women more com-
`monly affected, there are regional variations in incidence
`and prevalence (Ebers and Sadovnick, 1993, Magnano et al
`2004). Incidence in the general population is approximately 6
`per 100 000 per year with a prevalence of nearly 85/100 000.
`
`An increased in risk of MS is reported in individuals with
`affected fi rst degree relatives (Sadovnick et al 1993).
`MS is uncommon. Furthermore, assessments may be
`complicated as not all instances of magnetic resonance
`imaging (MRI) fi ndings consistent with demyelination are
`MS (Koller et al 2005a). Instances of chronic, infl ammatory,
`demyelinating polyneuropathy are usually peripheral,
`but may include cortical and optic nerve demyelination.
`Interestingly, MS appears to be a Th1 disorder mediated by
`cytokines including TNF and evinces many of the pathogenic
`pathways active in psoriasis (Koller et al 2005b).
`The putative relationships between TNF, TNF-antagonism,
`and MS are not obvious. Many theoretical reasons support
`TNF-antagonist activity induces demyelization (Magnano
`et al 2004). Equally supportive arguments support the
`contrary: TNF-antagonism does not increase the risk of MS
`and may potentially be of therapeutic value (Magnano et al
`2004). Reporting bias of case reports (Magnano et al 2004)
`and potential association with other autoimmune diseases
`(Midgard et al 1996) confound the role of TNF-antagonist
`therapy in the onset or exacerbation of MS.
`Much of the current concern over TNF-antagonism
`and MS results from a single study evaluating lenercept; a
`p55, recombinant, soluble TNFR1 receptor protein, for the
`treatment of relapsing-remitting and secondarily progressive
`MS (LMS-UBC 1999). The study found no increase in new
`or active lesions as demonstrated on MRI. However, there
`was a signifi cant dose-related increase of attack frequency
`though not attack severity nor attack duration. The contrary
`response of MS patients treated with lenercept underscores
`the diffi culty in extrapolating results from animal models.
`Antibodies to lenercept did not affect clinical response but
`did increase rate of drug clearance (Wiendl and Hohlfeld
`2002). Two cases of MS patients treated with infl iximab
`demonstrated increased MRI activity but no clinical
`worsening (van Oosten et al 1996).
`The rarity of MS in psoriasis patients treated with
`etanercept is highlighted by the rarity of case reports (Sukal
`et al 2006). From the rheumatology literature, there are
`cases temporally related to TNF-suppression, some of which
`resolved upon withdrawal of treatment (Mohan et al 2001).
`MS remains rare and of uncertain causal association with
`anti-TNF therapy (Mohan et al 2001; Magnano et al 2004).
`Many but not all patients develop recurrent symptoms on
`re-challenge (Mohan et al 2001; Cisternas et al 2002).
`There is growing concern among some groups of
`neurologists who suggest pretreatment MRI scans for all
`
`250
`
`Therapeutics and Cl nical Risk Management 2007:3(2)
`
`Biogen Exhibit 2119
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 6 of 14
`
`

`

`patients about to receive a TNF-antagonists (Bellesi et al
`2006). Currently, etanercept should be avoided in patients
`with a personal history of any central nervous system
`demyelinating disorder and used with caution in patients
`with a family history of these disorders. Pretreatment MRI
`may be considered in patients with equivocal histories of
`neuropathy or signs or symptoms of demyelination and fi rst
`degree family histories of MS.
`
`Hematological events
`Rare cases of pancytopenia and aplastic anemia are reported
`in association with etanercept (Lebwohl 2002). A fatal
`outcome is reported in some cases (Jarvis and Faulds 1999;
`Khanna et al 2004). Nonetheless, it is important to review
`drug and medical history to exclude other potential causes
`of myelosuppression (Baumelou et al 1993; Marshall et al
`2006).
`
`Hepatitis
`TNF plays a role in host response to hepatitis B and C (Herbein
`and O’Brien 2000). With 1.8% of the world population
`infected with hepatitis C virus and 5% infected with hepatitis
`B, the potential for exacerbation of viral hepatitis associated
`with TNF-antagonist therapy is noteworthy. While TNF does
`play a role in viral hepatic infection, the importance of TNF
`in maintaining suppression of viral replication is not clear
`(Calabrese et al 2004).
`Mixed reports on response of patients with hepatitis B
`treated with anti-TNF agents correlates with the natural,
`variable history of the infection (Khanna et al 2003;
`Calabrese et al 2004; Khanna et al 2004; Lok and McMahon
`2004). Nonetheless, TNF plays a primary role in sustaining a
`normal response to infection with hepatitis B (Schlaak et al
`1999; Michel et al 2003; Ostuni et al 2003). Recently, Health
`Canada issued an advisory related to possible reactivation
`of hepatitis B in patients receiving anti-TNF therapy. The
`advisory is based upon a single case report (HPB Canada
`2006).
`The mechanisms relating to reactivation of hepatitis B
`are uncertain. Equally uncertain is the association between
`treatment with TNF-antagonists or other immunosuppressant
`and hepatitis B reactivation. More likely is reaction upon drug
`discontinuation or rebound with replication of hepatitis B
`in hepatocytes upon discontinuation of immunosuppressive
`therapy (Herbein and

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