ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Evaluation and prevention of infections associated with biologic agents and JAK inhibitors in adults

Evaluation and prevention of infections associated with biologic agents and JAK inhibitors in adults
Literature review current through: Jan 2024.
This topic last updated: Jan 31, 2024.

INTRODUCTION — Biologic agents are a rapidly expanding therapeutic strategy for a wide range of medical conditions. These targeted approaches have improved outcomes for many chronic diseases but, at the same time, induce unique immunologic deficits that may increase the risk for common and opportunistic infections.

This topic provides an overview of the various classes of biologic agents along with janus kinase (JAK) inhibitors, their associated infections, and approaches to mitigate risk. The number of biologic agents is constantly expanding; the more commonly used nononcologic biologic agents are discussed here.

A more detailed overview of the mechanism of action and the secondary immunodeficiency caused by many of these agents is found elsewhere. (See "Overview of biologic agents in the rheumatic diseases".)

GENERAL PRINCIPLES

Immunologic mechanisms — Biologic agents are designed to interfere with the biological activity of a component of the immune system, typically a cytokine or a cellular receptor. Cytokines are soluble proteins that interact with immune cells to control proinflammatory responses, cellular activation, and cell migration. Cell receptors reside on the surface of immune cells and can initiate cellular activation or drive cell migration upon recognizing their respective cognate ligand. By interfering with the normal activities of these molecules, biologic agents disrupt signaling pathways that drive activation and migration of immune cells to sites of infection, thereby interfering with the host immune response and increasing risk for infection.

Many biologic therapies are associated with an increased risk of opportunistic infections. As an example, in a meta-analysis of 70 randomized controlled trials that included over 32,000 patients with rheumatoid arthritis, use of a biologic agent (tumor necrosis factor [TNF]-alpha inhibitors, anakinra, tocilizumab, abatacept, and rituximab) was associated with higher incidence of opportunistic infections compared with placebo (odds ratio [OR] 1.79, 95% CI 1.17-2.74; one additional opportunistic infection for every 582 patients treated with a biologic agent) [1]. Consequently, we test and/or vaccinate for certain latent and/or chronic infections prior to the start of these biologic agents.

Pretreatment infectious testing — Biologic agents can increase the risk for serious and potentially fatal infections from bacteria, viruses, fungi, and occasionally parasites [1-5]. The risk varies depending on the type of immunosuppressive agent, the condition being treated, and the concomitant use of other immunosuppressants (eg, corticosteroids). For some conditions, there may be additional infectious testing necessary (eg, cytomegalovirus [CMV] in select cases of ulcerative colitis) for diagnostic purposes.

Baseline testing – Baseline testing for some latent infections is recommended prior to initiating therapy with many of the medications in this section. In general, most of the biologic agents discussed here warrant testing for the following infections:

Tuberculosis infection (TBI), previously known as latent tuberculosis infection (table 1 and algorithm 1 and algorithm 2) (see "Tuberculosis infection (latent tuberculosis) in adults: Approach to diagnosis (screening)")

Hepatitis B virus (HBV) (algorithm 3) (see "Hepatitis B virus: Screening and diagnosis in adults", section on 'Approach to screening and testing')

Hepatitis C virus (HCV) (algorithm 4) (see "Screening and diagnosis of chronic hepatitis C virus infection")

For patients who do not have a negative human immunodeficiency virus (HIV) test documented in their records or are at increased risk of acquiring HIV (eg, men who have sex with men, engagement in sex work), the pretreatment infectious testing process is a good opportunity to provide routine HIV screening prior to the initiation of immunosuppression. (See "Screening and diagnostic testing for HIV infection", section on 'Preferred approach'.)

Specific testing recommendations for each biologic agent are discussed in the corresponding sections below.

Management of identified infections – If any infections are discovered on pretreatment testing, we treat the underlying infection and hold off on starting the biologic agent until the infection is controlled, if possible. Close monitoring is paramount to make sure there is no worsening of the underlying infection once the biologic agent is started.

Tuberculosis infection – Those who test positive for TBI warrant tuberculosis (TB) treatment prior to initiating the biologic agent. (See "Treatment of tuberculosis infection (latent tuberculosis) in nonpregnant adults without HIV infection".)

Hepatitis B – Those who test positive for chronic HBV infection may require antiviral prophylaxis to prevent HBV reactivation while they are on the biologic agent. (See "Hepatitis B virus reactivation associated with immunosuppressive therapy".)

Timing of vaccinations

Prior to initiating biologics — We make sure that all patients initiating biologics have age-appropriate routine vaccinations (including severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2] vaccine series and annual influenza vaccination, if applicable) completed prior to starting therapy when feasible, because some vaccinations may be less effective during and after therapy or may be contraindicated (eg, live vaccines) (figure 1) [6-9]. Routine vaccinations should be administered at least two weeks before starting these immunomodulatory therapies if possible, and timing should be extended to at least four weeks for live vaccines (table 2) [10,11]. Although concrete efficacy data are limited, therapeutic agents that target down-regulation of immune responses (eg, antigen presenting cells [dendritic cells and macrophages], T cells, and B cells) could potentially impair the immunogenicity and/or effectiveness of newly administered vaccines.

Additional recommended vaccinations for some biologic agents may include:

Pneumococcal vaccination (see "Immunizations in autoimmune inflammatory rheumatic disease in adults", section on 'Pneumococcal vaccines' and "Pneumococcal vaccination in adults", section on 'Approach to vaccination')

Varicella/herpes zoster vaccination – Generally, assessing for prior varicella zoster virus (VZV) infection by history and/or medical records is sufficient to document prior exposure to VZV. If a patient has been exposed to VZV (either by vaccination or by natural infection), we administer the nonlive recombinant zoster vaccine (RZV) to adult patients at least two weeks prior to initiating immunosuppressive therapy.

If a patient has never been exposed to varicella, some experts administer the live VZV vaccine, barring any contraindications, with the goal to complete the whole series at least four weeks prior to initiating immunosuppressive therapy, while others administer RZV irrespective of prior exposure history. (See "Vaccination for the prevention of shingles (herpes zoster)", section on 'Immunocompromised persons' and "Vaccination for the prevention of chickenpox (primary varicella infection)", section on 'Immunocompromised hosts'.)

During treatment with biologics — Vaccination with nonlive vaccines during therapy may be considered if the benefit of vaccination outweighs the risk (eg, annual influenza vaccination) [12,13]. Although vaccine efficacy may be attenuated, the humoral response often remains intact, and some protective antibody levels can be achieved.

When possible, vaccination should be given at the nadir of immunosuppression. This is often achieved by administering the nonlive vaccine midcycle of each dose interval (eg, at two weeks for a four-week dose interval). In general, we do not hold back treatments required for managing the associated chronic medical condition in order to optimize immune responses. However, with some treatments, such as rituximab, one can consider temporary discontinuation of the drug [9]. If treatment is discontinued, the length of time should be guided by the drug's pharmacokinetics and the expected time to immune recovery, as there are limited real-world data to provide specific guidance for this practice for any given drug.

Live vaccinations are typically contraindicated while receiving biologic agents. Specific vaccination recommendations for each biologic agent are discussed in the corresponding sections below.

Further detailed discussion of vaccine administration timing while the patient is taking immunosuppressive therapy for rheumatic diseases is found elsewhere. (See "Immunizations in autoimmune inflammatory rheumatic disease in adults".)

Active infections during treatment — In the event of a new serious infection while on treatment, the biologic agent should be stopped while the infection is treated [14]. Therapy can resume after the active infection is controlled, with close monitoring. If active M. tuberculosis infection is diagnosed during therapy, many of the drugs discussed in this section should be discontinued while treatment is initiated [15-21]. This is discussed in detail elsewhere. (See "Risk of mycobacterial infection associated with biologic agents and JAK inhibitors", section on 'Management'.)

Differentiating symptoms of infection versus medication adverse effects — Often, it may be difficult to distinguish symptoms of infection from symptoms caused by adverse effects of the biologic agent [22]. Since many drugs discussed here predispose a patient to infections, it is important to complete a comprehensive evaluation for infection prior to attributing the patient's symptoms to the medication.

TUMOR NECROSIS FACTOR (TNF)-ALPHA INHIBITORS (TNFI) — TNF is a pleiotropic cytokine that is an important component of innate immunity. TNF receptor binding leads to the release of proinflammatory cytokines, chemokines, and adhesion factors. The primary TNFi that have been US Food and Drug Administration (FDA)-approved are monoclonal antibodies (mAbs; eg, infliximab, adalimumab, certolizumab, and golimumab) and engineered antibody fragments (certolizumab pegol), which function by binding and inhibiting soluble and membrane-bound TNF. Etanercept is a fusion protein that acts as a soluble TNF receptor to bind TNF. TNFi therapies disrupt the immune system's capacity to clear intracellular pathogens and those sequestered in granulomas. (See "Overview of biologic agents in the rheumatic diseases", section on 'TNF inhibition' and "Treatment of Crohn disease in adults: Dosing and monitoring of tumor necrosis factor-alpha inhibitors".)

Risk of infection – Treatment with TNFi predisposes individuals to the following infections (table 3) [23-29]:

Tuberculosis (TB) and other mycobacterial infections

Listeria monocytogenes, Nocardia spp, and Legionella spp

Hepatitis B virus (HBV) and hepatitis C virus (HCV)

Herpes zoster virus

Endemic mycoses

Candidal infections

Cryptococcal infection

Aspergillosis

Toxoplasmosis (rare)

Common viral and bacterial infections (sinopulmonary infections, pneumonia, urinary tract infections, cellulitis)

TNFi particularly predispose an individual to acquisition and/or reactivation of mycobacterial infections (including TB), hepatitis B, and endemic mycoses such as histoplasmosis. Detailed discussions of the risk of infections seen with TNFi are discussed separately. (See "Tumor necrosis factor-alpha inhibitors: Bacterial, viral, and fungal infections" and "Risk of mycobacterial infection associated with biologic agents and JAK inhibitors".)

Pretreatment infectious testing – We test patients initiating TNF inhibitors for tuberculosis infection (TBI) (algorithm 1), HBV (algorithm 3), and HCV (algorithm 4 and table 3). In regions endemic to specific mycoses, we obtain history of symptoms that would suggest active or recent infection in the past two years and inquire about potential exposures. In patients with risk factors of being exposed to histoplasmosis in the last two years, we obtain a chest radiograph (see "Tumor necrosis factor-alpha inhibitors: Bacterial, viral, and fungal infections", section on 'Histoplasmosis'). In coccidioidomycosis endemic regions, we obtain coccidioides serology to test for previous exposure to Coccidioides spp. (See "Management considerations, screening, and prevention of coccidioidomycosis in immunocompromised individuals and pregnant patients", section on 'Patients receiving immunomodulatory agents'.)

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for individuals starting TNFi, as discussed above (table 3) (see 'Timing of vaccinations' above). Vaccine responses may be attenuated while on therapy.

ABATACEPT — Abatacept is an anti-T cell therapy that inhibits the costimulatory signal by competitively interacting with CD80/CD86 on antigen-presenting cells, blocking binding to T cell-expressed CD28. This prevents T cell activation and proinflammatory cytokine release. (See "Treatment of rheumatoid arthritis in adults resistant to initial biologic DMARD therapy", section on 'Abatacept'.)

Risk of infection – Although abatacept is not thought to be severely immunocompromising, it may predispose individuals to the following infections [26-29] (table 3):

Tuberculosis (TB) and other mycobacterial infections

Hepatitis B virus (HBV) and hepatitis C virus (HCV)

Epstein-Barr virus and cytomegalovirus (CMV) infection

Common viral and bacterial infections (sinopulmonary infections, pneumonia, urinary tract infections, cellulitis)

Abatacept does not appear to greatly increase the risk of infectious complications in most patients. A 2009 meta-analysis of five trials that included 2945 patients found similar rates of serious infection with abatacept compared with placebo (2.5 versus 1.7 percent) [30]. In another compilation of eight clinical trials of patients with rheumatoid arthritis, there was no difference in the infection rate between patients receiving abatacept versus placebo [31]. However, case reports and small retrospective analyses have demonstrated reactivation of hepatitis B with use of abatacept [26-29]. Using a Medicare database of patients with rheumatoid arthritis who had prior treatment with a biologic agent, the risk of infections in hospitalized patients was significantly higher for tumor necrosis factor-alpha inhibitors (TNFi) and rituximab compared with subjects receiving abatacept [32]. (See 'Tumor necrosis factor (TNF)-alpha inhibitors (TNFi)' above and 'Anti-B cell agents' below.)

The safety of abatacept was assessed in several trials [5,33]. Serious infections occurred at an incidence rate of 4.3 per 100 person-years. The most frequent infections were pneumonia, bronchitis, cellulitis, and urinary tract infections. The increased risk of respiratory tract infections mostly occurred in patients with underlying pulmonary disease (eg, chronic obstructive pulmonary disease).

Pretreatment infectious testing – We test patients initiating abatacept for TBI (algorithm 1), HBV (algorithm 3), and HCV (algorithm 4 and table 3).

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for individuals starting abatacept, as discussed above (table 3) (see 'Timing of vaccinations' above). Vaccine responses may be attenuated while on therapy.

ANTI-B CELL AGENTS — Rituximab and other anti-B cell antibodies (eg, ocrelizumab, ofatumumab) are monoclonal antibodies (mAbs) that bind to the CD20 receptor on B cells [34], resulting in B cell depletion. Belimumab confers less risk of infection because it prevents activation of B cells instead of depleting them [35].

Class-wide risk assessment

Risk of infection – Treatment with anti-B cell agents predisposes individuals to the following infections [36-42] (table 4):

Tuberculosis (TB) and other mycobacterial infections

Hepatitis B virus (HBV) [43,44] and hepatitis C virus (HCV)

Herpes zoster virus

Epstein-Barr virus and cytomegalovirus (CMV) infection

Severe coronavirus disease 2019 (COVID-19) [45-47] (except belimumab)

Progressive multifocal leukoencephalopathy (PML; mostly rituximab, although has been reported with belimumab)

Endemic mycoses

Cryptococcal infection

Pneumocystis pneumonia (only rituximab) [48]

Common viral and bacterial infections (sinopulmonary infections, pneumonia, urinary tract infections, cellulitis) [49]

Out of all the anti-B cell agents, rituximab is associated with the highest risk of serious infections, while belimumab is associated with the lowest risk for serious infections.

Risk is highest for hepatitis B reactivation. For patients with evidence of active HBV (positive surface antigen) or inactive HBV (positive core antibody, negative surface antigen), we try to avoid anti-B cell agents when possible. If treatment with an anti-B cell agent is necessary, HBV prophylaxis should be initiated prior to treatment to prevent HBV reactivation (see "Hepatitis B virus reactivation associated with immunosuppressive therapy", section on 'Preventing HBV reactivation'). If HBV reactivation is detected, therapy with the CD20 inhibitor should be stopped temporarily until antiviral therapy is initiated and HBV deoxyribonucleic acid (DNA) and alanine aminotransferase decrease to lower levels. (See "Hepatitis B virus reactivation associated with immunosuppressive therapy".)

Infection with SARS-CoV-2 has been a serious complication for those on anti-CD20 therapies, which increases the risk of COVID-19-related hospitalization and death [45,46]. Patients with SARS-CoV-2 infection should be promptly evaluated for treatment. (See "COVID-19: Management of adults with acute illness in the outpatient setting", section on 'Treatment with COVID-19-specific therapies'.)

People with HIV receiving anti-CD20 therapies should be closely monitored and strictly adhere to antiviral therapy; guidelines recommend withholding anti-CD20 therapies in patients with a CD4 cell counts ≤50 cells/microL [50].

The risk of infection is increased when CD20 inhibitors are used in the context of hematological malignancies [51] or solid organ transplants [52,53] compared with the treatment of autoimmune disorders [54]. There is also a risk of developing hypogammaglobulinemia while taking rituximab, which can further increase infection risk. (See "Secondary immunodeficiency induced by biologic therapies".)

Pretreatment infectious testing – We test patients initiating anti-B cell agents for tuberculosis infection (TBI) (algorithm 1), HBV (algorithm 3), and HCV (algorithm 4 and table 4).

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for patients starting anti-B cell agents, as discussed above (table 4) (see 'Timing of vaccinations' above). Vaccine responses may be attenuated while on therapy and for 6 to 12 months after cessation of therapy. When administering nonlive attenuated vaccines (with the exception of the annual influenza vaccine) during rituximab therapy, one can optimize immunogenicity by administering the vaccine at the time the rituximab dose is due and holding rituximab for at least two weeks after vaccination, if disease activity allows [9].

Drug-specific risks

Rituximab — Rituximab is a mAb that binds to the CD20 receptor on B cells [34], resulting in B cell depletion. It also disrupts B and T cell interactions, resulting in impaired cellular immunity and increased risk of viral reactivation [55] (see "Rituximab: Principles of use and adverse effects in rheumatoid arthritis"). Usage in the setting of malignancy is reviewed in other chapters. (See "Treatment protocols for lymphoma".)

Out of all the anti-B cell agents, rituximab is associated with the highest risk of serious infections, especially with HBV reactivation. Some experts choose to initiate Pneumocystis spp prophylaxis in patients taking rituximab monotherapy, although the absolute risk is low (see "Treatment and prevention of Pneumocystis pneumonia in patients without HIV", section on 'Indications'). PML has been observed in patients taking rituximab, but the absolute risk is quite low [56]. There are no recommendations to test for John Cunningham virus (JCV) antibody prior to initiating rituximab. Rituximab should be discontinued if PML is suspected and/or confirmed.

In addition to increasing risk of bacterial, viral, and fungal infections, rituximab is also associated with attenuated response to vaccines administered during and after discontinuation of the drug [57]. This phenomenon is due to the prolonged time needed for the repletion of B cells within the body. On average, it typically requires six to nine months after discontinuing rituximab for B cells numbers to return to normal [48]. In a study of 120 patients with different types of vasculitides, B cell recovery occurred over a range of 8 to 44 months [58]. Even after normalization of B cell numbers, vaccine response may be attenuated. In a study of 75 patients who received four injections of rituximab or placebo over one month, titers to vaccines administered 12 months after rituximab therapy were lower in patients who received rituximab versus placebo [59].

Pre-existing immunity to vaccinations administered previously is not affected by treatment. As an example, in a study of 75 patients with type 1 diabetes treated with four injections of rituximab or placebo over one month and then followed for 12 months (ie, a period sufficient for B cell recovery), measles, mumps, and rubella titers before rituximab and after were unchanged between the rituximab group and placebo [59].

Additionally, some degree of transient hypogammaglobulinemia is common and rituximab can also cause "late-onset" neutropenia, appearing one to five months after the end of therapy, which is reviewed separately. (See "Drug-induced neutropenia and agranulocytosis", section on 'Rituximab' and "Secondary immunodeficiency induced by biologic therapies".)

Ocrelizumab — Ocrelizumab is a humanized monoclonal anti-CD20 antibody therapy that triggers antibody-dependent cellular cytolysis. This will deplete circulating immature and mature B cells, not the CD20-negative plasma cells [60,61].

Ocrelizumab has been associated with a higher risk of infection in comparison to interferon-beta and placebo for treatment of multiple sclerosis. In phase II trials, ocrelizumab was associated with higher rates of nonserious infections, including upper respiratory tract infections, nasopharyngitis, urinary tract infections, skin infections, and herpesvirus infections [43,49]. Cases of osteomyelitis, urosepsis, cystitis, and pyelonephritis have been reported [49]. Additionally, there is a possible correlation between ocrelizumab administration and more severe COVID-19 outcomes [47]. Opportunistic infections appear relatively infrequently, except for serious HBV infections [43]. At least one case of PML was associated with ocrelizumab monotherapy in a patient without prior immunosuppressive therapy [62].

Ofatumumab — Ofatumumab is a fully human monoclonal anti-CD20 monoclonal antibody, which targets a distinct epitope on the CD20 molecules of B cells and induces apoptosis [63]. Limited information is available from clinical studies on the immune effects of this drug. In one uncontrolled trial of 33 patients with chronic lymphocytic leukemia, 51 percent experienced infections, although most were mild or moderate [40]. Reactivation of hepatitis B has also been reported.

Belimumab — Belimumab, a mAb that inhibits B cell activating factor, reduces B cell development and survival [64]. Compared with the other anti-B cell treatments and rheumatic disease immunomodulators, belimumab is less likely to increase the risk for serious infections, because it prevents activation of B cells but does not deplete them [35]. Belimumab treatment does not appear to substantially impact the risk for severe COVID-19 [65] or reduce the immune response to vaccination [66]. A low incidence of PML has been observed in patients taking belimumab [56].

Others — Obinutuzumab and inebilizumab are other anti-B cell agents. Although data on these agents are limited, we expect similar risks for infection compared with other anti-B cell agents and provide the same pretreatment testing and vaccinations for patients initiating these agents as other anti-B cell agents.

JANUS KINASE (JAK) INHIBITORS — The JAK-signal transducer and activator of transcription pathway transduces cytokine signaling, making it critical to inflammatory responses [67-69]. The JAK family is comprised of four members: JAK1, JAK2, JAK3, and tyrosine kinase 2. Each cytokine receptor requires a pair of two associated JAKs to signal through the cytokine receptor. These kinases are important for proper immune function and hematopoiesis due to their effect on the signaling of numerous cytokines and growth factors [70]. JAK inhibitors include ruxolitinib, baricitinib, tofacitinib, upadacitinib, and filgotinib. (See "Overview of the Janus kinase inhibitors for rheumatologic and other inflammatory disorders".)

Class-wide risk assessment

Risk of infection Treatment with JAK inhibitors predisposes individuals to the following infections [71-76] (table 3):

Tuberculosis (TB) [77,78] and other mycobacterial infections

Hepatitis B virus (HBV) [79] and hepatitis C virus (HCV)

Herpes zoster virus

Epstein-Barr virus and cytomegalovirus (CMV)

Progressive multifocal leukoencephalopathy (PML)

Endemic mycoses

Cryptococcal infection

Pneumocystis pneumonia

Common viral and bacterial infections (sinopulmonary infections, pneumonia, urinary tract infections, cellulitis) [68,78]

Pretreatment infectious testing – We test patients initiating JAK inhibitors for tuberculosis infection (TBI) (algorithm 1), HBV (algorithm 3), and HCV (algorithm 4 and table 3). In coccidioidomycosis-endemic regions, we also obtain Coccidioides serology if the individual will be starting ruxolitinib, based on observational data that this agent predisposes patients to reactivation of previous Coccidioides infection. (See "Management considerations, screening, and prevention of coccidioidomycosis in immunocompromised individuals and pregnant patients", section on 'Patients receiving immunomodulatory agents'.)

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for patients initiating JAK inhibitors, as discussed above (table 3). (See 'Timing of vaccinations' above.)

Drug-specific risks

Ruxolitinib — Ruxolitinib inhibits JAK1 and JAK2, which disrupts cytokine and growth factor signaling to attenuate immune responses.

Serious bacterial, viral, and fungal infections have been reported in patients treated with ruxolitinib, including mycobacterial infections, reactivation of HBV, PML, herpes zoster, and coccidioidomycosis [69,80]. Neutropenia may occur, and patients should be closely monitored during treatment.

Baricitinib — Baricitinib, like ruxolitinib, is a JAK1/JAK2 inhibitor that blocks plasmablast, Th1, and Th17 differentiation and innate stimulation of T cells [81].

Patients receiving baricitinib most commonly report respiratory infections, bronchitis, and urinary tract infections [71-73]. Herpes zoster occurred more frequently in patients receiving baricitinib compared with placebo in clinical trials [71-73].

Tofacitinib — Tofacitinib inhibits JAK1, JAK2, and JAK 3 to disrupt cytokine and growth factor signaling, thereby reducing lymphocyte activation, differentiation, and function [68].

Infections reported in patients treated with tofacitinib include pneumonia, cellulitis, herpes zoster, and urinary tract infections [74-76]. Other common infections include upper respiratory tract infections [68,78]. Opportunistic infections have been reported, including M. tuberculosis, P. jirovecii pneumonia, and cryptococcosis, as well as the reactivation of other viruses. Patients with HBV and HCV were excluded from clinical trials, but HBV reactivation has been observed [79]. It is unclear if risk of TB reactivation is lower with JAK inhibitors compared with tumor necrosis factor (TNF)-alpha inhibitor or other biologic agents.

Decreased responses to pneumococcal immunization, especially in combination with methotrexate, have been observed [82].

INTERLEUKIN-1 INHIBITORS — Interleukin (IL)-1 inhibitors prevent the activation of potent proinflammatory cytokine cascades. (See "Interleukin 1 inhibitors: Biology, principles of use, and adverse events".)

Class-wide risk assessment

Risk of infection – Treatment with IL-1 inhibitors predisposes individuals to the following infections (table 5):

Tuberculosis (TB) and other mycobacterial infections

Hepatitis B virus (HBV) and hepatitis C virus (HCV)

Herpes zoster virus

Candidal infections

Cryptococcal infection

Aspergillosis

Common viral and bacterial infections (sinopulmonary infections, pneumonia, urinary tract infections, cellulitis)

Since risk of infections with IL-1 inhibitors seems to be lower than with some other biologic agents (eg, tumor necrosis factor-alpha inhibitors [TNFi]), these agents can be a useful treatment option for patients who are undergoing evaluation for occult infections but would benefit from starting a biologic agent earlier than the time frame in which the evaluation for infection can be completed. Nevertheless, IL-1 inhibitors should not be initiated in patients with known active infections and should be held in the context of active infections that arise during treatment. IL-1 inhibitors should not be used in combination with TNFi [83].

Pretreatment infectious testing – We test patients initiating IL-1 inhibitors for tuberculosis infection (TBI) (algorithm 1), HBV (algorithm 3), and HCV (algorithm 4 and table 5).

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for patients starting IL-1 inhibitors, as discussed above (table 5). (See 'Timing of vaccinations' above.)

Drug-specific risks

Anakinra — Anakinra is an IL-1 receptor antagonist that prevents IL-1 from binding to IL-1 receptors [84]. IL-1 has a multitude of signaling responsibilities in the cell, and blocking this leads to reduced proinflammatory cytokine cascades.

The most commonly observed infections among patients treated with anakinra include upper respiratory tract infections, cellulitis, nasopharyngitis, and bone and joint infections [84-86]. Additional opportunistic infections reported in postmarketing studies include fungal, mycobacterial, and bacterial infections. Reactivation of M. tuberculosis infection has been reported but appears infrequent when appropriate testing and treatment are implemented [86]. The risk for HBV reactivation in patients receiving anakinra has not been studied.

Canakinumab — Canakinumab is a humanized monoclonal antibody (mAb) directed against IL-1 beta. Canakinumab specifically binds to IL-1 beta and inhibits its downstream effector functions by blocking its ability to bind to IL-1 receptors [87]. This signal inhibition decreases the proinflammatory immune response.

The most common infections seen in patients on canakinumab include nasopharyngitis, upper respiratory tract infections, and influenza [88-90]. Opportunistic infections with aspergillosis, mycobacterial infections, herpes zoster, and cytomegalovirus (CMV) have been reported. Patients with TBI, HBV, and HCV were excluded from all canakinumab trials, and the risk for reactivation of chronic bacterial and viral infections is unclear [89,90].

Rilonacept — Rilonacept is a bispecific humanized mAb directed against IL-1 beta to limit IL-1 signaling and reduce inflammatory responses [91,92].

Upper respiratory tract infection is the most common infection in patients undergoing treatment. However, many other serious infections can occur due to the consequences of preventing the IL-1 signaling pathways, such as M. tuberculosis infection [91-93]. Injection site reactions are common and should be monitored for infection.

INTERLEUKIN-4 INHIBITORS (DUPILUMAB) — Dupilumab is a humanized monoclonal antibody (mAb) directed against interleukin (IL)-4 receptor subunit alpha. Dupilumab inhibits the downstream signaling of both IL-4 and IL-13, critical drivers of the type II inflammatory response [94,95]. Studies are ongoing to determine the safety of long-term usage [96]. (See "Treatment of severe asthma in adolescents and adults", section on 'Anti-lL-4 receptor alpha subunit antibody (dupilumab)' and "Evaluation and management of severe refractory atopic dermatitis (eczema) in adults", section on 'Dupilumab'.)

Risk for infection – Generally, dupilumab is associated with less infectious risk compared with other immunomodulators, such as methotrexate [97] (table 6). Serious infections leading to drug discontinuation are infrequent, but patients should be monitored closely [98]. Herpes simplex virus infections have been reported in patients undergoing therapy with dupilumab. Most conjunctivitis seen in patients taking dupilumab is noninfectious, although bacterial and viral conjunctivitis can rarely occur [97]. Infection severity due to SARS-CoV-2 does not appear to be increased [99-101].

The impact of dupilumab on infections due to helminths and chronic infections with hepatitis B virus (HBV), hepatitis C virus (HCV), and HIV are unknown, as patients with these conditions were excluded from clinical trials [95]. However, subsequent case reports of patients with chronic HBV and HCV treated with dupilumab have been reassuring [102,103].

Pretreatment infectious testing – Testing for tuberculosis infection (TBI) (algorithm 1), HBV (algorithm 3), and HCV (algorithm 4) can be decided on a case-by-case basis, based on the patient's risk factors and prior testing history for each of these infections, since there are no data indicating increased risk of reactivation with TBI and HBV in patients taking dupilumab (table 6) [102-104]. Since dupilumab selectively suppresses Th2 immune responses, it is unlikely to cause reactivation of HBV, HCV, or tuberculosis (TB). In a case series of five patients with atopic dermatitis and positive hepatitis B surface antigen who were treated with dupilumab, there were no cases of hepatitis B reactivation after two years of treatment [102]. (See 'Pretreatment infectious testing' above.)

Vaccinations – No vaccinations are necessary prior to initiating dupilumab (table 6).

INTERLEUKIN-5 AND IMMUNOGLOBULIN E INHIBITORS — Eosinophil and mast cell activation are critical aspects of the immune response to allergens and parasitic pathogens, causing inflammation and tissue damage. Interleukin (IL)-5, secreted by Th2 cells and mast cells, is a critical regulator of eosinophil differentiation, stimulation, and survival; activation and signaling occur upon IL-5-specific binding to its receptor (IL-5R). Monoclonal antibodies (mAbs) designed to inhibit this interaction by binding circulating IL-5 (mepolizumab, reslizumab) or competitively binding to IL-5R (benralizumab) have been shown to deplete blood and tissue eosinophil counts [105-107]. Omalizumab is a competitive inhibitor of immunoglobulin E (IgE), leading to inhibition of mast cell activation and release of inflammatory proteins [108]. (See "Treatment of severe asthma in adolescents and adults", section on 'Persistently uncontrolled asthma'.)

Class-wide risk assessment

Risk of infection – Due to the known effect of IL-5 and IgE signaling on helminth clearance, there is a theoretical increased risk for parasitic infections (table 6). Thus, in a patient presenting with signs and symptoms of infection, we consider the possibility and evaluate for parasitic infections, especially within an appropriate clinical context. If a parasitic infection is discovered following the start of therapy, treatment should be stopped until the infection is resolved.

Due to the theoretical increased risk for parasitic infections, the major clinical trials for benralizumab [70,107,109-112], mepolizumab [106,113-119], reslizumab [105,120-122], and omalizumab [123-128] excluded patients with active parasitic infections and we do not know how much of an increased risk to parasitic infections these drugs confer. In a small randomized, double-blinded study of 137 participants in Brazil who were at high risk for helminth infection, there was a statistically nonsignificant trend toward higher incidence of parasitic infection in those who received omalizumab compared with placebo (50 versus 41 percent, OR 1.47, 95% CI 0.74-2.95) [129].

Pretreatment infectious testing – For patients at increased risk for parasitic infections (eg, patients who live in or recently traveled from endemic countries), we test for possible quiescent parasitic infections with a Strongyloides spp immunoglobulin G (IgG) antibody and stool microscopy for ova, cysts, and helminths prior to initiation of these agents (table 6). If any parasitic infections are found, they should be treated prior to the initiation of IL-5 inhibitor or anti-IgE monoclonal antibody agents. (See "Strongyloidiasis", section on 'Screening' and "Approach to stool microscopy".)

Vaccinations – We administer the herpes zoster vaccine to patients (especially those above the age of 50) starting mepolizumab, given the increased risk for herpes zoster infections (table 6).

Drug-specific risks

Benralizumab — Benralizumab is a humanized cytolytic IgG1 mAb directed against the IL-5R alpha subunit. Due to a direct apoptotic effect on eosinophils via antibody-mediated cellular cytotoxicity [130], benralizumab results in greater depletion of circulating and tissue-resident eosinophils compared with drugs that bind IL-5 directly [131]. In a review of the Vigibase database, parasitic infections were more prevalent in patients taking benralizumab (0.23 percent) compared with mepolizumab (0.07 percent), omalizumab (0.06 percent), dupilumab (0.04 percent), or placebo (0.02 percent) [132]. However, in most clinical trials, infections occurred at a similar rate in the benralizumab and placebo arms [70,107,109-112].

Mepolizumab — Mepolizumab is a humanized IgG1 kappa mAb directed against IL-5. The overall incidence of infections was similar in clinical trials of patients receiving mepolizumab compared with placebo [106,113-116,118,119]. However, multiple studies report cases of herpes zoster in patients taking mepolizumab [106,113,114]. Based on a small number of case studies, there is no clear evidence of increased COVID-19 severity in patients taking mepolizumab [133,134]. Nonetheless, the risk of hospitalization may be elevated in patients infected with SARS-CoV-2 [135].

Reslizumab — Reslizumab is a humanized IgG4 kappa mAb directed against IL-5. The incidence of infections in clinical trials was similar among patients receiving reslizumab and placebo [105,120-122].

Omalizumab — Omalizumab is a humanized IgG1 mAb that targets circulating IgE. Most studies have demonstrated a similar incidence of infections in patients receiving omalizumab compared with placebo [123-128]. A few case studies suggest that omalizumab does not increase the risk for SARS-CoV-2 infection or the development of severe COVID-19 [136]. In a randomized controlled trial exploring the rate of helminth infection in patients receiving omalizumab, there was a small increase in the incidence of helminth infections but no apparent difference in the severity of infection [129]. Omalizumab has also been associated with adverse effects that mimic infections, such as fever, arthralgias, and urticaria [22,137]. These symptoms have been reported in some individuals even after a year into the course of therapy [22].

OTHER INTERLEUKIN INHIBITORS (IL-6, IL-12, IL-17, IL-23) — Interleukin (IL)-6, IL-12, IL-17, and IL-23 are all cytokines involved in inflammatory and immune response pathways. The discussed monoclonal antibodies (mAbs) are directed against these cytokines and interfere with downstream signaling processes, preventing the release of proinflammatory cytokines. These cytokines are produced in a vast array of cells, including those involved in T cell activation and stimulation of hematopoietic precursor cell proliferation and differentiation. The cytokines can also be found locally at sites of infection and inflammation. Continual production of these cytokines is involved in the pathogenesis of some autoimmune diseases, and targeting these cytokines through neutralization or antagonization is an effective strategy to treat these diseases [138]. (See "Overview of biologic agents in the rheumatic diseases", section on 'Biologic cytokine inhibitors'.)

Sarilumab and tocilizumab — Sarilumab and tocilizumab bind to soluble and membrane-bound IL-6 receptors and interfere with IL-6 signaling via the receptor [139,140] (table 5). (See "Overview of biologic agents in the rheumatic diseases", section on 'IL-6 inhibition'.)

Risk of infection – Patients treated with IL-6 inhibitors are at increased risk of the following infections:

Tuberculosis (TB) and other mycobacterial infections

Hepatitis B virus (HBV) and hepatitis C virus (HCV)

Herpes zoster virus [141]

Candidal infections

Pneumocystis pneumonia

Common viral and bacterial infections (sinopulmonary infections, pneumonia, urinary tract infections, cellulitis) [142,143]

Long-term studies suggest that risk for infections is similar to other biologic agents, including tumor necrosis factor (TNF)-alpha inhibitors [144-148]. However, many of the opportunistic bacterial, viral, and fungal infections often occur during coadministration with other immunosuppressive medications [149,150]. Patients with HBV and HCV were excluded from trials using sarilumab and tocilizumab; however, reactivation of HBV has been reported [151].

Pretreatment infectious testing – For patients starting IL-6 inhibitors, we test for tuberculosis infection (TBI) (algorithm 1), HBV (algorithm 3), and HCV (algorithm 4 and table 5).

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for patients starting IL-6 inhibitors, as discussed above (table 5). (See 'Timing of vaccinations' above.)

Ixekizumab, secukinumab, brodalumab, and bimekizumab — These mAbs are directed against IL-17. In particular, ixekizumab and secukinumab selectively bind IL-17A and inhibit its ability to interact with the IL-17 receptor [15,16]. Brodalumab binds IL-17RA and prevents its interactions with other IL-17-related proteins [17]. These actions prevent cytokine signaling and the subsequent release of proinflammatory cytokines. Bimekizumab is not available in the United States. (See "Overview of biologic agents in the rheumatic diseases", section on 'IL-17 inhibition'.)

Risk of infections – Patients treated with IL-17 inhibitors are at increased risk of the following infections (table 5):

TB and other mycobacterial infections [15-20]

Candidal infections, especially oral candidiasis [18]

Dermatologic fungal infections (eg, tinea)

Common viral and bacterial pathogens (eg, nasopharyngitis, upper respiratory tract infections, urinary tract infections) [19,20]

Based on case reports, secukinumab has been used in patients with HBV and HCV infection without viral reactivation [152]. Very few cases of TB and other mycobacterial infections have been reported with these agents, and the overall risk is likely very low [153].

Pretreatment infectious testing – For patients initiating IL-17 inhibitors, we test for TBI (algorithm 1) prior to initiating treatment (table 5).

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for patients starting IL-17 inhibitors, as discussed above (table 5). (See 'Timing of vaccinations' above.)

Guselkumab, ustekinumab, and others — Guselkumab is a mAb directed against IL-23 to prevent binding to its receptor, which inhibits downstream signaling to dampen proinflammatory cytokine responses [154]. Ustekinumab is a mAb directed against IL-12 and IL-23, interfering with natural killer cell activation and CD4+ T cell differentiation [155]. Risankizumab and mirikizumab are other IL-23 inhibitors with similar mechanism of action to guselkumab. (See "Overview of biologic agents in the rheumatic diseases", section on 'IL-12/23 blockade'.)

Risk for infections – Patients treated with IL-23 inhibitors are at increased risk of candidal and dermatologic fungal infections (eg, tinea) (table 5).

Although the drug labels for these agents warns that the risk of TBI and HBV reactivation may be increased, postmarketing data have not shown a significantly increased risk of infection with these agents [153,156]. Nevertheless, we continue to test for these infections, because the benefit of testing outweighs the harms.

Risankizumab (an IL-23 inhibitor) appears to have a lower risk for HBV and HCV reactivation compared with guselkumab or ustekinumab.

Pretreatment infectious testing – We test for TBI (algorithm 1), HBV (algorithm 3), and HCV (algorithm 4) in patients initiating guselkumab or ustekinumab (table 5). Since there are no reports of HBV or HCV reactivation in patients taking isankizumab, we do not test patients starting isankizumab for HBV or HCV.

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for patients starting IL-23 inhibitors, as discussed above (table 5). (See 'Timing of vaccinations' above.)

INTEGRIN INHIBITORS

Vedolizumab — Vedolizumab is a monoclonal antibody (mAb) that binds alpha 4 beta 7 integrin and prevents its interaction with MAdCAM-1, which hinders the migration of memory T cells into the gastrointestinal tract [157]. (See "Medical management of moderate to severe Crohn disease in adults" and "Management of moderate to severe ulcerative colitis in adults", section on 'Vedolizumab'.)

Risk of infection – In clinical trials and postmarketing studies, infection rates were not significantly higher in patients receiving vedolizumab compared with placebo [157-161]. However, certain serious infections have been rarely reported in those taking vedolizumab, including [71] (table 7):

Tuberculosis (TB) and other mycobacterial infections

Cytomegalovirus (CMV) disease (eg, colitis) and Epstein-Barr virus

Progressive multifocal leukoencephalopathy (PML)

Common viral and bacterial pathogens (eg, nasopharyngitis, sinopulmonary infections, gastroenteritis, perirectal abscesses) [162,163]

Vedolizumab does not seem to affect the incidence of complications due to SARS-CoV-2 infection [164].

Vedolizumab treatment should not be started in patients with severe infections nor be administered concomitantly with natalizumab or tumor necrosis factor (TNF)-alpha inhibitors due to additive infection risk [14].

Vedolizumab appears to carry a low risk for opportunistic infections. In a pooled analysis of six trials that included 2830 patients, there was no increased risk of serious infection associated with vedolizumab exposure compared with placebo [157]. Both TB and CMV infections were reported in 1 person per 1000 person-years in the vedolizumab group.

Pretreatment infectious testing – Prior to administering vedolizumab, we test for tuberculosis infection (TBI) (algorithm 1), as the benefits of TBI preemptive treatment in those who test positive outweigh the risks associated with testing (table 7). (See "Tuberculosis infection (latent tuberculosis) in adults: Approach to diagnosis (screening)".)

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) for patients starting vedolizumab, as discussed above (table 7) (see 'Timing of vaccinations' above). Administration of live vaccines before initiating vedolizumab is preferred. Live vaccines can be administered during therapy if the benefit outweighs the risk [14]. Vedolizumab does not appear to reduce parenterally delivered vaccine responses, but the response to oral vaccines may be lower [165,166].

Natalizumab — Natalizumab is a selective adhesion molecule inhibitor, functioning as a humanized mAb that binds to alpha 4 beta 1 integrins on nonneutrophil leukocytes. This prevents alpha 4 beta 1 from interacting with vascular cell adhesion molecule 1 (VCAM-1) found in blood vessel lumens. By blocking the interaction of alpha 4 beta 1 to VCAM-1, leukocyte migration across the blood-brain barrier and interactions with extracellular matrix proteins are inhibited [167,168].

Risk for infections – Patients treated with natalizumab are at increased risk of the following infections (table 7):

TB [169] and other mycobacterial infections

PML and John Cunningham virus (JCV; can occur in both seropositive and seronegative patients) [167,169,170]

Herpes simplex virus meningoencephalitis [169]

Varicella zoster virus (VZV) and other herpesviruses (eg, acute retinal necrosis) [171,172]

Hepatitis B virus (HBV) and hepatitis C virus (HCV) [173]

Aspergillosis [169]

Cryptococcal infection [169]

Common viral and bacterial infections (nasopharyngitis, upper respiratory tract infections, urinary tract infections) [169,174,175]

Natalizumab should not be administered to patients with severe infections nor in combination with other immunomodulatory therapies. Corticosteroids should be discontinued prior to natalizumab initiation. (See "Clinical use of monoclonal antibody disease-modifying therapies for multiple sclerosis", section on 'Natalizumab'.)

In a nationwide Swedish cohort of 1573 patients taking natalizumab, the risk of developing a serious infection within the first six years of treatment was 11.4 per 1000 person-years (95% CI 8.3-15.3) compared with the general population [170]. Although the risk is elevated, it was still lower than the infection risk associated with use of fingolimod or rituximab in this study. (See 'Sphingosine-1-phosphate receptor modulators (fingolimod)' below and 'Rituximab' above.)

Although rare, PML is a significant concern in patients taking natalizumab due to its high mortality rate and moderate to severe disability among survivors. The risk of natalizumab-associated PML for an individual patient varies according to JCV antibody status, prior immunosuppressant treatment, and the duration of natalizumab exposure (table 8). (See "Progressive multifocal leukoencephalopathy (PML): Treatment and prognosis", section on 'Morbidity and mortality'.)

Pretreatment infectious testing – We test for TBI (algorithm 1), HBV (algorithm 3), HCV (algorithm 4), and JCV prior to starting natalizumab (table 7) [169,176,177]. We also obtain baseline brain magnetic resonance imaging (MRI) prior to initiating therapy, continue to monitor JCV antibodies every six months, and repeat the brain MRI annually during therapy. If JCV antibody is positive at any time, some experts opt to not initiate or discontinue natalizumab due to the increased risk of PML incidence [167,169,170,178]. As an example, in a multinational observational study of over 6500 patients started on natalizumab, 44 (0.7 percent) developed PML over the five-year follow-up period [178]. Half of these patients had positive JCV antibodies recorded in the previous six months, while another 47 percent did not have JCV antibody results recorded.

Surveillance for PML in patients with a positive JCV antibody who remain on natalizumab is discussed separately. (See "Clinical use of monoclonal antibody disease-modifying therapies for multiple sclerosis", section on 'Surveillance for PML'.)

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and varicella/herpes zoster vaccines) in patients starting natalizumab, as discussed above (table 7). (See 'Timing of vaccinations' above.) Live vaccines should be avoided during the treatment period and for four months following the cessation of therapy [11].

Others — Etrolizumab is similar to vedolizumab in mechanism of action and increased susceptibility to infections (see 'Vedolizumab' above). Efalizumab is similar to natalizumab and also increases the risk of PML. It is no longer available in the United States or Canada due to fatal PML infections [179].

SPHINGOSINE-1-PHOSPHATE RECEPTOR MODULATORS (FINGOLIMOD) — Drugs belonging to the sphingosine-1-phosphate (S1P) modulators class include fingolimod, ozanimod, siponimod, and ponesimod. All of them have similar risk of infection; patients taking S1P receptor modulators other than fingolimod should be managed similar to patients taking fingolimod. (See "Overview of dosing and monitoring of biologic agents and small molecules for treating ulcerative colitis in adults", section on 'Sphingosine 1-phosphate (S1P) receptor modulators' and "Clinical use of oral disease-modifying therapies for multiple sclerosis", section on 'S1PR modulators'.)

Fingolimod binds to S1P receptors, downregulating S1P signaling to inhibit the exit of naive T cells and central memory T cells from lymph nodes [180,181]. Expression of cytokines on CD4+ T cells is also inhibited [181,182].

Risk of infection – Patients taking fingolimod are at increased risk for the following infections [183,184] (table 7):

Tuberculosis (TB) and other mycobacterial infections

Varicella zoster virus (VZV) and herpes simplex virus infections

Progressive multifocal leukoencephalopathy (PML)

Human herpesvirus-8-associated tumors

Cryptococcal infection

Common viral and bacterial infections (sinopulmonary infections, pneumonia, urinary tract infections, cellulitis)

A significant increase in risk for lower respiratory tract infections and herpesvirus infections was identified in clinical trials, with two fatal cases of disseminated VZV reported [185-188]. Patients should be carefully monitored during treatment for lower respiratory infections and herpesvirus infections [189,190].

Although risk of PML with fingolimod is lower than with natalizumab, cases of PML have been reported [191,192]. No fingolimod dose-dependent increase in infection risk has been noted [181]. Fingolimod should not be used with natalizumab due to increased risk of PML. (See 'Natalizumab' above and "Progressive multifocal leukoencephalopathy (PML): Epidemiology, clinical manifestations, and diagnosis".)

Pretreatment infectious testing – For patients starting S1P receptor modulators, we test for tuberculosis infection (TBI) (algorithm 1), hepatitis B virus (HBV) (algorithm 3), and hepatitis C virus (HCV) (algorithm 4 and table 7). We prefer to test for John Cunningham virus (JCV), similar to the approach used in patients receiving natalizumab, due to the increased risk for PML in patients receiving fingolimod [193]. However, testing for JCV is not routine among all clinicians, and no guidelines are available to guide testing. Fingolimod should be held if any signs or symptoms of PML develop while further work-up is pursued [193]. (See "Clinical use of monoclonal antibody disease-modifying therapies for multiple sclerosis", section on 'Surveillance for PML'.)

Vaccinations – We administer the routine and additional vaccinations (pneumococcal and herpes zoster vaccines) for patients starting S1P receptor modulators, as discussed above (table 7). (See 'Timing of vaccinations' above.)

COMPLEMENT PATHWAY INHIBITORS (ECULIZUMAB) — Eculizumab is a monoclonal antibody (mAb) directed against protein C5 of the membrane attack complex (MAC). The binding of eculizumab to C5 reduces the number of terminal complement components (C5b-9) that form the MAC, thereby decreasing the cytolysis of targeted encapsulated bacteria [194-196].

Risk of infection – Patients treated with eculizumab are at increased risk for (table 9):

Serious infections due to encapsulated bacteria (eg, Neisseria meningitidis, S. pneumoniae, and Haemophilus influenzae type b (Hib) [194,197]), particularly severe or fatal meningococcal infections [194,195]

Common bacterial and viral infections (eg, upper respiratory infections, urinary tract infections) [197]

The severity of SARS-CoV-2 infection does not appear to be higher in patients treated with eculizumab [198].

Pretreatment infectious testing – No testing is necessary for patients prior to starting eculizumab (table 9).

Vaccinations – Due to the high risk of bacterial infections, initial or booster vaccinations for N. meningitidis (both for ACWY serogroups and serogroup B) and S. pneumoniae are recommended at least two weeks before the start of treatment or as soon as possible (table 9). Children receiving eculizumab treatment may be at particularly increased risk for infection with S. pneumoniae or Hib, and age-appropriate vaccinations should be administered according to standard guidelines. (See "Pneumococcal vaccination in children" and "Pneumococcal vaccination in adults" and "Prevention of Haemophilus influenzae type b infection" and "Meningococcal vaccination in children and adults", section on 'Immunization of persons at increased risk'.)

Other considerations – Antibacterial prophylaxis with penicillin, or a macrolide if penicillin allergic, can be considered while receiving this treatment, but the risks and benefits are not well evaluated [197]. This is discussed in further detail elsewhere. (See "Treatment and prevention of meningococcal infection", section on 'Patients receiving C5 inhibitors'.)

Close monitoring for serious infections is warranted in all patients receiving eculizumab since infection is still possible (although less likely) in a patient who has been vaccinated and/or is receiving antimicrobial prophylaxis [195].

MONOCLONAL ANTIBODIES WITH NO KNOWN EFFECT ON THE IMMUNE SYSTEM — Certain monoclonal antibodies (mAbs) are designed to not affect the immune system and thus do not predispose the patients taking these agents to infections. Agents in this category include evolocumab, abciximab, and calcitonin gene-related peptide antagonists (eg, erenumab, galcanezumab, atogepant).

SUMMARY AND RECOMMENDATIONS

Immunologic mechanisms – Biologic agents are designed to interfere with the biological activity of a component of the immune system, typically a cytokine or a cellular receptor. By interfering with the normal activities of these molecules, biologic agents disrupt signaling pathways that drive activation and migration of immune cells to sites of infection, thereby interfering with the host immune response and increasing risk for infection. (See 'Immunologic mechanisms' above.)

Pretreatment testing for infections – Baseline testing for some latent infections is recommended prior to initiating therapy with many of the medications in this section (table 3 and table 4 and table 5 and table 6 and table 9 and table 7). In general, most of the biologic agents discussed here warrant testing for tuberculosis infection (TBI), hepatitis B virus (HBV), and hepatitis C virus (HCV). If any infections are discovered on pretreatment testing, we treat the underlying infection and hold off on starting the biologic agent until the infection is controlled. Close monitoring is paramount to make sure there is no worsening of the underlying infection once the biologic agent is started. (See 'Pretreatment infectious testing' above.)

Pretreatment vaccinations

We administer age-appropriate routine vaccinations (including SARS-CoV-2 vaccine series and annual influenza vaccination) to all patients starting biologic agents or a janus kinase (JAK) inhibitor prior to initiation of therapy when feasible, because vaccination may be less effective during and after therapy or may be contraindicated (eg, live vaccines) (table 3 and table 4 and table 5 and table 6 and table 9 and table 7).

For some drugs, pneumococcal and herpes zoster virus vaccines should also be administered prior to initiation of biologic agent, if possible (table 3 and table 4 and table 5 and table 6 and table 9 and table 7).

Live vaccines should generally be avoided during treatment with immunomodulatory agents. (See 'Prior to initiating biologics' above.)

Administration of vaccines during treatment – Vaccination with nonlive vaccines during therapy may be considered if the benefit of vaccination outweighs the risk. When possible, vaccination should be given at the nadir of immunosuppression. (See 'During treatment with biologics' above.)

  1. Kourbeti IS, Ziakas PD, Mylonakis E. Biologic therapies in rheumatoid arthritis and the risk of opportunistic infections: a meta-analysis. Clin Infect Dis 2014; 58:1649.
  2. Murdaca G, Spanò F, Contatore M, et al. Infection risk associated with anti-TNF-α agents: a review. Expert Opin Drug Saf 2015; 14:571.
  3. Komano Y, Tanaka M, Nanki T, et al. Incidence and risk factors for serious infection in patients with rheumatoid arthritis treated with tumor necrosis factor inhibitors: a report from the Registry of Japanese Rheumatoid Arthritis Patients for Longterm Safety. J Rheumatol 2011; 38:1258.
  4. Rahier JF, Magro F, Abreu C, et al. Second European evidence-based consensus on the prevention, diagnosis and management of opportunistic infections in inflammatory bowel disease. J Crohns Colitis 2014; 8:443.
  5. Weinblatt M, Combe B, Covucci A, et al. Safety of the selective costimulation modulator abatacept in rheumatoid arthritis patients receiving background biologic and nonbiologic disease-modifying antirheumatic drugs: A one-year randomized, placebo-controlled study. Arthritis Rheum 2006; 54:2807.
  6. Jyssum I, Kared H, Tran TT, et al. Humoral and cellular immune responses to two and three doses of SARS-CoV-2 vaccines in rituximab-treated patients with rheumatoid arthritis: a prospective, cohort study. Lancet Rheumatol 2022; 4:e177.
  7. van Kempen ZLE, Wieske L, Stalman EW, et al. Longitudinal humoral response after SARS-CoV-2 vaccination in ocrelizumab treated MS patients: To wait and repopulate? Mult Scler Relat Disord 2022; 57:103416.
  8. Boekel L, Steenhuis M, Hooijberg F, et al. Antibody development after COVID-19 vaccination in patients with autoimmune diseases in the Netherlands: a substudy of data from two prospective cohort studies. Lancet Rheumatol 2021; 3:e778.
  9. Bass AR, Chakravarty E, Akl EA, et al. 2022 American College of Rheumatology Guideline for Vaccinations in Patients With Rheumatic and Musculoskeletal Diseases. Arthritis Care Res (Hoboken) 2023; 75:449.
  10. Park JK, Lee MA, Lee EY, et al. Effect of methotrexate discontinuation on efficacy of seasonal influenza vaccination in patients with rheumatoid arthritis: a randomised clinical trial. Ann Rheum Dis 2017; 76:1559.
  11. Schwob JM, Samer CF, Lalive PH, Eperon GA. Live vaccines and immunosuppressive monoclonal antibodies: weighing up the benefit-risk assessment for natalizumab. J Travel Med 2021; 28.
  12. Tashtoush B, Okafor NC, Ramirez JF, Smolley L. Follicular Bronchiolitis: A Literature Review. J Clin Diagn Res 2015; 9:OE01.
  13. Heijstek MW, Pileggi GC, Zonneveld-Huijssoon E, et al. Safety of measles, mumps and rubella vaccination in juvenile idiopathic arthritis. Ann Rheum Dis 2007; 66:1384.
  14. Takeda Pharmaceuticals. Entyvio. Highlights of Prescribing Information 2014. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/125476s000lbl.pdf (Accessed on January 18, 2023).
  15. Food and Drug Administration. Secukinumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  16. Food and Drug Administration. Ixekizumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  17. Food and Drug Administration. Brodalumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  18. van de Kerkhof PC, Griffiths CE, Reich K, et al. Secukinumab long-term safety experience: A pooled analysis of 10 phase II and III clinical studies in patients with moderate to severe plaque psoriasis. J Am Acad Dermatol 2016; 75:83.
  19. Nash P, Kirkham B, Okada M, et al. Ixekizumab for the treatment of patients with active psoriatic arthritis and an inadequate response to tumour necrosis factor inhibitors: results from the 24-week randomised, double-blind, placebo-controlled period of the SPIRIT-P2 phase 3 trial. Lancet 2017; 389:2317.
  20. Papp KA, Leonardi C, Menter A, et al. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N Engl J Med 2012; 366:1181.
  21. Singh JA, Furst DE, Bharat A, et al. 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken) 2012; 64:625.
  22. Food and Drug Administration. Omalizumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  23. Loomba R, Liang TJ. Hepatitis B Reactivation Associated With Immune Suppressive and Biological Modifier Therapies: Current Concepts, Management Strategies, and Future Directions. Gastroenterology 2017; 152:1297.
  24. Cowley NJ, Owen A, Shiels SC, et al. Safety and Efficacy of Antiviral Therapy for Prevention of Cytomegalovirus Reactivation in Immunocompetent Critically Ill Patients: A Randomized Clinical Trial. JAMA Intern Med 2017; 177:774.
  25. 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; 57:1431.
  26. Kim PS, Ho GY, Prete PE, Furst DE. Safety and efficacy of abatacept in eight rheumatoid arthritis patients with chronic hepatitis B. Arthritis Care Res (Hoboken) 2012; 64:1265.
  27. Talotta R, Atzeni F, Sarzi Puttini P. Reactivation of occult hepatitis B virus infection under treatment with abatacept: a case report. BMC Pharmacol Toxicol 2016; 17:17.
  28. Germanidis G, Hytiroglou P, Zakalka M, Settas L. Reactivation of occult hepatitis B virus infection, following treatment of refractory rheumatoid arthritis with abatacept. J Hepatol 2012; 56:1420.
  29. Fanouriakis A, Vassilopoulos D, Repa A, et al. Hepatitis B reactivation following treatment with abatacept in a patient with past hepatitis B virus infection. Rheumatology (Oxford) 2014; 53:195.
  30. Salliot C, Dougados M, Gossec L. Risk of serious infections during rituximab, abatacept and anakinra treatments for rheumatoid arthritis: meta-analyses of randomised placebo-controlled trials. Ann Rheum Dis 2009; 68:25.
  31. Weinblatt ME, Moreland LW, Westhovens R, et al. Safety of abatacept administered intravenously in treatment of rheumatoid arthritis: integrated analyses of up to 8 years of treatment from the abatacept clinical trial program. J Rheumatol 2013; 40:787.
  32. Yun H, Xie F, Delzell E, et al. Comparative Risk of Hospitalized Infection Associated With Biologic Agents in Rheumatoid Arthritis Patients Enrolled in Medicare. Arthritis Rheumatol 2016; 68:56.
  33. Genovese MC, Schiff M, Luggen M, et al. Efficacy and safety of the selective co-stimulation modulator abatacept following 2 years of treatment in patients with rheumatoid arthritis and an inadequate response to anti-tumour necrosis factor therapy. Ann Rheum Dis 2008; 67:547.
  34. Onrust SV, Lamb HM, Balfour JA. Rituximab. Drugs 1999; 58:79.
  35. Wallace DJ, Navarra S, Petri MA, et al. Safety profile of belimumab: pooled data from placebo-controlled phase 2 and 3 studies in patients with systemic lupus erythematosus. Lupus 2013; 22:144.
  36. Chen DR, Cohen PL. Living life without B cells: is repeated B-cell depletion a safe and effective long-term treatment plan for rheumatoid arthritis? Int J Clin Rheumtol 2012; 7:159.
  37. Lanini S, Molloy AC, Prentice AG, et al. Infections in patients taking Rituximab for hematologic malignancies: two-year cohort study. BMC Infect Dis 2013; 13:317.
  38. Emery P, Rigby W, Tak PP, et al. Safety with ocrelizumab in rheumatoid arthritis: results from the ocrelizumab phase III program. PLoS One 2014; 9:e87379.
  39. Kappos L, Li D, Calabresi PA, et al. Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised, placebo-controlled, multicentre trial. Lancet 2011; 378:1779.
  40. Coiffier B, Lepretre S, Pedersen LM, et al. Safety and efficacy of ofatumumab, a fully human monoclonal anti-CD20 antibody, in patients with relapsed or refractory B-cell chronic lymphocytic leukemia: a phase 1-2 study. Blood 2008; 111:1094.
  41. Marcus R, Davies A, Ando K, et al. Obinutuzumab for the First-Line Treatment of Follicular Lymphoma. N Engl J Med 2017; 377:1331.
  42. Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 2009; 113:4834.
  43. Epstein DJ, Dunn J, Deresinski S. Infectious Complications of Multiple Sclerosis Therapies: Implications for Screening, Prophylaxis, and Management. Open Forum Infect Dis 2018; 5:ofy174.
  44. Garcia-Rodriguez MJ, Canales MA, Hernandez-Maraver D, Hernandez-Navarro F. Late reactivation of resolved hepatitis B virus infection: an increasing complication post rituximab-based regimens treatment? Am J Hematol 2008; 83:673.
  45. Simpson-Yap S, De Brouwer E, Kalincik T, et al. Associations of Disease-Modifying Therapies With COVID-19 Severity in Multiple Sclerosis. Neurology 2021; 97:e1870.
  46. Andersen KM, Bates BA, Rashidi ES, et al. Long-term use of immunosuppressive medicines and in-hospital COVID-19 outcomes: a retrospective cohort study using data from the National COVID Cohort Collaborative. Lancet Rheumatol 2022; 4:e33.
  47. Hughes R, Whitley L, Fitovski K, et al. COVID-19 in ocrelizumab-treated people with multiple sclerosis. Mult Scler Relat Disord 2021; 49:102725.
  48. Kimby E. Tolerability and safety of rituximab (MabThera). Cancer Treat Rev 2005; 31:456.
  49. Seery N, Sharmin S, Li V, et al. Predicting Infection Risk in Multiple Sclerosis Patients Treated with Ocrelizumab: A Retrospective Cohort Study. CNS Drugs 2021; 35:907.
  50. Brunnberg U, Hentrich M, Hoffmann C, et al. HIV-Associated Malignant Lymphoma. Oncol Res Treat 2017; 40:82.
  51. Aksoy S, Dizdar O, Hayran M, Harputluoğlu H. Infectious complications of rituximab in patients with lymphoma during maintenance therapy: a systematic review and meta-analysis. Leuk Lymphoma 2009; 50:357.
  52. Grim SA, Pham T, Thielke J, et al. Infectious complications associated with the use of rituximab for ABO-incompatible and positive cross-match renal transplant recipients. Clin Transplant 2007; 21:628.
  53. Kamar N, Milioto O, Puissant-Lubrano B, et al. Incidence and predictive factors for infectious disease after rituximab therapy in kidney-transplant patients. Am J Transplant 2010; 10:89.
  54. van Vollenhoven RF, Fleischmann RM, Furst DE, et al. Longterm Safety of Rituximab: Final Report of the Rheumatoid Arthritis Global Clinical Trial Program over 11 Years. J Rheumatol 2015; 42:1761.
  55. Mikulska M, Lanini S, Gudiol C, et al. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Agents targeting lymphoid cells surface antigens [I]: CD19, CD20 and CD52). Clin Microbiol Infect 2018; 24 Suppl 2:S71.
  56. Kapoor T, Mahadeshwar P, Hui-Yuen J, et al. Prevalence of progressive multifocal leukoencephalopathy (PML) in adults and children with systemic lupus erythematosus. Lupus Sci Med 2020; 7.
  57. 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; 62:75.
  58. Thiel J, Rizzi M, Engesser M, et al. B cell repopulation kinetics after rituximab treatment in ANCA-associated vasculitides compared to rheumatoid arthritis, and connective tissue diseases: a longitudinal observational study on 120 patients. Arthritis Res Ther 2017; 19:101.
  59. Pescovitz MD, Torgerson TR, Ochs HD, et al. Effect of rituximab on human in vivo antibody immune responses. J Allergy Clin Immunol 2011; 128:1295.
  60. Sorensen PS, Blinkenberg M. The potential role for ocrelizumab in the treatment of multiple sclerosis: current evidence and future prospects. Ther Adv Neurol Disord 2016; 9:44.
  61. Mulero P, Midaglia L, Montalban X. Ocrelizumab: a new milestone in multiple sclerosis therapy. Ther Adv Neurol Disord 2018; 11:1756286418773025.
  62. Patel A, Sul J, Gordon ML, et al. Progressive Multifocal Leukoencephalopathy in a Patient With Progressive Multiple Sclerosis Treated With Ocrelizumab Monotherapy. JAMA Neurol 2021; 78:736.
  63. Zhang B. Ofatumumab. MAbs 2009; 1:326.
  64. Bossen C, Schneider P. BAFF, APRIL and their receptors: structure, function and signaling. Semin Immunol 2006; 18:263.
  65. Ugarte-Gil MF, Alarcón GS, Izadi Z, et al. Characteristics associated with poor COVID-19 outcomes in individuals with systemic lupus erythematosus: data from the COVID-19 Global Rheumatology Alliance. Ann Rheum Dis 2022; 81:970.
  66. Fabris M, De Marchi G, Domenis R, et al. High T-cell response rate after COVID-19 vaccination in belimumab and rituximab recipients. J Autoimmun 2022; 129:102827.
  67. O'Shea JJ, Kontzias A, Yamaoka K, et al. Janus kinase inhibitors in autoimmune diseases. Ann Rheum Dis 2013; 72 Suppl 2:ii111.
  68. Food and Drug Administration. Tofacitinib Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  69. Food and Drug Administration. MedWatch: The FDA Safety Information and Adverse Event Reporting Program https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  70. FitzGerald JM, Bleecker ER, Nair P, et al. Benralizumab, an anti-interleukin-5 receptor α monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2016; 388:2128.
  71. Taylor PC, Keystone EC, van der Heijde D, et al. Baricitinib versus Placebo or Adalimumab in Rheumatoid Arthritis. N Engl J Med 2017; 376:652.
  72. Dougados M, van der Heijde D, Chen YC, et al. Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: results from the RA-BUILD study. Ann Rheum Dis 2017; 76:88.
  73. Genovese MC, Kremer J, Zamani O, et al. Baricitinib in Patients with Refractory Rheumatoid Arthritis. N Engl J Med 2016; 374:1243.
  74. Curtis JR, Xie F, Yun H, et al. Real-world comparative risks of herpes virus infections in tofacitinib and biologic-treated patients with rheumatoid arthritis. Ann Rheum Dis 2016; 75:1843.
  75. Winthrop KL, Wouters AG, Choy EH, et al. The Safety and Immunogenicity of Live Zoster Vaccination in Patients With Rheumatoid Arthritis Before Starting Tofacitinib: A Randomized Phase II Trial. Arthritis Rheumatol 2017; 69:1969.
  76. Winthrop KL, Curtis JR, Lindsey S, et al. Herpes Zoster and Tofacitinib: Clinical Outcomes and the Risk of Concomitant Therapy. Arthritis Rheumatol 2017; 69:1960.
  77. Hopman RK, Lawrence SJ, Oh ST. Disseminated tuberculosis associated with ruxolitinib. Leukemia 2014; 28:1750.
  78. Iwamoto N, Tsuji S, Takatani A, et al. Efficacy and safety at 24 weeks of daily clinical use of tofacitinib in patients with rheumatoid arthritis. PLoS One 2017; 12:e0177057.
  79. Chen YM, Huang WN, Wu YD, et al. Reactivation of hepatitis B virus infection in patients with rheumatoid arthritis receiving tofacitinib: a real-world study. Ann Rheum Dis 2018; 77:780.
  80. Kusne Y, Kimes KE, Feller FF, et al. Coccidioidomycosis in Patients Treated With Ruxolitinib. Open Forum Infect Dis 2020; 7:ofaa167.
  81. Kubo S, Nakayamada S, Sakata K, et al. Janus Kinase Inhibitor Baricitinib Modulates Human Innate and Adaptive Immune System. Front Immunol 2018; 9:1510.
  82. Winthrop KL, Silverfield J, Racewicz A, et al. The effect of tofacitinib on pneumococcal and influenza vaccine responses in rheumatoid arthritis. Ann Rheum Dis 2016; 75:687.
  83. Genovese MC, Cohen S, Moreland L, et al. Combination therapy with etanercept and anakinra in the treatment of patients with rheumatoid arthritis who have been treated unsuccessfully with methotrexate. Arthritis Rheum 2004; 50:1412.
  84. Food and Drug Administration. Anakinra Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  85. Mertens M, Singh JA. Anakinra for rheumatoid arthritis: a systematic review. J Rheumatol 2009; 36:1118.
  86. Fleischmann RM, Tesser J, Schiff MH, et al. Safety of extended treatment with anakinra in patients with rheumatoid arthritis. Ann Rheum Dis 2006; 65:1006.
  87. Food and Drug Administration. Canakinumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  88. Lachmann HJ, Lauwerys B, Miettunen P, et al. Canakinumab improves patient-reported outcomes in children and adults with autoinflammatory recurrent fever syndromes: results from the CLUSTER trial. Clin Exp Rheumatol 2021; 39 Suppl 132:51.
  89. Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, et al. Use of canakinumab in the cryopyrin-associated periodic syndrome. N Engl J Med 2009; 360:2416.
  90. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med 2017; 377:1119.
  91. Gillespie J, Mathews R, McDermott MF. Rilonacept in the management of cryopyrin-associated periodic syndromes (CAPS). J Inflamm Res 2010; 3:1.
  92. Hoffman HM, Throne ML, Amar NJ, et al. Efficacy and safety of rilonacept (interleukin-1 Trap) in patients with cryopyrin-associated periodic syndromes: results from two sequential placebo-controlled studies. Arthritis Rheum 2008; 58:2443.
  93. Food and Drug Administration. Rilonacept Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  94. Thibodeaux Q, Smith MP, Ly K, et al. A review of dupilumab in the treatment of atopic diseases. Hum Vaccin Immunother 2019; 15:2129.
  95. Food and Drug Administration. Dupilumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  96. Blauvelt A, Guttman-Yassky E, Paller AS, et al. Long-Term Efficacy and Safety of Dupilumab in Adolescents with Moderate-to-Severe Atopic Dermatitis: Results Through Week 52 from a Phase III Open-Label Extension Trial (LIBERTY AD PED-OLE). Am J Clin Dermatol 2022; 23:365.
  97. Schneeweiss MC, Kim SC, Wyss R, et al. Dupilumab and the risk of conjunctivitis and serious infection in patients with atopic dermatitis: A propensity score-matched cohort study. J Am Acad Dermatol 2021; 84:300.
  98. Simpson EL, Bieber T, Guttman-Yassky E, et al. Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N Engl J Med 2016; 375:2335.
  99. Patruno C, Stingeni L, Fabbrocini G, et al. Dupilumab and COVID-19: What should we expect? Dermatol Ther 2020; 33:e13502.
  100. El-Qushayri AE, Mahmoud MA, Salman S, et al. Dupilumab therapy in atopic dermatitis is safe during COVID-19 infection era: A systematic review and meta-analysis of 1611 patients. Dermatol Ther 2022; 35:e15476.
  101. Ungar B, Glickman JW, Golant AK, et al. COVID-19 Symptoms Are Attenuated in Moderate-to-Severe Atopic Dermatitis Patients Treated with Dupilumab. J Allergy Clin Immunol Pract 2022; 10:134.
  102. Matsutani M, Imai Y, Nakatani-Kusakabe M, et al. Dupilumab in atopic dermatitis patients with chronic hepatitis B. Journal of Cutaneous Immunology and Allergy 2021; 5:65.
  103. Mota F. Atopic Dermatitis Patient With Hepatitis C Treated With Dupilumab-A Case Report. Actas Dermosifiliogr 2023.
  104. Adam DN, Gooderham MJ, Beecker JR, et al. Expert consensus on the systemic treatment of atopic dermatitis in special populations. J Eur Acad Dermatol Venereol 2023; 37:1135.
  105. Castro M, Zangrilli J, Wechsler ME, et al. Reslizumab for inadequately controlled asthma with elevated blood eosinophil counts: results from two multicentre, parallel, double-blind, randomised, placebo-controlled, phase 3 trials. Lancet Respir Med 2015; 3:355.
  106. Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet 2012; 380:651.
  107. Ferguson GT, FitzGerald JM, Bleecker ER, et al. Benralizumab for patients with mild to moderate, persistent asthma (BISE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med 2017; 5:568.
  108. Navinés-Ferrer A, Serrano-Candelas E, Molina-Molina GJ, Martín M. IgE-Related Chronic Diseases and Anti-IgE-Based Treatments. J Immunol Res 2016; 2016:8163803.
  109. Bleecker ER, FitzGerald JM, Chanez P, et al. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting β2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial. Lancet 2016; 388:2115.
  110. Busse WW, Bleecker ER, FitzGerald JM, et al. Long-term safety and efficacy of benralizumab in patients with severe, uncontrolled asthma: 1-year results from the BORA phase 3 extension trial. Lancet Respir Med 2019; 7:46.
  111. Harrison TW, Chanez P, Menzella F, et al. Onset of effect and impact on health-related quality of life, exacerbation rate, lung function, and nasal polyposis symptoms for patients with severe eosinophilic asthma treated with benralizumab (ANDHI): a randomised, controlled, phase 3b trial. Lancet Respir Med 2021; 9:260.
  112. Nair P, Wenzel S, Rabe KF, et al. Oral Glucocorticoid-Sparing Effect of Benralizumab in Severe Asthma. N Engl J Med 2017; 376:2448.
  113. Haldar P, Brightling CE, Hargadon B, et al. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med 2009; 360:973.
  114. Ortega HG, Liu MC, Pavord ID, et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N Engl J Med 2014; 371:1198.
  115. Bel EH, Wenzel SE, Thompson PJ, et al. Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. N Engl J Med 2014; 371:1189.
  116. Jackson DJ, Bacharier LB, Gergen PJ, et al. Mepolizumab for urban children with exacerbation-prone eosinophilic asthma in the USA (MUPPITS-2): a randomised, double-blind, placebo-controlled, parallel-group trial. Lancet 2022; 400:502.
  117. Pavord ID, Chanez P, Criner GJ, et al. Mepolizumab for Eosinophilic Chronic Obstructive Pulmonary Disease. N Engl J Med 2017; 377:1613.
  118. Chupp GL, Bradford ES, Albers FC, et al. Efficacy of mepolizumab add-on therapy on health-related quality of life and markers of asthma control in severe eosinophilic asthma (MUSCA): a randomised, double-blind, placebo-controlled, parallel-group, multicentre, phase 3b trial. Lancet Respir Med 2017; 5:390.
  119. Han JK, Bachert C, Fokkens W, et al. Mepolizumab for chronic rhinosinusitis with nasal polyps (SYNAPSE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med 2021; 9:1141.
  120. Bjermer L, Lemiere C, Maspero J, et al. Reslizumab for Inadequately Controlled Asthma With Elevated Blood Eosinophil Levels: A Randomized Phase 3 Study. Chest 2016; 150:789.
  121. Corren J, Weinstein S, Janka L, et al. Phase 3 Study of Reslizumab in Patients With Poorly Controlled Asthma: Effects Across a Broad Range of Eosinophil Counts. Chest 2016; 150:799.
  122. Murphy K, Jacobs J, Bjermer L, et al. Long-term Safety and Efficacy of Reslizumab in Patients with Eosinophilic Asthma. J Allergy Clin Immunol Pract 2017; 5:1572.
  123. Busse W, Corren J, Lanier BQ, et al. Omalizumab, anti-IgE recombinant humanized monoclonal antibody, for the treatment of severe allergic asthma. J Allergy Clin Immunol 2001; 108:184.
  124. Busse WW, Morgan WJ, Gergen PJ, et al. Randomized trial of omalizumab (anti-IgE) for asthma in inner-city children. N Engl J Med 2011; 364:1005.
  125. Maurer M, Rosén K, Hsieh HJ, et al. Omalizumab for the treatment of chronic idiopathic or spontaneous urticaria. N Engl J Med 2013; 368:924.
  126. Solèr M, Matz J, Townley R, et al. The anti-IgE antibody omalizumab reduces exacerbations and steroid requirement in allergic asthmatics. Eur Respir J 2001; 18:254.
  127. Milgrom H, Berger W, Nayak A, et al. Treatment of childhood asthma with anti-immunoglobulin E antibody (omalizumab). Pediatrics 2001; 108:E36.
  128. Holgate ST, Chuchalin AG, Hébert J, et al. Efficacy and safety of a recombinant anti-immunoglobulin E antibody (omalizumab) in severe allergic asthma. Clin Exp Allergy 2004; 34:632.
  129. Cruz AA, Lima F, Sarinho E, et al. Safety of anti-immunoglobulin E therapy with omalizumab in allergic patients at risk of geohelminth infection. Clin Exp Allergy 2007; 37:197.
  130. Wu Y, Li JJ, Kim HJ, et al. A Neutralizing Antibody Assay Based on a Reporter of Antibody-Dependent Cell-Mediated Cytotoxicity. AAPS J 2015; 17:1417.
  131. Ghassemian A, Park JJ, Tsoulis MW, Kim H. Targeting the IL-5 pathway in eosinophilic asthma: a comparison of mepolizumab to benralizumab in the reduction of peripheral eosinophil counts. Allergy Asthma Clin Immunol 2021; 17:3.
  132. Lifar P, Montastruc F, Reber LL, et al. Parasitic Infections and Biological Therapies Targeting Type 2 Inflammation: A VigiBase Study. Am J Respir Crit Care Med 2023; 207:1253.
  133. Licari A, Castagnoli R, Votto M, et al. Biologic Use in Allergic and Asthmatic Children and Adolescents During the COVID-19 Pandemic. Pediatr Allergy Immunol Pulmonol 2020; 33:155.
  134. Aksu K, Yesilkaya S, Topel M, et al. COVID-19 in a patient with severe asthma using mepolizumab. Allergy Asthma Proc 2021; 42:e55.
  135. Papaioannou AI, Fouka E, Tzanakis N, et al. SARS-Cov-2 Infection in Severe Asthma Patients Treated With Biologics. J Allergy Clin Immunol Pract 2022; 10:2588.
  136. Passante M, Napolitano M, Dastoli S, et al. Safety of omalizumab treatment in patients with chronic spontaneous urticaria and COVID-19. Dermatol Ther 2021; 34:e15111.
  137. Xolair. https://www.xolairhcp.com/?adobe_mc=MCMID%3D25061105514458683721135041519440244321%7CMCORGID%3DDF784CF658BD66380A495D3E%2540AdobeOrg%7CTS%3D1685127355 (Accessed on May 29, 2023).
  138. Lai Y, Dong C. Therapeutic antibodies that target inflammatory cytokines in autoimmune diseases. Int Immunol 2016; 28:181.
  139. Food and Drug Administration. Sarilumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  140. Food and Drug Administration. Tocilizumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 20, 2023).
  141. Fleischmann R, Genovese MC, Lin Y, et al. Long-term safety of sarilumab in rheumatoid arthritis: an integrated analysis with up to 7 years' follow-up. Rheumatology (Oxford) 2020; 59:292.
  142. Emery P, Rondon J, Parrino J, et al. Safety and tolerability of subcutaneous sarilumab and intravenous tocilizumab in patients with rheumatoid arthritis. Rheumatology (Oxford) 2019; 58:849.
  143. Morel J, Constantin A, Baron G, et al. Risk factors of serious infections in patients with rheumatoid arthritis treated with tocilizumab in the French Registry REGATE. Rheumatology (Oxford) 2017; 56:1746.
  144. De Benedetti F, Brunner HI, Ruperto N, et al. Randomized trial of tocilizumab in systemic juvenile idiopathic arthritis. N Engl J Med 2012; 367:2385.
  145. Burmester GR, Rubbert-Roth A, Cantagrel A, et al. A randomised, double-blind, parallel-group study of the safety and efficacy of subcutaneous tocilizumab versus intravenous tocilizumab in combination with traditional disease-modifying antirheumatic drugs in patients with moderate to severe rheumatoid arthritis (SUMMACTA study). Ann Rheum Dis 2014; 73:69.
  146. Schiff MH, Kremer JM, Jahreis A, et al. Integrated safety in tocilizumab clinical trials. Arthritis Res Ther 2011; 13:R141.
  147. Stone JH, Tuckwell K, Dimonaco S, et al. Trial of Tocilizumab in Giant-Cell Arteritis. N Engl J Med 2017; 377:317.
  148. Lauper K, Kearsley-Fleet L, Galloway JB, et al. Evaluation of serious infections, including Mycobacterium tuberculosis, during treatment with biologic disease-modifying antirheumatic drugs: does line of therapy matter? Rheumatology (Oxford) 2023.
  149. Genovese MC, Fleischmann R, Kivitz AJ, et al. Sarilumab Plus Methotrexate in Patients With Active Rheumatoid Arthritis and Inadequate Response to Methotrexate: Results of a Phase III Study. Arthritis Rheumatol 2015; 67:1424.
  150. Huizinga TW, Fleischmann RM, Jasson M, et al. Sarilumab, a fully human monoclonal antibody against IL-6Rα in patients with rheumatoid arthritis and an inadequate response to methotrexate: efficacy and safety results from the randomised SARIL-RA-MOBILITY Part A trial. Ann Rheum Dis 2014; 73:1626.
  151. Katelani S, Fragoulis GE, Bakasis AD, et al. HBV reactivation in patients with rheumatoid arthritis treated with anti-interleukin-6: a systematic review and meta-analysis. Rheumatology (Oxford) 2023; 62:SI252.
  152. Bevans SL, Mayo TT, Elewski BE. Safety of secukinumab in hepatitis B virus. J Eur Acad Dermatol Venereol 2018; 32:e120.
  153. Nogueira M, Warren RB, Torres T. Risk of tuberculosis reactivation with interleukin (IL)-17 and IL-23 inhibitors in psoriasis - time for a paradigm change. J Eur Acad Dermatol Venereol 2021; 35:824.
  154. Food and Drug Administration. Guselkumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  155. Food and Drug Administration. Stelara (Ustekinumab) Injection, for Subcutaneous or Intravenous. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  156. Papp KA, Griffiths CE, Gordon K, et al. Long-term safety of ustekinumab in patients with moderate-to-severe psoriasis: final results from 5 years of follow-up. Br J Dermatol 2013; 168:844.
  157. Colombel JF, Sands BE, Rutgeerts P, et al. The safety of vedolizumab for ulcerative colitis and Crohn's disease. Gut 2017; 66:839.
  158. Feagan BG, Bhayat F, Khalid M, et al. Respiratory Tract Infections in Patients With Inflammatory Bowel Disease: Safety Analyses From Vedolizumab Clinical Trials. J Crohns Colitis 2018; 12:905.
  159. Feagan BG, Rutgeerts P, Sands BE, et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2013; 369:699.
  160. Sandborn WJ, Feagan BG, Rutgeerts P, et al. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med 2013; 369:711.
  161. Sands BE, Feagan BG, Rutgeerts P, et al. Effects of vedolizumab induction therapy for patients with Crohn's disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology 2014; 147:618.
  162. Cohen RD, Bhayat F, Blake A, Travis S. The Safety Profile of Vedolizumab in Ulcerative Colitis and Crohn's Disease: 4 Years of Global Post-marketing Data. J Crohns Colitis 2020; 14:192.
  163. Loftus EV Jr, Feagan BG, Panaccione R, et al. Long-term safety of vedolizumab for inflammatory bowel disease. Aliment Pharmacol Ther 2020; 52:1353.
  164. Agrawal M, Zhang X, Brenner EJ, et al. The Impact of Vedolizumab on COVID-19 Outcomes Among Adult IBD Patients in the SECURE-IBD Registry. J Crohns Colitis 2021; 15:1877.
  165. Wyant T, Leach T, Sankoh S, et al. Vedolizumab affects antibody responses to immunisation selectively in the gastrointestinal tract: randomised controlled trial results. Gut 2015; 64:77.
  166. Alexander JL, Kennedy NA, Ibraheim H, et al. COVID-19 vaccine-induced antibody responses in immunosuppressed patients with inflammatory bowel disease (VIP): a multicentre, prospective, case-control study. Lancet Gastroenterol Hepatol 2022; 7:342.
  167. Hutchinson M. Natalizumab: A new treatment for relapsing remitting multiple sclerosis. Ther Clin Risk Manag 2007; 3:259.
  168. Yaldizli O, Putzki N. Natalizumab in the treatment of multiple sclerosis. Ther Adv Neurol Disord 2009; 2:115.
  169. Singer BA. The role of natalizumab in the treatment of multiple sclerosis: benefits and risks. Ther Adv Neurol Disord 2017; 10:327.
  170. Luna G, Alping P, Burman J, et al. Infection Risks Among Patients With Multiple Sclerosis Treated With Fingolimod, Natalizumab, Rituximab, and Injectable Therapies. JAMA Neurol 2020; 77:184.
  171. Fine AJ, Sorbello A, Kortepeter C, Scarazzini L. Central nervous system herpes simplex and varicella zoster virus infections in natalizumab-treated patients. Clin Infect Dis 2013; 57:849.
  172. Sood AB, Kumar G, Robinson J. Bilateral acute retinal necrosis in a patient with multiple sclerosis on natalizumab. J Ophthalmic Inflamm Infect 2016; 6:26.
  173. Hillen ME, Cook SD, Samanta A, et al. Fatal acute liver failure with hepatitis B virus infection during nataluzimab treatment in multiple sclerosis. Neurol Neuroimmunol Neuroinflamm 2015; 2:e72.
  174. Shirani A, Stüve O. Natalizumab: Perspectives from the Bench to Bedside. Cold Spring Harb Perspect Med 2018; 8.
  175. Kapoor R, Ho PR, Campbell N, et al. Effect of natalizumab on disease progression in secondary progressive multiple sclerosis (ASCEND): a phase 3, randomised, double-blind, placebo-controlled trial with an open-label extension. Lancet Neurol 2018; 17:405.
  176. Benkert TF, Dietz L, Hartmann EM, et al. Natalizumab exerts direct signaling capacity and supports a pro-inflammatory phenotype in some patients with multiple sclerosis. PLoS One 2012; 7:e52208.
  177. Bloomgren G, Richman S, Hotermans C, et al. Risk of natalizumab-associated progressive multifocal leukoencephalopathy. N Engl J Med 2012; 366:1870.
  178. Foley J, Carrillo-Infante C, Smith J, et al. The 5-year Tysabri global observational program in safety (TYGRIS) study confirms the long-term safety profile of natalizumab treatment in multiple sclerosis. Mult Scler Relat Disord 2020; 39:101863.
  179. Major EO. Progressive multifocal leukoencephalopathy in patients on immunomodulatory therapies. Annu Rev Med 2010; 61:35.
  180. Ingwersen J, Aktas O, Kuery P, et al. Fingolimod in multiple sclerosis: mechanisms of action and clinical efficacy. Clin Immunol 2012; 142:15.
  181. Zhao Z, Ma CL, Gu ZC, et al. Incidence and Risk of Infection Associated With Fingolimod in Patients With Multiple Sclerosis: A Systematic Review and Meta-Analysis of 8,448 Patients From 12 Randomized Controlled Trials. Front Immunol 2021; 12:611711.
  182. Chun J, Hartung HP. Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis. Clin Neuropharmacol 2010; 33:91.
  183. Grebenciucova E, Reder AT, Bernard JT. Immunologic mechanisms of fingolimod and the role of immunosenescence in the risk of cryptococcal infection: A case report and review of literature. Mult Scler Relat Disord 2016; 9:158.
  184. Samudralwar RD, Spec A, Cross AH. Case Report: Fingolimod and Cryptococcosis: Collision of Immunomodulation with Infectious Disease. Int J MS Care 2019; 21:275.
  185. Grebenciucova E, Pruitt A. Infections in Patients Receiving Multiple Sclerosis Disease-Modifying Therapies. Curr Neurol Neurosci Rep 2017; 17:88.
  186. Cohen JA, Barkhof F, Comi G, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med 2010; 362:402.
  187. Winkelmann A, Loebermann M, Reisinger EC, et al. Disease-modifying therapies and infectious risks in multiple sclerosis. Nat Rev Neurol 2016; 12:217.
  188. Issa NP, Hentati A. VZV encephalitis that developed in an immunized patient during fingolimod therapy. Neurology 2015; 84:99.
  189. Fazekas F, Berger T, Fabjan TH, et al. Fingolimod in the treatment algorithm of relapsing remitting multiple sclerosis: a statement of the Central and East European (CEE) MS Expert Group. Wien Med Wochenschr 2012; 162:354.
  190. Signoriello E, Bonavita S, Sinisi L, et al. Is antibody titer useful to verify the immunization after VZV Vaccine in MS patients treated with Fingolimod? A case series. Mult Scler Relat Disord 2020; 40:101963.
  191. Gyang TV, Hamel J, Goodman AD, et al. Fingolimod-associated PML in a patient with prior immunosuppression. Neurology 2016; 86:1843.
  192. Faulkner M. Risk of progressive multifocal leukoencephalopathy in patients with multiple sclerosis. Expert Opin Drug Saf 2015; 14:1737.
  193. McGuigan C, Craner M, Guadagno J, et al. Stratification and monitoring of natalizumab-associated progressive multifocal leukoencephalopathy risk: recommendations from an expert group. J Neurol Neurosurg Psychiatry 2016; 87:117.
  194. McNamara LA, Topaz N, Wang X, et al. High Risk for Invasive Meningococcal Disease Among Patients Receiving Eculizumab (Soliris) Despite Receipt of Meningococcal Vaccine. MMWR Morb Mortal Wkly Rep 2017; 66:734.
  195. Parikh SR, Lucidarme J, Bingham C, et al. Meningococcal B Vaccine Failure With a Penicillin-Resistant Strain in a Young Adult on Long-Term Eculizumab. Pediatrics 2017; 140.
  196. Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med 2013; 368:2169.
  197. Food and Drug Administration. Eculizumab Highlights of Prescribing Information. https://www.fda.gov/safety/medwatch-fda-safety-information-and-adverse-event-reporting-program (Accessed on January 18, 2023).
  198. Mastellos DC, Pires da Silva BGP, Fonseca BAL, et al. Complement C3 vs C5 inhibition in severe COVID-19: Early clinical findings reveal differential biological efficacy. Clin Immunol 2020; 220:108598.
Topic 127104 Version 1.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟