Biologic response modifiers (BRMs) are substances that interact with and modify the host immune system. BRMs that dampen the immune system are used to treat conditions such as juvenile idiopathic arthritis, psoriatic arthritis, or inflammatory bowel disease and often in combination with other immunosuppressive agents, such as methotrexate and corticosteroids. Cytokines that are targeted include tumor necrosis factor α; interleukins (ILs) 6, 12, and 23; and the receptors for IL-1α (IL-1A) and IL-1β (IL-1B) as well as other molecules. Although the risk varies with the class of BRM, patients receiving immune-dampening BRMs generally are at increased risk of infection or reactivation with mycobacterial infections (Mycobacterium tuberculosis and nontuberculous mycobacteria), some viral (herpes simplex virus, varicella-zoster virus, Epstein-Barr virus, hepatitis B) and fungal (histoplasmosis, coccidioidomycosis) infections, as well as other opportunistic infections. The use of BRMs warrants careful determination of infectious risk on the basis of history (including exposure, residence, and travel and immunization history) and selected baseline screening test results. Routine immunizations should be given at least 2 weeks (inactivated or subunit vaccines) or 4 weeks (live vaccines) before initiation of BRMs whenever feasible, and inactivated influenza vaccine should be given annually. Inactivated and subunit vaccines should be given when needed while taking BRMs, but live vaccines should be avoided unless under special circumstances in consultation with an infectious diseases specialist. If the patient develops a febrile or serious respiratory illness during BRM therapy, consideration should be given to stopping the BRM while actively searching for and treating possible infectious causes.

Biologic response modifiers (BRMs) refer to substances that interact with the host immune system and modify it. Several BRMs, such as cytokines, chemokines, and antibodies, occur naturally in the body and help to protect against infections. Depending on the condition, synthetic BRMs mimicking natural cytokines or inhibitors, including humanized monoclonal antibodies against a cytokine or its receptor, have been used in treatment to restore, boost, or dampen the host immune response. The focus of this clinical report is on cytokine-inhibiting BRMs that dampen the immune response and treat immune-mediated conditions, such as juvenile idiopathic arthritis (JIA), inflammatory bowel disease, graft-versus-host disease associated with hematopoietic stem cell transplant (HSCT), and scleritis. Cytokines that have been targeted include tumor necrosis factor (TNF) α; interleukins (ILs) 6, 12, and 23; and the receptors for IL-1α (IL-1A) and IL-1β (IL-1B) as well as other molecules described below. These medications are increasingly being used in pediatric populations in combination with other immunosuppressive agents, such as methotrexate and corticosteroids. Although they have been very effective in treating the symptoms of the underlying immune-mediated conditions and lessening disability, the immunosuppressive effect of the cytokine-inhibiting BRMs can persist for weeks to months after the last dose.1 There is strong evidence of an association of the use of these drugs with an elevated risk of infection with viral and mycobacterial pathogens and weaker evidence for fungal and other intracellular pathogens.2,4 

This clinical report aims to summarize the infectious disease complications associated with BRMs to guide subspecialists and to familiarize pediatricians, family physicians, and other primary care practitioners who may care for, diagnose, and manage infections in children treated with BRMs. A summary of key points for primary care providers is given in Table 1. It should be noted that experience with the use of BRMs varies by product, with infliximab having the most data available for its use. Data for children are mostly extrapolated from studies in adults,5,9 with mostly small case series and cohort studies reported,2,10,15 and thus suggested practices are consensus driven on the basis of knowledge of the impact of BRMs on the immune system. Finally, patients are usually prescribed BRMs in conjunction with other immunosuppressive agents, such as methotrexate and prednisone, which also have immunomodulatory effects that must be considered when the data are being reviewed. In general, the combinations should be considered at least as immunosuppressive as the most immunosuppressive agent in the combination. Table 2 summarizes the BRMs that are currently approved by the US Food and Drug Administration (FDA) along with their mechanisms of action, route of administration, half-lives, and indications. There are also several other products under consideration or in clinical trials. Some currently licensed BRMs are being tested for many new indications, and unlicensed BRMs are also being tested for various indications either as sole agents or in combinations.

TABLE 1

Summary of Suggested Screening/Immunizations Before and After BRM Therapy

Suggested screening/immunizations before BRM started 
 Thorough history 
  Document previous vaccines, antibody testing when indicated (routine antibody testing not recommended for varicella) 
  Query about possible exposure and epidemiologic risk factors for histoplasmosis and coccidioidomycosis 
  Query about history of recurrent HSV 
  Consider serologic testing for EBV 
  Screen for past hepatitis B and determine need for vaccine (Table 3
 Routine immunizations 
  Follow current AAP, Centers for Disease Control and Prevention, and American Academy of Family Physicians guidelines16,17  
  Give recommended vaccines, inactivated, or subunit vaccines 2 weeks before initiation of BRM 
  Consider safety of giving live vaccine; if appropriate, give 4 weeks before initiating BRM18  
 TB 
  Test for latent TB and manage based on result (see ref 19 for algorithm) 
Suggested screening/immunizations after BRM started 
 Routine immunizations 
  May still receive routine inactivated, polysaccharide, recombinant, or subunit vaccinea 
  Give annual inactivated influenza vaccine18  
  Avoid live vaccines, unless under special circumstances with help of infectious diseases specialist18  
 Immunizing immunocompetent household contacts (before or during treatment)? 
  Follow AAP guidelines for immunizing household contacts of immunocompromised patients18  
 Risk of listeriosis 
  Avoid unpasteurized milk and milk products, uncooked meats20  
 Febrile or serious respiratory illness during BRM therapy 
  Consider stopping BRM and actively search for infections including bacterial, mycobacterial, and opportunistic infections 
Suggested screening/immunizations after BRM stopped 
 Routine immunizations 
  May still receive routine inactivated, polysaccharide, recombinant, or subunit vaccinea 
 Timing of giving live vaccines after stoppage of BRM ± other immunosuppressive agents? 
  Consult infectious diseases specialist 
Suggested screening/immunizations before BRM started 
 Thorough history 
  Document previous vaccines, antibody testing when indicated (routine antibody testing not recommended for varicella) 
  Query about possible exposure and epidemiologic risk factors for histoplasmosis and coccidioidomycosis 
  Query about history of recurrent HSV 
  Consider serologic testing for EBV 
  Screen for past hepatitis B and determine need for vaccine (Table 3
 Routine immunizations 
  Follow current AAP, Centers for Disease Control and Prevention, and American Academy of Family Physicians guidelines16,17  
  Give recommended vaccines, inactivated, or subunit vaccines 2 weeks before initiation of BRM 
  Consider safety of giving live vaccine; if appropriate, give 4 weeks before initiating BRM18  
 TB 
  Test for latent TB and manage based on result (see ref 19 for algorithm) 
Suggested screening/immunizations after BRM started 
 Routine immunizations 
  May still receive routine inactivated, polysaccharide, recombinant, or subunit vaccinea 
  Give annual inactivated influenza vaccine18  
  Avoid live vaccines, unless under special circumstances with help of infectious diseases specialist18  
 Immunizing immunocompetent household contacts (before or during treatment)? 
  Follow AAP guidelines for immunizing household contacts of immunocompromised patients18  
 Risk of listeriosis 
  Avoid unpasteurized milk and milk products, uncooked meats20  
 Febrile or serious respiratory illness during BRM therapy 
  Consider stopping BRM and actively search for infections including bacterial, mycobacterial, and opportunistic infections 
Suggested screening/immunizations after BRM stopped 
 Routine immunizations 
  May still receive routine inactivated, polysaccharide, recombinant, or subunit vaccinea 
 Timing of giving live vaccines after stoppage of BRM ± other immunosuppressive agents? 
  Consult infectious diseases specialist 

These are suggestions only. Each situation should be guided by clinical scenario, and the help of an infectious diseases consultant may be sought.

a

If receiving rituximab, may not respond.

TABLE 2

FDA-Approved BRMs and Indications

Generic Name (Year[s] FDA Approved for Indications)Trade NameMechanism of ActionUsual Route, Half-lifeFDA-Approved Indicationa
Infliximab (1999, 2009) Remicade21  TNF-α inhibitor (anti–TNF-α chimeric monoclonal IgG1κ antibody) IV, 7.5–9.5 d Crohn disease [RA, plaque psoriasis, psoriatic arthritis, ankylosing spondylitis, ulcerative colitis] 
Etanercept (1998) Enbrel22  TNF-α inhibitor (soluble TNF-α receptor fusion protein) SQ, 70–132 h JIA [RA, plaque psoriasis, psoriatic arthritis, ankylosing spondylitis] 
Adalimumab (2002) Humira23  TNF-α inhibitor (anti–TNF-α humanized monoclonal IgG1 antibody) SQ, 10–20 d JIA [RA, plaque psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn disease] 
Golimumab (2009) Simponi24  TNF-α inhibitor (anti–TNF-α IgG1κ antibody) SQ, 7–20 d [RA, psoriatic arthritis, ankylosing spondylitis] 
Certolizumab pegol (2009) Cimzia25  TNF-α inhibitor (PEGylated human Fab antigen binding) SQ, 14 d [RA, Crohn disease] 
Abatacept (2005, 2009) Orencia26  Anti-CTLA4 selective T-cell costimulation modulator protein (blocks TNF-α, IL-2, and interferon-γ production) IV or SQ, 8–25 d JIA [RA] 
Anakinra (2001) Kineret27  Recombinant anti-IL-1 receptor antagonist SQ, 4–6 h [RA] 
Rituximab (2006) Rituxan28  Anti-CD20 therapy IV, 14–62 d [RA, non-Hodgkin’s lymphoma] 
Tocilizumab (2010) Actemra29  Anti-IL-6 humanized monoclonal antibody IV, 8–14 d [RA] 
Ustekinumab (2013) Stelara30  Anti-IL-12 and IL-23 humanized monoclonal antibody SQ, 20–24 d [Psoriatic arthritis, plaque psoriasis] 
Canakinumab (2009, 2013) Ilaris31,32  Anti-IL-1B human monoclonal antibody SQ, 26 d CAPS, JIA 
Natalizumab (2008, 2013) Tysabri33  Humanized anti–integrin α 4 subunit monoclonal antibody (reduces leukocyte adhesion and transmigration) IV, 11 d [Crohn disease, multiple sclerosis] 
Belimumab (2011) Benlysta34  Human IgG1λ monoclonal antibody against soluble human B lymphocyte stimulator protein IV, 19 d [Systemic lupus erythematosus] 
Rilonacept (2008, orphan drug) Arcalyst35  IL-1 receptor fusion protein SQ, 8.6 d CAPS 
Tofacitinib (2012) Xeljanz36  Small molecule protein kinase inhibitor of JAK3 and JAK1 Oral, 3 h [RA
Generic Name (Year[s] FDA Approved for Indications)Trade NameMechanism of ActionUsual Route, Half-lifeFDA-Approved Indicationa
Infliximab (1999, 2009) Remicade21  TNF-α inhibitor (anti–TNF-α chimeric monoclonal IgG1κ antibody) IV, 7.5–9.5 d Crohn disease [RA, plaque psoriasis, psoriatic arthritis, ankylosing spondylitis, ulcerative colitis] 
Etanercept (1998) Enbrel22  TNF-α inhibitor (soluble TNF-α receptor fusion protein) SQ, 70–132 h JIA [RA, plaque psoriasis, psoriatic arthritis, ankylosing spondylitis] 
Adalimumab (2002) Humira23  TNF-α inhibitor (anti–TNF-α humanized monoclonal IgG1 antibody) SQ, 10–20 d JIA [RA, plaque psoriasis, psoriatic arthritis, ankylosing spondylitis, Crohn disease] 
Golimumab (2009) Simponi24  TNF-α inhibitor (anti–TNF-α IgG1κ antibody) SQ, 7–20 d [RA, psoriatic arthritis, ankylosing spondylitis] 
Certolizumab pegol (2009) Cimzia25  TNF-α inhibitor (PEGylated human Fab antigen binding) SQ, 14 d [RA, Crohn disease] 
Abatacept (2005, 2009) Orencia26  Anti-CTLA4 selective T-cell costimulation modulator protein (blocks TNF-α, IL-2, and interferon-γ production) IV or SQ, 8–25 d JIA [RA] 
Anakinra (2001) Kineret27  Recombinant anti-IL-1 receptor antagonist SQ, 4–6 h [RA] 
Rituximab (2006) Rituxan28  Anti-CD20 therapy IV, 14–62 d [RA, non-Hodgkin’s lymphoma] 
Tocilizumab (2010) Actemra29  Anti-IL-6 humanized monoclonal antibody IV, 8–14 d [RA] 
Ustekinumab (2013) Stelara30  Anti-IL-12 and IL-23 humanized monoclonal antibody SQ, 20–24 d [Psoriatic arthritis, plaque psoriasis] 
Canakinumab (2009, 2013) Ilaris31,32  Anti-IL-1B human monoclonal antibody SQ, 26 d CAPS, JIA 
Natalizumab (2008, 2013) Tysabri33  Humanized anti–integrin α 4 subunit monoclonal antibody (reduces leukocyte adhesion and transmigration) IV, 11 d [Crohn disease, multiple sclerosis] 
Belimumab (2011) Benlysta34  Human IgG1λ monoclonal antibody against soluble human B lymphocyte stimulator protein IV, 19 d [Systemic lupus erythematosus] 
Rilonacept (2008, orphan drug) Arcalyst35  IL-1 receptor fusion protein SQ, 8.6 d CAPS 
Tofacitinib (2012) Xeljanz36  Small molecule protein kinase inhibitor of JAK3 and JAK1 Oral, 3 h [RA

CAPS, cryopyrin-associated periodic syndromes (consisting of familial cold autoinflammatory syndrome and Muckle-Wells syndrome); IV, intravenous; SQ, subcutaneous.

a

For underlined conditions, safety and efficacy have been established in children <18 y; for bracketed indications, safety and efficacy have only been shown in adults. Infliximab, etanercept, and adalimumab have been used off-label for scleritis, but none are FDA approved for this condition.

Two classes of TNF-α blocking agents are currently used in managing rheumatologic conditions: (1) monoclonal anti-TNF antibodies (includes infliximab [Remicade; Janssen Biotech, Horsham, PA],21,37,39 adalimumab [Humira; AbbVie, North Chicago, IL],38,40,43 golimumab [Simponi; Janssen Biotech, Horsham, PA],24,44,45 and certolizumab pegol [Cimzia; UCB, Smyrna, GA]25) and (2) soluble TNF receptors (etanercept [Enbrel; Immunex, Thousand Oaks, CA]).22,46,47 Infliximab, the first to be licensed in its class, is a chimeric mouse/human protein, and the other monoclonal anti-TNF agents are fully composed of human amino acid sequences. Certolizumab pegol is unique in also being pegylated, which improves its pharmacokinetics and bioavailability.48 Etanercept is composed of 2 extracellular domains of human TNF-R2 fused to the fragment-crystallizable (Fc) fragment of human immunoglobulin (Ig) G1 (Fig 1).48 

FIGURE 1

Structures of the TNF-α antagonists. (Adapted from Wallis RS. Biologics and infections: lessons from tumor necrosis factor blocking agents. Infect Dis Clin North Am. 2011;25(4):896, with permission from Elsevier Limited.) Fab, Fragment antibody binding; mAb, monoclonal antibody; sTNFR, soluble TNF receptor.

FIGURE 1

Structures of the TNF-α antagonists. (Adapted from Wallis RS. Biologics and infections: lessons from tumor necrosis factor blocking agents. Infect Dis Clin North Am. 2011;25(4):896, with permission from Elsevier Limited.) Fab, Fragment antibody binding; mAb, monoclonal antibody; sTNFR, soluble TNF receptor.

Close modal

The other classes of BRMs consist of monoclonal antibodies and other proteins that antagonize IL-1, IL-6, IL-12, and IL-23 or other molecules, which are important in the inflammatory cascade.

Monoclonal Antibodies

Tocilizumab (Actemra [IL-6]; Genentech, South San Francisco, CA), ustekinumab (Stelara [IL-12 and IL-23]; Janssen Biotech, Horsham, PA), and canakinumab (Ilaris [IL-1B]; Novartis Pharma, Basil, Switzerland) are all humanized monoclonal antibodies against the interleukins noted. Natalizumab (Tysabri, Biogen, Cambridge, MA) is a recombinant human monoclonal antibody against the cell adhesion molecule α-4-integrin, which acts by preventing the adhesion of leukocytes to endothelial cells. Rituximab (Rituxan; Genentech, South San Francisco, CA) is a chimeric monoclonal antibody against the CD20 protein, primarily found on the surface of B cells, which acts by inducing B-cell destruction through mechanisms that could include complement-dependent cytotoxicity, apoptosis, or antibody-dependent cytotoxicity. Belimumab (Benlysta; GlaxoSmithKline, Rockville, MD) is a human IgG1-λ monoclonal antibody against soluble human B lymphocyte stimulator protein (BLyS; also known as BAFF and TNFSF13B). It does not bind B cells directly but blocks the binding of soluble BLyS to its receptors on B cells, thereby inhibiting B-cell survival and differentiation into immunoglobulin-producing plasma cells.34 

Recombinant IL1 Antagonist

Anakinra (Kineret; Swedish Orphan Biovitrum AB, Stockholm, Sweden) is a recombinant, nonglycosylated human IL-1 receptor (IL-1R) antagonist that blocks the biologic activity of both IL-1A and IL-1B by competitively inhibiting IL-1 binding to the IL-1 type 1 receptor found in a wide range of organs and tissues.

Fusion Proteins

Abatacept (Orencia; Bristol Myers Squibb, Princeton, NJ) and rilonacept (Arcalyst; Regeneron Pharmaceuticals, Tarrytown, NY) are both fusion proteins composed of Fc regions of IgG1 fused with another molecule. Abatacept is a fusion protein of the Fc region of IgG1 fused to the extracellular domain of cytotoxic T-lymphocyte antigen 4. Abatacept has strong affinity and binds to the B7 protein on the antigen-presenting cells, which would normally bind to the CD28 protein on T cells, preventing the antigen-presenting cells from delivering the costimulatory signals needed to fully activate the T cells. Rilonacept (Arcalyst; Regeneron Pharmaceuticals, Tarrytown, NY) is a dimeric fusion protein made of the Fc region of human IgG1 that binds IL-1 linked to the ligand binding domains of the extracellular portions of human IL-1R1 and the IL-1R accessory protein (IL-1RAcP).

Tofacitinib (Xeljanz; Pfizer, New York, NY) is an oral, small-molecule protein kinase inhibitor of the enzyme Janus kinase (JAK) 3 and 1 (JAK3 and JAK1, respectively) that interferes with the JAK-STAT signaling pathway, which transmits extracellular information into the cell nucleus, influencing DNA transcription. Functionally, tofacitinib affects both innate and adaptive immune responses by inhibiting pathogenic T helper (Th) 17 cells and Th-1 and Th-2 cell differentiation (Table 2).49,52 

BRM use has been associated with an increased risk of developing certain infections. In addition to the immunosuppressive effects of these agents, concomitant use of other immunosuppressive agents, such as steroids and methotrexate, and the underlying inflammatory disease likely contribute to increased infectious risk.47,53,54 

A 2011 Cochrane review examined adverse events identified in randomized controlled trials, controlled clinical trials, and open-label extension studies involving >60 000 participants (primarily adults) receiving 9 BRMs (adalimumab, certolizumab, etanercept, golimumab, infliximab, anakinra, tocilizumab, abatacept, and rituximab) for treatment of either rheumatoid arthritis (RA) or cancer.55 Serious infections were defined as infections associated with death, hospitalization, and/or use of intravenous antibiotics. The review identified an overall increased risk of serious infections (odds ratio [OR]: 1.37; 95% confidence interval [CI]: 1.04–1.82) among participants receiving BRMs compared with placebo recipients.55 The drugs most commonly associated with serious infection were certolizumab (OR: 4.75; 95% CI: 1.52–18.5) and anakinra (OR: 4.05; 95% CI: 1.22–16.8). Overall, patients receiving TNF-α inhibitors had a greater risk of developing a serious infection versus patients receiving other BRMs (OR: 1.4; 95% CI: 1.13–1.75). However, patients receiving any of the other BRMs also had an increased overall risk of serious infection when compared with those receiving placebo. There was evidence of an increased risk of reactivation of tuberculosis (TB) among patients receiving BRMs compared with those receiving placebo (see section titled “TB”).

Although the risk of serious infections is increased, there is no clear evidence of overall increased risk of bacterial infections, other than mycobacteria, with the use of BRMs. Before the BRM era, the rate of serious bacterial infections requiring hospitalization or parenteral antibiotics in patients with RA (primarily adults) was reported to be between 0.02 and 0.12 per patient-year.56,57 In contrast, the incidence of infection noted for patients with RA treated with etanercept has ranged from 0.017 to 0.050 per patient-year, a rate that is not significantly different from the general population.58 One hypothesis would be that even though BRMs temporarily increase the risk of serious infections, their profound positive effects on the underlying disease counterbalances this effect.

In head-to-head trials, the rates of upper respiratory infections (20%) were similar among recipients of etanercept and control patients with RA.47 Among studies involving children alone, etanercept has not been shown to increase the risk of serious nonmycobacterial infection compared with etanercept plus conventional disease-modifying antirheumatic drugs (DMARDs) such as methotrexate,22,46,59,66 with rates ranging from 0.007 to 0.035 per patient-year. Children receiving infliximab12,31,61,67,70 and adalimumab38,42,43,64,71 had higher rates of serious nonmycobacterial infections, ranging from 0.027 to 0.086 per patient-year, but there were no data presented to suggest that these rates were higher than those observed for patients receiving DMARDs alone or DMARDs in combination with the BRMs. Infections related to these BRMs were primarily lower respiratory tract and gastrointestinal infections, sepsis, and abscesses. Although there is no clear increased risk compared with patients receiving DMARDs alone, case reports of sepsis and even death among patients receiving these drugs necessitate caution in instituting therapy in patients who have active infections.

Studies involving children treated with the other classes of BRMs (IL-1 [anakinra], IL-6 [tocilizumab], and T-cell costimulation [abatacept] inhibitors) are limited and generally showed few serious infections. Similarly, infection rates have been low in adult patients treated with ustekinumab, canakinumab, and rilonacept. When infections are identified, they have primarily been viral, gastrointestinal tract, or skin infections. However, there are no data to suggest that the overall incidence of infection is increased compared with the incidence in patients taking DMARDs alone. More studies are needed on the rates of infections for patients receiving these classes of BRMs.

Rituximab-associated neutropenia is prolonged neutropenia described during or after the completion of therapy, primarily in adult patients being treated for lymphomas.72,80 The incidence ranges mostly from 0.02% to 6%, although it has been described to be as high as 25% in 1 series and has involved admission to the hospital with fever and neutropenia and sometimes sepsis with bacterial pathogens including Pseudomonas species. The duration of the neutropenia has ranged from 4 days to 1 year. The mechanism is unknown, but circulating antibodies against neutrophils were postulated in 1 study.80 In 1 open-label cohort study in which rituximab was used to treat JIA in 55 children who had failed to respond to infliximab and other DMARDs, 8 serious infections, all lower respiratory tract infections including Pneumocystis jirovecii and Mycoplasma pneumonia, were identified during a 96-week follow-up period.81 Of interest, neutropenia occurred in 17 (31%) of these patients between weeks 6 and 28 of treatment. The neutrophil count did not exceed 1500 per µL in 9 (16%) of the patients, was <1000 per µL in 5 (9%) of the patients, and was <500 per µL in 3 (6%) of the patients. All patients were treated with granulocyte-macrophage colony–stimulating factor (5 µg/kg), and no cases of sepsis or febrile neutropenia were described. Not surprisingly given the mechanism of action (anti-CD20 monoclonal antibody), 80% of the patients had a complete depletion of their B cells (CD20+ cells), which remained low throughout the 96 weeks of follow-up, but the level or duration of depletion did not correlate with any specific infection. Eleven (20%) patients also had a reduction in their serum immunoglobulin (IgM and IgG) levels below the age-related lower limits of normal. The most common (mild) infections noted were ear, nose, and throat (14%) as well as skin (11%) infections. Four cases of herpetic infections were also reported among the mild infections.

TB

Reactivation infections caused by organisms in the Mycobacterium tuberculosis complex have been associated with use of TNF inhibitors. TNF-α has been shown in mouse models to contribute to granuloma formation and to induce and maintain latency of TB infection.82 In contrast, blockage of this cytokine results in failure to control bacillary growth and form protective granulomata.83,86 In the Cochrane review by Singh et al,55 the overall OR of TB reactivation was 4.7 (95% CI: 1.2–18.6) among patients receiving BRMs compared with those receiving placebo, with the absolute risk measured at 20 cases per 10 000 compared with 4 per 10 000 patients receiving placebo. However, there were insufficient data in that study to compare risk of TB activation in patients receiving 1 BRM versus another.

In other studies conducted in adults, the risk of TB reactivation appeared to be related to the class of TNF inhibitor. TNF antibodies (eg, infliximab and adalimumab) are associated with the highest risk, whereas soluble TNF receptor antibodies (eg, etanercept) appear to have the lowest risks.87 One study counted the rates of opportunistic infections associated with infliximab use by analysis of the Adverse Event Reporting System (AERS) of MedWatch, a spontaneous reporting system of the FDA.4 Although the baseline rate of TB in adults was estimated at 6.2 per 100 000, the rate among the estimated 147 000 patients receiving infliximab was found to range from ∼10 per 100 00088 to 24.4 per 100 000.4 In contrast, there were many fewer cases of TB reported in relation to etanercept, with 9 cases identified among ∼102 000 patients treated in the same database. Similar results were reported by Dixon et al,89 based on analysis of 10 712 patients treated with anti-TNF BRMs (3913 etanercept, 3295 infliximab, 3504 adalimumab). The rates of TB were much higher in patients receiving either of the monoclonal antibodies (adalimumab: 144 events per 100 000 person-years; infliximab: 136 per 100 000 person-years) versus those taking etanercept (39 per 100 000 person-years). Other data suggest that adalimumab is associated with TB in patients with RA in a dose-related manner, with higher doses associated with greater risk.90 

Although the risk appears to be lower with etanercept, since its approval many cases of TB have been described among patients receiving the drug. The estimated incidence noted in studies of 10 to 14.3 cases per 100 000 etanercept-treated patients is more than the Centers for Disease Control and Prevention–estimated rate of TB in the general US population of 6.2 per 100 000.88,90,91 Most cases have localized disease, although disseminated infections have been reported. Other cohort92 and case-control93,94 studies noted rates of 6 to 39 cases of TB for etanercept, compared with 71 to 100 cases for infliximab and adalimumab per 100 000 patient-years.Risk of Extrapulmonary and Disseminated TB.

Of note is the significant increase in the incidence of extrapulmonary and disseminated disease among the TB cases detected in adult patients receiving BRMs, with many cases requiring an invasive procedure for diagnosis.95 For example, in the Keane et al study,4 56% of patients receiving infliximab who developed TB had extrapulmonary TB, and 24% had disseminated disease, compared with an expected background rate of 18% and 2%, respectively, in patients with non–HIV-related TB. Similarly, in the study by Dixon et al,89 62% of all cases were extrapulmonary TB, with 28% with disseminated disease. A high proportion of patients do not develop granulomas, which is consistent with diminished host response against the mycobacteria. In general, the median time to presentation is fastest with infliximab (12 weeks) versus adalimumab (30 weeks) and etanercept (46 weeks), suggesting different pharmacokinetics or pharmacodynamics or different levels of modulation of the immune system for each medication.94 

Although there are fewer data available in children, the increased risk of TB reactivation with infliximab and etanercept has been corroborated in several case reports88,96,97 and at least 1 randomized controlled trial.31,68 However, it is important to note that the risk of TB reactivation associated with the underlying autoimmune/inflammatory conditions alone without medications has been estimated to be twice the baseline rate for the normal population.98Screening to Reduce TB Infections During BRM Therapy.

There is fair evidence that screening for TB and treating before starting BRMs substantially reduces the risk of TB reactivation. In a small case series, 36 children from Turkey with JIA were treated with etanercept for a median duration of 11.5 months after careful screening with the use of chest radiography, tuberculin skin test (TST), clinical histories, family screening, and physical examination.99 It was noted that Turkey has a moderate baseline prevalence of TB of 27 per 100 000 population. Patients with TST results of >10 mm who had received bacille Calmette-Guérin (BCG) vaccine or those suspected of having latent TB for other reasons underwent computed tomography of the chest and/or had cultures performed if specimens were available. Those identified as having only latent TB (no evidence of abnormalities on computed tomography and/or sputum specimens) received 4 to 8 weeks of “treatment doses” of isoniazid followed by 9 months of “lower prophylactic doses.” The investigators did not indicate the actual doses used for either isoniazid treatment or prophylaxis, and North American authorities do not normally make a distinction between prophylaxis and treatment doses. Treatment with etanercept was started only during the “prophylaxis stage” of the treatment in 7 of the 36 children meeting the criteria, and none of the 36 developed active TB. Similarly, among 2210 adult patients with different rheumatologic conditions who were treated worldwide with golimumab, no cases of TB developed among the 317 who were assessed and treated for latent TB with isoniazid, whereas 5 cases developed in patients not assessed and treated by week 52.44 

Nontuberculous Mycobacteria

Although M tuberculosis has received the most attention, some reports suggest that nontuberculous mycobacteria (NTM) may be twice as common as TB in adult patients receiving BRMs.100,,102 With the use of the FDA MedWatch database, 239 patients with NTM were identified among patients taking BRMs.102 Although pulmonary disease was most common (56%), a substantial number of presentations were extrapulmonary (44%), which is similar to the cases seen for patients developing TB while receiving BRMs. Most of the cases occurred during treatment with infliximab (75%), but this finding may be more indicative of the overall usage of this agent compared with the others. Mycobacterium avium was the organism most commonly isolated. Actual rates of disease could not be calculated from the data presented because the total numbers of patients in the database were not reported. Even though NTM may be common, there is no consensus on baseline screening, because there is no accurate (sensitive and specific) screening test.102 Furthermore, the treatment of NTM is not as clearly delineated as for TB.

Varicella and Herpes Zoster Infections

Varicella-zoster virus (VZV) infections (primary or reactivated) are a frequently reported complication among patients receiving BRMs. For patients receiving anti–TNF-α BRMs, reactivation of VZV is the most common infection.2,60,63,103 However, primary VZV infections have also been reported.Varicella.

In a retrospective cohort study of etanercept use among 25 Italian children younger than 4 years treated for a mean period of 23 months, 2 unimmunized patients developed primary VZV infections at 40 and 24 months, respectively,60 with 1 case being complicated with necrotizing fasciitis. Similarly, Lovell et al63 described 3 patients (ages 13, 10, and 8 years) with primary VZV among 58 children (ages 4–17 years) enrolled in a North American open-label trial of treatment of JIA with etanercept. Although there was no report of their immunization status, all 3 were antibody negative at the time of diagnosis and seroconverted after infection, suggesting that these were primary wild-type infections. One of these 3 patients’ courses was complicated by aseptic meningitis, but all 3 recovered with acyclovir therapy and discontinuation of etanercept. These data support the importance of appropriate varicella immunization and the recognition of exposures in patients taking all BRMs.Herpes Zoster.

There is mixed evidence of an increase in zoster infections with the use of BRMs. The data for this evidence are almost exclusively from adult patients. In a large German registry in which 5040 patients receiving anti–TNF-α BRMs or DMARDs were enrolled prospectively to monitor their outcomes, there was a significant increase in the risk of herpes zoster infections in patients treated with monoclonal antibodies (infliximab or adalimumab) but no increase noted for those receiving etanercept or DMARDs.104 Similarly, Smitten et al,105 Galloway et al,106 and Winthrop et al,107 analyzing large databases in the United States and United Kingdom, found an increased risk of VZV reactivation in patients receiving BRMs compared with those receiving DMARDs alone. In contrast, another study108 involving the National Data Bank for Rheumatic Diseases did not find an increased risk of zoster for infliximab, etanercept, or adalimumab. Encephalitis and meningitis attributable to herpes simplex virus (HSV) and VZV have also been described in postmarketing surveillance of patients receiving natalizumab.33Herpes Simplex.

Although uncommon, HSV encephalitis, disseminated cutaneous HSV, and localized disease have been described primarily in case series as a complication of treatment with TNF-α inhibitors.5,109,,,,113 There is at least 1 case report in which infliximab was successfully used along with valacyclovir prophylaxis in a girl with Crohn disease who had relapsing peribuccal herpes labialis attributable to HSV type 1.114 

Hepatitis B

There have been reports of an increased risk of hepatitis B reactivation for patients receiving BRMs.115,116 As expected, the risk is greatest in patients who are hepatitis B surface antigen (HBsAg) positive. However, the presence of hepatitis B core antibody (HBcAb) in HBsAg-negative patients (indicating immunity on the basis of infection rather than vaccine) has been associated with HBV reactivation in some patients up to 2 years after immunosuppression with BRMs.117 In 1 study involving 257 patients infected with hepatitis B and treated with TNF inhibitors, 39% of patients who were HBsAg positive and 5% of those who were HBcAb positive had reactivation of hepatitis B.45 Increased risk of reactivation correlates with low levels of hepatitis B surface antibody (HbsAb) for patients taking rituximab or TNF-α antagonists,115,116 suggesting that reactivation is a direct result of the BRM decreasing antibody levels.

Endemic Mycoses and Other Fungi

Fungal infections (Aspergillus,111,113,118Coccidioides,119,,121Histoplasma, Cryptococcus, Sporothrix, and Candida)3,5,70,101,111,118,122,,124 have been noted, primarily in case reports or series during treatment with BRMs. Histoplasma is the most common invasive fungal organism identified3,122,123 and is especially important to diagnose, because the symptoms, signs, and chest radiography findings are virtually indistinguishable from those of acute TB (cough, fever, chills, night sweats, weight loss, and possible skin or mouth lesions). In at least 1 series, histoplasmosis was diagnosed 3 times more commonly than TB among recipients of BRMs.101 The presentation of histoplasmosis generally includes fever of unknown origin with disseminated disease in approximately three-fourths of patients, and pulmonary disease is less common. Because the signs and symptoms are nonspecific, diagnosis is usually more challenging. Furthermore, the case fatality rate of patients with disseminated disease receiving BRMs is very high (50%).122 Pancytopenia and liver dysfunction are often noted. Antigen detection in urine and serum is most useful for diagnosis, although culture of bone marrow could be considered.

Other fungi are less common. In an observational study of AERS data by Keane et al,4 although actual incidence rates were not given, there were 12 patients with P jirovecii pneumonia (PCP), 7 with histoplasmosis, 6 with aspergillosis, and 7 with severe Candida infections in association with infliximab treatment among the cohort of ∼147 000 patients treated.

HIV

There have been no studies showing an increased rate of HIV infections among patients receiving BRMs.

Progressive Multifocal Leukoencephalopathy

Progressive multifocal leukoencephalopathy (PML), an opportunistic viral infection of the brain associated with JC virus (a human polyomavirus named after the initials of the first patient in whom it was identified) infection in immunocompromised hosts, has been reported in adult patients receiving natalizumab, either as monotherapy or in conjunction with other immunomodulators or immunosuppressors.33 Risk factors associated with the development of PML include the duration of therapy, previous use of immunosuppressants, and presence of anti–JC virus antibodies. For this reason, natalizumab is only available through a restricted distribution program that also involves close monitoring known as the TOUCH prescribing program (https://www.touchprogram.com/TTP/).33 

Other Infections

Other opportunistic-type infections that have been noted (mostly case reports or case series) during treatment with BRMs used alone or with other immunosuppressive agents include viral (cytomegalovirus, Epstein-Barr virus [EBV]),111 protozoal (Pneumocystis),4,111 and bacterial (Listeria125,,127 and Legionella128,129) during postmarketing surveillance47 or in national or regional databases. In studies in which EBV or cytomegalovirus viral load was measured or polymerase chain reaction assay was performed before and during infliximab treatment over a period of 6 weeks to 5 years, there was no evidence of viral reactivation.130,131 In the adult US population older than 60 years, the annual risk of Listeria was identified as 43 per 1 000 000 among people receiving anti-TNF regimens versus 13 million in the general population.127 Similarly, a Spanish registry identified rates of 26.5 per 100 000 patient-years versus 0.34 per 100 000 patient-years in the general population.126 

P jirovecii colonization has been identified in 16% to 29% of adult patients with systemic autoimmune inflammatory diseases.132,,134 In mouse models, B cells play an antibody-independent role in clearing a murine form of Pneumocystis (Pneumocystis carinii f sp muris) by presenting antigen to CD4 T cells.135 Rituximab depletes B cells, and there have been reports of an increased risk of PCP in the population of patients receiving this BRM, primarily those patients receiving it for hematologic malignancies.136 Despite this report, the benefit of using induced sputum to screen for P jirovecii or chemoprophylaxis for PCP in patients receiving rituximab is unclear.133,137 Although some experts have recommended PCP prophylaxis in the setting of rituximab therapy,138 there are concerns that the PCP risk is lower than the target of 3.5% for the benefits to outweigh the adverse effects of chemoprophylaxis in non–HIV-infected patients.139,140 PCP infections have also been identified in 84 patients receiving infliximab identified through the AERS database141 and in those receiving other BRMs.142,143 However, virtually all of the patients were also receiving concomitant immunosuppressive agents that included methotrexate, prednisone, azathioprine, 6-mercaptopurine, and cyclosporine. As a result, it is unclear whether there is an increased risk for PCP beyond that attributable to the underlying DMARDs.

The indications for intervening with biologics are often to reverse a serious clinical condition. Thus, deferring BRMs to provide protection against vaccine-preventable diseases often involves trading risks. Although, at times, it is clearly appropriate to defer BRMs so the patient can receive an immunization, it may not always be the case, and deferral of BRMs should generally be undertaken in consultation with the relevant specialist managing the patient (eg, rheumatologist, gastroenterologist, dermatologist). However, some basic principles can be applied to reduce the risk of serious infections for patients taking BRMs. All patients with a newly diagnosed rheumatologic or immune-mediated condition, including Crohn disease and graft-versus-host disease after HSCT should be considered as a current or future candidate for a BRM and should be screened for potential opportunistic infections at the time of diagnosis before initiation of any immunosuppressive agents. Although the guidelines in this statement are applicable, patients with HSCT with or without graft-versus-host disease generally need to have several other considerations before and after their transplantation that are beyond the scope of this statement, and consultation with an infectious diseases specialist is warranted. These agents should not be given to any patients while they have clinically significant acute infections. Furthermore, restraint should be shown in prescribing BRMs to children who are being treated for chronic infections, those with a history of recurrent infections, or those with conditions (including HIV infection) that predispose them to such infections.89 For such children, it is prudent to involve an infectious diseases specialist to help guide and advocate that all risks are assessed and that appropriate screening tests before treatment and monitoring during and after treatment occur. Risks associated with potential opportunistic infections that are endemic in the area of residence (eg, histoplasmosis, coccidioidomycosis, etc) should be thoroughly considered before starting a BRM. Part of the consideration for such children will include the determination of underlying previous exposure to the infectious agent and associated risk of reactivation or infection during treatment with the BRM. If a decision is made to treat such children with a BRM, close monitoring will be required, ideally in conjunction with an infectious diseases specialist. Special consideration should always be given to the risk of TB. The development of symptoms suggestive of an opportunistic infection should lead to immediate cessation of the BRM pending confirmation of the infection, appropriate management of the infection, and resolution of symptoms. Serum neutrophil counts should be monitored regularly (weekly) in patients receiving rituximab, and episodes of recurrent ear, nose, or throat infections in these patients should lead to measurement of serum immunoglobulin concentrations.

Immunization Before a BRM Is Started

Immunizations of children about to receive a BRM should follow the current recommendations of the American Academy of Pediatrics (AAP), Centers for Disease Control and Prevention, and American Academy of Family Physicians for persons 0 through 18 years of age16 (available at: http://aapredbook.aappublications.org/site/resources/IZSchedule.pdf144). Before the initiation of BRM therapy, a thorough history should be taken for documentation of previous receipt of appropriate inactivated and live vaccines or history of having had the disease, with testing for specific antibody concentrations where documentation is inadequate. For varicella, if there is a reported history of vaccination but not confirmed by documentation, serologic testing is not indicated and vaccination is recommended if there is sufficient time to safely administer it (4 weeks minimum) before initiating the BRM (Table 2).

All recommended inactivated vaccines should be given at least 2 weeks before the initiation of BRMs to enhance immunogenicity. Live vaccines (rotavirus, live-attenuated influenza, varicella, measles, mumps, and rubella) should be given a minimum of 4 weeks in advance of beginning BRMs. The measles-mumps-rubella (MMR) vaccine can be given on the same day as a TST is administered. If the MMR vaccine is given before administering the TST, the latter should be delayed for 4 to 6 weeks because of temporary suppression of the TST test by the MMR vaccine.145 Inactivated vaccines, polysaccharide vaccines, and recombinant or subunit vaccines and toxoids do not interfere with TST interpretation. The determination of which vaccines are safe to give for patients receiving steroids should be made in concert with an infectious diseases specialist. The Committee on Infectious Diseases of the AAP recommends that children who have diseases such as systemic lupus erythematosus, which are, in themselves, considered to be suppressive of the immune system, and/or who are receiving immunosuppressive medications other than corticosteroids and who are receiving systemic or locally administered corticosteroids should not be given live-virus vaccines except in special circumstances.18 Patients receiving higher doses of steroids (≥2 mg/kg per day of prednisone or its equivalent or ≥20 mg/day for ≥14 days for those weighing >10 kg) should not receive any live-virus vaccines until corticosteroid therapy has been discontinued for at least 1 month.18 

Immunization After a BRM Is Started

If the patient is already receiving a BRM, he or she may still receive any inactivated, polysaccharide, recombinant, or subunit vaccine that is due. There is evidence that adult patients receiving BRM therapy develop a diminished but adequate response to such vaccines, including the annual inactivated influenza immunizations.146,,,149 A possible exception to this may be if the child is receiving rituximab, which has been shown to significantly impair antibody response, especially to pneumococcal vaccines.150,,,,154 These impairments may persist for up to 6 months after the discontinuation of rituximab and highlight the importance of immunizing such patients who are to receive this or other BRMs before they begin BRM therapy. Where there is a test available for a known antibody correlate of protection in patients who are immunized while receiving a BRM, specific postimmunization serum antibody titers can be measured 4 to 6 weeks after immunization to assess immune response and to guide further immunization and management of future exposures.18 All children receiving BRMs should receive annual immunization with inactivated influenza vaccine. Live vaccines, including the live-attenuated influenza vaccines, should not be given to any patients receiving BRMs or other immunosuppressive drugs during treatment because of the risk of dissemination and adverse outcomes, except in special circumstances as recommended by an infectious diseases specialist.18 

Immunization After a BRM is Stopped

Because patients receiving BRMs may have prolonged periods of immunosuppression after the discontinuation of the agent, there are currently no clear data to guide how soon a live vaccine may be administered, and studies of duration of suppression are needed.18 Any consideration for giving a live vaccine after the cessation of a BRM should be made in consultation with an infectious diseases specialist.

Immunizing Immunocompetent Household Contacts of Patients Receiving BRMs

The immunization history of household members of patients receiving BRMs should also be assessed, because they may pose a risk of transmitting vaccine-preventable conditions. Household members should all be up to date with the recommended inactivated vaccines. The principles of vaccination of household contacts of patients receiving BRMs should follow the same principles recommended by the AAP for household contacts of immunocompromised patients.18 All household contacts aged ≥6 months should also receive influenza immunization annually.18 With regard to live vaccines, oral polio vaccine (no longer given in the United States) should not be administered to household contacts of persons receiving BRMs. However, susceptible household contacts can and should receive both MMR and rotavirus vaccines, because the vaccine-strain viruses are rarely transmitted. Similarly, varicella vaccine should be given to susceptible household contacts, because vaccine-strain virus is also rarely transmitted. In the event a household contact of a patient receiving a BRM who is thought to be susceptible to VZV develops a vesicular rash after receiving the varicella vaccine, efforts should be made to separate him or her from the person receiving a BRM for the duration of the rash. However, because the risk of transmission of the vaccine strain is very low and disease associated with the virus is expected to be mild, VariZIG (varicella-zoster immune globulin) is not indicated if contact does occur. For further guidance on immunizing parents and other household contacts in the pediatric office setting, including medico-legal, financial, and logistic considerations, please see the AAP’s technical report “Immunizing Parents and Other Households Contacts in the Pediatric Office Setting” (http://pediatrics.aappublications.org/content/129/1/e247.full; accessed September 10, 2015).

As soon as a patient is diagnosed with a rheumatologic or inflammatory/autoimmune condition for which BRMs may be later needed for treatment, TB screening and appropriate treatment should be initiated155,,157 using the principles outlined as follows:

  • All patients, regardless of specific TB risk factors, who will be taking an immunomodulating biologic agent should be tested for latent TB infection (LTBI) before starting the therapy.

  • There are 2 options that should be considered for screening, depending on the clinical scenario: either TST or use of an interferon-γ release assay (IGRA) or both. There are 2 choices for IGRA, the QuantiFERON-TB Gold In-Tube assay (Cellestis/Qiagen, Carnegie, Australia) and the T-SPOT.TB assay (Oxford Immunotec, Abingdon, United Kingdom). Both are considered acceptable.19,157 

  • For children without risk factors for LTBI (previous history of TB, previous history of positive TST result, history of exposure to someone with active TB, travel to an area with endemic TB in the past 12 months, or foreign-born patient or parents from area with endemic TB) and without any symptoms suggestive of TB, the decision as to which screening method to use at baseline should be based on age. Children younger than 5 years, in general, should be screened with TST, whereas an IGRA is preferred for children older than 5 years.19 

  • For patients with any risk factor for LTBI (previous history of TB, previous history of positive TST result, history of exposure to someone with active TB, or travel to an area with endemic TB in the past 12 months) or any symptoms suggestive of TB, both the TST and an IGRA should be performed and appropriate treatment of LTBI should be started if either test result is positive once TB disease has been ruled out.19 Most experts do not currently use an IGRA when testing for LTBI for children younger than 2 years because of a lack of data for this age group and a high risk of progression to disease but will still use an IGRA in children 2 years or older, especially if they have received a bacille Calmette-Guerin vaccine.19 Although most of the data available are for children 5 years and older, there are increasing data also available for children 2 to 5 years of age.19 

  • Patients with a positive TST or an IGRA result or any risk factor as stated previously for LTBI should also have chest radiography (postanterior and lateral) performed.

  • Routine annual TST or IGRA is not recommended, but patients should be asked at least annually about TB symptoms and risk factors.

  • If there is a change in risk or new symptoms suggestive of TB infection while receiving treatment, there should be further evaluation and risk assessment, including a repeat of both TST and IGRA (if the baseline was negative) as well as chest radiography.

  • If LTBI is diagnosed, the patient needs treatment to prevent TB disease.155,156 

  • Patients suspected of having active TB should have their BRM and other immunosuppressive agents discontinued until TB is ruled out or under control. Guidance from an infectious diseases specialist is recommended in this setting for possible isolation precautions and management. Risk factors for drug-resistant TB (previous history of treatment of TB disease, contact with a patient with known drug-resistant TB, country of origin or current residence in geographic area with a high prevalence of drug-resistant TB, or contact with a source case who has positive smears for acid-fast bacilli or cultures after 2 months of appropriate anti-TB therapy) should be considered in formulating empirical therapy.158 There will need to be an intensive investigation to examine for disseminated or extrapulmonary disease guided by an expert in TB management.

There are no recommendations or tests at the present time for screening for NTM. However, NTM should be considered in the differential diagnosis of patients receiving BRMs with febrile illnesses, cervical or unexplained lymphadenitis, or other focal infections or anytime TB is being considered.

All patients should be queried about possible exposure and epidemiologic risk factors for potentially invasive fungal infections, especially histoplasmosis and coccidioidomycosis, which have symptoms and signs that significantly overlap with TB. The main epidemiologic determinants of risk for histoplasmosis include the following122,124,159,,161: geographic location of residence near river beds (Mississippi River Valley, Ohio River Valley, Chesapeake Bay area, eastern Oklahoma, and eastern Texas). Anyone who lives in a histoplasmosis belt is potentially infected; estimates are that 90% of residents acquire histoplasmosis and self-resolve before adulthood; thus, routine laboratory screening is not indicated. Furthermore, negative serologic test results before the initiation of BRMs does not predict patients at risk of development of histoplasmosis, especially among patients already receiving DMARDs.122 Immunosuppression and extremes of age are also major risk factors. Residence in geographic areas where coccidioidomycosis is endemic (eg, southwestern United States and southern California) is an epidemiologic risk factor for developing coccidioidomycosis,119,120,161,,,164 and Filipino, Hispanic, black, American Indian, and Asian persons and pregnant women during the third trimester and women in the immediate postpartum period are at risk for more severe disease. Any form of immunosuppression also leads to an increased risk of invasive and disseminated disease. Up to 2% of patients receiving BRMs will develop coccidioidomycosis if they reside in an at-risk region.

Patients suspected of having signs or symptoms compatible with acute histoplasmosis122,124,159,160 or coccidioidomycosis119,,121,162,,164 during therapy with BRMs should have the BRM discontinued immediately and require evaluation with a combination of chest radiography and serologic, antigen detection, and culture tests. These tests and treatment options are best conducted in consultation with an infectious diseases expert.

VZV and HSV

As noted previously in the immunization section, all patients should be screened by history, immunization, and possibly laboratory serologic records to facilitate documentation that they are either immune or have received the age-appropriate dose(s) of varicella vaccine at presentation. Routine testing for VZV is not recommended because of the variability in sensitivity of the assays, particularly in assessing immunity after VZV immunization; and where there is doubt about immunity on the basis of history or inadequacy of records, vaccination is recommended. If VZV vaccine is needed, because it is a live-virus vaccine it should be given at least 4 weeks before the initiation of BRM therapy.165 

Patients suspected of having VZV or HSV infection during BRM therapy should stop taking the BRM. Diagnosis should be sought by a combination of clinical, serologic, and polymerase chain reaction tests from skin lesions (ie, vesicles or papules), and acyclovir or valacyclovir therapy should be started pending confirmation of diagnosis. Patients with recurrent HSV who are being considered for BRM therapy should be given consideration for prophylaxis with valacyclovir or acyclovir.

EBV

Consideration should be given to baseline serologic testing for EBV at the time of diagnosis of the underlying rheumatologic condition. Further serologic testing should be considered if the patient develops signs or symptoms that have been associated with EBV, including mononucleosis-like illness or excessive fatigue.

Hepatitis B

Patients being considered for BRM therapy should be screened for past hepatitis B infection with both immunization records and serologic testing for HBsAg, HbcAb (total and IgM), and quantitative HBsAb166 (Table 3). It is advisable to obtain baseline liver function tests (LFTs) at the same time.167,,,,,172 Patients who are negative for HBsAg, HBcAb, and HBsAb are considered uninfected and nonimmune. They should be immunized with the first dose of hepatitis B vaccine at least 2 weeks before initiation of the BRM.173 Consideration of whether the full series of HBV vaccinations is given before initiation of the BRM should be made in conjunction with the specialist initiating the BRM, weighing the risks and benefit of delaying BRM therapy. Patients who are only HBsAb positive are likely immune and can be safely treated with BRMs. If their antibody titer is low (<10 mIU/mL), strong consideration should be given to giving a booster dose of hepatitis B vaccine before initiation of treatment.148,166 Recent receipt of Immune Globulin Intravenous (IGIV) may be associated with a transiently positive serologic test result. Patients who have acute HBV infection (HBsAg positive, total HBcAb positive, hepatitis B core IgM positive, and elevated LFTs) should not receive a BRM. Patients who show evidence of chronic HBV infection (HBsAg positive, HBcAb positive, hepatitis B core IgM negative, and persistently or intermittently high LFT results) should be evaluated by either an infectious diseases specialist or a hepatologist for further testing to determine their status.169 This testing will likely involve measuring HBV DNA levels to determine the likelihood of clearance with antiviral treatment. In general, chronic carriers with a high viral load (HBV DNA >105 copies per mL) and abnormal LFTs will need to be treated with antiviral agents effective against HBV, but guidance by an infectious diseases specialist or hepatologist is strongly recommended. Patients who are HBsAg negative, HBsAb positive, and HBcAb positive are considered to have “resolved HBV infection” and can likely be treated with close monitoring by an infectious diseases expert or hepatologist. In this scenario, it is advisable to first obtain a baseline HBV DNA quantitative level as well as LFTs that can be monitored in follow-up. Finally, some patients will only be positive for HBcAb and are generally considered to have “occult HBV.” Some of these patients may also have a false-positive test result or may have resolving infection. They may or may not be at risk of HBV reactivation. Most can be treated with the BRM under the guidance of an infectious diseases expert or a hepatologist.

TABLE 3

Screening Tests for HBV Infection, Interpretation, and Recommendations Before BRM Therapy

HBV InfectionHBsAgHBsAbHBcAbHBcIgMAbnormal LFTs and/or SymptomsAdditional TestingManagementComments
Uninfected, nonimmune – – – – – Not needed Safely treat with BRM Administer hepatitis B vaccine before BRM therapy 
Vaccinated – – – – Not needed Safely treat with BRM For HBsAb <10 mIU/mL, suggest HBV booster 
Acute – Not needed Defer BRM therapy  
Chronic – – +/– HBeAg, HBeAb, HBV DNA Treat with BRM in consultation with ID consultant or hepatologist  
Resolved – – – HBV DNA Treat with BRM in consultation with ID consultant or hepatologist  
Occult – – – – HBV DNA Treat with BRM in consultation with ID consultant or hepatologist  
HBV InfectionHBsAgHBsAbHBcAbHBcIgMAbnormal LFTs and/or SymptomsAdditional TestingManagementComments
Uninfected, nonimmune – – – – – Not needed Safely treat with BRM Administer hepatitis B vaccine before BRM therapy 
Vaccinated – – – – Not needed Safely treat with BRM For HBsAb <10 mIU/mL, suggest HBV booster 
Acute – Not needed Defer BRM therapy  
Chronic – – +/– HBeAg, HBeAb, HBV DNA Treat with BRM in consultation with ID consultant or hepatologist  
Resolved – – – HBV DNA Treat with BRM in consultation with ID consultant or hepatologist  
Occult – – – – HBV DNA Treat with BRM in consultation with ID consultant or hepatologist  

Adapted with permission from ref 117. HBcIgM, hepatitis B core IgM antibody; HBeAb, hepatitis B core e antibody; HBeAg, hepatitis B core e antigen; ID, infectious diseases; –, negative; +, positive.

Long-term Monitoring for HBV

All patients at risk of HBV reactivation (chronic, resolved, or occult) should have regular (every 1–3 months, depending on underlying HBV infection) monitoring of their LFTs, HBsAg, hepatitis B e antigen, and HBV DNA counts. This monitoring should continue for at least 6 months after termination of the BRM.

PML

All patients receiving natalizumab should be monitored regularly (3 and 6 months after the first infusion and every 6 months after and for at least 6 months after discontinuation) for PML, and the medication should be withheld immediately with any signs or symptoms of the condition.174 Diagnosis of PML involves a gadolinium-enhanced MRI brain scan and cerebrospinal fluid analysis for JC viral DNA when indicated. An algorithm has been proposed for risk profiling and management of PML for patients receiving natalizumab.174 

BRMs have become an important component of effective management of patients with a variety of autoimmune/inflammatory conditions. However, there is an increased risk of certain serious and opportunistic infections for patients receiving these biologic agents, especially the risks of TB and viral infections. It is important for pediatricians, family physicians, and other primary care practitioners managing patients who receive these important treatments to be aware of the potential infections complications and to practice anticipatory guidance between visits to reduce the risk of occurrence or of negative outcomes if the complications do occur. In most scenarios, a close working relationship among the pediatrician, the pediatric medical subspecialist prescribing the BRM, and an infectious diseases specialist is warranted.

AAP

American Academy of Pediatrics

AERS

Adverse Event Reporting System

BRM

biologic response modifier

CI

confidence interval

DMARD

disease-modifying antirheumatic drug

EBV

Epstein-Barr virus

Fc

fragment-crystallizable

FDA

Food and Drug Administration

HBcAb

hepatitis B core antibody

HBsAb

hepatitis B surface antibody

HBsAg

hepatitis B surface antigen

HBV

hepatitis B virus

HSCT

hematopoietic stem cell transplant

HSV

herpes simplex virus

Ig

immunoglobulin

IGRA

interferon-γ release assay

IL

interleukin

JAK

Janus kinase

JIA

juvenile idiopathic arthritis

LFT

liver function test

LTBI

latent tuberculosis infection

MMR

measles-mumps-rubella

NTM

nontuberculous mycobacteria

OR

odds ratio

PCP

Pneumocystis jirovecii (previously carinii) pneumonia

PML

progressive multifocal leukoencephalopathy

RA

rheumatoid arthritis

TB

tuberculosis

Th

T helper

TNF

tumor necrosis factor

TST

tuberculin skin test

VZV

varicella-zoster virus

This document is copyrighted and is property of the American Academy of Pediatrics and its Board of Directors. All authors have filed conflict of interest statements with the American Academy of Pediatrics. Any conflicts have been resolved through a process approved by the Board of Directors. The American Academy of Pediatrics has neither solicited nor accepted any commercial involvement in the development of the content of this publication.

Clinical reports from the American Academy of Pediatrics benefit from expertise and resources of liaisons and internal (AAP) and external reviewers. However, clinical reports from the American Academy of Pediatrics may not reflect the views of the liaisons or the organizations or government agencies that they represent.

The guidance in this report does not indicate an exclusive course of treatment or serve as a standard of medical care. Variations, taking into account individual circumstances, may be appropriate.

All clinical reports from the American Academy of Pediatrics automatically expire 5 years after publication unless reaffirmed, revised, or retired at or before that time.

FUNDING: No external funding.

1
Saag
KG
,
Teng
GG
,
Patkar
NM
, et al;
American College of Rheumatology
.
American College of Rheumatology 2008 recommendations for the use of nonbiologic and biologic disease-modifying antirheumatic drugs in rheumatoid arthritis.
Arthritis Rheum
.
2008
;
59
(
6
):
762
784
[PubMed]
2
Toussi
SS
,
Pan
N
,
Walters
HM
,
Walsh
TJ
.
Infections in children and adolescents with juvenile idiopathic arthritis and inflammatory bowel disease treated with tumor necrosis factor-α inhibitors: systematic review of the literature.
Clin Infect Dis
.
2013
;
57
(
9
):
1318
1330
[PubMed]
3
Giles
JT
,
Bathon
JM
.
Serious infections associated with anticytokine therapies in the rheumatic diseases.
J Intensive Care Med
.
2004
;
19
(
6
):
320
334
[PubMed]
4
Keane
J
,
Gershon
S
,
Wise
RP
, et al
.
Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent.
N Engl J Med
.
2001
;
345
(
15
):
1098
1104
[PubMed]
5
Ford
AC
,
Peyrin-Biroulet
L
.
Opportunistic infections with anti-tumor necrosis factor-α therapy in inflammatory bowel disease: meta-analysis of randomized controlled trials.
Am J Gastroenterol
.
2013
;
108
(
8
):
1268
1276
[PubMed]
6
Lichtenstein
GR
,
Feagan
BG
,
Cohen
RD
, et al
.
Serious infections and mortality in association with therapies for Crohn’s disease: TREAT registry.
Clin Gastroenterol Hepatol
.
2006
;
4
(
5
):
621
630
[PubMed]
7
Rahier
JF
,
Ben-Horin
S
,
Chowers
Y
, et al;
European Crohn’s and Colitis Organisation
.
European evidence-based consensus on the prevention, diagnosis and management of opportunistic infections in inflammatory bowel disease.
J Crohn’s Colitis
.
2009
;
3
(
2
):
47
91
[PubMed]
8
Rahier
JF
,
Yazdanpanah
Y
,
Colombel
JF
,
Travis
S
.
The European (ECCO) consensus on infection in IBD: what does it change for the clinician?
Gut
.
2009
;
58
(
10
):
1313
1315
[PubMed]
9
Viget
N
,
Vernier-Massouille
G
,
Salmon-Ceron
D
,
Yazdanpanah
Y
,
Colombel
JF
.
Opportunistic infections in patients with inflammatory bowel disease: prevention and diagnosis.
Gut
.
2008
;
57
(
4
):
549
558
[PubMed]
10
Beukelman
T
,
Patkar
NM
,
Saag
KG
, et al
.
2011 American College of Rheumatology recommendations for the treatment of juvenile idiopathic arthritis: initiation and safety monitoring of therapeutic agents for the treatment of arthritis and systemic features.
Arthritis Care Res (Hoboken)
.
2011
;
63
(
4
):
465
482
[PubMed]
11
Beukelman
T
,
Xie
F
,
Baddley
JW
, et al;
SABER Collaboration
.
Brief report: incidence of selected opportunistic infections among children with juvenile idiopathic arthritis.
Arthritis Rheum
.
2013
;
65
(
5
):
1384
1389
[PubMed]
12
Hyams
J
,
Walters
TD
,
Crandall
W
, et al
.
Safety and efficacy of maintenance infliximab therapy for moderate-to-severe Crohn’s disease in children: REACH open-label extension.
Curr Med Res Opin
.
2011
;
27
(
3
):
651
662
[PubMed]
13
Le Saux
N
;
Canadian Paediatric Society, Infectious Diseases and Immunization Committee
.
Biologic response modifiers to decrease inflammation: focus on infection risks.
Paediatr Child Health
.
2012
;
17
(
3
):
147
154
[PubMed]
14
Veereman-Wauters
G
,
de Ridder
L
,
Veres
G
, et al;
ESPGHAN IBD Porto Group
.
Risk of infection and prevention in pediatric patients with IBD: ESPGHAN IBD Porto Group commentary.
J Pediatr Gastroenterol Nutr
.
2012
;
54
(
6
):
830
837
[PubMed]
15
Woerner
A
,
Ritz
N
.
Infections in children treated with biological agents.
Pediatr Infect Dis J
.
2013
;
32
(
3
):
284
288
[PubMed]
16
Centers for Disease Control and Prevention, Advisory Committee on Immunization Practices;
.
Recommended immunization schedules for persons aged 0 through 18 years—United States, 2013.
MMWR Morb Mortal Wkly Rep
.
2013
;
62
(
1
):
2
8
17
Centers for Disease Control and Prevention, Advisory Committee on Immunization Practices
;
American Academy of Pediatrics
;
American Academy of Family Physicians
.
Recommended immunization schedules for persons aged 0 through 18 years--United States, 2013.
MMWR Morb Mortal Wkly Rep
.
2013
;
62
(
1
):
1
19
18
American Academy of Pediatrics
.
Immunization in special clinical circumstances
. In:
Pickering
LK
,
Baker
CJ
,
Kimberlin
DW
,
Long
SS
, eds.
Red Book: 2012 Report of the Committee on Infectious Diseases
.
Elk Grove Village, IL
:
American Academy of Pediatrics
;
2012
:
69
109
19
Starke
JR
;
Committee on Infectious Diseases
.
Interferon-γ release assays for diagnosis of tuberculosis infection and disease in children.
Pediatrics
.
2014
;
134
(
6
). Available at: www.pediatrics.org/cgi/content/full/134/6/e1763
[PubMed]
20
American Academy of Pediatrics
.
Listeria monocytogenes infections (listeriosis)
. In:
Pickering
LK
,
Baker
CJ
,
Kimberlin
DW
,
Long
SS
, eds.
Red Book: 2012 Report of the Committee on Infectious Diseases
.
Elk Grove Village, IL
:
American Academy of Pediatrics
;
2012
:
471
474
21
Remicade (infliximab) [package insert]. Horsham, PA: Janssen Biotech;
2013
. Available at: www.remicade.com/shared/product/remicade/prescribing-information.pdf. Accessed October 22, 2014
22
Horneff
G
,
De Bock
F
,
Foeldvari
I
, et al;
German and Austrian Paediatric Rheumatology Collaborative Study Group
.
Safety and efficacy of combination of etanercept and methotrexate compared to treatment with etanercept only in patients with juvenile idiopathic arthritis (JIA): preliminary data from the German JIA Registry.
Ann Rheum Dis
.
2009
;
68
(
4
):
519
525
[PubMed]
23
Humira (adalimunab) [package insert]. North Chicago, IL: AbbVie;
2013
. Available at: www.rxabbvie.com/pdf/humira.pdf. Accessed October 22, 2014
24
Simponi (golimumab) [package insert]. Horsham, PA: Jannsen Biotech;
2013
. Available at: www.simponiaria.com/shared/product/simponiaria/prescribing-information.pdf. Accessed October 22, 2014
25
Cimzia (certolizumab pegol) [package insert]. Smyrna, GA: UCB;
2012
. Available at: www.cimzia.com/assets/pdf/Prescribing_Information.pdf. Accessed October 22, 2014
26
Orencia (abatacept) [package insert]. Princeton, NJ: Bristol Myers Squibb;
2013
. Available at: http://packageinserts.bms.com/pi/pi_orencia.pdf. Accessed October 22, 2014
27
Kineret (anakinra) [package insert]. Stockholm, Sweden: Swedish Orphan Biovitrum AB;
2013
. Available at: www.kineretrx.com/fileadmin/user_upload/kineretus/documents/Kineret_Full_Prescribing_Information.pdf. Accessed October 22, 2014
28
Rituxan (rituximab) [package insert]. South San Francisco, CA: Genentech;
2013
. Available at: www.gene.com/download/pdf/rituxan_prescribing.pdf. Accessed October 22, 2014
29
Actemra (tocilizumab) [package insert]. South San Francisco, CA: Genentech;
2013
. Available at: www.gene.com/download/pdf/actemra_prescribing.pdf. Accessed October 22, 2014
30
Stelara (ustekinumab) [package insert]. Horsham, PA: Janssen Biotech;
2013
. Available at: www.stelarainfo.com/pdf/PrescribingInformation.pdf. Accessed October 22, 2014
31
Ruperto
N
,
Lovell
DJ
,
Cuttica
R
, et al;
Paediatric Rheumatology International Trials Organisation
;
Pediatric Rheumatology Collaborative Study Group
.
A randomized, placebo-controlled trial of infliximab plus methotrexate for the treatment of polyarticular-course juvenile rheumatoid arthritis.
Arthritis Rheum
.
2007
;
56
(
9
):
3096
3106
[PubMed]
32
Ilaris (canakinumab) [package insert]. East Hanover, NJ: Novartis;
2013
. Available at: www.pharma.us.novartis.com/product/pi/pdf/ilaris.pdf. Accessed October 22, 2014
33
Tysabri (natalizumab) [package insert]. Cambridge, MA: Biogen Idec;
2013
. Available at: www.tysabri.com/pdfs/I61061-13_PI.pdf. Accessed October 22, 2014
34
Benlysta (belimumab) [package insert]. GlaxoSmithKline, Mississauga, Ontatrio, Canada;
2014
. Available at: www.gsksource.com/gskprm/en/US/adirect/gskprm?cmd=ProductDetailPage&product_id=1300455676143&featureKey=602522&tab=tab1&footer=yes&pTab=one#nlmhighlights. Accessed October 22, 2014
35
Arcalyst (rilonacept) [package insert]. Tarrytown, NY: Regeneron Pharmaceuticals;
2013
. Available at: www.arcalyst.com/images/pdf/ARCALYST-fpi.pdf?phpMyAdmin=e99e09ae33c1fdbbafd69c8883f97963. Accessed October 22, 2014
36
Xeljanz (tofacitinib) [package insert]. New York, NY: Pfizer;
2014
. Available at: http://labeling.pfizer.com/ShowLabeling.aspx?id=959. Accessed October 22, 2014
37
Bray
V
.
The safety of infliximab (Remicade(R)) infusion in clinical practice
[abstract 153]
.
Arthritis Rheum
.
2001
;(
suppl 9
):
S83
38
Hyams
JS
,
Griffiths
A
,
Markowitz
J
, et al
Safety and efficacy of adalimumab for moderate to severe Crohn's disease in children.
Gastroenterology
.
2012
;143(2):365.e2–374.e2
39
Lipsky
PE
,
van der Heijde
DM
,
St Clair
EW
, et al;
Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group
.
Infliximab and methotrexate in the treatment of rheumatoid arthritis.
N Engl J Med
.
2000
;
343
(
22
):
1594
1602
[PubMed]
40
Furst
D
,
Schiff
M
,
Fleischmann
R
, et al
.
Safety and efficacy of adalimumab (D2E7), a fully human anti-TNF-alpha monoclonal antibody, given in combination with standard antirheumatic therapy: safety trial of adalimumab in rheumatoid arthritis (RA)
[abstract]
.
Arthritis Rheum
.
2002
;
46
(
suppl
):
S572
41
Keystone
E
,
Kavanaugh
A
,
Sharp
J
, et al
.
Adalimumab (D2E7), a fully human anti-TNF-alpha monoclonal antibody, inhibits the progression of structural joint damage in patients with active RA despite concomitant methotrexate therapy
[abstract]
.
Arthritis Rheum
.
2002
;
46
(
suppl 9
):
S205
42
Lovell
DJ
,
Ruperto
N
,
Goodman
S
, et al;
Pediatric Rheumatology Collaborative Study Group
;
Pediatric Rheumatology International Trials Organisation
.
Adalimumab with or without methotrexate in juvenile rheumatoid arthritis.
N Engl J Med
.
2008
;
359
(
8
):
810
820
[PubMed]
43
Trachana
M
,
Pratsidou-Gertsi
P
,
Pardalos
G
,
Kozeis
N
,
Badouraki
M
,
Kanakoudi-Tsakalidou
F
.
Safety and efficacy of adalimumab treatment in Greek children with juvenile idiopathic arthritis.
Scand J Rheumatol
.
2011
;
40
(
2
):
101
107
[PubMed]
44
Hsia
EC
,
Cush
JJ
,
Matteson
EL
, et al
.
Comprehensive tuberculosis screening program in patients with inflammatory arthritides treated with golimumab, a human anti-tumor necrosis factor antibody, in phase III clinical trials.
Arthritis Care Res (Hoboken)
.
2013
;
65
(
2
):
309
313
[PubMed]
45
Pérez-Alvarez
R
,
Díaz-Lagares
C
,
García-Hernández
F
, et al;
BIOGEAS Study Group
.
Hepatitis B virus (HBV) reactivation in patients receiving tumor necrosis factor (TNF)-targeted therapy: analysis of 257 cases.
Medicine (Baltimore)
.
2011
;
90
(
6
):
359
371
[PubMed]
46
Giannini
EH
,
Ilowite
NT
,
Lovell
DJ
, et al;
Pediatric Rheumatology Collaborative Study Group
.
Long-term safety and effectiveness of etanercept in children with selected categories of juvenile idiopathic arthritis.
Arthritis Rheum
.
2009
;
60
(
9
):
2794
2804
[PubMed]
47
Enbrel (etanercept) [package insert]. Thousand Oaks, CA: Immunex;
2013
. Available at: http://pi.amgen.com/united_states/enbrel/derm/enbrel_pi.pdf. Accessed October 22, 2014
48
Wallis
RS
.
Biologics and infections: lessons from tumor necrosis factor blocking agents.
Infect Dis Clin North Am
.
2011
;
25
(
4
):
895
910
[PubMed]
49
Cutolo
M
.
The kinase inhibitor tofacitinib in patients with rheumatoid arthritis: latest findings and clinical potential.
Ther Adv Musculoskelet Dis
.
2013
;
5
(
1
):
3
11
[PubMed]
50
Ghoreschi
K
,
Laurence
A
,
O’Shea
JJ
.
Selectivity and therapeutic inhibition of kinases: to be or not to be?
Nat Immunol
.
2009
;
10
(
4
):
356
360
[PubMed]
51
Ghoreschi
K
,
Laurence
A
,
O’Shea
JJ
.
Janus kinases in immune cell signaling.
Immunol Rev
.
2009
;
228
(
1
):
273
287
[PubMed]
52
Riese
RJ
,
Krishnaswami
S
,
Kremer
J
.
Inhibition of JAK kinases in patients with rheumatoid arthritis: scientific rationale and clinical outcomes.
Best Pract Res Clin Rheumatol
.
2010
;
24
(
4
):
513
526
[PubMed]
53
Bongartz
T
,
Sutton
AJ
,
Sweeting
MJ
,
Buchan
I
,
Matteson
EL
,
Montori
V
.
Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials.
JAMA
.
2006
;
295
(
19
):
2275
2285
[PubMed]
54
Thompson
AE
,
Rieder
SW
,
Pope
JE
.
Tumor necrosis factor therapy and the risk of serious infection and malignancy in patients with early rheumatoid arthritis: a meta-analysis of randomized controlled trials.
Arthritis Rheum
.
2011
;
63
(
6
):
1479
1485
[PubMed]
55
Singh
JA
,
Wells
GA
,
Christensen
R
, et al
.
Adverse effects of biologics: a network meta-analysis and Cochrane overview.
Cochrane Database Syst Rev
.
2011
;
2
:
CD008794
[PubMed]
56
Doran
MF
,
Crowson
CS
,
Pond
GR
,
O’Fallon
WM
,
Gabriel
SE
.
Predictors of infection in rheumatoid arthritis.
Arthritis Rheum
.
2002
;
46
(
9
):
2294
2300
[PubMed]
57
Jeurissen
ME
,
Boerbooms
AM
,
van de Putte
LB
, et al
.
Methotrexate versus azathioprine in the treatment of rheumatoid arthritis: a forty-eight-week randomized, double-blind trial.
Arthritis Rheum
.
1991
;
34
(
8
):
961
972
[PubMed]
58
Immunex Corporation.
Enbrel: serious and opportunistic infection rates
.
Thousand Oaks, CA
:
Immunex
;
2013
. Available at: www.enbrel.com/RheumPro/infection-rates.jspx. Accessed October 22, 2014
59
Beukelman
T
,
Xie
F
,
Chen
L
, et al;
SABER Collaboration
.
Rates of hospitalized bacterial infection associated with juvenile idiopathic arthritis and its treatment.
Arthritis Rheum
.
2012
;
64
(
8
):
2773
2780
[PubMed]
60
Bracaglia
C
,
Buonuomo
PS
,
Tozzi
AE
, et al
.
Safety and efficacy of etanercept in a cohort of patients with juvenile idiopathic arthritis under 4 years of age.
J Rheumatol
.
2012
;
39
(
6
):
1287
1290
[PubMed]
61
Gerloni
V
,
Pontikaki
I
,
Gattinara
M
,
Fantini
F
.
Focus on adverse events of tumour necrosis factor alpha blockade in juvenile idiopathic arthritis in an open monocentric long-term prospective study of 163 patients.
Ann Rheum Dis
.
2008
;
67
(
8
):
1145
1152
[PubMed]
62
Lovell
DJ
,
Giannini
EH
,
Reiff
A
, et al;
Pediatric Rheumatology Collaborative Study Group
.
Etanercept in children with polyarticular juvenile rheumatoid arthritis.
N Engl J Med
.
2000
;
342
(
11
):
763
769
[PubMed]
63
Lovell
DJ
,
Giannini
EH
,
Reiff
A
, et al;
Pediatric Rheumatology Collaborative Study Group
.
Long-term efficacy and safety of etanercept in children with polyarticular-course juvenile rheumatoid arthritis: interim results from an ongoing multicenter, open-label, extended-treatment trial.
Arthritis Rheum
.
2003
;
48
(
1
):
218
226
[PubMed]
64
Lovell
DJ
,
Reiff
A
,
Ilowite
NT
, et al;
Pediatric Rheumatology Collaborative Study Group
.
Safety and efficacy of up to eight years of continuous etanercept therapy in patients with juvenile rheumatoid arthritis.
Arthritis Rheum
.
2008
;
58
(
5
):
1496
1504
[PubMed]
65
Lovell
DJ
,
Reiff
A
,
Jones
OY
, et al;
Pediatric Rheumatology Collaborative Study Group
.
Long-term safety and efficacy of etanercept in children with polyarticular-course juvenile rheumatoid arthritis.
Arthritis Rheum
.
2006
;
54
(
6
):
1987
1994
[PubMed]
66
Prince
FH
,
Twilt
M
,
ten Cate
R
, et al
.
Long-term follow-up on effectiveness and safety of etanercept in juvenile idiopathic arthritis: the Dutch national register.
Ann Rheum Dis
.
2009
;
68
(
5
):
635
641
[PubMed]
67
Hyams
J
,
Damaraju
L
,
Blank
M
, et al;
T72 Study Group
.
Induction and maintenance therapy with infliximab for children with moderate to severe ulcerative colitis.
Clin Gastroenterol Hepatol
.
2012
;
10
(
4
):
391
–39
9.e1
[PubMed]
68
Ruperto
N
,
Lovell
DJ
,
Cuttica
R
, et al;
Paediatric Rheumatology INternational Trials Organization (PRINTO)
;
Pediatric Rheumatology Collaborative Study Group (PRCSG)
.
Long-term efficacy and safety of infliximab plus methotrexate for the treatment of polyarticular-course juvenile rheumatoid arthritis: findings from an open-label treatment extension.
Ann Rheum Dis
.
2010
;
69
(
4
):
718
722
[PubMed]
69
Stephens
MC
,
Shepanski
MA
,
Mamula
P
,
Markowitz
JE
,
Brown
KA
,
Baldassano
RN
.
Safety and steroid-sparing experience using infliximab for Crohn’s disease at a pediatric inflammatory bowel disease center.
Am J Gastroenterol
.
2003
;
98
(
1
):
104
111
[PubMed]
70
Tschudy
J
,
Michail
S
.
Disseminated histoplasmosis and pneumocystis pneumonia in a child with Crohn disease receiving infliximab.
J Pediatr Gastroenterol Nutr
.
2010
;
51
(
2
):
221
222
[PubMed]
71
Imagawa
T
,
Takei
S
,
Umebayashi
H
, et al
.
Efficacy, pharmacokinetics, and safety of adalimumab in pediatric patients with juvenile idiopathic arthritis in Japan.
Clin Rheumatol
.
2012
;
31
(
12
):
1713
1721
[PubMed]
72
Chaiwatanatorn
K
,
Lee
N
,
Grigg
A
,
Filshie
R
,
Firkin
F
.
Delayed-onset neutropenia associated with rituximab therapy.
Br J Haematol
.
2003
;
121
(
6
):
913
918
[PubMed]
73
Dunleavy
K
,
Hakim
F
,
Kim
HK
, et al
.
B-cell recovery following rituximab-based therapy is associated with perturbations in stromal derived factor-1 and granulocyte homeostasis.
Blood
.
2005
;
106
(
3
):
795
802
[PubMed]
74
Fukuno
K
,
Tsurumi
H
,
Ando
N
, et al
.
Late-onset neutropenia in patients treated with rituximab for non-Hodgkin’s lymphoma.
Int J Hematol
.
2006
;
84
(
3
):
242
247
[PubMed]
75
Hofer
S
,
Viollier
R
,
Ludwig
C
.
Delayed-onset and long-lasting severe neutropenia due to rituximab.
Swiss Med Wkly
.
2004
;
134
(
5-6
):
79
80
[PubMed]
76
Mitsuhata
N
,
Fujita
R
,
Ito
S
,
Mannami
M
,
Keimei
K
.
Delayed-onset neutropenia in a patient receiving rituximab as treatment for refractory kidney transplantation.
Transplantation
.
2005
;
80
(
9
):
1355
[PubMed]
77
Nitta
E
,
Izutsu
K
,
Sato
T
, et al
.
A high incidence of late-onset neutropenia following rituximab-containing chemotherapy as a primary treatment of CD20-positive B-cell lymphoma: a single-institution study.
Ann Oncol
.
2007
;
18
(
2
):
364
369
[PubMed]
78
Terrier
B
,
Ittah
M
,
Tourneur
L
, et al
.
Late-onset neutropenia following rituximab results from a hematopoietic lineage competition due to an excessive BAFF-induced B-cell recovery.
Haematologica
.
2007
;
92
(
2
):
e20
e23
[PubMed]
79
Voog
E
,
Morschhauser
F
,
Solal-Céligny
P
.
Neutropenia in patients treated with rituximab.
N Engl J Med
.
2003
;
348
(
26
):
2691
2694; discussion: 2691–2694
[PubMed]
80
Weissmann-Brenner
A
,
Brenner
B
,
Belyaeva
I
,
Lahav
M
,
Rabizadeh
E
.
Rituximab associated neutropenia: description of three cases and an insight into the underlying pathogenesis.
Med Sci Monit
.
2011
;
17
(
11
):
CS133
CS137
[PubMed]
81
Alexeeva
EI
,
Valieva
SI
,
Bzarova
TM
, et al
.
Efficacy and safety of repeat courses of rituximab treatment in patients with severe refractory juvenile idiopathic arthritis.
Clin Rheumatol
.
2011
;
30
(
9
):
1163
1172
[PubMed]
82
Flynn
JL
,
Goldstein
MM
,
Chan
J
, et al
.
Tumor necrosis factor-alpha is required in the protective immune response against Mycobacterium tuberculosis in mice.
Immunity
.
1995
;
2
(
6
):
561
572
[PubMed]
83
Wagner
TE
,
Huseby
ES
,
Huseby
JS
.
Exacerbation of Mycobacterium tuberculosis enteritis masquerading as Crohn’s disease after treatment with a tumor necrosis factor-alpha inhibitor.
Am J Med
.
2002
;
112
(
1
):
67
69
[PubMed]
84
Fallahi-Sichani
M
,
Schaller
MA
,
Kirschner
DE
,
Kunkel
SL
,
Linderman
JJ
.
Identification of key processes that control tumor necrosis factor availability in a tuberculosis granuloma.
PLOS Comput Biol
.
2010
;
6
(
5
):
e1000778
[PubMed]
85
Marino
S
,
Myers
A
,
Flynn
JL
,
Kirschner
DE
.
TNF and IL-10 are major factors in modulation of the phagocytic cell environment in lung and lymph node in tuberculosis: a next-generation two-compartmental model.
J Theor Biol
.
2010
;
265
(
4
):
586
598
[PubMed]
86
Mata-Espinosa
DA
,
Mendoza-Rodríguez
V
,
Aguilar-León
D
,
Rosales
R
,
López-Casillas
F
,
Hernández-Pando
R
.
Therapeutic effect of recombinant adenovirus encoding interferon-gamma in a murine model of progressive pulmonary tuberculosis.
Mol Ther
.
2008
;
16
(
6
):
1065
1072
[PubMed]
87
Salgado
E
,
Gómez-Reino
JJ
.
The risk of tuberculosis in patients treated with TNF antagonists.
Expert Rev Clin Immunol
.
2011
;
7
(
3
):
329
340
[PubMed]
88
Mohan
AK
,
Coté
TR
,
Block
JA
,
Manadan
AM
,
Siegel
JN
,
Braun
MM
.
Tuberculosis following the use of etanercept, a tumor necrosis factor inhibitor.
Clin Infect Dis
.
2004
;
39
(
3
):
295
299
[PubMed]
89
Dixon
WG
,
Hyrich
KL
,
Watson
KD
, et al;
BSRBR Control Centre Consortium
;
BSR Biologics Register
.
Drug-specific risk of tuberculosis in patients with rheumatoid arthritis treated with anti-TNF therapy: results from the British Society for Rheumatology Biologics Register (BSRBR).
Ann Rheum Dis
.
2010
;
69
(
3
):
522
528
[PubMed]
90
Fleischmann
R
,
Yocum
D
.
Does safety make a difference in selecting the right TNF antagonist?
Arthritis Res Ther
.
2004
;
6
(
suppl 2
):
S12
S18
[PubMed]
91
Dye
C
,
Scheele
S
,
Dolin
P
,
Pathania
V
,
Raviglione
MC
.
Global burden of tuberculosis: estimated incidence, prevalence, and mortality by country
[consensus statement]. WHO Global Surveillance and Monitoring Project.
JAMA
.
1999
;
282
(
7
):
677
686
[PubMed]
92
Dixon
WG
,
Watson
K
,
Lunt
M
,
Hyrich
KL
,
Silman
AJ
,
Symmons
DP
;
British Society for Rheumatology Biologics Register
.
Rates of serious infection, including site-specific and bacterial intracellular infection, in rheumatoid arthritis patients receiving anti-tumor necrosis factor therapy: results from the British Society for Rheumatology Biologics Register.
Arthritis Rheum
.
2006
;
54
(
8
):
2368
2376
[PubMed]
93
Tubach
F
,
Salmon
D
,
Ravaud
P
, et al;
Research Axed on Tolerance of Biotherapies Group
.
Risk of tuberculosis is higher with anti-tumor necrosis factor monoclonal antibody therapy than with soluble tumor necrosis factor receptor therapy: the three-year prospective French Research Axed on Tolerance of Biotherapies Registry
[published correction appears in
Arthritis Rheum
. 2009;60(8):2540.
Arthritis Rheum
.
2009
;
60
(
7
):
1884
1894
[PubMed]
94
Imperato
AK
,
Smiles
S
,
Abramson
SB
.
Long-term risks associated with biologic response modifiers used in rheumatic diseases.
Curr Opin Rheumatol
.
2004
;
16
(
3
):
199
205
[PubMed]
95
Gómez-Reino
JJ
,
Carmona
L
,
Angel Descalzo
M
;
Biobadaser Group
.
Risk of tuberculosis in patients treated with tumor necrosis factor antagonists due to incomplete prevention of reactivation of latent infection.
Arthritis Rheum
.
2007
;
57
(
5
):
756
761
[PubMed]
96
Myers
A
,
Clark
J
,
Foster
H
.
Tuberculosis and treatment with infliximab.
N Engl J Med
.
2002
;
346
(
8
):
623
626
[PubMed]
97
Armbrust
W
,
Kamphuis
SSM
,
Wolfs
TWF
, et al
.
Tuberculosis in a nine-year-old girl treated with infliximab for systemic juvenile idiopathic arthritis.
Rheumatology (Oxford)
.
2004
;
43
(
4
):
527
529
[PubMed]
98
Askling
J
,
Fored
CM
,
Brandt
L
, et al
.
Risk and case characteristics of tuberculosis in rheumatoid arthritis associated with tumor necrosis factor antagonists in Sweden.
Arthritis Rheum
.
2005
;
52
(
7
):
1986
1992
[PubMed]
99
Ayaz
NA
,
Demirkaya
E
,
Bilginer
Y
, et al
.
Preventing tuberculosis in children receiving anti-TNF treatment.
Clin Rheumatol
.
2010
;
29
(
4
):
389
392
[PubMed]
100
Winthrop
KL
,
Daley
CL
,
Griffith
D
.
Nontuberuclous mycobacterial disease: updated diagnostic criteria for an under-recognized infectious complication of anti-tumor necrosis factor therapy.
Nat Clin Pract Rheumatol
.
2007
;
3
(
10
):
E1
[PubMed]
101
Winthrop
KL
,
Yamashita
S
,
Beekmann
SE
,
Polgreen
PM
;
Infectious Diseases Society of America Emerging Infections Network
.
Mycobacterial and other serious infections in patients receiving anti-tumor necrosis factor and other newly approved biologic therapies: case finding through the Emerging Infections Network.
Clin Infect Dis
.
2008
;
46
(
11
):
1738
1740
[PubMed]
102
Winthrop
KL
,
Chang
E
,
Yamashita
S
,
Iademarco
MF
,
LoBue
PA
.
Nontuberculous mycobacteria infections and anti-tumor necrosis factor-alpha therapy.
Emerg Infect Dis
.
2009
;
15
(
10
):
1556
1561
[PubMed]
103
Cullen
G
,
Baden
RP
,
Cheifetz
AS
.
Varicella zoster virus infection in inflammatory bowel disease.
Inflamm Bowel Dis
.
2012
;
18
(
12
):
2392
2403
[PubMed]
104
Strangfeld
A
,
Listing
J
,
Herzer
P
, et al
.
Risk of herpes zoster in patients with rheumatoid arthritis treated with anti-TNF-alpha agents.
JAMA
.
2009
;
301
(
7
):
737
744
[PubMed]
105
Smitten
AL
,
Choi
HK
,
Hochberg
MC
, et al
.
The risk of herpes zoster in patients with rheumatoid arthritis in the United States and the United Kingdom.
Arthritis Rheum
.
2007
;
57
(
8
):
1431
1438
[PubMed]
106
Galloway
JB
,
Mercer
LK
,
Moseley
A
, et al
.
Risk of skin and soft tissue infections (including shingles) in patients exposed to anti-tumour necrosis factor therapy: results from the British Society for Rheumatology Biologics Register.
Ann Rheum Dis
.
2013
;
72
(
2
):
229
234
[PubMed]
107
Winthrop
KL
,
Baddley
JW
,
Chen
L
, et al
.
Association between the initiation of anti-tumor necrosis factor therapy and the risk of herpes zoster.
JAMA
.
2013
;
309
(
9
):
887
895
[PubMed]
108
Wolfe
F
,
Michaud
K
,
Chakravarty
EF
.
Rates and predictors of herpes zoster in patients with rheumatoid arthritis and non-inflammatory musculoskeletal disorders.
Rheumatology (Oxford)
.
2006
;
45
(
11
):
1370
1375
[PubMed]
109
Bradford
RD
,
Pettit
AC
,
Wright
PW
, et al
.
Herpes simplex encephalitis during treatment with tumor necrosis factor-alpha inhibitors.
Clin Infect Dis
.
2009
;
49
(
6
):
924
927
[PubMed]
110
Justice
EA
,
Khan
SY
,
Logan
S
,
Jobanputra
P
.
Disseminated cutaneous herpes simplex virus-1 in a woman with rheumatoid arthritis receiving infliximab: a case report.
J Med Case Reports
.
2008
;
2
:
282
[PubMed]
111
Salmon-Ceron
D
,
Tubach
F
,
Lortholary
O
, et al;
RATIO Group
.
Drug-specific risk of non-tuberculosis opportunistic infections in patients receiving anti-TNF therapy reported to the 3-year prospective French RATIO registry.
Ann Rheum Dis
.
2011
;
70
(
4
):
616
623
[PubMed]
112
Sciaudone
G
,
Pellino
G
,
Guadagni
I
,
Selvaggi
F
.
Education and imaging: gastrointestinal: herpes simplex virus-associated erythema multiforme (HAEM) during infliximab treatment for ulcerative colitis.
J Gastroenterol Hepatol
.
2011
;
26
(
3
):
610
[PubMed]
113
van der Klooster
JM
,
Bosman
RJ
,
Oudemans-van Straaten
HM
,
van der Spoel
JI
,
Wester
JP
,
Zandstra
DF
.
Disseminated tuberculosis, pulmonary aspergillosis and cutaneous herpes simplex infection in a patient with infliximab and methotrexate.
Intensive Care Med
.
2003
;
29
(
12
):
2327
2329
[PubMed]
114
Checchin
D
,
Buda
A
,
Sgarabotto
D
,
Sturniolo
GC
,
D’Incà
R
.
Successful prophylaxis with valaciclovir for relapsing HSV-1 in a girl treated with infliximab for moderate Crohn’s disease.
Eur J Gastroenterol Hepatol
.
2009
;
21
(
9
):
1095
1096
[PubMed]
115
Carroll
MB
.
The impact of biologic response modifiers on hepatitis B virus infection.
Expert Opin Biol Ther
.
2011
;
11
(
4
):
533
544
[PubMed]
116
Zachou
K
,
Sarantopoulos
A
,
Gatselis
NK
, et al
.
Hepatitis B virus reactivation in hepatitis B virus surface antigen negative patients receiving immunosuppression: a hidden threat.
World J Hepatol
.
2013
;
5
(
7
):
387
392
[PubMed]
117
Motaparthi
K
,
Stanisic
V
,
Van Voorhees
AS
,
Lebwohl
MG
,
Hsu
S
;
Medical Board of the National Psoriasis Foundation
.
From the Medical Board of the National Psoriasis Foundation: recommendations for screening for hepatitis B infection prior to initiating anti-tumor necrosis factor-alfa inhibitors or other immunosuppressive agents in patients with psoriasis.
J Am Acad Dermatol
.
2014
;
70
(
1
):
178
186
[PubMed]
118
Tsiodras
S
,
Samonis
G
,
Boumpas
DT
,
Kontoyiannis
DP
.
Fungal infections complicating tumor necrosis factor alpha blockade therapy.
Mayo Clin Proc
.
2008
;
83
(
2
):
181
194
[PubMed]
119
Ampel
NM
.
Coccidioidomycosis: a review of recent advances.
Clin Chest Med
.
2009
;
30
(
2
):
241
251
[PubMed]
120
Bergstrom
L
,
Yocum
DE
,
Ampel
NM
, et al
.
Increased risk of coccidioidomycosis in patients treated with tumor necrosis factor alpha antagonists.
Arthritis Rheum
.
2004
;
50
(
6
):
1959
1966
[PubMed]
121
Taroumian
S
,
Knowles
SL
,
Lisse
JR
, et al
.
Management of coccidioidomycosis in patients receiving biologic response modifiers or disease-modifying antirheumatic drugs.
Arthritis Care Res (Hoboken)
.
2012
;
64
(
12
):
1903
1909
[PubMed]
122
Hage
CA
,
Bowyer
S
,
Tarvin
SE
,
Helper
D
,
Kleiman
MB
,
Wheat
LJ
.
Recognition, diagnosis, and treatment of histoplasmosis complicating tumor necrosis factor blocker therapy.
Clin Infect Dis
.
2010
;
50
(
1
):
85
92
[PubMed]
123
Winthrop
KL
,
Chiller
T
.
Preventing and treating biologic-associated opportunistic infections.
Nat Rev Rheumatol
.
2009
;
5
(
7
):
405
410
[PubMed]
124
Lee
JH
,
Slifman
NR
,
Gershon
SK
, et al
.
Life-threatening histoplasmosis complicating immunotherapy with tumor necrosis factor alpha antagonists infliximab and etanercept.
Arthritis Rheum
.
2002
;
46
(
10
):
2565
2570
[PubMed]
125
Bodro
M
,
Paterson
DL
.
Listeriosis in patients receiving biologic therapies.
Eur J Clin Microbiol Infect Dis
.
2013
;
32
(
9
):
1225
1230
[PubMed]
126
Peña-Sagredo
JL
,
Hernández
MV
,
Fernandez-Llanio
N
, et al;
Biobadaser Group
.
Listeria monocytogenes infection in patients with rheumatic diseases on TNF-alpha antagonist therapy: the Spanish Study Group experience.
Clin Exp Rheumatol
.
2008
;
26
(
5
):
854
859
[PubMed]
127
Slifman
NR
,
Gershon
SK
,
Lee
JH
,
Edwards
ET
,
Braun
MM
.
Listeria monocytogenes infection as a complication of treatment with tumor necrosis factor alpha-neutralizing agents.
Arthritis Rheum
.
2003
;
48
(
2
):
319
324
[PubMed]
128
Lanternier
F
,
Tubach
F
,
Ravaud
P
, et al;
Research Axed on Tolerance of Biotherapies Group
.
Incidence and risk factors of Legionella pneumophila pneumonia during anti-tumor necrosis factor therapy: a prospective French study.
Chest
.
2013
;
144
(
3
):
990
998
[PubMed]
129
Tubach
F
,
Ravaud
P
,
Salmon-Céron
D
, et al;
Recherce Axée sur la Tolérance des Biothérapies Group
.
Emergence of Legionella pneumophila pneumonia in patients receiving tumor necrosis factor-alpha antagonists.
Clin Infect Dis
.
2006
;
43
(
10
):
e95
e100
[PubMed]
130
Torre-Cisneros
J
,
Del Castillo
M
,
Castón
JJ
,
Castro
MC
,
Pérez
V
,
Collantes
E
.
Infliximab does not activate replication of lymphotropic herpesviruses in patients with refractory rheumatoid arthritis.
Rheumatology (Oxford)
.
2005
;
44
(
9
):
1132
1135
[PubMed]
131
Balandraud
N
,
Guis
S
,
Meynard
JB
,
Auger
I
,
Roudier
J
,
Roudier
C
.
Long-term treatment with methotrexate or tumor necrosis factor alpha inhibitors does not increase Epstein-Barr virus load in patients with rheumatoid arthritis.
Arthritis Rheum
.
2007
;
57
(
5
):
762
767
[PubMed]
132
Fritzsche
C
,
Riebold
D
,
Munk-Hartig
A
,
Klammt
S
,
Neeck
G
,
Reisinger
E
.
High prevalence of Pneumocystis jirovecii colonization among patients with autoimmune inflammatory diseases and corticosteroid therapy.
Scand J Rheumatol
.
2012
;
41
(
3
):
208
213
[PubMed]
133
Mekinian
A
,
Durand-Joly
I
,
Hatron
PY
, et al
.
Pneumocystis jirovecii colonization in patients with systemic autoimmune diseases: prevalence, risk factors of colonization and outcome.
Rheumatology (Oxford)
.
2011
;
50
(
3
):
569
577
[PubMed]
134
Mori
S
,
Cho
I
,
Sugimoto
M
.
A cluster of Pneumocystis jirovecii infection among outpatients with rheumatoid arthritis.
J Rheumatol
.
2010
;
37
(
7
):
1547
1548
[PubMed]
135
Lund
FE
,
Hollifield
M
,
Schuer
K
,
Lines
JL
,
Randall
TD
,
Garvy
BA
.
B cells are required for generation of protective effector and memory CD4 cells in response to Pneumocystis lung infection.
J Immunol
.
2006
;
176
(
10
):
6147
6154
[PubMed]
136
Martin-Garrido
I
,
Carmona
EM
,
Specks
U
,
Limper
AH
.
Pneumocystis pneumonia in patients treated with rituximab.
Chest
.
2013
;
144
(
1
):
258
265
[PubMed]
137
Mori
S
,
Cho
I
,
Sugimoto
M
.
A followup study of asymptomatic carriers of Pneumocystis jiroveci during immunosuppressive therapy for rheumatoid arthritis.
J Rheumatol
.
2009
;
36
(
8
):
1600
1605
[PubMed]
138
Specks
U
.
Biologic agents in the treatment of granulomatosis with polyangiitis.
Cleve Clin J Med
.
2012
;
79
(
suppl 3
):
S50
S53
[PubMed]
139
Besada
E
,
Nossent
JC
.
Should Pneumocystis jiroveci prophylaxis be recommended with rituximab treatment in ANCA-associated vasculitis?
Clin Rheumatol
.
2013
;
32
(
11
):
1677
1681
[PubMed]
140
Green
H
,
Paul
M
,
Vidal
L
,
Leibovici
L
.
Prophylaxis of Pneumocystis pneumonia in immunocompromised non-HIV-infected patients: systematic review and meta-analysis of randomized controlled trials.
Mayo Clin Proc
.
2007
;
82
(
9
):
1052
1059
[PubMed]
141
Kaur
N
,
Mahl
TC
.
Pneumocystis jiroveci (carinii) pneumonia after infliximab therapy: a review of 84 cases.
Dig Dis Sci
.
2007
;
52
(
6
):
1481
1484
[PubMed]
142
Tanaka
M
,
Sakai
R
,
Koike
R
, et al
.
Pneumocystis jirovecii pneumonia associated with etanercept treatment in patients with rheumatoid arthritis: a retrospective review of 15 cases and analysis of risk factors.
Mod Rheumatol
.
2012
;
22
(
6
):
849
858
[PubMed]
143
Watanabe
K
,
Sakai
R
,
Koike
R
, et al
.
Clinical characteristics and risk factors for Pneumocystis jirovecii pneumonia in patients with rheumatoid arthritis receiving adalimumab: a retrospective review and case-control study of 17 patients.
Mod Rheumatol
.
2013
;
23
(
6
):
1085
1093
[PubMed]
144
Rubin
LG
,
Levin
MJ
,
Ljungman
P
, et al;
Infectious Diseases Society of America
.
2013 IDSA clinical practice guideline for vaccination of the immunocompromised host.
Clin Infect Dis
.
2014
;
58
(
3
):
309
318
[PubMed]
145
American Academy of Pediatrics
.
Testing for Mycobacterium tuberculosis infection
. In:
Pickering
LK
,
Baker
CJ
,
Kimberlin
DW
,
Long
SS
, eds.
Red Book: 2012 Report of the Committee on Infectious Diseases
.
Elk Grove Village, IL
:
American Academy of Pediatrics
;
2012
:
39
146
Elkayam
O
,
Bashkin
A
,
Mandelboim
M
, et al
.
The effect of infliximab and timing of vaccination on the humoral response to influenza vaccination in patients with rheumatoid arthritis and ankylosing spondylitis.
Semin Arthritis Rheum
.
2010
;
39
(
6
):
442
447
[PubMed]
147
Glück
T
,
Müller-Ladner
U
.
Vaccination in patients with chronic rheumatic or autoimmune diseases.
Clin Infect Dis
.
2008
;
46
(
9
):
1459
1465
[PubMed]
148
Moses
J
,
Alkhouri
N
,
Shannon
A
, et al
.
Hepatitis B immunity and response to booster vaccination in children with inflammatory bowel disease treated with infliximab.
Am J Gastroenterol
.
2012
;
107
(
1
):
133
138
[PubMed]
149
Visvanathan
S
,
Keenan
GF
,
Baker
DG
,
Levinson
AI
,
Wagner
CL
.
Response to pneumococcal vaccine in patients with early rheumatoid arthritis receiving infliximab plus methotrexate or methotrexate alone.
J Rheumatol
.
2007
;
34
(
5
):
952
957
[PubMed]
150
Bingham
CO
 III
,
Looney
RJ
,
Deodhar
A
, et al
.
Immunization responses in rheumatoid arthritis patients treated with rituximab: results from a controlled clinical trial.
Arthritis Rheum
.
2010
;
62
(
1
):
64
74
[PubMed]
151
Crnkic Kapetanovic
M
,
Saxne
T
,
Jönsson
G
,
Truedsson
L
,
Geborek
P
.
Rituximab and abatacept but not tocilizumab impair antibody response to pneumococcal conjugate vaccine in patients with rheumatoid arthritis.
Arthritis Res Ther
.
2013
;
15
(
5
):
R171
[PubMed]
152
Nazi
I
,
Kelton
JG
,
Larché
M
, et al
.
The effect of rituximab on vaccine responses in patients with immune thrombocytopenia.
Blood
.
2013
;
122
(
11
):
1946
1953
[PubMed]
153
van Assen
S
,
Holvast
A
,
Benne
CA
, et al
.
Humoral responses after influenza vaccination are severely reduced in patients with rheumatoid arthritis treated with rituximab.
Arthritis Rheum
.
2010
;
62
(
1
):
75
81
[PubMed]
154
van der Kolk
LE
,
Baars
JW
,
Prins
MH
,
van Oers
MH
.
Rituximab treatment results in impaired secondary humoral immune responsiveness.
Blood
.
2002
;
100
(
6
):
2257
2259
[PubMed]
155
Centers for Disease Control and Prevention
. Latent tuberculosis infection: a guide for primary health care providers. Atlanta, GA: Centers for Disease Control and Prevention;
2013
. Available at: www.cdc.gov/tb/publications/ltbi/diagnosis.htm. Accessed October 22, 2014
156
Centers for Disease Control and Prevention
. TB guidelines, treatment. Atlanta, GA: Centers for Disease Control and Prevention;
2013
. Available at: www.cdc.gov/tb/publications/guidelines/treatment.htm. Accessed October 22, 2014
157
Mazurek
GH
,
Jereb
J
,
Vernon
A
,
LoBue
P
,
Goldberg
S
,
Castro
K
;
IGRA Expert Committee
;
Centers for Disease Control and Prevention
.
Updated guidelines for using interferon gamma release assays to detect Mycobacterium tuberculosis infection—United States, 2010.
MMWR Recomm Rep
.
2010
;
59
(
RR-5
):
1
25
[PubMed]
158
American Academy of Pediatrics
.
Tuberculosis
. In:
Pickering
LK
,
Baker
CJ
,
Kimberlin
DW
,
Long
SS
, eds.
Red Book: 2012 Report of the Committee on Infectious Diseases
.
Elk Grove Village, IL
:
American Academy of Pediatrics
;
2012
:
736
759
159
Chamany
S
,
Mirza
SA
,
Fleming
JW
, et al
.
A large histoplasmosis outbreak among high school students in Indiana, 2001.
Pediatr Infect Dis J
.
2004
;
23
(
10
):
909
914
[PubMed]
160
National Institute for Occupational Safety and Health
. Histoplasmosis, protecting workers at risk. Atlanta, GA: National Institute for Occupational Safety and Health, Department of Health and Human Services;
2004
. Available at: www.cdc.gov/niosh/docs/2005-109/. Accessed October 22, 2014
161
Jassal
MS
,
Bishai
WR
.
The risk of infections with tumor necrosis factor-alpha inhibitors.
J Clin Rheumatol
.
2009
;
15
(
8
):
419
426
[PubMed]
162
Chiller
TM
,
Galgiani
JN
,
Stevens
DA
.
Coccidioidomycosis.
Infect Dis Clin North Am
.
2003
;
17
(
1
):
41
57, viii
[PubMed]
163
Crum
NF
,
Lederman
ER
,
Stafford
CM
,
Parrish
JS
,
Wallace
MR
.
Coccidioidomycosis: a descriptive survey of a reemerging disease—clinical characteristics and current controversies.
Medicine (Baltimore)
.
2004
;
83
(
3
):
149
175
[PubMed]
164
Rosenstein
NE
,
Emery
KW
,
Werner
SB
, et al
.
Risk factors for severe pulmonary and disseminated coccidioidomycosis: Kern County, California, 1995-1996.
Clin Infect Dis
.
2001
;
32
(
5
):
708
715
[PubMed]
165
American Academy of Pediatrics
.
Varicella-zoster infections
. In:
Pickering
LK
,
Baker
CJ
,
Kimberlin
DW
,
Long
SS
, eds.
Red Book: 2012 Report of the Committee on Infectious Diseases
.
Elk Grove Village, IL
:
American Academy of Pediatrics
;
2012
:
774
789
166
Centers for Disease Control and Prevention
. Hepatitis B information for health professionals. Atlanta, GA: Centers for Disease Control and Prevention;
2013
. Available at: www.cdc.gov/hepatitis/hbv/hbvfaq.htm. Accessed October 22, 2014
167
Carroll
MB
,
Bond
MI
.
Use of tumor necrosis factor-alpha inhibitors in patients with chronic hepatitis B infection.
Semin Arthritis Rheum
.
2008
;
38
(
3
):
208
217
[PubMed]
168
Kato
M
,
Atsumi
T
,
Kurita
T
, et al
.
Hepatitis B virus reactivation by immunosuppressive therapy in patients with autoimmune diseases: risk analysis in hepatitis B surface antigen-negative cases.
J Rheumatol
.
2011
;
38
(
10
):
2209
2214
[PubMed]
169
Lok
AS
,
McMahon
BJ
.
Chronic hepatitis B.
Hepatology
.
2007
;
45
(
2
):
507
539
[PubMed]
170
Post
A
,
Nagendra
S
.
Reactivation of hepatitis B: pathogenesis and clinical implications.
Curr Infect Dis Rep
.
2009
;
11
(
2
):
113
119
[PubMed]
171
Yeo
W
,
Chan
PK
,
Zhong
S
, et al
.
Frequency of hepatitis B virus reactivation in cancer patients undergoing cytotoxic chemotherapy: a prospective study of 626 patients with identification of risk factors.
J Med Virol
.
2000
;
62
(
3
):
299
307
[PubMed]
172
Yeo
W
,
Johnson
PJ
.
Diagnosis, prevention and management of hepatitis B virus reactivation during anticancer therapy.
Hepatology
.
2006
;
43
(
2
):
209
220
[PubMed]
173
Rahier
JF
,
Moutschen
M
,
Van Gompel
A
, et al
.
Vaccinations in patients with immune-mediated inflammatory diseases.
Rheumatology (Oxford)
.
2010
;
49
(
10
):
1815
1827
[PubMed]
174
Hunt
D
,
Giovannoni
G
.
Natalizumab-associated progressive multifocal leucoencephalopathy: a practical approach to risk profiling and monitoring.
Pract Neurol
.
2012
;
12
(
1
):
25
35
[PubMed]

Carrie L. Byington, MD, FAAP, Chairperson

Yvonne A. Maldonado, MD, FAAP, Vice Chairperson

Elizabeth D. Barnett, MD, FAAP

H. Dele Davies, MD, FAAP

Kathryn M. Edwards, MD, FAAP

Ruth Lynfield, MD, FAAP

Flor M. Munoz-Rivas, MD, FAAP

Dawn L. Nolt, MD, FAAP

Ann-Christine Nyquist, MD, MSPH, FAAP

Mobeen H. Rathore, MD, FAAP

Mark H. Sawyer, MD, FAAP

William J. Steinbach, MD, FAAP

Tina Q. Tan, MD, FAAP

Theoklis E. Zaoutis, MD, MSCE, FAAP

Dennis L. Murray, MD, FAAP

Gordon E. Schutze, MD, FAAP

Rodney E. Willoughby, MD, FAAP

Henry H. Bernstein, DO, MHCM, FAAP – Red Book Online Associate Editor

Michael T. Brady, MD, FAAP – Red Book Associate Editor

Mary Anne Jackson, MD, FAAP, Red Book Associate Editor

David W. Kimberlin, MD, FAAP – Red Book Editor

Sarah S. Long, MD, FAAP – Red Book Associate Editor

H. Cody Meissner, MD, FAAP – Visual Red Book Associate Editor

Douglas Campos-Outcalt, MD, MPH – American Academy of Family Physicians

Karen M. Farizo, MD – Food and Drug Administration

Marc Fischer, MD, FAAP – Centers for Disease Control and Prevention

Bruce Gellin, MD, MPH – National Vaccine Program Office

Richard L. Gorman, MD, FAAP – National Institutes of Health

Natasha B. Halasa, MD, MPH, FAAP – Pediatric Infectious Diseases Society

Joan L. Robinson, MD – Canadian Paediatric Society

Marco Aurelio Palazzi Safadi, MD – Sociedad Latinoamericana de Infectologia Pediatrica

Jane F. Seward, MBBS, MPH, FAAP – Centers for Disease Control and Prevention

Geoffrey Simon, MD, FAAP – Committee on Practice Ambulatory Medicine

Jeffrey R. Starke, MD, FAAP – American Thoracic Society

Jennifer M. Frantz, MPH

Competing Interests

POTENTIAL CONFLICT OF INTEREST: The author has indicated he has no potential conflicts of interest to disclose.

FINANCIAL DISCLOSURE: The author has indicated he does not have a financial relationship relevant to this article to disclose.