Therapeutics: Biologics and Small Molecules


Biologics or biological disease-modifying antirheumatic drugs (DMARDs) are different from nonbiologic or synthetic DMARDs in that they are produced by biological processes rather than chemical syntheses. Biological DMARDs target specific, well-defined molecules expressed on cells or secreted into the extracellular space. , The terms nonbiologic , synthetic , or conventional are used to identify the agents discussed in Chapter 13 . This chapter focuses on the mechanism of action, pharmacokinetics, pharmacodynamics, dosing, and safety of various biological DMARDs (b-DMARDs) , as well as targeted synthetic therapies also known as small molecules . For information related to efficacy of these agents in pediatric rheumatic diseases, please consult the chapter that discusses the specific disorder.

General Considerations

In general, affinity to the drug target is a major determinant of the pharmacokinetic and pharmacodynamic profile of monoclonal antibodies (mAbs) and fusion proteins used for treatment of rheumatic diseases. , Higher affinity allows efficacy to be maintained at lower serum concentrations. That is why biological DMARDs with similar half-lives and sizes can have very different durations of efficacy. Essentially, all biological agents are immunogenic because they are “nonself.” Even humanized and fully human mAbs and fusion molecules can elicit antibody responses. The effects of human antichimeric antibodies (HACAs) and human antihuman antibodies (HAHAs) include reduction in serum levels, anaphylactoid reactions, neutralization of biological activity with loss of clinical efficacy, and adverse drug reactions. The interactions with Fc receptors also affect the pharmacodynamics of those mAbs and fusion proteins that include the Fc portion of immunoglobulin (Ig)G. The system of naming monoclonal and receptor biologics is summarized Table 14.1 . Information regarding the major mAbs and fusion proteins used in pediatric rheumatology is shown in Table 14.2 .

TABLE 14.1
Naming Conventions for Monoclonal Antibody and Receptor Biological Drugs Encountered in Rheumatology
Adapted from World Health Organization’s International Nonproprietary Names (INN) Scheme for Pharmaceutical Substances, Guidelines on the Use of International Nonproprietary Names (INNs) for Pharmaceutical Substances
Suffix Source Target or Action: Example
Monoclonal antibody suffix
-ximab Chimeric: the variable regions are of murine origins whereas the constant regions are human Immunomodulator: infliximab, rituximab
-zumab Humanized: mostly derived from a human source except for the part of the antibody which binds to its target Tumor (other): epratuzumab
Lymphocyte: certolizumab pegol, daclizumab, eculizumab, ocrelizumab, omalizumab, tocilizumab, vedolizumab
-umab Human: entirely derived from a human source Interleukin: canakinumab, guselkumab, secukinumab, sirukumab, ustekinumab
Immune modulator: adalimumab anifrolumab, belimumab, golimumab, sifalimumab
Fusion proteins Cytotoxic T lymphocyte-associated antigen 4 receptors: abatacept
B-cell: Atacicept
TNF receptors: etanercept
Interleukin receptor antagonist: rilonacept, anakinra
TNF, Tumor necrosis factor.

Modified form of the physiological human protein interleukin-1 receptor antagonist (IL-1Ra), lacking glycosylation and added methionine residue at the amino terminus.

TABLE 14.2
Molecule, Target, Half-Life, Route of Administration, Indications, Dose, and Major Toxicities of Biological Agents in Pediatric Rheumatology
Generic Name Molecule Target Drug Half-Life Indication for Use Off-Label Use in Pediatric Rheumatology Preparations Boxed Warnings Comments
Abatacept CTLA4-Ig Selective costimulation modulator, which inhibits T-cell activation by binding to CD80/CD86, thereby blocking T-cell interaction with CD28 IV: 13 days
SC: 14.3 days
RA, PsA
JIA (min age: 2 years)
Uveitis Single-use vial:
250 mg lyophilized powder for reconstitution/dilution prior to IV use
Prefilled syringe for SC use
50 mg/0.4 mL
87.5 mg/0.7 mL
125 mg/1.0mL
Autoinjector for SC use
125 mg/mL solution in a single dose
Don’t use with other biologics
Adalimumab mAb to TNFα TNF specific inhibitor, which blocks TNF interaction with the p55 and p75 cell surface TNFR; in vitro adalimumab also lyses surface TNF expressing cells when complement is present 2 weeks RA, PsA, Ps, AS, Hidradenitis Suppurativa (min age 12 years)
JIA (min age 2 years)
Uveitis (min age 2 years) Peds CD (min age 6 years)
Sarcoidosis Prefilled syringe for SC use
80 mg/0.8 mL
40 mg/0.8 mL
40 mg/0.4 mL
20 mg/0.4 mL
20 mg/0.2 mL
10 mg/0.2 mL
10 mg/0.1 mL
Autoinjector for SC use
80 mg/0.8 mL
40 mg/0.8 mL
40 mg/0.4 mL
Serious infections, TB, lymphoma, and other malignancies. Postmarketing cases of fatal HSTC lymphoma With or without MTX
Anakinra IL-1Ra IL-1 4–6 hours RA sJIA, CAPS (8 months of age at least 10 kg) Don’t use with other biologics Don’t use with other biologics
Belimumab mAb to BLyS B-lymphocyte stimulator (BLyS)-specific inhibitor that blocks the binding of soluble BLyS, a B-cell survival factor, to its receptors on B cells IV: 19.4 days
SC: 18.3 days
SLE (min age: 18 years) cSLE IV Infusion
120 mg or 400 mg lyophilized powder in single-dose vials for reconstitution and dilution prior to IV use
Prefilled syringe for SC use
200 mg/mL single-dose
Autoinjector for SC use
200 mg/mL single-dose
Observe for 1 hour after infusion is complete
Canakinumab mAb to IL-1 IL-1 26 days adults
23–26 days child
CAPS, sJIA, TRAPs, MSKD, FMF Don’t use with other biologics Don’t use with other biologics
Certolizumab Pegylated mAb to TNF Recombinant, humanized antibody Fab’ fragment, with specificity for human TNF-α, conjugated to an approximately 40-kDa polyethylene glycol 14 days CD, RA, PsA, AS, Ps JIA pCD SC injection
200 mg lyophilized powder in a single-dose vial
Prefilled syringe for SC use
200 mg/mL
Serious infections
TB
lymphoma, and other malignancies
With or without MTX
Etanercept TNFRII/FcIgG1 Dimeric fusion protein consisting of the extracellular ligand-binding portion of the human 75 kDa (p75) TNFR linked to the Fc portion of human IgG1, resulting in blockage of TNF α and β 4.25 days RA, PsA, Ps, AS, JIA (min age: 2 years)
Peds Ps (min age: 4 years)
JDM Prefilled syringe for SC use
25 mg/0.5 mL
50 mg/mL
Autoinjector for single SC use
50 mg/mL single dose
Reusable autoinjector
50 mg/mL cartridge single dose
Serious infections
TB
lymphoma and other malignancies
With or without MTX
Golimumab (ARIA) mAb to TNF mAb specific to human TNF-α (ARIA IV form is preservative free) 2 weeks RA, PsA, AS,
JIA (in Europe for patients >40 kg)
JIA
Uveitis
Intravenous infusion
50 mg/4 mL solution in a single-dose vial
Prefilled syringe for SC use
50 mg/0.5 mL
100 mg/1 mL
Autoinjector for single SC use
50 mg/0.5 mL single dose
100 mg/mL single dose
Serious infections
TB
lymphoma and other malignancies
With MTX
Infliximab mAb to TNF mAb that neutralizes the biological activity of TNF-α by binding with high affinity to the soluble and transmembrane forms of TNF-α and inhibits binding of TNF-α with its receptors. 8–9.5 days CD, Peds CD, UC, Peds UC, RA, PsA, Ps, AS Uveitis
JIA
Sarcoidosis
Peds Ps
Kawasaki Disease
Intravenous infusion
100 mg of lyophilized infliximab in a 20 mL vial
Serious infections
TB
lymphoma and other malignancies
Postmarketing cases of fatal hepatosplenic T-cell lymphoma
With MTX premed with corticosteroid, acetaminophen, antihistamine
Rilonacept IL-1R/IL-1AcP/FCIgG1 IL-1 7.72 days CAPS, sJIA, FMS 4.4 mg/kg (max 320 mg) loading dose then 2.2 mg/kg q week (max 160 mg) Don’t use with b-DMARDs Don’t use with b-DMARDs
Rituximab mAb to CD20 mAb that targets the CD20 antigen expressed on the surface of pre-B and mature B-lymphocytes. Upon binding to CD20, rituximab mediates B-cell lysis. 32 days RA, GPA, MPA, Pemphigus vulgaris cSLE (skin, joints, brain, kidney)
RF-positive JIA
GPA/MPA
JDM
Scleroderma
Injection:
100 mg/10 mL (for lymphoma therapy)
Single use vials:
500 mg/50 mL
Fatal infusion reactions
Severe mucocutaneous reactions
Hep B reactivation
PML
With MTX
premed with glucocorticoid, acetaminophen, antihistamine
Tocilizumab mAb to IL-6 receptor IL-6 11–13 days RA, sJIA, pJIA Intravenous Infusion: 20mg/1mL in vials of 80mg, 200mg or 400 mg Prefilled syringe for SC use: 162 mg/0.9 mL Autoinjector for SC use: 162 mg/0.9 mL Infection, TB, malignancy, GI perforation, hypersensitivity reaction, anaphylaxis/anaphylactoid reactions
MS, lipids
With or without MTX; dose interruptions for liver toxicity, neutropenia, thrombocytopenia
Please also see medication package inserts approved by the regulatory agencies for medication licensure for more detailed information.
AS, Ankylosing spondylitis; CAPS, cryopyrin-associated periodic syndrome; CD, Crohn disease; FMF, familial Mediterranean fever; GI, gastrointestinal; GPA, granulomatosis with polyangiitis; HSTC, hepatosplenic T-cell; Ig, immunoglobulin; IL, interleukin; IV, intravenous infusion; JDM, juvenile dermatomyositis; JIA, polyarticular course juvenile idiopathic arthritis; mAb, monoclonal antibody; MPA, microscopic polyangiitis; MS, multiple sclerosis/demyelinating disease; MSKD, musculoskeletal disorders; MTX, methotrexate; PML, progressive multifocal leukoencephalopathy; Ps, psoriasis; PsA, psoriatic arthritis; RA, rheumatoid arthritis; RF, rheumatoid factor; SC, subcutaneous injection; sJIA, systemic JIA; TB, tuberculosis; TNF, tumor necrosis factor; TNFR, tumor necrosis factor receptor; TRAPs, TNF receptor-associated periodic syndrome; UC, ulcerative colitis.

In addition to complications due to HACAs or HAHAs, there are concerns that targeting the immune system may result in an increase in serious infections, malignancies, and autoimmune disease. Clinical trials are powered for efficacy rather than to exactly quantify rare safety events that may be associated with treatment (e.g., malignancy, serious infections, autoimmune diseases). Pharmacovigilance, the pharmacological science relating to the collection, detection, assessment, monitoring, and prevention of adverse effects with medicines after they have been licensed for use, is generally done to capture previously unreported adverse reactions. Such safety information is best collected in disease-specific, long-term, large-scale registries. There are several national and international registries, including the German BIKER (Biologika in der Kinderrheumatologie) Registry, the Swedish Registry, the Childhood Arthritis and Rheumatology Research Alliance (CARRA) Registry, and the PharmaChild Registry. , Current rates of serious safety events with b-DMARDs are summarized in Table 14.3 .

TABLE 14.3
Rates of Serious Infections Malignancies, Autoimmune Diseases, Antibiological DMARD Antibodies Used in Pediatric Rheumatology Per 100 Patient-Years of Exposure ,
Adapted from J.F. Swart, S. de Roock, N.M. Wulffraat, Arthritis Res. Ther. 15 (2013) 213.
Serious Infections Malignancies Autoimmune Diseases Antidrug Antibodies
Normal children 1.0 0.032 0.0069 new-onset uveitis
0.0083 IBD
0.00015 optic neuritis
0.0001 MS
JIA without MTX, steroids, or anti-TNF 2.2 0.025 2.5 new-onset uveitis
JIA with MTX 3.3 0.033–0.046 0.83 uveitis
JIA with steroids 6.9 ND ND
Abatacept 1.3 ND 0.22 uveitis flare
0.22 MS
23 no adverse events
Adalimumab 2.9 ND 0 7.6 with MTX within 1 year
25.3 without MTX within 1 year
Anakinra 8.7 ND ND 75.0 no neutralizing within 1 year
81.8 after 1 year
6.3 neutralizing within 1 year
0 neutralizing after 1 year
Etanercept 2.7 0.015 0.44 new-onset uveitis
0.57 flares of uveitis
0.31 new-diagnosis IBD
0.15 new cSLE
0.64 new-diagnosis sarcoid
2.9 no neutralizing
Infliximab 1.0 ND 5.1 new-onset uveitis
25.9 new ANA ≥ 1:320 no symptoms
6.6 new anti-dsDNA no symptoms
36.6 positive
32.4 inconclusive
Infusion reaction related
Rituximab 14.5 ND ND ND
Tocilizumab 11.6 ND ND 7.1
1 anaphylactoid reaction
ANA, antinuclear antibodies; cSLE , childhood-onset systemic lupus erythematosus; IBD, inflammatory bowel disease; JIA, juvenile idiopathic arthritis; MS, multiple sclerosis/demyelinating disease; MTX, methotrexate; ND , not determined.

General rules for children with rheumatic disease treated with a b-DMARD are as follows:

  • Do not start or give b-DMARDs to a patient with an active infection.

  • Treatment with b-DMARDs and also synthetic DMARDs is associated with an increased risk of infections.

  • Prior to initiation of a b-DMARD, screening must be done for the presence of tuberculosis, infectious hepatitis, and, depending on local recommendations, fungal diseases such as histoplasmosis, coccidiomycosis, or leishmaniasis.

  • Surveillance for infections must occur during b-DMARD therapy.

  • While on treatment with a b-DMARD, vaccination with live or live-attenuated vaccines remains contraindicated, despite some reports of the potential safety of these vaccines when given to a child on b-DMARD therapy.

  • All b-DMARDS can be combined with synthetic DMARDs (e.g. methotrexate [MTX], leflunomide, or sulfasalazine).

Regarding the formation of antibodies against b-DMARDs, a systematic review and meta-analysis identified 26 studies and 2354 juvenile idiopathic arthritis (JIA) patients available for the analysis. Prevalence of antidrug antibodies ranged from 0% to 82% across nine biological agents. Overall pooled prevalence of antidrug antibodies was 16.9% (95% confidence interval [CI], 9.5 to 25.9). Qualitative analysis of included studies indicated that antibodies to infliximab, adalimumab, anakinra, and tocilizumab were associated with reduced effectiveness and/or hypersensitivity reactions. Concomitant use of MTX uniformly reduced the risk of antibody formation during adalimumab treatment (risk ratio 0.33; 95% CI, 0.21 to 0.52).

In adults, anticytokine combination therapy has been so far avoided because in the trials that first studied such a combination, severe infectious side effects occurred. No studies are currently available in children with the combination of biological drugs.

In considering the use of b-DMARDs, one should weigh the risk of the wild-type infection against the possible side effects of vaccination and the risk of disease flare should treatment be withheld for a period after vaccination. Children who are considered for treatment with a b-DMARD should be evaluated for latent tuberculosis with a tuberculosis skin test (purified protein derivative [PPD] or Mantoux test) or a blood-based diagnostic assay (e.g., QuantiFERON-Gold). The latter may have higher specificity particularly in patients who have had bacillus Calmette-Guérin (BCG) vaccination. A chest radiograph is probably unnecessary unless the PPD result is positive. If the skin test or blood-based assay is positive, a thorough investigation of the patient and family for active tuberculosis must be undertaken. If latent tuberculosis is present, standard treatment (i.e., isoniazid for 9 months) should be initiated. , Typically, patients receive latent tuberculosis therapy for at least 1 month before starting the anti–tumor necrosis factor (TNF) regimen or a Janus kinase inhibitor, which can be used provided that local rates of primary multidrug-resistant tuberculosis (TB) infection are less than 5%. ,

Before the initiation of immunosuppressive therapy, it is recommended that an individual without a history of or vaccination against varicella zoster virus or measles virus be tested for protective antibody levels and, if possible, vaccination should be given if required. If possible, live or live-attenuated vaccinations should be given at least 3 weeks prior to starting b-DMARDs. The safety, immunogenicity, and effects of vaccines on the underlying rheumatic disease remain a concern in patients on b-DMARDs. , Immunization responses may be decreased compared with those observed in healthy pediatric populations. Notably, immunization schedules of children treated with DMARDs for their pediatric rheumatic disease should mirror those for children with altered immunocompetence. The 2011 European League Against Rheumatism (EULAR) recommendations state that nonlive vaccines are safe and can be administered to patients on corticosteroids and b-DMARD therapy. This includes the human papilloma virus (HPV) vaccine series, which is safe and effective to prevent HPV infection and genital cancer. There is no contraindication to HPV vaccination for adolescents with pediatric rheumatic diseases. As in the general population, timing is key for the efficacy of the HPV vaccine as the goal is to vaccinate prior to sexual debut and exposure to HPV, although vaccine responses may be somewhat lower than in normal children. ,

Perioperative management of b-DMARDs has been extensively discussed. Various national and international societies have produced recommendations focused on adults with rheumatic diseases. Depending on the type of surgical intervention, it is recommended that b-DMARDs should be held prior to surgical procedures for two to five terminal half-lives (see Table 14.2 ). As such, b-DMARDs should be held for two half-lives when surgery is performed in a sterile environment (e.g., cataract) and five half-lives for surgeries performed in a septic environment (e.g., colon) or in septic risk situations (e.g., joint prosthesis). If there is no evidence of infection and wound healing is satisfactory, b-DMARDs may be restarted postoperatively.

Intravenous Immunoglobulin

Intravenous immunoglobulin (IVIG) is a preparation of purified human plasma that had been sterilized from thousands of healthy donors. It consists of 95% intact IgG, IgA, and traces of IgM molecules. IVIG doses used in inflammatory conditions are many-fold higher than the doses used for replacement therapy in immunocompromised patients. The total dose of IVIG per infusion is usually 2 g/kg and is administered over a period of 1 to 2 days. Upon intravenous administration, IgG enters the vascular compartment at high concentration, redistributes rapidly into tissue compartments (i.e., α phase involving rapid lysosomal degradation as a result of saturation of Fc receptors), and then is slowly catabolized (i.e., β phase when the neonatal Fc receptors are not saturated and IVIG is recycled back to the surface of the cell). The mechanisms whereby IVIG exerts its therapeutic effects are not entirely clear and may differ in each disease state. Potential mechanisms are listed in Table 14.4 . IVIG is relatively safe but anaphylactoid reactions, thromboembolic events, renal complications including osmotic nephrosis and renal failure, hemolysis, and acute meningeal inflammation can occur. , Both intravenous and subcutaneous administration of Igs are possible. Because patients with IgA deficiency are often considered to be at increased risk of allergic reactions or anaphylaxis when they receive IVIG that contain some IgA, selective deficiency of IgA should be excluded before administration. Anaphylactoid, thromboembolic, hemolytic, and meningeal (aseptic meningitis) events can be minimized by slower infusion or changing preparations; renal complications can be minimized by better hydration and use of sugar-free stabilizers. Current preparation protocols purify the product so that it is not contaminated with, for example, human immunodeficiency virus or hepatitis C virus. However, there is always a risk of transmission of as-yet unidentified pathogens. A possible schema for IVIG administration is shown in Table 14.5 .

TABLE 14.4
Proposed Immunomodulating Mechanisms of Intravenous Immunoglobulins
  • Saturation and modulation of the expression of Fc gamma receptors

  • Saturation of neonatal Fc receptors

  • Modulation of dendritic cells

  • Expansion of regulatory T cells

  • Decreasing proinflammatory effects of monocytes

  • Decreasing the interferon-alpha response

  • Inhibition of the complement activation cascade

  • Neutralization of chemokines and/or cytokines

  • Inhibition of apoptosis

  • Neutralization of autoantibodies based on in vitro studies

  • -

    Anti-factor VII

  • -

    Anti-DNA

  • -

    Antierythroblast

  • -

    Anti-C1 inhibitor

  • -

    Anticardiolipin

  • -

    Antiplatelet

  • -

    Antiintrinsic factor

  • -

    Antimitochondrial antigens

  • -

    Antithyroglobulin

  • -

    Antineutrophil cytoplasmic antigens (ANCA)

  • -

    Antiendothelial cell

  • -

    Anti-S retinal antigen

  • -

    Antigangliosides (anti-Gm1)

  • -

    Antiglomerular basement membrane

  • -

    Anti-C3 nephritic factor

  • -

    Antiacetylcholine receptor

TABLE 14.5
Schema for Intravenous Immunoglobulin Administration
Dose & Preparation
  • 5% or 10% preparation (use lower concentration initially)

  • Use IgA depleted preparation for severe IgA deficiency or primary immune deficiencies

  • Up to 2 g/kg (max 75 g); supplied as 2.5–5-g vials

Administration
  • Start IV with normal saline

  • Give premedication as needed (e.g., acetaminophen, diphenhydramine, methylprednisolone)

  • Give IVIG at rate of 0.5 mL/hr for 30 minutes, then 1.0 mL/kg/hr for 30 minutes, then 2.0 mL/kg/hr until complete, if tolerated

Clinical Monitoring
  • Vital signs every 15 minutes for first hour, every 30 minutes for second hour, every 60 minutes thereafter

  • Observe 30 minutes after infusion completion

  • Monitor infusion reactions ; stop infusion, check oxygen saturation

  • Headache, vomiting 18–36 hours after infusion (aseptic meningitis)

Laboratory Monitoring

  • Immunoglobulin level IgA at baseline

IgA, Immunoglobulin A; IVIG, intravenous immunoglobulin.

Vital signs include all of the following: temperature, respiratory rate, heart rate, blood pressure.

Signs of infusion reaction include fever, chills, pruritus, urticaria, chest pain, shortness of breath, hypotension, or hypertension.

Inhibition Of T Cell Costimulation

CTLA-4 Ig (Abatacept)

In order to effectively activate resting T cells, two molecular signals are required : (1) the interaction of the T-cell receptor with processed peptide, bound to the appropriate major histocompatibility complex molecule; and (2) interaction of CD28 on T cells with CD80/86 on the surface of the antigen-presenting cell. Another high-affinity receptor, cytotoxic T lymphocyte–associated antigen-4 (CTLA-4), can also bind to CD80/86 with a higher avidity than CD28, thereby preventing the second signal required for T-cell activation (see Chapter 5 ). Abatacept is a fully human, soluble fusion protein comprising the extracellular domain of CTLA-4 and the Fc component of IgG1, which selectively inhibits the costimulatory signal necessary for full T-cell activation ( Fig. 14.1 ). As a selective costimulation modulator, abatacept inhibits T-cell activation by binding to CD80 and CD86, thereby blocking their interaction with CD28. This interaction provides a costimulatory signal necessary for full activation of T lymphocytes.

Fig. 14.1, Biological therapies currently used in JIA and uveitis. A, Mechanism of action of abatacept. Abatacept binds the CD80 and CD86 family of B proteins on antigen-presenting cells, thereby preventing the costimulatory signaling needed to activate T cells via CD28. B, Structure of the three anti-TNF biologics used in the treatment of JIA and uveitis. Adalimumab golimumab, and infliximab are anti-TNF monoclonal antibodies (mAbs) but differ in that infliximab is a chimeric antibody whose antigen-binding region uses a mouse component. Etanercept is a fusion protein containing a human FcIgG1 linked with a TNF receptor. C, Mechanism of action of etanercept. TNF exerts its action on immunoinflammatory cells by binding to cell-bound TNF receptors, resulting in cell activation. Etanercept binds circulating TNF, thus preventing cellular binding and cell activation. D, Mechanism of action of anakinra. Anakinra binds to the IL-1 receptor, blocking binding of IL-1α or IL-β, and thereby prevents binding of the IL-1 receptor accessory protein (IL-1R-AcP) and subsequent cell signaling. E, Mechanism of action of tocilizumab. Tocilizumab is a genetically engineered, humanized mAb to the IL-6 receptor that is produced by grafting the complementarity-determining region of mouse antihuman IL-6 receptor antibody to human IgG1. Tocilizumab competes with both the soluble and the membrane-bound IL-6 receptor preventing cell signaling. F , Canakinumb directly binds to IL-1β, neutralizes IL-1β and prevents it from binding to IL-1β receptor. Figure F from Lachmann HJ, et al. The emerging role of interleukin‐1β in autoinflammatory diseases. Arthritis Rheum. 2011;63:314–24.

There are two preparations of abatacept, one for intravenous administration and one for subcutaneous administration. Maltose is contained in the abatacept preparation for intravenous administration and can give falsely elevated blood glucose readings with certain blood glucose monitors on the day of abatacept infusion.

In an international, multicenter prospective study of 190 subjects with polyarticular course JIA using a randomized, double-blind, placebo-controlled withdrawal design, adverse events were recorded in 37 abatacept recipients (62%) and 34 (55%) placebo recipients ( P = 0.47). Abatacept was well tolerated in this trial, and there were no serious adverse events. In the open-label extension of this study, there were 1.33 serious infections per 100 patient-years among 153 patients. Five patients developed six infections, including dengue fever, erysipelas, gastroenteritis, herpes zoster, bacterial meningitis, and pyelonephritis. Subcutaneous abatacept was tested in a 24-month open-label international trial of 219 subjects with polyarticular course JIA with age at enrollment as young as 2 years. The effectiveness of subcutaneous abatacept was comparable to what was observed with intravenous abatacept in children with JIA. Across weight groups, abatacept trough levels prior to the next injection (Cmin) were comparable to what is observed in rheumatoid arthritis (RA). Local injection-site reactions were of mild or moderate intensity and occurred in around 7% of subjects. The frequency of serious adverse events was 4.7%. There were four serious infections (upper respiratory, pneumonia, pyelonephritis, and sepsis) during the 380 patient-years of follow-up (1.1%). There was one subject found to have stage III ovarian germ cell teratoma, which was thought to be unrelated to the study drug. Systematic reviews with meta-analyses suggest that abatacept therapy is associated with few treatment emergent infections, with similar responses to therapy and reduction of flare risk in polyarticular course JIA compared with other b-DMARDs used in JIA. , In line with data from trials in RA, abatacept monotherapy and combination therapy with MTX yielded comparable improvement of polyarticular course JIA. In adult RA, the time to response to abatacept and TNF inhibitors were similar, , raising the expectation that the same might hold true for JIA. Pregnancy outcomes with abatacept exposures were reported for 161 pregnancies, 151 pregnancies after maternal exposure and 10 after paternal exposure, respectively. Seven of 86 (8.1%) live births after maternal exposure had congenital anomalies: cleft lip/cleft palate, congenital aortic anomaly, meningocele, pyloric stenosis, skull malformation, ventricular septal defect/congenital arterial malformation, and Down syndrome with premature rupture of membranes at 17 weeks that resulted in a live birth via cesarean section and subsequent infant death. In addition, 59 of 151 (39.0%) cases with maternal exposure resulted in abortions (40 spontaneous and 19 elective). Of the 10 pregnancies with paternal exposure, there were nine live births and one elective abortion; there were no congenital abnormalities identified or fetal deaths. Based on these data, there does not seem to be a specific pattern of congenital abnormalities after maternal or paternal exposure to abatacept. EULAR has recommended that abatacept be replaced before conception by other medications and only be used during pregnancy when there is no other pregnancy-compatible drug that can effectively control maternal disease. A schema for abatacept use in children with JIA is shown in Table 14.6 .

TABLE 14.6
Schema for Use of Abatacept in the Treatment of JIA
Dose & Preparation
Intravenous

  • <75 kg: 10 mg/kg intravenously on weeks 0, 2, 4, then every 4 weeks

  • 75 kg to 100 kg: 750 mg intravenously on weeks 0, 2, 4, then every 4 weeks

  • >100 kg: 1000 mg intravenously on weeks 0, 2, 4, then every 4 weeks

Subcutaneous

  • 20–<25 kg: 50 mg/ week subcutaneously

  • 25–50 kg: 87.5 mg/ week subcutaneously

  • >50 kg: 125 mg / week subcutaneously

Infusion
  • Give premedication as needed (e.g., acetaminophen, diphenhydramine, methylprednisolone)

  • Given over 30 minutes

  • Vital signs every 30 minutes during infusion until 30 minutes afterward

  • Monitor infusion reactions – if present stop infusion, check oxygen saturation and possibly restart at lower infusion speed, depending on clinical assessment

Clinical Monitoring
  • Hold for suspected bacterial infection, varicella, or measles

  • Document absence of latent or active tuberculosis and hepatitis B before starting and while on abatacept treatment

  • Improvement can be seen by 2–4 months but may be delayed

  • Monitor disease status every 1–2 months initially, then every 3–6 months, depending on course

Laboratory Monitoring
  • CBC with differential; AST, ALT, albumin at baseline and then every 12 weeks

  • Glucose monitoring for IV abatacept for patients with diabetes or glucose intolerance

  • Evaluation for tuberculosis or infectious hepatitis at baseline and intermittently during use

  • Monitor for pregnancy as indicated.

ALT, Alanine aminotransferase; AST, aspartate aminotransferase; CBC , complete blood count; JIA, juvenile idiopathic arthritis.

Vital signs include all of the following: temperature, respiratory rate, heart rate, blood pressure.

Signs of infusion reaction include fever, chills, pruritus, urticaria, chest pain, shortness of breath, hypotension, and hypertension.

B-Cell Targeted biological Dmards

Rituximab

Rituximab is a chimeric mAb that targets the CD20 antigen expressed on the surface of pre-B and mature B-lymphocytes but not plasma cells or stem cells. Upon binding to CD20, rituximab mediates B-cell lysis. Possible mechanisms of cell lysis include complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). Although the antibody-producing plasma cells are not removed, B cells that may act as antigen-presenting cells, produce cytokines, and infiltrate tissues are removed for a prolonged period. Memory B cells, which are also responsible for antibody production, may be removed as well (see Chapter 5 ). Rituximab is theoretically beneficial in diseases in which autoantibodies may be pathogenic.

Rituximab dosing is influenced by the indication of use and the stage of therapy. Higher doses are given for induction as opposed to maintenance therapy or disease relapse. Rituximab induces a profound depletion of all peripheral blood B-cell populations in patients with RA. Relapse of RA tends to occur upon the return of B cells and repopulation is accompanied by higher numbers of memory B cells. ,

Prolonged hypogammaglobulinemia is not uncommon after rituximab. Serious (including fatal) bacterial and fungal infections can occur during and after rituximab-based therapy. The same holds true for de novo or reactivation of viral infections including cytomegalovirus, herpes simplex virus, parvovirus B19, varicella zoster virus, and hepatitis B and C virus. Supplementation with IVIG to maintain low-normal IgG levels may be needed to avoid recurrent infections in the setting of rituximab-associated hypogammaglobinemia.

Based primarily on adult data, approaches to off-label use of rituximab in children with pediatric rheumatic diseases are shown in Table 14.7 . Most pediatric studies of rituximab are in childhood-onset systemic lupus erythematosus, , where its use has been associated with serious infection rate at 6.6 to 12.6 per 100 patient-years of follow-up. Other reported uses are for the treatment of antiphospholipid syndrome, antineutrophil cytoplasmic antigens (ANCA)-associated vasculitis, Sjögren syndrome, Henoch–Schönlein purpura, uveitis, juvenile dermatomyositis, systemic sclerosis, and pediatric autoimmune encephalitis. HACAs may develop in one-third of patients after treatment and are associated with lower serum levels of rituximab at 2 months and less effective B-cell depletion. In a pooled analysis of placebo-controlled studies in RA, acute infusion-related reactions were experienced by 27% of rituximab-treated patients after their first infusion. Acute infusion-related reactions required dose modification (stopping, slowing, or interruption of the infusion) in 10% of the patients after the first course. Although there were no clear benefits from oral glucocorticoids, the administration of intravenous glucocorticoids prior to rituximab infusions reduced the incidence and severity of such reactions. Patients in clinical studies also received antihistamines and acetaminophen prior to rituximab infusions. Considering the experience with rituximab in 2578 RA patients, the rate of serious infections is 4.31 per 100 patient-years, mainly pneumonia, lower respiratory tract infections, cellulitis, and urinary tract infections.

TABLE 14.7
Schema for Use of Rituximab
Dose & Preparation
Intravenous

  • 500 mg/m 2 (max 1000 mg) intravenously on weeks 0 and 2

  • 375 mg/m 2 weekly for 4 doses at 50 mg/hr initially

Infusion
  • Patient to be under visual observation during all dose increases and for 30 minutes after infusion completed

  • Vital signs every 15 minutes ×2 times then every 1 hour during infusion until 30 minutes post infusion completion

  • Premedicate with methylprednisolone 100 mg intravenously 30 minutes prior to infusion

  • Give other premedication as needed (e.g., acetaminophen, diphenhydramine, ranitidine)

  • Infusion rate for first infusion

    • Initiate at 0.2 mL/kg/hr (max 12.5 mL/hr) for 30 minutes

    • Increase by 0.2 mL/kg/hr (max 12.5 mL/hr) every 30 minutes as tolerated to a maximum of 1.4 mL/kg/hr (max 100 mL/hr) until completed

  • Infusion rate for subsequent infusions (if no infusion reaction occurred)

    • Initiate at 0.4 mL/kg/hr (max 25 mL/hr) for 30 minutes

    • Increase by 0.4 mL/kg/hr (max 25 mL/hr) every 30 minutes as tolerated to a maximum of 1.4 mL/kg/hr (max 100 mL/hr) until completed

  • For infusion reaction, stop infusion and treat with corticosteroids and antihistamines possibly restart at a lower rate based on the severity of reaction

  • Consider withholding antihypertensive medication for 12 hours prior to infusion as hypotension can occur

Clinical Monitoring
  • Improvement should be seen within 1–2 months of initial infusion

  • Hold for suspected bacterial infection, fungal infection, varicella, or measles

  • Discontinue for suspected progressive multifocal leukoencephalopathy

  • Monitor every 1–2 months initially, then every 3 months, depending on course

Laboratory Monitoring
  • Screen for tuberculosis and infectious hepatitis

  • Check B-cell numbers before and 1 month after infusion

  • Quantitative immunoglobulins every 3 months

  • CBC, AST, ALT, GGT every 4–12 weeks

  • Evaluation for tuberculosis or infectious hepatitis at baseline and intermittently during use

  • Monitor for pregnancy as indicated

ALT, Alanine aminotransferase; AST, aspartate aminotransferase; CBC , complete blood count; GGT , gamma-glutamyl transferase.

Vital signs include all of the following: temperature, respiratory rate, heart rate, blood pressure.

Signs of infusion reaction include fever, chills, pruritus, urticaria, chest pain, shortness of breath, hypotension, and hypertension

Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease of the central nervous system resulting from reactivation of the Creutzfeldt–Jakob virus. PML is extremely uncommon despite the high prevalence of the Creutzfeldt–Jakob virus in the general population. The U.S. Food and Drug Administration (FDA) issued an alert in December 2006 after documentation of two fatal cases of PML in patients with systemic lupus erythematosus (SLE) after rituximab treatment. In total, 26 cases of PML were identified in patients concomitantly treated with rituximab for rheumatic disease. Nevertheless, rituximab-associated PML is a rare adverse event. It occurs in less than 1 in 20,000 rituximab-treated RA patients and the risk may be higher in SLE. However, PML can occur in patients with SLE even without rituximab exposure, with the incidence ranging from 1.0 to 2.4 per 100,000 person-years. No specific treatment of PML is available, and the prognosis is poor. Baseline screening for Creutzfeldt–Jakob virus is not recommended or required. Rituximab also has been classified as a drug associated with a high risk for hepatitis B virus reactivation in hepatitis B surface antigen (HbsAg)-negative/anti-hepatitis B core (HBc) positive patients.

Using the rituximab global drug safety database, Chakravarty et al. identified 231 pregnancies associated with maternal rituximab exposure. Maternal indications included lymphoma, autoimmune cytopenias, and other autoimmune diseases. Most cases were confounded by concomitant use of potentially teratogenic medications and severe underlying disease. Of 153 pregnancies with known outcomes, 90 resulted in live births with 23 infants born prematurely, and one neonatal death at 6 weeks. Eleven neonates had hematological abnormalities; none had corresponding infections. Four neonatal infections were reported (fever, bronchiolitis, cytomegalovirus hepatitis, and chorioamnionitis). Two congenital malformations were identified: clubfoot in one twin and cardiac malformation in a singleton birth. One maternal death from preexisting autoimmune thrombocytopenia occurred. The authors concluded that, although few congenital malformations or neonatal infections were seen among exposed neonates, women should be counseled to avoid pregnancy for at least 12 months after rituximab exposure. EULAR has recommended that rituximab be replaced before conception by other pregnancy-compatible drugs whenever possible. Ofatumumab, a fully human mAb targeting CD20, is commercially available but its clinical trial program in RA has been discontinued. Potentially, given the absence of HACAs, infusion reactions from ofatumumab may be less frequent than with rituximab in rheumatic diseases.

Belimumab

Belimumab is a human IgG1 neutralizing mAb against B-lymphocyte stimulating factor (also known as B-lymphocyte stimulator [BLyS ]). BLyS, a member of the TNF ligand superfamily, exists in both a 285-amino acid membrane-bound form and a cleaved 152-amino acid soluble form. Expressed on monocytes, macrophages, and monocyte-derived dendritic cells, BLyS is upregulated in response to interferon (IFN)-γ and interleukin (IL)-10, enhancing B-cell proliferation and Ig secretion. Belimumab binds with high affinity to BLyS and inhibits binding of BLyS to its three receptors, inhibiting BLyS-induced proliferation of B cells and decreasing survival of autoreactive B cells, which are particularly dependent on soluble BLyS. , Treatment results in decreasing serum anti–double-stranded DNA antibody levels.

Belimumab is indicated for adult and pediatric patients with active, autoantibody-positive SLE despite receiving standard therapy. The efficacy of belimumab for the treatment of severe active lupus nephritis or severe active central nervous system lupus has recently been established, the benefit of combination therapy with other b-DMARDs or intravenous cyclophosphamide is still under investigation.

Safety data from 2133 adult SLE patients from one phase II study and two phase III studies were analyzed. The most common serious adverse events with belimumab therapy were serious infections (6.0%), most frequently pneumonia, urinary tract infection, cellulitis, and bronchitis. Infections leading to discontinuation of treatment or death occurred in less than 1.0% of patients treated with intravenous belimumab. The most commonly reported adverse events, occurring in greater than or equal to 5% of patients in clinical trials were nausea, diarrhea, pyrexia, nasopharyngitis, bronchitis, insomnia, pain in extremities, depression, migraine, and pharyngitis. The proportion of patients who discontinued treatment due to any adverse reaction during the controlled clinical trials was 6.2% for patients receiving belimumab plus standard therapy.

The frequency of mostly mild to moderate injection-site reactions was 6.1% with subcutaneous belimumab, leading to discontinuation of the drug in 6% of those with injection-site reactions. Anti-belimumab antibodies with intravenous administration are very rare (<0.7%) and were not observed during the clinical trial of subcutaneous belimumab in 556 patients. In the controlled clinical trials of belimumab administered intravenously, hypersensitivity reactions (occurring on the same day of infusion) were reported in 13% of patients receiving belimumab, with anaphylaxis occurring in 0.6% these patients. In the controlled trials, malignancies including nonmelanoma skin cancers were similarly frequent in the belimumab and the placebo groups (0.4%).

There are only a few case reports on pregnancy outcomes with belimumab exposures, none of which supports a special pattern of birth defects. EULAR has recommended that belimumab be replaced by other medications before conception and only be used during pregnancy when there is no other pregnancy-compatible drug that can effectively control maternal disease.

A trial in childhood-onset SLE (cSLE) is under way and results are available for the 52 week endpoints. Ninety-three children with cSLE aged 5 to 17 years who continued standard of care therapy were randomized to receive IV belimumab (10 mg/kg) every 4 weeks or placebo. At week 52, there were 28/53 (52.8%) Systemic Lupus International Collaborating Clinics (SLICC) responder index 4 (SRI4) responders in the belimumab group versus 17/39 (43.6%) in the placebo group (odds Ratio [OR] 1.49 [95% CI, 0.64 to 3.46]). A greater proportion of belimumab patients were ACR/Pediatric Rheumatology International Trials Organization (PRINTO) responders (belimumab 32/53 [60.4%] vs. placebo 14/40 [35%]). The authors concluded that the benefit/risk profile of IV belimumab in the children with cSLE population is generally consistent with that of adult study populations with either intravenous or subcutaneous administration.

Current recommendations for the use of belimumab are shown in Table 14.8 .

TABLE 14.8
Schema for Use of Belimumab in the Treatment of Systemic Lupus Erythematosus ,
Dose & Preparation
Intravenous

  • 10 mg/kg intravenously on weeks 0, 2, 4, then every 4 weeks

Subcutaneous

  • 200 mg SC every 1 week for adult-sized size

Infusion
  • Give premedication as needed (e.g., acetaminophen, diphenhydramine, methylprednisolone)

  • Dilute in 250 mL of normal saline and give over 60 minutes

  • Vital signs every 30 minutes during infusion until 30 minutes afterward

  • Monitor infusion reactions – if present stop infusion, check oxygen saturation and possibly restart at lower infusion speed, depending on clinical assessment

Clinical Monitoring
  • Hold for suspected bacterial infection, fungal infection, varicella, or measles

  • Discontinue for suspected progressive multifocal leukoencephalopathy

  • Document absence of latent or active tuberculosis and hepatitis B before starting and while on belimumab treatment

  • Improvement can be seen by 4–5 months but may be delayed

  • Monitor disease status every 1–2 months initially, then every 3 months, depending on course

Laboratory Monitoring
  • CBC with differential; AST, ALT, albumin every 4–12 weeks

  • Evaluation for tuberculosis or infectious hepatitis at baseline and intermittently during use

  • Monitor for pregnancy as indicated.

  • Monitor for leukopenia and elevated transaminases with each infusion

ALT, Alanine aminotransferase; AST, aspartate aminotransferase. CBC , complete blood count.

Vital signs include all of the following: temperature, respiratory rate, heart rate, blood pressure.

Signs of infusion reaction include fever, chills, pruritus, urticaria, chest pain, shortness of breath, hypotension, and hypertension.

Interference With Cytokines

Tumor Necrosis Factor Inhibitors

General Considerations

TNF is an important cytokine implicated in the pathogenesis of JIA and can induce expression of other proinflammatory cytokines such as IL-1, IL-6, and IL-8, leading to an overall protracted inflammatory response. In the normal physiological state, proinflammatory cytokines, including TNF-α, are maintained in equilibrium with antiinflammatory cytokines, such as IL-10. Among others, TNF levels correlate with disease activity and TNF inhibition results in suppression of IL-6 and IL-8 in parallel with clinical improvement in JIA. TNF inhibitors have been proven to be effective in the treatment of different inflammatory conditions, including polyarticular course JIA, psoriatic arthritis, ankylosing spondylitis, and inflammatory bowel disease, whereas effectiveness in systemic JIA is limited ( Table 14.2 ). Anti-TNF agents or TNF inhibitors currently in use or under study in children are etanercept, infliximab, adalimumab, golimumab, and certolizumab.

Generally, the efficacy of the various TNF inhibitors is comparable in JIA. An exception is etanercept, which is not recommended for use in uveitis. Combination treatment with MTX seems to improve the response to etanercept, adalimumab, and infliximab in JIA besides decreasing the occurrence of antibodies against these b-DMARDs.

With the increasing use of TNF inhibitors, several concerns have arisen, one of which is the increased risk of infection, particularly tuberculosis, fungal infections such as histoplasmosis, and opportunistic infections. Children with JIA had a higher rate of opportunistic infections, such as coccidiomycosis, salmonellosis, listeriosis, and herpes zoster. Earlier research supports that the rate of infection is not substantially increased with MTX or TNF-inhibitor use in JIA, in contrast to the use of high doses of corticosteroids. It is reassuring that a recent meta-analysis that compared children with JIA treated with a TNF-inhibitors with those who were not treated with a TNF-inhibitor showed similar risks of infections (OR, 1.13; 95% CI, 0.76 to 1.69; P =0.543). ,

Before starting treatment with any TNF-inhibitor, screening for the presence of latent or active tuberculosis based on local recommendations should be performed. If latent tuberculosis is diagnosed, the patient should be given isoniazid. Treatment with anti-TNF agents may be initiated 1 month after starting isoniazid. The FDA advises close monitoring of patients for signs and symptoms of potential fungal infection, especially in endemic areas, both during and after treatment with TNF inhibitors. Patients in whom fever, malaise, weight loss, sweats, cough, dyspnea, pulmonary infiltrates on chest radiographs, or serious systemic illness develop should undergo a complete diagnostic workup appropriate for immunocompromised patients. The decision to initiate empiric antifungal therapy in at-risk symptomatic patients should be made in conjunction with an infectious disease specialist, considering both the risk for severe infection and the risks of antifungal therapy.

TNF inhibitors should be withheld for serious infection or sepsis and not initiated in patients with active infection. Mild upper respiratory tract or urinary tract infections can be a reason to hold TNF inhibitors intermittently or temporarily but not to permanently discontinue anti-TNF therapy. Patients with hepatitis B who were treated with a TNF inhibitor experienced worsening symptoms, viral load, or hepatic function. Although hepatitis B reactivation has been added to the label, concomitant antiviral treatment can be given.

Concern remains regarding the development of malignancy, particularly lymphoma, with TNF-inhibitor therapy. In information obtained from manufacturers of TNF inhibitors approved for use in children, 48 cases of malignancies in children and adolescents were identified. It was estimated that 14,837 children received infliximab, 9200 received etanercept, and 2636 received adalimumab during the studied period. Approximately half of the malignancies were lymphomas, and others included leukemia, melanoma, and solid organ cancers. Eleven of the patients died mainly from T cell lymphoma. The rates of malignancy were higher with infliximab than expected, but the primary use of infliximab was for inflammatory bowel disease in contrast to patients with JIA who were mostly treated with etanercept. Eighty-eight percent of cases were in patients that were also taking other immunosuppressive medications such as azathioprine, 6-mercaptopurine, or MTX. Based on the above data it was concluded that there is an increased risk of malignancy with TNF-inhibitor exposure, but the strength of the association, or a definite causal relationship, could not be assigned. Some major problems with these conclusions were that the precise denominator for users was not known, and treated patients were not compared with a control group of JIA patients not treated with b-DMARDs. Bernatsky et al., using data from three Canadian centers, did not find an increased incidence of malignancy in patients with JIA. In contrast, using comprehensive administrative data from Sweden, Simard et al. reported that, compared with the preceding two decades, the incidence of malignancy in patients with JIA had increased in the years from 1987 to 1999, prior to the use of b-DMARDs. This is supported by a study of 7812 children with a total follow-up time of 12,614 person‐years, including 1484 children treated with TNF inhibitors (2922 person‐years of exposure), which reported that children with JIA have an increased rate of incident malignancy compared with children without JIA. For all children with JIA versus children without JIA, the standardized incidence rate was 4.4 (95% CI, 1.8 to 9.0) for probable or highly probable malignancies. The treatment for JIA, including TNF inhibitors and/or MTX, did not appear to be significantly associated with the development of malignancy. Nonetheless, the risk of malignancy with different types of b-DMARDs and when used in conjunction or in sequence remains a topic of intense research and the risk of malignancy in patients with JIA remains ill defined. For now, patients must be observed closely for the occurrence of malignancies.

Concerns exist also for the development of demyelinating syndromes, especially multiple sclerosis, with anti-TNF therapy. In a study which retrieved information from the US Food and Drug Administration Adverse Event Reporting System (FAERS) over a 13-year period, 3,226 reports of multiple sclerosis were retrieved. Antineoplastic and immunomodulating drugs' (33% of total reports) were the most frequently reported, including etanercept (445 cases; ROR: 2.48; 95% Cl: 2.24–2.74), adalimumab (329; 2.05; 1.83–2.30), and infliximab (119 cases; OR = 2.25; 95% Cl, 1.87 to 2.70). Besides multiple sclerosis, other demyelinating diseases have been reported with TNF-inhibitor use such as optic neuritis and Guillain-Barré syndrome. TNF-inhibitor use should be avoided in patients with a history of demyelinating syndromes or those with a strong family history of demyelinating diseases. If TNF-inhibitor use is nonetheless deemed necessary, then a baseline central nervous system magnetic resonance imaging (MRI) should be considered in patients with a family history of multiple sclerosis.

TNF-inhibitor in RA therapy has been also associated with SLE-like syndromes and antiphospholipid antibody syndrome, leukocytoclastic vasculitis, , new-onset psoriasis, , Crohn disease, and congestive heart failure in patients with no previous risk factors. A systematic review showed that TNF inhibition does not lead to significant changes in intima-media thickness, endothelial function, or lipid profiles over 52 weeks. ,

TNF inhibitors are classified as a category B medication, meaning that there is no evidence of risk in pregnancy outcome in women with rheumatic diseases. The impact of TNF inhibitors on pregnancy outcomes has been evaluated in a meta-analysis that included 13 studies of women treated with a TNF-inhibitor. There were trends toward reduced rates of live birth, increased risk of preterm birth (OR, 2.62; 95% CI, 2.12 to 3.23; P < 0.0001) with spontaneous abortion (OR, 4.08; 95% CI, 1.12 to 14.89; P = 0.033) and low birth weight (OR, 5.95; 95% CI, 1.17 to 30.38; P = 0.032) among females treated with TNF inhibitors compared with the general population. However, there was no increased risk of birth anomalies (OR, 1.46; 95% CI, 0.84 to 2.56; P = 0.18). Comparing users of TNF inhibitors with nonusers of TNF inhibitors, there were no significant differences in the rates of live birth and pregnancy-related complications. EULAR recommends considering continuation of TNF inhibitors during the first part of pregnancy in women with inflammatory arthritis. Etanercept and certolizumab may be considered for use throughout pregnancy because of low rate of transplacental passage. It has been recommended that women with inflammatory bowel disease generally continue TNF inhibitors during pregnancy. Further, it is strongly recommended to withhold live vaccinations for newborns of women who were treated with TNF inhibitors during pregnancy within the first 6 months of life. Breast-feeding appears to be safe.

Etanercept

Etanercept is a fully human, dimeric fusion protein consisting of the extracellular ligand-binding portion of the human 75 kilodalton (p75) TNF receptor linked to the Fc portion of human IgG1. Etanercept is produced by recombinant DNA and is a rather large molecule consisting of 934 amino acids and an apparent molecular weight of approximately 150 kDa. Although soluble forms of the TNF receptor occur naturally, they are generally inadequate to block TNF activity in systemic inflammatory disorders. Etanercept also binds lymphotoxin (previously called TNF-β ) and the relevance of this for JIA therapy remains unknown. Because of the initial report of its effectiveness in 2000, etanercept has been used extensively in children with JIA. It is currently indicated for children age 2 and above for polyarticular course JIA in both North America and Europe. In Europe it is also approved for use in children ages 12 to 17 years with juvenile psoriatic arthritis or enthesitis-related arthritis. The placebo-controlled study showed an increase in symptoms of upper respiratory tract infection, and 7% of children with JIA treated with etanercept reported generally mild injection-site reactions during the first 3 months of treatment. A registry of nearly 600 children with JIA followed for 3 years and smaller studies of long-term continuous use for up to 10 years support the long-term safety of etanercept in JIA. In general, adverse reactions in pediatric patients are similar in frequency and type as those seen in adult patients. In open-label clinical studies of children with JIA, adverse reactions reported in those ages 2 to 4 years were similar to adverse reactions reported in older children. A recent report from the German Biologics Registry suggests that etanercept monotherapy, but not combination therapy with MTX, may be associated with an increased risk of newly developing inflammatory bowel disease and uveitis, an observation that requires further investigation. Additional information about the use of etanercept is shown in Table 14.9 .

TABLE 14.9
Schema for Use of Etanercept in the Treatment of Juvenile Idiopathic Arthritis
Dose
Subcutaneous

  • 0.4 mg/kg twice weekly or 0.8 mg/kg per week subcutaneously

  • Maximum 50 mg/week

Clinical Monitoring
  • Document absence of latent or active tuberculosis and hepatitis B before starting and while on treatment

  • Hold for suspected bacterial infection, fungal infection, varicella, or measles

  • Improvement should be seen by the third or fourth dose

  • Monitor every 1–2 months initially, then every 3–6 months, depending on course

  • Monitor for pregnancy as indicated

Laboratory Monitoring
  • CBC with differential; AST, ALT, albumin at baseline and then every 12 weeks

ALT, Alanine aminotransferase; AST, aspartate aminotransferase; CBC , complete blood count.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here