biologics monograph

Upload: sundar-ramanathan

Post on 10-Apr-2018

249 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/8/2019 Biologics Monograph

    1/14

    Biologic Therapies:Clinical Implications for Rheumatologists,Gastroenterologists, Allied Health Practitioners

    R.J. Fasenmyer Center for Clinical Immunology

  • 8/8/2019 Biologics Monograph

    2/14

    Spondyloarthritis: TherapeuticImplications o Advances in PathogenesisThree prevalent forms of inammatory arthritis include rheu-

    matoid arthritis (RA), psoriatic arthritis (PsA), and ankylosingspondylitis (AS). Although they share certain characteristics,

    they are distinguished by the specic sites of joint inammation

    associated with each: the synovium in RA; the bone, enthesis,

    and synovium in PsA; and the bone and enthesis in AS. The

    location of inammation is an important factor to consider

    when making treatment decisions. For example, an agent that is

    effective for peripheral synovitis may not have the same level of

    therapeutic response in axial disease or enthesitis.

    Osteoclast precursors as a biomarker

    Osteoclast precursors are monocyte effector cells in the circulation.

    An abundance of osteoclast precursors is observed in patients with

    certain phenotypes of erosive immune-mediated inammatory

    disorders, namely RA, PsA, and psoriasis (see Figure 1). Of note,

    osteoclast precursors are strikingly high in a subset of patients

    with psoriasis without musculoskeletal signs and symptoms who

    later develop inammatory arthritis, which suggests that osteo-

    clast precursors may be useful as a biomarker for susceptibility

    to arthritis in this population.

    Fig 1. Osteoclast precursors in erosiveIMID phenotypes

    Control AS Crohns RA PsA Ps

    OCPpermillionmonocy

    tes

    100 --

    80 --

    60 --

    40 --

    20 --

    0 ---- -- -- -- -- -- --

    IMID = immune-mediated inflammatory disorders

    Therapeutic opportunityAn opportunity to arrest the development of a severely

    inammatory phenotype of arthritis lies in the identication of

    patients with psoriasis and subclinical joint inammation and

    patients with early inammatory disease. This will enable clini-

    cians to intervene with disease-modifying antirheumatic drugs

    and/or biologic agents at an early stage.

    T-cell pathways in spondyloarthropathies

    Activation of precursor T helper cells in response to signal-

    ing through transforming growth factor-beta-1 and interleu-

    kin (IL)-6 is considered a promoter of the Th17 phenotype, an

    especially inammatory phenotype. The activator of the Th17

    response is the dendritic cell, a master cell that releases IL-23

    which drives and perpetuates the Th17 phenotype. Th17 cells

    are a subset of CD4+ T cells that produce IL-17. The Th17

    cells release a number of cytokines that are integral to thehyperproliferation of keratinocytes that is observed in psoriasis.

    In addition, Th17 cells encourage osteoclastogenesis by three

    mechanisms:

    1) The release of IL-17 by Th17 cells induces the expression o

    receptor activator NF-B ligand (RANKL) on synovial

    broblasts,

    2) Th17 cells express RANKL on the cell surface which binds

    to RANK on OCPS and promotes the osteoclastogenesis, and

    3) Th17 produces tumor necrosis factor (TNF), a cytokine that

    potentiates the formation of osteoclasts.1,2

    Other evidence implicating the IL-17 subset in an inammatoryphenotype is the observed association between psoriasis, PsA

    and AS with IL-23 alleles; the presence of increased levels of IL-

    17 in the blood and synovial uid of patients with PsA, and the

    effectiveness of anti-p40 antibodies in the treatment of psoriasis

    and PsA.

    Therapeutic targets

    Th17 represents a therapeutic target for joint destruction asso

    ciated with T cell activation. Other logical therapeutic targets

    include IL-23 (a perpetuator of the Th17 phenotype), IL-17, IL

    21, dendritic cells that express IL-23, and dendritic cell specic

    transmembrane protein (DC-STAMP).Anti-p40 antibody was associated with an improvement in the

    American College of Rheumatology (ACR) 20 response com

    pared with placebo in patients with PsA, but the ACR20, ACR50

    and ACR70 response achieved was somewhat inferior to that of

    other biologic agents tested in randomized, placebo-controlled

    trials, and typically lower than that achieved in the treatment of

    plaque psoriasis.3

    Dendritic cell specic transmembrane protein is a 7-transmem

    brane protein that is present on monocytes. Its role in osteoclas

    togenesis is as a regulator of cell to cell fusion in the formation

    of a polykaryon. When a monocyte forms an osteoclast, it must

    form a multinucleated cell, called a polykaryon. DC-STAMP iscritical to polykaryon formation.

    DC-STAMP may therefore be a therapeutic target; DC-STAMP

    antibodies added to culture have been shown to prevent polykaryon

    formation. DC-STAMP is but one signaling pathway required

    for osteoclast formation and activation. Osteoclast activation can

    also be blocked at RANK or in accessory pathways downstream

    of RANK, such as Syk. Molecules that bind to Syk have shown

    impressive efcacy in the treatment of RA.

    Osteoblasts and osteoclasts offer different therapeutic targets

    The Wnt signaling pathway is a critical pathway responsible for

    osteoblast formation and development of new bone. The Dick

    Rationale or the use o biologic therapies in rheumatologyChristopher Ritchlin, MD, MPH, Elaine Husni, MD, MPH, Carol A. Langord, MD, MHS

  • 8/8/2019 Biologics Monograph

    3/14

    kopf (DKK) family of proteins acts as inhibitors of the Wnt path-

    way and may interfere with new bone formation by interrupting

    osteoblast differentiation. DKK-1 is highly elevated in patients

    with RA, which may explain the relative absence of bone repara-

    tive response associated with RA.

    The effect of TNF inhibition on new bone formation has been a

    concern, but two phase 3 clinical trials with etanercept and

    adalimumab revealed no decline in radiographic progression

    (when compared to progression in a historical cohort).4-6

    New models of AS consider that osteoproliferation may be

    uncoupled from inammation, such that new bone formation

    may be inuenced by pathways that are separate from those that

    regulate bone resorption. The possibility that TNF may actually

    inhibit new bone formation has also been suggested, and in this

    scenario, TNF blockade may actually promote the development

    of a pathologic bone formation phenotype.

    Biologics and Systemic VasculitisThe investigation of biologic agents to treat vasculitis is ongo-

    ing. The use of biologic agents must be evaluated in the context

    of standard therapies, as no biologic agent has been proven to beeffective in this patient population.

    Theoretical advantages and objectives with the use of biologic

    agents in vasculitis are the reduction in the toxicity of therapy,

    the reduction in the incidence of disease relapse, and the induc-

    tion of disease remission permitted by specic immunologic tar-

    geting. Of concern, however, is that such specic targeting may

    be insufcient to modulate the disease and reduce the potential

    for disease-specic toxicities.

    In considering the use of a biologic agent for a patient with sys-

    temic vasculitis, several issues must be contemplated, including

    the goals of treatment, the effectiveness of current standard thera-

    py, and clinical data on biologic agents drawn from the particular

    form of vasculitis being treated. For this discussion, these issues

    will be examined in the settings of Wegeners granulomatosis

    and giant cell arteritis.

    Wegeners granulomatosis

    Wegeners granulomatosis is a potentially life-threatening dis-

    ease, and protection of patient survival is the first priority.

    Because of the effectiveness of current treatment options, the

    goals may be broadened to include the following:

    1) Induction of remission, dened as the absence of disease

    activity;

    2) Avoidance of disease relapse, dened as the return of

    disease activity after achieving remission; and

    3) Minimization of therapeutic toxicity.

    The current treatment approach to Wegeners granulomatosis is

    based on two phases: an induction phase in which active disease

    is placed into remission and a maintenance of remission phase

    (see Figure 2). Wegeners granulomatosis is a disease that has a

    wide spectrum of severity ranging from localized upper airway

    disease to acute multisystem disease involvement. The choice

    of therapy has traditionally been based on the degree of disease

    severity, with cyclophosphamide induction followed by less toxic

    maintenance therapy (eg, methotrexate, azathioprine, mycophe

    nolate mofetil) for severe disease and methotrexate for induction

    and maintenance of less severe active disease.

    These regimens have been proven to prolong survival and to

    induce remission in more than 85% of patients, but relapse

    occurs in 50% to 75% of patients and therapy is potentially toxic

    As a result, other treatment options, particularly biologic agents

    have been investigated.

    Fig 2. Wegeners granulomatosis: Summary ocurrent treatment

    Induction

    Cyclophosphamide

    Methotrexate

    Maintenance

    Methotrexate

    Azathioprine

    (Mycophenolate mofetil)

    Methotrexate

    prolong survivalremission in >85% of patients

    high frequence of relapsepotential toxicity

    }}

    Severe Disease

    Non-severe Disease

    Interfering with the pathway leading to granulomatous inam

    mation may represent a therapeutic strategy in Wegeners granu

    lomatosis. Information gleaned from other human diseases and

    murine models indicates that CD4+ T cells with the Th1 cytokine

    pattern play an important role in the initiation and maintenance

    of granuloma formation. The primacy of this cytokine pattern

    in Wegeners granulomatosis has been supported by evidencegained from the laboratory.

    Based on these data, therapies that interrupt TNF production gen

    erated initial excitement in Wegeners granulomatosis. However

    a clinical trial of 180 patients randomized to etanercept or placebo

    failed to support the use of this agent for the induction or mainte

    nance of remission in patients with Wegeners granulomatosis.

    There were six malignancies documented in patients receiving

    etanercept (none with placebo); all malignancies occurred in

    patients receiving concurrent cyclophosphamide.8 Although this

    occurred in a small population, it raises caution against the use of

    cyclophosphamide concurrently with any anti-TNF agent.

    An open-label trial of iniximab in 32 patients with Wegenersgranulomatosis or microscopic polyangiitis achieved an 88%

    remission rate, but all patients were also receiving standard ther

    apy concomitantly.9 The rate of relapse was 18%, the mortality

    rate was 6%, and 21% of patients experienced serious infec

    tions. In the absence of a sufciently powered trial, the efcacy

    of other anti-TNF agents cannot be determined. Current evi

    dence, however, does not support the use of any anti-TNF agen

    in Wegeners granulomatosis.

    Another avenue for intervention in Wegeners granulomatosis

    is interference with T-cell activation. Abatacept is an agent that

    acts on this pathway, although there is currently no experience

    Rationale or the use o biologic therapies in rheumatology

  • 8/8/2019 Biologics Monograph

    4/14

    with it in the treatment of Wegeners granulomatosis. A clinical

    trial of abatacept in the treatment of mild, relapsing Wegeners

    granulomatosis by the Vasculitis Clinical Research Consortium

    is ongoing.

    B-celldepleting therapies were rst explored based on the

    rationale of interfering with the production of antineutrophil

    cytoplasmic antibodies (ANCA). The role of ANCA in disease

    pathogenesis remains unclear, although bodies of evidence from

    the laboratory have supported a means through which ANCA-induced neutrophil-mediated vascular injury might occur. Two

    open-label studies of rituximab combined with glucocorticoids

    in patients with refractory ANCA-associated vasculitis resulted

    in undetectable levels of B cells and induction of remission in

    all patients.10,11 All patients remained in remission as long as B

    lymphocytes were depleted; 10% to 20% relapsed in association

    with a return of B cells and ANCA. Although ANCA produc-

    tion declined in all patients, it remained positive in up to 40% of

    patients, which raises the possibility that rituximab may exert its

    action through other mechanisms.

    In addition to its potential efcacy, important questions that

    remain with B-celldepleting therapy are whether its effectsdiffer between organs and whether any disease impact occurs

    through ANCA or other B-celldependent mechanisms. These

    questions are being explored through the ongoing Rituximab in

    ANCA-associated Vasculitis (RAVE) trial.

    Giant cell arteritis

    Giant cell arteritis is the most common form of vasculitis, and it

    carries many phenotypes. It is most commonly associated with

    cranial disease (temporal arteritis) and a risk of loss of vision, but

    it also includes polymyalgia rheumatica and potential involve-

    ment of large vessels of the aorta and its branch vessels.

    Treatment goals for giant cell arteritis are to prevent seriousmorbidity or mortality, reduce symptoms that affect quality of

    life, lessen disease relapse, and avoid therapeutic toxicity. Pred-

    nisone and aspirin remain the standard treatments (see Figure 3).

    Relapse, however, occurs in 75% to 90% of treated patients, and

    toxicity is common.

    The introduction of biologic therapies offered great potential,

    particularly in the inhibition of TNF. However, Hoffman et al12

    found no difference between placebo and iniximab on relapse

    through week 22 in patients with newly diagnosed giant cell

    arteritis, all of whom also received standard doses of pred-

    nisone. More patients randomized to iniximab had infec-

    tion than placebo recipients. This nding was replicated in aplacebo-controlled study of iniximab in patients with newly

    diagnosed polymyalgia rheumatica, all of whom were also

    treated with prednisone.13

    Intervention with T-cell activation, as with abatacept, is a

    potential therapeutic target in giant cell arteritis. The T cell is one

    cell that appears to play a role in the pathogenesis of giant cell

    arteritis. Activated T cells in the arterial wall secrete cytokines

    that initiate and maintain granulomatous inammation. Granulo-

    mas in the vessel wall, formed by interferon-gamma-producing

    CD4+ T cells and macrophages, are a characteristic feature of

    giant cell arteritis.

    Figure 3. Giant cell arteritis treatment

    1950

    Prednisone+

    Aspirin

    2009

    Prednisone+

    Aspirin

    Improves symptoms

    Reduces risks of blindness

    Relapse 75-90%

    Toxicity 35-86%

    PsA: QOL Issues and the Role o BiologicsUp to 30% of patients with psoriasis can have inammatory joint

    disease or arthritis.

    Features that distinguish PsA from RA are seronegativity and

    associated dactylitis and enthesitis. The arthritis in a patient with

    PsA can range from mild nondestructive disease to a severely

    rapid and destructive arthropathy. In addition to joint involve

    ment, the tendons, ligaments, and bone are affected in PsA

    (see Table 1).

    Table 1. Psoriatic arthritis conditions17-21

    Musculoskeletal

    Arthritis

    Dactylitis/sausage digits

    Enthesitis

    Tenosynovitis

    Sacroliitis/spondylitis

    Cutaneous and extraarticular maniestations

    Psoriasis

    Nail pitting

    Onycholysis

    Conjunctivitis/iritis/uveitis

    Radiographic characteristics

    Erosions

    Pencil-in-cup deformity

    Ray distribution

    Osteolysis

    Nonmarginal asymmetric syndesmophytes

    Asymmetric sacroiliitis

    Novel applications are being used in various disease states

    including PsA, to assess the impact of disease on QOL and to mea

    sure patient preferences associated with health-related QOL.

    Measuring QOL

    In public health and in medicine, the concept of health-related

    QOL refers to a person or groups perceived physical and men

    tal health over time. Tracking health-related QOL in differen

    populations can identify subgroups with poor physical or menta

    health and can help guide interventions to improve their health

    Quality of life has not been a standard outcome measure in the

    recent past in studies of biologic therapies. However, as the bar is

    being raised regarding how much treatment effect can be gained

  • 8/8/2019 Biologics Monograph

    5/14

    with biologic agents, an effort is being made to quantify more

    outcomes measures for patients suffering from PsA.

    Health-related QOL is a measure of the effects of chronic ill-

    ness with the goal of obtaining a better understanding of how an

    illness interferes with a persons day-to-day life. Patients with

    arthritis have signicantly worse QOL than those without

    arthritis; they report twice the number of unhealthy days and

    three times as many days with limitations in activity monthly.14

    Scores on health-related QOL and life satisfaction question-naires are similar between patients with RA and PsA despite

    greater peripheral joint damage, inammation, and physical

    disability associated with RA.15 This suggests that the QOL

    impact of skin disease in patients with PsA is more profound

    than previously believed.

    The Patient Reported Outcomes Measurement Information Sys-

    tem (PROMIS) is a cooperative network of seven institutions and

    the National Institutions of Health that was designed to quantify

    patient-reported symptoms (eg, pain, fatigue, aspects of health-

    related QOL) in clinical practice across several disease states.

    Perceived changes in such symptoms is often the best way that

    patients can judge the effectiveness of treatments.

    Instruments that measure health-related QOL must be valid,

    reliable, and responsive to change. Patient-recorded QOL

    outcomes scales can be general or disease-specic. Examples

    of general health measures are the SF-36, the Euro-QOL,

    the Nottingham Health Prole, and the Health Assessment

    Questionnaire (HAQ), which is a measure of disease activity

    and long-term disability. Instruments specic to PsA include

    the PsA QOL, the expanded HAQ (HAQ-SK), and the Psoriatic

    Arthritis Response Criteria (PsARC).

    Willingness to pay as a measure o impact

    A novel technique to further extrapolate on the QOL issues isthe concept of willingness to pay. This is a method to measure

    the value of stated preferences in terms of dollars, with a higher

    willingness to pay associated with stronger preference. It is being

    used increasingly in health care to gauge the impact of a chronic

    disease on health-related QOL. Willingness to pay is able to have

    the patient take all the possible factors (education, severity of ill-

    ness, income, beliefs) that are important to them and balance this

    in their ultimate decision for a health benet or risk.

    A recent study was done to validate this in a cohort of psori -

    atic arthritis patients. The impact of psoriasis on health-related

    QOL was measured in 59 patients with PsA using eight QOL

    domains: intimacy, physical comfort, social comfort, self-care,ability to work, ability to concentrate, ability to sleep, and emo -

    tional health.16 Nearly 90% of patients were willing to pay to

    fully restore physical function. Also, the emotional, sleep, and

    work domains were considered important to cure by a majority.

    References

    1. Sato K, Suematsu A, Okamoto K, et al. Th17 functions as an osteoclastogenic

    helper T cell subset that links T cell activation and bone destruction.J Exp Med

    2006;203:2673-82.

    2. Jandus C, Bioley G, Rivals JP, et al. Increased numbers of circulating poly-

    functional Th17 memory cells in patients with seronegative spondylarthritides.

    Arthritis Rheum 2008;58:2307-17.

    3. Gottlieb A, Menter A, Mendelsohn A, et al. Ustekinumab, a human interleukin

    12/23 monoclonal antibody, for psoriatic arthritis: a randomised, double-blind,

    placebo-controlled, crossover trial.Lancet2009;373:633-40.

    4. van der Heijde D, Pangan AL, Schiff MH, et al. Adalimumab effectively

    reduces the signs and symptoms of active ankylosing spondylitis in patients with

    total spinal ankylosis.Ann Rheum Dis 2008;67:1218-21.

    5. Kavanaugh A, Klareskog L, van der Heijde D et al. Improvements in clinical

    response between 12 and 24 weeks in patients with rheumatoid arthritis on etaner-

    cept therapy with or without methotrexate.Ann Rheum Dis 2008;67:1444-7.

    6. Mease, PJ, Gladman DD, Ritchline CT, et al, for the Adalimumab Effective-

    ness in Psoriatic Arthritis Trial Study Group. Adalimumab for the treatment ofpatients with moderately to severely active psoriatic arthritis.Arthritis Rheum

    2005;52:3279-89.

    7. Wegeners Granulomatosis Etanercept Trial (WGET) Research Group.

    Etanercept plus standard therapy for Wegeners granulomatosis.N Engl J Med

    2005;352:351-61.

    8. Stone JH, Holbrook JT, Marriott MA, et al. Solid malignancies among patients in

    the Wegeners Granulomatosis Etanercept Trial.Arthritis Rheum 2006;54:1608-18.

    9. Booth A, Harper L, Hammad T, et al. Prospective study of TNF-alpha blockade

    with iniximab in anti-neutrophil cytoplasmic antibody-associated systemic

    vasculitis.J Am Soc Nephrol2004;15:717-21.

    10. Keogh KA, Wylam ME, Stone JH, Specks U. Induction of remission by B

    lymphocyte depletion in eleven patients with refractory antineutrophil cytoplas-

    mic antibody-associated vasculitis.Arthritis Rheum 2005;52:262-8.

    11. Keogh KA, Ytterberg SR, Fervenza FC, Carlson KA, Schroeder DR, SpecksU. Rituximab for refractory Wegeners granulomatosis: report of a prospective,

    open-label pilot trial.Am J Resp Crit Care Med2006;173:180-7.

    12. Hoffman GS, Cid MC, Rendt-Zagar KE, et al. Iniximab for maintenance of

    glucocorticosteroid-induced remission of giant cell arteritis: a randomized trial.

    Ann Intern Med2007;146:621-30.

    13. Salvarani C, Macchioni P, Manzini C, et al. Iniximab plus prednisone or

    placebo plus prednisone for the initial treatment of polymyalgia rheumatica:

    a randomized trial.Ann Intern Med2007;146:631-9.

    14. Mili F, Helmick CG, Moriarty DG. Health related quality of life among adults

    reporting arthritis: analysis of data from the Behavioral Risk Factor Surveillance

    System, US, 1996-99.J Rheumatol2003;30:160-6.

    15. Borman P, Toy GG, Babaoglu S, et al. A comparative evaluation of quality of

    life and life satisfaction in patients with psoriatic and rheumatoid arthritis.

    Clin Rheumatol2007;26:330-4.

    16. Hu SW, Holt EW, Husni ME, Qureshi AA. Willingness-to-pay stated

    preferences for 8 health-related quality-of-life domains in psoriatic arthritis:

    a pilot study. Semin Arthritis Rheum 2008;Dec 16 [Epub ahead of print].

    17. Mease P, Goffe BS. Diagnosis and treatment of psoriatic arthritis.

    J Am Acad Dermatol2005;52:1-19.

    18. Brockbank JE, Stein M, Schentag CT, Gladman DD. Dactylitis in psoriatic

    arthritis: a marker for disease severity?Ann Rheum Dis 2005;64:188-90

    [Epub ahead of print].

    19. Taylor WJ. Epidemiology of psoriatic arthritis. Curr Opin Rheumatol

    2002;14:98-103.

    20. Gladman D. Psoriatic arthritis.Rheum Dis Clin North Am 1998;24:829-44.

    21. Brockbank J, Gladman D. Diagnosis and management of psoriatic arthritis.

    Drugs 2002;62:2447-57.

    Rationale or the use o biologic therapies in rheumatolog

  • 8/8/2019 Biologics Monograph

    6/14

    Benets and Risks o Biologics inInfammatory Bowel Disease:

    Critical Management DecisionsThe treatment goals in inammatory bowel disease (IBD) are

    to manage symptoms, which entails induction of remission

    (rapid control of symptoms) and maintenance of remission.

    Other goals are to change the natural history of the disease

    through endoscopic remission, thereby improving long-term out-

    comes; minimize complications from treatment; and reduce the

    need for surgeries and hospitalizations.

    Prior to the era of biologic therapies, less than 40% of treated

    patients with Crohns disease achieved remission, and approxi-

    mately one-third had high disease activity. Furthermore, toxicities

    associated with several classes of nonbiologic agents (eg, cortico-

    steroids, methotrexate) preclude their long-term use.

    Four biologic agents have been approved for the treatment of

    Crohns disease:

    Iniximab, a chimeric mouse (25%)-human (75%) IgG1

    monoclonal antibody;

    Adalimumab, a fully human IgG1 monoclonal antibody;

    Certolizumab pegol, a humanized Fab fragment of the

    anti-TNF antibody that is linked to two polyethylene

    glycol molecules;

    Natalizumab, a humanized antibody to alpha 4 beta 7 integrins.

    Randomized controlled trials have shown that anti-TNF therapy

    is effective in patients for moderate to severe Crohns disease.

    In several trials, remission and maintenance of remission was

    achieved signicantly more often in patients assigned to anti-TNF

    therapy (iniximab, adalimumab, and certolizumab) than in pla-

    cebo recipients.1-5 The trials also showed signicant improvement

    in maintenance of remission with biologic agents as well as lower

    rates of Crohns disease-related hospitalizations, consistent with

    an improvement in the natural history of the disease.

    The potential mechanisms for secondary loss of response to

    anti-TNF therapy are the formation of neutralizing antibodies,

    increased clearance of biologic agents, and noninammatory

    complications of the bowel.

    About one-third of patients with Crohns disease treated with

    iniximab will require dose escalation.2 In patients with sec-

    ondary failure to iniximab, response rates of about 80% are

    obtained with dose escalation, a rate similar to that with placebo.

    About one-third of patients who do not achieve remission with

    adalimumab at week 4 will need escalation from every other

    week to weekly therapy to achieve and maintain remission.6

    In patients who initially responded to iniximab therapy but lost

    responsiveness, switching them to adalimumab achieved signi-

    cant clinical response and remission rates compared with placebo.7

    However, the absolute likelihood of response to a second anti-TNF

    agent is lower than the response in treatment-naive patients.3,5

    Loss of response to anti-TNF therapy should prompt an investiga

    tion to conrm ongoing active inammation, as not all recurrent

    symptoms are caused by disease activity. If the presence of inam

    mation is documented, measurement of antibodies is warranted

    Switching treatments within the anti-TNF class is reasonable if

    antibodies are present. Dose escalation is reasonable if antibodies

    and serum drug levels are absent. Switching to natalizumab is rea

    sonable if antibodies are absent and serum drug levels are adequate

    Factors involved in the choice o therapy

    Choosing between biologic therapies is complicated because

    different trial designs, patient populations, and denitions of

    response and remission prevent valid comparisons of results

    Taking into consideration the heterogeneity in the denition of

    response, all clinical trials of TNF inhibitors have resulted in a

    fairly uniform response rate of approximately 60% at week 4.With efcacy established for the biologic agents in the treatmen

    of Crohns disease, attention shifts to selecting the best treatmen

    and improving outcomes.

    Risk o immunogenicity

    Immunogenicity is a concern with the use of biologics. Factors

    that contribute to immunogenicity are the molecular sequence and

    structure of the agent, the treatment regimen (ongoing or episodic)

    and concomitant therapy. Episodic therapy with long intervals

    between iniximab infusions is associated with the formation o

    anti-iniximab antibodies (ATIs). The development of ATIs corre

    lates with an increased risk of infusion reactions, lower serum con

    centrations of iniximab, and a shorter duration of response.1,8-1The formation of ATIs is approximately halved by concomitant

    use of immunosuppressive agents.1,8-11 Therefore, it is now clear

    that azathioprine, methotrexate, or 6-mercaptopurine should be

    used concomitantly with iniximab, and that administering inix

    imab on a scheduled basis with the recommended induction regi

    men will reduce formation of ATIs.

    Antibody formation is not unique to iniximab. In the treatment

    of patients with rheumatoid arthritis, anti-adalimumab antibodies

    developed in 1% of patients on methotrexate and 12% of patients

    not on methotrexate. In another trial, 8% of patients developed

    antibodies to certolizumab pegol and a similar proportion o

    patients form antibodies when treated with fontolizumab.Step up or top down strategies

    Two competing strategies for the treatment of Crohns disease are

    the step up and top down strategies. The step up strategy is the

    traditional practice in which steroids are used for induction of dis

    ease remission; thiopurine or methotrexate is added for maintenance

    of remission. Once steroid dependence or failure is encountered, a

    biologic agent is added. The top down strategy starts with a combi

    nation of a thiopurine, methotrexate, and a biologic agent, with use

    of corticosteroids if loss of response occurs (see Figure 1).

    A top down strategy starting with iniximab and azathioprine was

    superior to a step up strategy at inducing remission without corti

    The rationale or and clinicalexperience with biologic agents in gastroenterology

    Aaron Brzezinski, MD, Bret Lashner, MD, Arthur McCullough, MD, Nizar Zein, MD

  • 8/8/2019 Biologics Monograph

    7/14

    costeroids and without bowel surgery in patients with Crohns dis-

    ease diagnosed in the previous 4 years.12 Relapse happened later for

    patients who started with iniximab and azathioprine than in those

    who started on steroids. Serious adverse events were similar in each

    treatment group. The rate of pancreatitis was not signicantly differ-

    ent between groups, and all cases of pancreatitis occurred in patients

    on azathioprine. The top down strategy was associated with a non-

    signicant trend toward more eczema and rash.

    Figure 1. Top down vs step up strategies12

    Steroids

    Steroids Steroids

    + AZAMTX

    + IFX IFX + AZA

    + (episodic) IFX

    Step up Top down

    Selecting therapy

    The choice of biologic agent accounts for efcacy and toxicity as well

    as patient preference and the willingness to cooperate with treatment.

    Although efcacy in achieving and maintaining remission betweenTNF inhibitors appears similar in controlled clinical trials, deci-

    phering toxicity in IBD is complicated by the use of concomitant

    medications, side effects that may be related to the disease itself,

    the under-recognition of adverse events during controlled trials and

    clinical practice, and the uncertain reliability of the information.

    Three biologic agents iniximab, natalizumab, and certoli-zumab are infusion therapies. Adalimumab, in contrast, can

    be self-administered, and although it offers greater convenience,

    it also requires the patient to ll the prescription.

    The patients willingness to accept the potential adverse events

    in exchange for clinical benets must also be considered in the

    choice of treatment. In a survey of patients with chronic activedisease, greater risk acceptance correlated with greater symptom

    improvement (see Figure 2).13

    Figure 2. Risks vs benets: Patient acceptance

    1.0 --

    0.9 --

    0.8 --

    0.7 --

    0.6 --

    0.5 --

    0.4 --

    0.3 --

    0.2 --

    0.1 --

    0 --Moderate

    to MildModerate toRemission

    Severe toModerate

    Severeto Mild

    Severe toRemission

    Observedrisk

    MAR

    (Annual%R

    isk)

    PML

    Serious infection

    Lymphoma

    The Role and Risks o Natalizumab inthe Treatment o IBD

    Natalizumab may be considered for difcult-to-treat patients with

    Crohns disease. It is approved for treating moderately to severely

    active Crohns disease. The benet-to-risk ratio of natalizumab

    appears favorable in appropriately selected patients. Contraindica-

    tions include hypersensitivity (if severe), active infection, and a his

    tory of progressive multifocal leukoencephalopathy.

    Natalizumab has been shown effective for induction and main

    tenance of remission, and it is steroid sparing.14 In patients in

    whom anti-TNF agents had failed, the remission rate was 17%

    with natalizumab versus 5% with placebo (P < 0.05). In twolarge clinical studies,15 the incidence of all-cause and Crohns

    disease-related hospitalizations was reduced signicantly in the

    natalizumab group in treatment-naive patients and anti-TNF failures.

    Progressive multifocal leukoencephalopathy (PML) is an infec

    tious disease caused by the JC virus. The infection is asymptomatic

    occurs in childhood, and remains latent; however, immunosuppres

    sive medications have been associated with reactivation of the virus

    If not recognized early, PML is almost uniformly fatal.

    Seven cases of PML have been conrmed among the 39,000

    patients worldwide who have been treated with natalizumab, onlyone of whom was being treated for Crohns disease. This patient

    received eight doses of natalizumab, was taking azathioprine con

    comitantly, and had a history of reduced white blood cell counts.

    Natalizumab distribution is restricted to pharmacies and infusion

    centers certied through the monitoring program developed in

    accordance with the US Food and Drug Administration. Figure 3shows the timeline for natalizumab marketing approvals.

    Figure 3. Natalizumab approval dates

    2004 2005 2006 2007 2008 2009| | | | | |

    November 2004:Approval by US FDA

    February 2005:Voluntarily withdrawn by

    manufacturer due to PML

    June 2006:Re-approved by USFDA and approved

    by EuropeanMedicines Agency

    January 2008:Approved by US FDA

    for moderately to

    severely activeCrohns Disease

    Current Use: 35,500patients worldwide

    September 2006:Approved byHealth Canada

    US Approval:- Monotherapy- Relapsing forms of MS to delay disability

    and reduce frequence of relapses- Mandatory registryto receive natalizumab

    EU Approval:- Monotherapy, relapsing

    remitting MS- No required registry

    The Potential Role or Biologic Agents

    in the Management o Nonalcoholic andAlcoholic Fatty Liver DiseaseNonalcoholic fatty liver disease represents a wide spectrum o

    liver disease, ranging from steatosis (relatively benign fatty liv

    er) to nonalcoholic steatohepatitis (NASH) and cirrhosis. Only

    a minority with nonalcoholic fatty liver progress to NASH, and

    few of those progress to cirrhosis.

    The histologic picture of alcoholic fatty liver disease is much the same

    as nonalcoholic fatty liver disease (accumulation of fat in liver cells

    inammation of the liver, necrosis of liver cells, and possibly brosis),

    but the progression from alcoholic liver disease to hepatitis and cirrho

    sis is much greater. Whereas about 1% of patients with nonalcoholic

    The rationale or and clinical experience with biologic agents in gastroenterolog

  • 8/8/2019 Biologics Monograph

    8/14

    steatosis will progress over their lifetime to cirrhosis and liver-related

    death, alcoholic hepatitis will progress to cirrhosis and one-fth will

    die from liver-related injury. However, the prevalence of nonalco-

    holic steatohepatitis is much higher than that for alcoholic hepatitis.

    Alcoholic atty liver: The rationale or biologics

    The basis for the use of biologic agents in alcoholic fatty liver

    disease lies in the histology of liver injury. The Kupffer cell is

    the resident macrophage in the liver. These cells are the critical

    controllers of the stellate cell, which can cause scarring and cir-rhosis when activated. Alcohol abuse increases the production of

    reactive oxygen species and endotoxemia, upregulating nuclear

    factor kappa beta to increase the production of TNF, leading to

    bacterial translocation and oxidative tissue injury.

    Tumor necrosis factor signaling through activation of Kupffer cells

    is crucial to the development of hepatotoxicity. The liver is normally

    resistant to TNF, but Kupffer cells undergo priming and activation

    during alcoholic steatohepatitis to increase production of TNF and

    sensitization to TNF. Nearly all experimental models of liver disease

    are associated with elevated levels of plasma or tissue TNF.

    The Kupffer cell is critical in activating the cytokine response

    in alcoholic liver disease. By inactivating Kupffer cells throughthe administration of gadolinium chloride, ethanol-induced

    increases in AST can be blunted.16 Fatty liver, inammation, and

    brosis have been prevented by knocking out TNF-R1 and by

    administration of TNF-alpha antibodies.

    Clinical data with biologics in alcoholic atty liver

    Etanercept and iniximab have been studied in alcoholic liver

    disease. A standard dose of iniximab results in reductions in

    ALT and total bilirubin, but mortality in severe alcoholic hep-

    atitis was increased with the addition of iniximab to steroids

    compared with steroids alone.17

    In a placebo-controlled study of etanercept in patients with alcoholic

    hepatitis, six doses of etanercept over 30 days increased mortality

    signicantly at 30 days but not at 60 days relative to placebo.18

    Pentoxyfylline, an inhibitor of TNF synthesis, appears to be the

    most promising of the biologic agents in the treatment of alcoholic

    fatty liver disease. In addition to inhibiting cytokine synthesis, it

    decreases blood viscosity and improves renal microcirculation.

    In severe alcoholic hepatitis, pentoxyfylline was associated with a

    signicant 40% reduction in mortality compared with placebo.19The

    difference in mortality occurred after 2 to 3 weeks of therapy and

    was due to a reduction in the development of hepatorenal syndrome.

    Nonalcoholic atty liver disease:

    The rationale or biologicsThe basis for the use of biologic agents in nonalcoholic fatty liver

    disease lies in adipocyte production and release of cytokines as

    fat mass increases. In addition to TNF, the release of non-TNF

    cytokines, such as transdermal growth factor-beta and IL-8, has

    an important role in the genesis of nonalcoholic fatty liver disease.

    When administered to animals on a methionine-choline decient

    diet, anti-TNF therapy produced signicant reductions in the

    percentage of steatotic cells, inammation, necrotic focus, bro-

    sis, and activated stellate cells compared with placebo.

    In experimental nonalcoholic fatty liver disease, iniximab

    decreased total liver fat and levels of TNF, IL-8, and other

    cytokines. Inhibition of TNF-alpha using pentoxyfylline

    reduced ALT signicantly with a reduction in insulin resistance in

    patients with nonalcoholic steatohepatitis.20

    No current role or biologics

    Biologic agents have no proven value in alcoholic fatty liver dis

    ease, at least in the doses studied, and the safety data are concern

    ing. In nonalcoholic fatty liver disease (NAFLD), their role is mut

    ed since lifelong therapy would be required unless patients lose a

    substantial amount of weight. Although invasive, the only therapythat interrupts the natural history of NAFLD is bariatric surgery.

    References

    1. Hanauer S, Feagan BG, Lichtenstein GR, et al. Maintenance iniximab for Crohnsdisease: the ACCENT I randomised trial.Lancet2002;359:1541-9.

    2. Rutgeerts P, Feagan BG, Lichtenstein GR, et al. Comparison of scheduled andepisodic treatment strategies of iniximab in Crohns disease.Gastroenterology 2004;126:402-13.

    3. Colombel JF, Sandborn WJ, Rutgeerts P, et al. Adalimumab for maintenance ofclinical response and remission in patients with Crohns disease: the CHARM trial.Gastroenterology 2007;132:52-65.

    4. Feagan BG, Panaccione R, Sandborn WJ, et al. Effects of adalimumab therapy onincidence of hospitalization and surgery in Crohns disease: results from the CHARMstudy. Gastroenterology 2008;135:1493-9.

    5. Schreiber S, Khalig-Kareemi M, Lawrence IC, et al. Maintenance therapy withcertolizumab pegol for Crohns disease.N Engl J Med2007;357:239-50.

    6. Sandborn WJ, Hanauer SB, Rutgeerts P, et al. Adalimumab for maintenancetreatment of Crohns disease: results of the CLASSIC II trial. Gut2007;56:1232-9.

    7. Sandborn WJ, Rutgeerts P, Enns R, et al. Adalimumab induction therapy forCrohns disease previously treated with iniximab: a randomized trial.

    Ann Intern Med2007;146:829-38.

    8. Baert F, Noman M, Vermeire S, et al. Inuence of immunogenicity on the long-termefcacy of iniximab in Crohns disease.N Engl J Med2003;348:601-8.

    9. Sands BE, Anderson FH, Bernstein CN, et al. Iniximab maintenance therapy forstulizing Crohns disease.N Engl J Med2004;350:876-85.

    10. Kugathasan S, Levy MB, Saeian K, et al. Iniximab retreatment in adults andchildren with Crohns disease: risk factors for the development of delayed severesystemic reaction.Am J Gastroenterol2002;97:1408-14.

    11. Farrell RJ, Alsahli M, Jeen YT, et al. Intravenous hydrocortisone premedicationreduces antibodies to iniximab in Crohns disease: a randomized controlled trial.Gastroenterology 2003;124:917-24.

    12. DHaens G, Baert F, van Assche G et al. Early combined immunosuppression orconventional management in patients with newly diagnosed Crohns disease:an open randomised trialLancet2008;371:660-7.

    13. Sands B, Siegel C, Hass S, et al. Crohns disease patients willingness to accept therisks of serious adverse events in exchange for clinical benet. Presentation atDigestive Disease Week; Los Angeles, CA; May 20-25, 2006:Abstract W1200.

    14. Targan SR, Feagan BG, Fedorak RN, et al. Natalizumab for the treatment of activeCrohns disease: results of the ENCORE Trial. Gastroenterology2007;132:1672-83.

    15. Sands BE, Siegel CA, Spencer M, Hass S. Natalizumab reduces the hospitalizationrate in moderate to severe Crohns disease patients: A pooled analysis of the ENACT-1and ENCORE studies. Presentation at Digestive Disease Week; San Diego, CA;May 17-22, 2008:Abstract 1039.

    16. Adachi Y, Bradford BU, Gao W, et al. Inactivation of Kupffer cells prevents earlyalcohol-induced liver injury.Hepatology 1994;20:453-60.

    17. Naveau S, Chollet-Martin S, Dharancy S, et al. A double-blind randomizedcontrolled trial of iniximab associated with prednisolone in acute alcoholic hepatitis.

    Hepatology 2004;39:1390-7.

    18. Menon KV, Stadheim L, Kamath PS, et al. A pilot study of the safety and tolerabilityof etanercept in patients with alcoholic hepatitis.Am J Gastroenterol2004;99:255-60.

    19. Akriviadis E, Botla R, Briggs W, et al. Pentoxifylline improves short-term survivalin severe acute alcoholic hepatitis: a double-blind, placebo-controlled trial.Gastroenterology2000;119:1637-48.

    20. Satapathy SK, Garg S, Chauhan R, et al. Benecial effects of tumor necrosis factor-alpha inhibition by pentoxifylline on clinical, biochemical, and metabolic parameters of

    patients with nonalcoholic steatohepatitis.Am J Gastroenterology 2004;99:1946-52.

  • 8/8/2019 Biologics Monograph

    9/14

    Immunology o Biologic Agents or

    NonimmunologistsThe immune system is an integrated system composed of the

    innate immune system and the adaptive immune system. Even

    though they are distinct systems, they are intimately linked.

    The innate immune response can be dened by several principles:

    It has no memory, so when assaulted, it responds as it did to

    previous assaults.

    The response occurs early and rapidly through quick

    mobilization of its cellular elements macrophages,

    polymorphonuclear leukocytes, dendritic cells, and the

    soluble components that complement acute-phase proteins.

    It operates by expressing the germ-line encoded receptors. It does not recognize every antigen; instead, it recognizes

    highly conserved structures.

    The role of the immune system is to protect against outside

    invasion. It does this by detecting danger in the form of infection,

    chemical insults (eg, environmental and self-inicted toxins),

    and physical damage (eg, tissue injury, cell death).

    In the case of an insult that punctures the skin, the rst layer

    of defense is the epithelium, which contains molecules (eg, beta

    defensins) that have antibacterial properties. Bacteria also reside

    in the subcutaneous space for insults that penetrate the epithelium.

    Dendritic cells in the epithelium (Langerhans cells in the epi -dermis) have a system of pattern recognition receptors, such

    as pathogen-associated molecular pattern (PAMP) receptors.

    When stimulated, dendritic cells secrete cytokines (eg, tumor

    necrosis factor, IL-1, IL-6). These cytokines serve as an early

    warning signal to the immune system. Resident tissue macro-

    phages respond to this danger signal by accumulating at the

    injury site. The cytokines prepare the endothelium to allow

    the invasion of more inammatory cells by upregulating the

    endotheliums adhesion molecules (selectins and integrins)

    They further upregulate the microbicidal activity of the residen

    phagocytic cells to allow them to kill the invader bacteria. Thiscytokine response occurs within a matter of hours, producing

    inammation and erythema.

    If the innate immune system fails, it must recruit additiona

    defenses, such as natural killer cells (see Figure 1). The acute

    phase response is an innate defense that involves increases in

    the production of acute-phase proteins in the serum, one being

    C-reactive proteins, which have the ability to bind to certain

    bacterial recognition agents and activate brinogen, hapto

    globin, and serum amyloid A, among others.

    If infection persists despite the efforts of the innate immune

    system, elements of the adaptive immune system are recruited

    Remaining bacteria migrate to the regional lymph nodes, where

    they interact with the adaptive immune system.

    The adaptive immune response is a delayed response to a spe

    cic antigen, demonstrating the features of specicity and

    memory, two elements lacking in innate immunity. The two

    types of adaptive immune response are cell-mediated and

    humoral. Whenever adaptive immunity is elicited, it creates

    cells that have immunologic memory to react quickly should

    the invader attack again. Antigens are taken up by macrophages

    and presented to helper T cells.

    Both antibodies and T cells have unique surface receptors. Each

    cell bears its own type of receptor that responds to a differenttype of antigen. When an antigen enters a cell, transport mole

    cules called major histocompatibility complex (MHC) molecules

    within the cell attach themselves to the cells surface, where they

    present the antigen to helper T cells so that the T cell can bind to

    the presented antigen. A second costimulatory signal is essentia

    to activate the T cell. These costimulatory signals are members

    of the B7 family and are expressed constitutively by T cells and

    react to ligands on antigen-presenting cells.

    Figure 1. Immune response stages

    Innate Immunity(immediate:0-4 hours)

    Early inducedresponse

    (early:4-96 hours)

    Adaptive immuneresponse

    Infection

    Infection

    Infection

    Recognition bypreformed,

    nonspecic effectors

    Recruitment ofeffector cells

    Transport ofantigen to

    Removal ofinfectious agent

    Recognition,activation ofeffector cells

    Recognitionby naive

    Removal ofinfectious agent

    Clonal expansionand differentiation

    Removal of

    The role o allied health proessionals in the administrationand monitoring o biologic therapies in rheumatology

    Leonard Calabrese, DO, Elizabeth Kirchner, CNP, Tiany Clark, RN, Carmen Gota, MD

  • 8/8/2019 Biologics Monograph

    10/14

    After the antigen has migrated to the regional lymph nodes and

    has found the appropriate helper T cells, which have become

    activated, the nal step in the adaptive response is deployment.

    One limb of the activated T cell forms active effector cells, which

    recognize cytokines and chemokines emanating from the area

    of infection, and another limb of the activated T cells turns into

    memory cells, which recognize the infection should it recur.

    At the same time, B cells that have differentiated in the bone mar-

    row migrate to the lymph nodes and search for the antigens thathave migrated from the skin. On the surface of every B cell are

    Y-shaped antibodies called immunoglobulins (IgG, IgM, IgD, IgA,

    and IgE) that have the capacity to recognize larger pieces of the

    antigen. This is part of the humoral immune response. The anti-

    body binds to the antigen at the arms of the Y.

    Activated B cells secrete cytokines such as macrophages, T

    cells, and dendritic cells. These cytokines and proteins cause

    inammation and joint destruction in the patient with rheu-

    matoid arthritis (RA). An important pathogenic inammatory

    cytokine produced directly by activated B cells is IL-6, which

    has a number of biological activities. Interleukin-6 is found

    in the highest concentration of all the cytokines released by Bcells. It is active in many tissues including those of the liver,

    bone, immune cells, fat, muscle, and kidney.

    Role o the Allied Health Proessionalin use o Biologic Therapies or RAEach limb of the immune system is a potential target for

    immune-based therapies. The infusible biologic agents with

    indications in rheumatology are iniximab, abatacept, and

    rituximab (see Table 1).

    Table 1. Indications or TNF inhibitors

    Disease Etanercept Infiximab Adalimumab

    Rheumatoid arthritis Yes Yes Yes

    Psoriatic arthritis Yes Yes Yes

    Ankylosing spondylitis Yes Yes Yes

    Juvenile idiopathic

    arthritis

    Yes In trials Yes

    Crohns disease No Yes Yes

    Ulcerative colitis No Yes In trials

    Infiximab

    Iniximab is an antibody that neutralizes the biological activ-

    ity of TNF-alpha by binding to its soluble and transmembrane

    forms and inhibiting TNF-alpha binding.

    Abatacept

    Abatacept is a fusion protein composed of an immunoglobulin

    fused to the extracellular domain of CTLA-4, a molecule

    capable of binding B7. Abatacept is a selective costimulation

    modulator as it inhibits the costimulation of T cells.

    Ordinarily, full T cell activation requires the following:

    1) Binding of the T-cell receptor to the antigen-MHC

    complex on the APC, and

    2) A costimulatory signal provided by the binding of the

    T cells CD28 protein to the B7 protein on the APC.

    Abatacept, which contains a high-afnity binding site for B7

    works by binding to the B7 protein on antigen-presenting cells

    and preventing them from delivering the costimulatory signato T cells, thus preventing the full activation of T cells.

    Rituximab

    Rituximab is a chimeric monoclonal antibody against the

    protein CD20, which is widely expressed on B cells, from

    early pre-B cells to later in differentiation. Rituximab destroys

    B lymphocytes, and thus, it is used to treat diseases that are

    characterized by having too many B cells, overactive B cells

    or dysfunctional B cells.

    Contraindications to the use o biologicdisease-modiying antirheumatic drugs

    Active infection and pneumonitis are contraindications to the

    use of biologics. Others are prior lymphoproliferative disease

    diagnosed and/or treated in the past 5 years, moderate to severe

    heart failure (New York Heart Association Class III-IV), acute

    hepatitis B virus (HBV) or hepatitis C virus (HCV) infection

    (or chronic HBV or HCV infection for those with signican

    liver injury), and multiple sclerosis or demyelinating disorders.

    In patients scheduled for surgery, the American College of

    Rheumatology recommends holding the medication for 1

    week before and after surgery. An attempt should be made

    to schedule the surgery in the middle of the infusion cycle

    (eg, week 4 for those on iniximab).

    Considerations beore starting anti-TNF therapy

    Hepatitis screening

    Rare reactivation of hepatitis B has occurred in chronic carriers

    of the virus; evaluate all patients prior to initiation and during

    treatment in patients at risk for hepatitis B infection.

    Malignancy screening

    Conrm that appropriate cancer screening is complete. Use of

    anti-TNF therapies may affect defenses against malignancies

    the impact on the development and course of malignancies is

    not fully dened. An increased risk of lymphoma has been not

    ed in clinical trials.

    Tuberculosis

    Data have associated TNF inhibitors with tuberculosis (TB)

    reactivation, so patients should be evaluated for latent TB

    infection with a tuberculin skin test before starting therapy

    If results are positive, TB therapy should be initiated before

    administering TNF inhibitors. Some patients who test negative

    prior to therapy will develop active TB infection; therefore, it is

    imperative to monitor for TB infection in all patients.

  • 8/8/2019 Biologics Monograph

    11/14

    Inections

    Serious and potentially fatal infections have been reported

    including bacterial sepsis and tuberculosis. Patients should be

    assessed for active infection prior to every infusion.

    Vaccination update

    All vaccines should be brought up to date before starting anti-TNF

    therapy. Immunosuppressed patients should not receive live vaccines.

    Pregnancy test

    Anti-TNF therapy has not been studied in pregnancy, so use of

    these therapies during pregnancy should be avoided.

    Nervous system disorders (demyelinating disease)

    Demyelinating disease, such as multiple sclerosis, seizures, or

    inammation of the nerves, have occurred in rare cases. Symp-

    toms include numbness or tingling, problems with vision, weak-

    ness in arms and legs, and dizziness. Patients should be screened

    for these types of symptoms prior to beginning therapy.

    Congestive heart ailure (CHF)

    An echocardiogram or examination to rule out signs and symp-toms of CHF should be performed prior to initiating anti-TNF

    therapy. Administration of anti-TNF agents has been linked to

    worsening or new onset CHF.

    Additional considerations

    Specic TNF inhibitors have additional factors to consider

    before starting therapy:

    Abatacept should not be used in combination with anakinra

    or TNF-blocking agents.

    Baseline measures of immunoglobulins are recommended

    because rituximab can deplete IgM and sometimes IgG.

    Although evidence does not indicated an increased frequency

    of TB in patients with lymphoma treated with rituximab, it

    is often used concomitantly with methotrexate, so TB screen-

    ing should be up to date.

    Monitoring therapy with biologic agents

    Abatacept

    Six infusions of abatacept (5 months of treatment) may be re-

    quired before its full effect is observed. A joint count and health

    assessment questionnaire can help measure efcacy without the

    need for a blood draw. Because patients must present to the

    ofce every 4 weeks for abatacept treatments, it provides anopportunity to assess for new or subtle symptoms.

    Monitoring is recommended approximately every 3 months.

    This frequency allows clinicians to obtain a disease activity

    score with three variables (DAS28-3), which requires a sedi-

    mentation rate evaluation.

    Rituximab

    Monitoring patients on rituximab is unlike monitoring patients

    on other biologics. Following rituximab infusion, 90% of the

    patients B cells are depleted, although patients may not report

    an improvement in RA symptoms for 6 weeks.

    Our practice (Clark/Kirchner) checks patients 2 to 4 weeks

    after they complete their second dose of rituximab, at which

    point patients are evaluated for signs of clinical efcacy a

    well as new symptoms or adverse events. Laboratory values

    are monitored, including a complete blood count, B cell count

    metabolic panel, and acute phase reactants. A 28-joint count

    (DAS28) is advised.

    Management o Inusion-Related IssuesThe ability to generate an adverse reaction to infusible biologic agents depends in part on the amount of antihuman protein

    contained. The immune system reacts to these antibodies as a

    foreign antigen and creates its own antibodies. Adverse effects

    are classied in three categories (see Figure 2).

    FIgure 2. Adverse eects o biologics

    Adverse effects to drugs

    Related directly to themechanism of action

    allergicidiosyncratic

    Infusion reactions to biologics can be either acute (occurring

    within 24 hours, most commonly 10 minutes to 4 hours) or

    delayed (occurring after 24 hours, most commonly 5 to 7 days)

    Acute reactions are classied based on their severity (mild to mod

    erate or severe). Mild to moderate reactions can consist of a change

    in systolic blood pressure (SBP) of less than 20 mm Hg, headache

    dizziness, pruritus, nausea, and palpitations. Severe reactions are

    those causing a change in systolic BP of more than 40 mm Hgushing, fever, and shortness of breath with wheezing.

    Delayed reactions mimic serum sickness and are often associated

    with joint pain, mild fever, headache, myalgia, and skin rash.

    Infusion reactions may have a hypersensitivity basis, but these

    are rare. The clinical manifestations of anaphylactic acute

    infusion reactions include bronchospasm and urticaria. Withou

    these symptoms, the reaction is probably not allergic.

    Infiximab

    In ve studies of patients with RA, infusion reactions to inix

    imab occurred in 15% to 20% of patients and in 3% of infusions.1-

    Most reactions were mild with only 0.5% severe. Approximately0.4% of reactions were delayed.2 Reactions were similar for trials

    in patients with Crohns disease (Table 2).6-10

    Management o acute inusion reactions

    The severity of the reaction guides the management strategie

    for acute iniximab infusion reactions. Slowing the rate of infu

    sion decreases the opportunity for immune complexes to form

    With hemodynamic changes (eg, hypotension), stopping the

    infusion may be necessary. Medications such as acetaminophen

    (for fever) and diphenhydramine (for pruritus or mild allergy)

    can be used on a symptomatic basis.

    The role o allied health proessionals in the administration and monitoring o biologic therapies in rheumatology

  • 8/8/2019 Biologics Monograph

    12/14

    For more severe reactions, methylprednisolone or hydrocorti-

    sone have been recommended. For anaphylactic reactions, epi-

    nephrine is advised.

    Monitoring vital signs is important. The frequency of monitor-

    ing depends on the severity of the reaction, which can range

    from every 10 minutes with mild reactions to every 2 minutes

    with very severe reactions.

    Once the reaction is stable, the infusion can be reattempted at

    a lower rate, following treatment with IV or oral diphenhydr-amine, 25 to 50 mg, and oral acetaminophen, 650 mg. Start

    with an iniximab test dose of 10 mL/hr for 15 minutes then

    gradually increase it to 125 mL/hr over 3 hours. A protocol has

    been developed for effectively managing iniximab infusion

    reactions.11

    Table 2. Inusion reactions with infiximab

    Study, disease Pts Inusions Inusion reactions

    Infiximab Placebo

    Rheumatoid Arthritis

    Maini1 340 20% pts 10%

    Maini2 259 3% 2%

    St Clair4 751 15%-20% pts 0

    Westhovens5 721 7592 2.6%

    Crohns Disease

    Kugathasan6 86 304 24% pts

    Baert7 125 487 27% pts

    Cheifetz8 165 479 6.1%

    Farrell9 53 199 28% pts

    Hanauer10 573 2863 3%-6%

    Premedication: Evidence or and against

    Preinfusion treatment with antihistamines or acetaminophen

    to reduce the likelihood of infusion reactions is controversial.

    Diphenhydramine pretreatment provided no benet to patients

    with previous iniximab infusion reactions.12

    Investigators developed a protocol for patients with prior

    severe infusion reactions in which patients were treated with

    oral diphenhydramine and oral acetaminophen 90 minutes

    before the infusion, in addition to oral prednisone (or alterna-

    tively IV hydrocortisone or IV methylprednisolone) 20 minutes

    before the infusion.13 This protocol allowed successful retreat-

    ment of three of four patients with severe acute reactions and

    all patients with prior mild or moderate acute reactions.

    Glucocorticoid pretreatment

    Glucocorticoid pretreatment may be reasonable in RA patients

    with prior moderate to severe iniximab infusion reactions or

    comorbidities, or when changing to another infusible biologic

    is not an option.

    Preinfusion treatment with IV hydrocortisone has been shown

    to reduce the development of antibodies to iniximab and the

    incidence of subsequent infusion reactions.14 Use of daily low-

    dose glucocorticoids has been found by some to be associated

    with fewer treatment-limiting immediate infusion reactions to

    iniximab but should not be used solely for this purpose.15 Pre-

    infusion treatment with IV betamethasone, however, was no

    found to reduce the risk of iniximab infusion reactions.16

    Anti-infiximab antibodies

    Iniximab infusion reactions are often attributed to anti-inix

    imab antibodies (ATIs). Their true incidence is unknown be

    cause measurement of ATIs depends on analytical technique

    timing of sampling, dosing regimen, circulating drug, and con

    comitant therapy.

    Development of ATIs has been associated with a greater risk

    of infusion reactions and tends to limit the long-term efcacy

    of iniximab.17 Hanauer et al18 showed that infusion reactions

    occurred in 16% of patients who were positive for ATIs compared

    with 8% who were negative. Higher titers of ATIs have also been

    associated with a greater frequency of infusion reactions.

    Anti-iniximab antibodies can develop at any time but are more

    likely to occur with sporadic infusions. Maser et al19 found tha

    compared with scheduled infusions of iniximab, episodic infu

    sions were associated with a greater rate of ATIs (39% vs 16%

    P= 0.036) and infusion reactions (50% vs 21%; P= 0.018)

    The development of ATIs was reduced only by scheduled

    maintenance treatment. Patients receiving other immunosup

    pressive therapy (eg, methotrexate) are much less likely to

    develop ATIs.19-23

    Even when treatment is optimized to avoid ATIs through

    scheduled maintenance or by concomitant immunomodulator

    therapy, approximately one-half of patients still need dose

    adjustment to treat recurrent symptoms and about one-fth lose

    response.

    Rituximab

    Rituximab infusion reactions are easily managed medically with

    acetaminophen and diphenhydramine before the rst admin

    istered dose of rituximab. Additional doses of acetaminophenhydrocortisone, and diphenhydramine can be used if fever or any

    type of bronchospastic symptom occurs. If rigors occur, they can

    be managed with meperidine. Most symptoms such as broncho

    spasm, rigor, fever, and hypotension will resolve if treatment is

    delayed for 30 minutes after additional medications.

    If a side effect occurs, recommendations are to stop the infusion

    for approximately 30 minutes, add IV saline, then resume the

    infusion at a rate one step slower than the rate at the time of the

    reaction. If the reaction abates, slowly increase the rate.

    Emery et al24 showed that premedication with IV glucocorti

    coids signicantly reduced both the incidence and severity of

    acute infusion reactions in rituximab recipients.

    Mechanism is multiactorial

    Antichimeric antibodies are very rarely encountered in ritux

    imab recipients. Rather, the mechanism of rituximab infusion

    reactions appears to be multifactorial. A cytokine release syn

    drome has been described in which increases in IL-6 and TNF-

    alpha have been documented in patients with non-Hodgkins

    lymphoma and chronic lymphocytic leukemia who have been

    treated with rituximab.25 Hypersensitivity reactions are also

    possible. In patients with Hodgkins lymphoma, some patients

    have developed tumor lysis syndrome.

  • 8/8/2019 Biologics Monograph

    13/14

    Abatacept

    Abatacept is a human fusion protein that has the ability to pre-

    vent the costimulation of T cells. The FDA classies abatacept

    infusion reactions as peri-infusional (within 24 hours) and

    acute infusion events (within the rst hour). The incidence of

    peri-infusional reactions is 22.8% and the incidence of acute

    infusion reactions is 8.9%. Treatment discontinuation due to

    infusion reactions is 0.6%. Most abatacept infusion reactions

    are mild to moderate in severity. The most common effects aredizziness, headaches, and nausea.

    Uncertain mechanism

    The mechanism of abatacept infusion reactions is uncertain. The

    immunogenicity potential is very low: antiabatacept antibodies

    have been observed in only 1.7% of infusions, with antibody

    titers being slightly higher with missed doses or long intervals

    between infusions.

    References

    1. Maini RN, Breedveld FC, Kalden JR, et al. Therapeutic efcacy of multiple

    intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody

    combined with low-dose weekly methotrexate in rheumatoid arthritis.

    Arthritis Rheum 1998;41:1552-63.

    2. Maini RN, Breedveld FC, Kalden JR, et al. Sustained improvement over two

    years in physical function, structural damage, and signs and symptoms among

    patients with rheumatoid arthritis treated with iniximab and methotrexate.

    Arthritis Rheum 2004;50:1051-65.

    3. Lipsky PE, van der Heijde DM, St Clair EW, et al for the Anti-Tumor

    Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study

    Group. Iniximab and methotrexate in the treatment of rheumatoid arthritis.

    N Engl J Med2000;343:1594-602.

    4. St Clair W, van der Heijde DM, Smolen JS, et al. Combination of iniximab

    and methotrexate therapy for early rheumatoid arthritis: a randomized,

    controlled trial.Arthritis Rheum 2004;50:3432-43.

    5. Westhovens R, Yocum D, Han J, et al. The safety of iniximab, combined

    with background treatments, among patients with rheumatoid arthritis and

    various comorbidities: a large, randomized, placebo-controlled trial.Arthritis Rheum 2006;54:1075-86.

    6. Kugathasan S, Levy MB, Saeian K, et al. Iniximab retreatment in adults

    and children with Crohns disease: risk factors for the development of delayed

    severe systemic reaction.Am J Gastroenterol2002;97:1408-14.

    7. Baert F, Noman M, Vermeire S, et al. Inuence of immunogenicity on the

    long-term efcacy of iniximab in Crohns disease.

    N Engl J Med2003;348:601-8.

    8. Cheifetz A, Smedley M, Martin S, et al. The incidence and management of

    infusion reactions to iniximab: a large center experience. Am J Gastroenterol

    2003;98:1315-24.

    9. Farrell RJ, Alsahli M, Jeen YT, et al. Intravenous hydrocortisone

    premedication reduces antibodies to iniximab in Crohns disease:

    a randomized controlled trial. Gastroenterology 20003;124:917-24.

    10. Hanauer SB, Feagan BG, Lichtenstein GR, et al, for the ACCENT I StudyGroup. Maintenance iniximab for Crohns disease: the ACCENT I randomized

    trial.Lancet2002;359:1541-9.

    11. Lequerr T, Vittecoq O, Klemmer N, et al. Management of infusion reactions

    to iniximab in patients with rheumatoid arthritis or spondyloarthritis:

    experience from an immunotherapy unit of rheumatology.

    J Rheumatol2006;33:1301-14.

    12. Wasserman MJ, Weber DA, Guthrie JA, et al. Infusion-related reactions to

    iniximab in patients with rheumatoid arthritis in a clinical practice setting:

    relationship to dose, antihistamine pretreatment, and infusion number.

    J Rheumatol2004;31:1912-7.

    13. Cheifetz A, Smedley M, Martin S, et al. The incidence and management of

    infusion reactions to iniximab: a large center experience.

    Am J Gastroenterol2003;98:1315-24.

    14. Farrell RJ, Alsahli M, Jeen YT, et al. Intravenous hydrocortisone pre -

    medication reduces antibodies to iniximab in Crohns disease: a randomized

    controlled trial. Gastroenterology 2003;124:917-24.

    15. Augustsson J, Eksborg S, Ernestam S, et al. Low-dose glucocorticoid

    therapy decreases risk for treatment-limiting infusion reaction to iniximab in

    patients with rheumatoid arthritis.Ann Rheum Dis 2007;44:1462-6.

    16. Sany J, Kaiser MJ, Jorgensen C, Trape G. Study of the tolerance of inix-

    imab infusions with or without betamethasone premedication in patients with

    active rheumatoid arthritis.Ann Rheum Dis 2005;64:1647-9.

    17. Baert F, Noman M, Vermeire S, et al. Inuence of immunogenicity

    on the long-term efcacy of iniximab in Crohns disease. N Engl J Med2003;348:601-8.

    18. Hanauer SB, Feagan BJ, Lichtenstein GR, et al. Maintenance iniximab for

    Crohns disease: the ACCENT I randomised trial. Lancet2002;359:1541-9.

    19. Maser EA, Villela R, Silverberg MS, Greenberg GR. Association of trough

    serum iniximab to clinical outcome after scheduled maintenance treatment for

    Crohns disease. Clin Gastroenterol Hepatol2006;4:1248-54.

    20. Braun J, Kstner P, Flaxenberg P, et al. Comparison of the clinical

    efcacy and safety of subcutaneous versus oral administration of methotrexate

    in patients with active rheumatoid arthritis: results of a six-month,

    multicenter, randomized, double-blind, controlled, phase IV trial.

    Arthritis Rheum 2008;58:73-81.

    21. Maini RN, Breedveld FC, Kalden JR, et al. Therapeutic efcacy of multiple

    intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody

    combined with low-dose weekly methotrexate in rheumatoid arthritis.Arthritis Rheum 1998;41:1552-63.

    22. Vermeire S, Noman M, Van Assche G, et al. Effectiveness of concomitant

    immunosuppressive therapy in suppressing the formation of antibodies to

    iniximab in Crohns disease. Gut2007;56:1226-31.

    23. Hanauer SB, Wagner CL, Bala M, et al. Incidence and importance of

    antibody responses to iniximab after maintenance or episodic treatment in

    Crohns disease. Clin Gastroenterol Hepatol2004;2:542-53.

    24. Emery P, Fleischmann R, Filipowicz-Sosnowska A, et al. The efcacy and

    safety of rituximab in patients with active rheumatoid arthritis despite

    methotrexate treatment: results of a phase IIB randomized, double-blind,

    placebo-controlled, dose-ranging trial.Arthritis Rheum 2006;54:1390-400.

    25. Winkler U, Jensen M, Manzke O, et al. Cytokine-release syndrome in

    patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts

    after treatment with an anti-CD20 monoclonal antibody

    (rituximab, IDEC-C2B8).Blood1999;94:2217-24.

    The role o allied health proessionals in the administration and monitoring o biologic therapies in rheumatology

  • 8/8/2019 Biologics Monograph

    14/14

    Produced and Distributed by Cleveland Clinic Center or Continuing Education

    Copyright 2009 The Cleveland Clinic Foundation. All Rights Reserved.