Acessibilidade / Reportar erro

Reducing infection risk in multiple sclerosis and neuromyelitis optica spectrum disorders: a Brazilian reference center's approach

Redução do risco de infeção em esclerose múltipla e doença do espectro neuromielite óptica: abordagem de um centro de referência brasileiro

Abstract

Background

Multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD) are the most common autoimmune diseases of the central nervous system (CNS). They present chronic relapsing courses that demand treatment with disease-modifying drugs (DMDs) to prevent inflammatory activity. Disease-modifying drugs lead to immunomodulation or immunosuppression through diverse mechanisms (e.g., shifting lymphocyte and cytokine profile, suppressing specific lymphocyte subpopulations). Thus, patients are more prone to infectious complications and associated worsening of disease.

Objective

To present feasible strategies for mitigating the infection risk of MS and NMOSD treated patients.

Methods

Targeted literature review concerning the management of infection risk with an emphasis on vaccination, therapy-specific measures, and particularities of the Brazilian endemic infectious diseases' scenario.

Conclusion

We propose a vaccination schedule, infectious screening routine, and prophylactic measures based on the current scientific evidence. Awareness of emergent tropical diseases is necessary due to evidence of demyelinating events and possible parainfectious cases of MS and NMOSD.

Keywords
Multiple Sclerosis; Neuromyelitis Optica; Vaccines

Resumo

Antecedentes

A esclerose múltipla (EM) e a doença do espectro neuromielite optica (NMOSD) são as doenças autoimunes mais comuns do sistema nervoso central (SNC). Ambas apresentam curso crônico com recaídas (surtos) e exigem tratamento com drogas modificadoras de doenças (DMDs) para a prevenção de atividade inflamatória. As DMDs levam à imunomodulação ou imunossupressão através de diversos mecanismos (por exemplo deslocando e/ou suprimindo subpopulações linfocitárias ou alterando perfil de produção de citocinas). Desta forma, os pacientes com EM ou NMOSD são mais propensos a complicações infecciosas, as quais podem levar ao agravamento de suas doenças de base.

Objetivo

Apresentar estratégias viáveis para mitigar o risco de infecção de pacientes com EM ou NMOSD sob tratamento.

Métodos

Revisão bibliográfica focada em manejo de risco de infecção com ênfase em vacinação, medidas específicas de tratamento e particularidades de doenças infecciosas endêmicas do Brasil.

Conclusão

Propomos um calendário de vacinação, rotina de triagem infecciosa e medidas profiláticas baseadas em evidências científicas atuais. A conscientização das doenças tropicais emergentes é necessária devido a evidências de eventos desmielinizantes e possíveis casos parainfecciosos de EM e NMOSD.

Palavras-chave
Esclerose Múltipla; Neuromielite Óptica; Vacinas

INTRODUCTION

Multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSDs) are the most common demyelinating diseases of the central nervous system.11 Leray E, Moreau T, Fromont A, Edan G. Epidemiology of multiple sclerosis. Rev Neurol (Paris) 2016;172(01):3–13 Relapses are the hallmark of clinical activity and add up to increasing disability, particularly in NMOSD, in which case they tend to be more severe and refractory to treatment. Available disease modifying drugs (DMDs) lead to immunomodulation or immunosuppression through diverse mechanisms, which halt disease activity but also impair the body's defense response against infections.

In patients with MS or NMOSD, infections may increase the risk of relapses and worsening of disease. While using DMDs, infections can be more frequent and severe; thus, adequate infection risk management is required. Feasible measures include adequate infectious screening prior to DMDs initiation, a well-suited approach to vaccination, and prophylactic measures during treatment.

In this article, we will present feasible strategies for mitigating the risk of infection of MS and NMOSD treated patients with an emphasis on vaccination, therapy-specific measures, and particularities of the Brazilian endemic infectious diseases' scenario.

METHODS

We performed a targeted literature review to address questions (Supplementary Material, available online only) determined by a panel of specialists in neuroimmunology and infectious diseases. Information from Pubmed, Web of Science, Cochrane, medication labels, and official reports from the Brazilian Society of Immunizations (SBIm) and the Brazilian Ministry of Health were searched, compiled, and reviewed by four junior neuroimmunologists from March to November 2020. All resulting evidence was assessed by a panel of three senior neuroimmunologists and two senior infectious disease specialists with a focus on the management of immunocompromised patients. Suggestions were elaborated through the consensus of all specialists.

Objective

The objective of this paper is to provide Brazilian neurologists with practical strategies for mitigating the risk of infection of MS and NMOSD treated patients.

Infection prevention

Infectious screening

Infection risk can be mitigated through vaccination and treatment of latent infections before the beginning of DMDs for the treatment of MS or NMOSD. At the first clinical visit, it is recommended to screen patients' serological status and vaccination records aiming to rule out differential diagnoses and identify lack of immunity against vaccine-preventable diseases (▸Table 1)22 Fragoso YD, Elso FG, Carrá A Differential diagnosis of multiple sclerosis in Latin America. Mult Scler J Exp Transl Clin 2017;3(03): 205521731771427966 Buonomo AR, Zappulo E, Viceconte G, Scotto R, Borgia G, Gentile I. Risk of opportunistic infections in patients treated with alemtuzumab for multiple sclerosis. Expert Opin Drug Saf 2018;17(07): 709–717 In addition, prior to switching DMDs, it is advisable to re-check immunity and infections against specific DMD-relevant infectious agents (▸Table 2).44 Arvin AM, Wolinsky JS, Kappos L, et al. Varicella-zoster virus infections in patients treated with fingolimod: risk assessment and consensus recommendations for management. JAMA Neurol 2015;72(01):31–3966 Buonomo AR, Zappulo E, Viceconte G, Scotto R, Borgia G, Gentile I. Risk of opportunistic infections in patients treated with alemtuzumab for multiple sclerosis. Expert Opin Drug Saf 2018;17(07): 709–717

Table 1
First appointment suggested screening in multiple sclerosis/ neuromyelitis optica spectrum disorders patients to reduce infection risk
Table 2
Disease modifying drugs specific screening suggestion

Vaccine Efficacy in MS and NMOSD Patients under Treatment

Influenza is the most studied vaccine in the MS and NMOSD treated populations. Vaccination response to other vaccines is often surmised based on the available knowledge of that vaccine. Studies have found that the influenza vaccine response is reduced in patients being treated with: fingolimod,77 Olberg HK, Eide GE, Cox RJ, et al. Antibody response to seasonal influenza vaccination in patients with multiple sclerosis receiving immunomodulatory therapy. Eur J Neurol 2018;25(03):527–534 natalizumab,77 Olberg HK, Eide GE, Cox RJ, et al. Antibody response to seasonal influenza vaccination in patients with multiple sclerosis receiving immunomodulatory therapy. Eur J Neurol 2018;25(03):527–534 rituximab,88 Kim W, Kim SH, Huh SY, et al. Reduced antibody formation after influenza vaccination in patients with neuromyelitis optica spectrum disorder treated with rituximab. Eur J Neurol 2013;20(06): 975–980 ocrelizumab,99 Bar-Or A, Calkwood JC, Chognot C, et al. Effect of ocrelizumab on vaccine responses in patients with multiple sclerosis: The VELOCE study. Neurology 2020;95(14):e1999–e2008. Doi: 10.1212/WNL.0000000000010380
https://doi.org/10.1212/WNL.000000000001...
and mycophenolate-mofetil.88 Kim W, Kim SH, Huh SY, et al. Reduced antibody formation after influenza vaccination in patients with neuromyelitis optica spectrum disorder treated with rituximab. Eur J Neurol 2013;20(06): 975–980 The influenza vaccine response has been found to be effective in patients under interferons1010 Schwid SR, Decker MD, Lopez-Bresnahan MRebif-Influenza Vaccine Study Investigators. Immune response to influenza vaccine is maintained in patients with multiple sclerosis receiving interferon beta-1a. Neurology 2005;65(12):1964–1966 and teriflunomide, though decreased response is seen in the latter.1111 Bar-Or A, Freedman MS, Kremenchutzky M, et al. Teriflunomide effect on immune response to influenza vaccine in patients with multiple sclerosis. Neurology 2013;81(06):552–558 Data are conflicting regarding glatiramer-acetate.1212 Olberg HK, Cox RJ, Nostbakken JK, Aarseth JH, Vedeler CA, Myhr KM. Immunotherapies influence the influenza vaccination response in multiple sclerosis patients: an explorative study. Mult Scler 2014;20(08):1074–1080,1313 Metze C, Winkelmann A, Loebermann M, et al. Immunogenicity and predictors of response to a single dose trivalent seasonal influenza vaccine in multiple sclerosis patients receiving disease-modifying therapies. CNS Neurosci Ther 2019;25(02):245–254 Influenza vaccine response in patients using dimethyl-fumarate, cladribine, azathioprine, and methotrexate has not been studied in the MS and NMOSD populations. However, data from kidney transplant patients indicates that the vaccine's response is maintained in azathioprine users.1414 Keshtkar-Jahromi M, Argani H, Rahnavardi M, et al. Antibody response to influenza immunization in kidney transplant recipients receiving either azathioprine or mycophenolate: a controlled trial. Am J Nephrol 2008;28(04):654–660 In addition, vaccine response during the use of dimethyl fumarate has been shown to be adequate for tetanus-diphtheria toxoid, pneumococcal, and meningococcal vaccine when compared with interferon.1515 von Hehn C, Howard J, Liu S, et al. Immune response to vaccines is maintained in patients treated with dimethyl fumarate. Neurol Neuroimmunol Neuroinflamm 2017;5(01):e409 Data on routine serological screening after immunizations in patients under DMDs is lacking and not widely recommended.

Timing of vaccines

Vaccines are more effective if administered before the beginning of immunosuppressive or immunomodulatory treatments. Therefore, as vaccines do not increase the risk of relapses in MS patients, vaccination prior to DMD initiation is recommended.1616 Lebrun C, Vukusic SFrench Group for Recommendations in Multiple Sclerosis (France4MS) and the Société Francophone de la Sclérose En Plaques (SFSEP) Immunization and multiple sclerosis: Recommendations from the French multiple sclerosis society. Mult Scler Relat Disord 2019;31:173–188,1717 Farez MF, Correale J, Armstrong MJ, et al. Practice guideline update summary: Vaccine-preventable infections and immunization in multiple sclerosis: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology 2019;93(13):584–594 Contrarily, NMOSD patients who are not on immunotherapy have a higher likelihood of presenting relapses following immunization. Thus, immediate initiation of DMDs following diagnosis and subsequent vaccination are recommended when safe in patients with NMOSD.1818 Mealy MA, Cook LJ, Pache F, et al. Vaccines and the association with relapses in patients with neuromyelitis optica spectrum disorder. Mult Scler Relat Disord 2018;23:78–82

To maintain vaccination efficacy, ensure protection before immunosuppression, and reduce the risk of side effects, following Brazilian Society of Immunizations (SBIm) and Brazilian Ministry of Health guidelines, we recommend starting DMDs at least 2 weeks after inactivated vaccines and at least 4 weeks after live-attenuated vaccines have been administered.1919 Sociedade Brasileira de Imunizações. Calendário de Vacinaçao Pacientes Especiais 2020-2021. 2020,2020 Ministério da Saúde Manual dos Centros de Referência para Imunobiológicos Especiais- 5a edição; 2019 If vaccination prior to drug initiation is impossible, inactivated vaccines may be administered during treatment; however, they will probably lead to compromised immune responses. In patients who are not severely immunocompromised, careful individual assessment should precede live-attenuated vaccines administration. Multiple sclerosis patients treated with cladribine may receive live vaccines after their leucocyte count resumes to normal.2121 Merck. Mavenclad - prescribing information; 2019

Vaccination schedule for MS and NMOSD patients

In 2019, the American Academy of Neurology (AAN) and the Société Francophone de la Sclérose en Plaques (SFSEP) issued recommendations regarding the immunization of MS patients. Both statements highlight the current evidence on vaccine safety and efficacy in this population. Vaccines are considered safe and efficient in MS patients without DMD use, similar to the general population, supporting the rationale to provide proper immunization prior to starting DMDs. The recommended vaccine schedule includes vaccines that are standard for the general population plus specific vaccines based on local epidemiology.1616 Lebrun C, Vukusic SFrench Group for Recommendations in Multiple Sclerosis (France4MS) and the Société Francophone de la Sclérose En Plaques (SFSEP) Immunization and multiple sclerosis: Recommendations from the French multiple sclerosis society. Mult Scler Relat Disord 2019;31:173–188,1717 Farez MF, Correale J, Armstrong MJ, et al. Practice guideline update summary: Vaccine-preventable infections and immunization in multiple sclerosis: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology 2019;93(13):584–594

For patients already using DMDs, each medication may require a specific approach. While immunomodulators such as betainterferons and glatiramer acetate seem to have less impact in vaccine safety and efficacy, most DMDs require well-timed vaccine administration for optimal efficacy. Special attention should be given to the administration of live-attenuated vaccines, which cannot be prescribed to patients under immunosuppressive drugs, including prednisone ≥ 20 mg/day.

In Brazil, the Brazilian Society of Immunizations (SBIm) has published schedules for special patients, including those with autoimmune diseases or immunosuppression.1919 Sociedade Brasileira de Imunizações. Calendário de Vacinaçao Pacientes Especiais 2020-2021. 2020 There are no schedules specific to MS or NMOSD patients. Our vaccine schedule suggestion for MS and NMOSD patients over the age of 10 is displayed in ▸Table 3.

Table 3
Vaccination schedule for multiple sclerosis and neuromyelitis optica spectrum disorders patients > 10 years of age

Close contact vaccination

The Infectious Disease Society of America (IDSA), SBIm and Brazilian Ministry of Health guidelines recommend healthy immunocompetent persons who live with immunocompromised patients should receive a broader vaccination schedule than the general population.1919 Sociedade Brasileira de Imunizações. Calendário de Vacinaçao Pacientes Especiais 2020-2021. 2020,2020 Ministério da Saúde Manual dos Centros de Referência para Imunobiológicos Especiais- 5a edição; 2019,2222 Armstrong C. IDSA releases recommendations on vaccinations in immunocompromised patients. Am Fam Physician 2014; 90:664–666 Close contacts of immunosuppressed patients should receive yearly influenza vaccine and be questioned about history of varicella and immunization status. They should be vaccinated for measles, mumps, rubella, and varicella, if susceptible. Oral polio vaccine should not be administered to people living with immunocompromised patients, as the living virus can be excreted in the feces. The inactivated vaccine is preferred.2222 Armstrong C. IDSA releases recommendations on vaccinations in immunocompromised patients. Am Fam Physician 2014; 90:664–666

Impact on the vaccination of newborns of mothers who received immunosuppressants

Experts recommend that live vaccine administration should be withheld in the first year of newborns that were exposed to rituximab in utero, particularly if exposed in the third trimester.2323 Berkhout A, Clark JE, Wen SC-H. In utero exposure to biologic disease-modifying anti-rheumatic drugs and effects to the infant: infectious complications, vaccine response, and safety of live vaccine administration. Expert Rev Vaccines 2019;18(05): 495–504 Though BCG, measles, and rubella vaccines can be postponed with minimal impact to infants, the rotavirus vaccine poses a challenge. The latter is a major cause of acute gastroenteritis associated with significant death and hospitalization rates in newborns and infants. Hence, the benefits of rotavirus vaccine may outweigh the risks in normal schedule. The SBIm considers that newborns of mothers who received rituximab should not receive BCG or rotavirus vaccines in the first 6 to 8 months of life as no consensus can be reached regarding optimal safety in this scenario. To the best of our knowledge, there is no literature addressing the impact of other DMDs on newborn vaccination. Nonetheless, we suggest caution when administering live vaccines to newborns of immunosuppressed mothers.

Vaccination in pediatric population diagnosed with MS and NMOSD

Pediatric onset MS and NMOSD are extremally rare, accounting for ∼ 3 to 10% of all cases.2424 Chitnis T. Pediatric Central Nervous System Demyelinating Diseases. Continuum (Minneap Minn) 2019;25(03):793–814 There are several reports of CNS inflammatory events following infections and vaccination in children.2424 Chitnis T. Pediatric Central Nervous System Demyelinating Diseases. Continuum (Minneap Minn) 2019;25(03):793–814 The link between exposure to infections and immune dysregulating triggering autoimmune disorders, including acquired demyelinating syndromes (ADS), is a special concern in the pediatric population.2525 Hughes AM, Ponsonby AL, Dear K, et al; Ausimmune Investigator Group. Childhood infections, vaccinations, and tonsillectomy and risk of first clinical diagnosis of CNS demyelination in the Ausimmune Study. Mult Scler Relat Disord 2020;42:102062,2626 Suleiman L, Waubant E, Aaen G, et al; Network of Pediatric Multiple Sclerosis Centers. Early infectious exposures are not associated with increased risk of pediatric-onset multiple sclerosis. Mult Scler Relat Disord 2018;22:103–107. Doi: 10.1016/j.msard.2018.03.015.Early
https://doi.org/10.1016/j.msard.2018.03....
No association between vaccination and acquired demyelinating syndromes (ADSs) has consistently been described, including pediatric-onset of MS.2727 Di Pietrantonj C, Rivetti A, Marchione P, Debalini MG, Demicheli V. Vaccines for measles, mumps, rubella, and varicella in children. Cochrane Database Syst Rev 2020;2020:•••,2828 Stowe J, Andrews N, Miller E. Do Vaccines Trigger Neurological Diseases? Epidemiological Evaluation of Vaccination and Neurological Diseases Using Examples of Multiple Sclerosis, Guillain-Barré Syndrome and Narcolepsy. CNS Drugs 2020;34(01): 1–8 Studies focusing on pediatric NMOSD are lacking. It is currently accepted that vaccines might act by accelerating the transition from subclinical to clinically relevant inflammatory events. However, the risk of developing vaccine-preventable infectious diseases is considered higher than the risk of developing vaccine-triggered demyelinating events.2929 Langer-Gould A, Qian L, Tartof SY, et al. Vaccines and the risk of multiple sclerosis and other central nervous system demyelinating diseases. JAMA Neurol 2014;71(12):1506–1513

Thus, recommendations for the pediatric population with neuroimmunologic disorders are similar to those of adults: (1) children with incomplete vaccination charts should be vaccinated according to age and serology prior to beginning immunosuppressive drugs; (2) all pediatric patients should receive the influenza vaccine annually; (3) live-attenuated vaccines are contraindicated in patients on immunosuppressive drugs. Infectious screening should be performed as in the adult population (▸Tables 1 and ▸2).

Other infection prevention measures

Immunoglobulin replacement

Rituximab has been associated with increased risk of infections.3030 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(02): 184–191 Hypogammaglobulinemia is a possible causal mechanism, which is observed in almost half of the patients treated with the drug3131 Barmettler S, Ong MS, Farmer JR, Choi H, Walter J. Association of Immunoglobulin Levels, Infectious Risk, and Mortality With Rituximab and Hypogammaglobulinemia. JAMA Netw Open 2018;1(07):e184169. Intravenous immunoglobulin (IgIV) replacement in patients with recurrent infections and low IgG levels (< 5 g/dl) should be performed, as it is associated with a reduction of the infection rate of patients receiving rituximab, at least in observational studies3131 Barmettler S, Ong MS, Farmer JR, Choi H, Walter J. Association of Immunoglobulin Levels, Infectious Risk, and Mortality With Rituximab and Hypogammaglobulinemia. JAMA Netw Open 2018;1(07):e184169 Although IgIV replacement is not suggested for all MS/NMOSD anti-CD20/CD19 treated patients, we recommend immunoglobulin dosing (IgG, IgA, and IgM), or protein electrophoresis, and infection monitoring every 6 months with immunoglobulin replacement, in case of recurrent infections.3131 Barmettler S, Ong MS, Farmer JR, Choi H, Walter J. Association of Immunoglobulin Levels, Infectious Risk, and Mortality With Rituximab and Hypogammaglobulinemia. JAMA Netw Open 2018;1(07):e184169

Antibiotic and antiviral prophylaxis

Prophylactic antibiotics and antivirals are routinely used in MS patients treated with alemtuzumab. Herpes infections occurred in 6% of alemtuzumab-treated patients in clinical trials and were serious in 0.1 to 0.2% of patients.3232 Hersh CM, Cohen JA. Alemtuzumab for the treatment of relapsing-remitting multiple sclerosis. Immunotherapy 2014;6(03):249–259 Acyclovir 200 mg twice a day is recommended for at least 60 days after last alemtuzumab administration and until CD4 lymphocyte count is above 200 mg/uL. However, only ∼ 50% of patients have return of CD4 count above 200 mg/uL in 9 months and 80% of patients in 12 months after alemtuzumab dose.3333 Lycke J, et al. Lymphocyte subset dynamics following alemtuzumab administration in the CARE-MS I trial. in 89; 2012 Hence, if CD4 levels are not monitored, we recommend prophylactic acyclovir for at least 2 years. Cotrimoxazole prophylaxis can also be prescribed to prevent both listeria infection and Pneumocystis jiroveci pneumonia until lymphocyte count is above 200 mg/uL.66 Buonomo AR, Zappulo E, Viceconte G, Scotto R, Borgia G, Gentile I. Risk of opportunistic infections in patients treated with alemtuzumab for multiple sclerosis. Expert Opin Drug Saf 2018;17(07): 709–717 Hygienic measures for patients using alemtuzumab include listeria-free diet and toxoplasmosis-free diet in toxoplasmosis IgG-negative patients.

Eculizumab, a terminal complement inhibitor, requires special attention to bacterial infections due to its mechanism of action tampering immune response to encapsulated bacteria. Particularly, patients may present severe meningococcal infections. Risk of meningococcal infection can increase up to 2.000 times during treatment. Vaccination decreases but does not eliminate this risk. Quadrivalent meningococcal vaccine plus vaccine against B serogroup should be administered at least 2 weeks before beginning of therapy. A booster dose of quadrivalent meningococcal vaccine after 5 years is recommended. Antibiotic prophylaxis should be given for at least 4 weeks after immunization, as to protect against infection during assembly of proper immune response to vaccination. Penicillin V potassium 250 mg 12/12h or ciprofloxacin 500 mg daily for at least 4 weeks after last vaccination are feasible regimens.3434 Benamu E, Montoya JG. Infections associated with the use of eculizumab: recommendations for prevention and prophylaxis. Curr Opin Infect Dis 2016;29(04):319–329

Patients living with MS/NMOSD frequently have neurogenic bladder after spinal cord lesions and may need clean intermittent self-catheterization (CISC) to empty their bladder. In this population, 25% of patients experience recurrent urinary tract infection (UTI). Based in the AnTIC trial,3535 Fisher H, Oluboyede Y, Chadwick T, et al. Continuous low-dose antibiotic prophylaxis for adults with repeated urinary tract infections (AnTIC): a randomised, open-label trial. Lancet Infect Dis 2018;18(09):957–968 patients using CISC with two episodes of symptomatic UTI in the last year or with one episode of UTI that leads to hospitalization should be submitted to prophylactic antibiotics to reduce the risk of a new symptomatic UTI. Options for prophylactic antibiotics include: 50 mg/d nitrofurantoin, 100 mg/d trimethoprim, or 250 mg/d cefalexin. Nevertheless, this strategy is associated with increased bacterial resistance in new events of UTI and, hence, trials of discontinuation of prophylactic antibiotics after 12 months may be considered to reduce this complication. We also recommend urological consultation for patients experiencing bladder disfunction and recurrent urinary tract infections (UTIs).

Hepatitis B reactivation management

Vaccination remains the most effective intervention to prevent hepatitis B virus (HBV) infection. However, previously infected individuals may present HBV reactivation during immunosuppressive treatment and require adequate monitoring. Particularly, B-cell depleting therapies have been associated to reactivation;3636 Ciardi MR, et al. Reactivation of Hepatitis B Virus with Immune-Escape Mutations after Ocrelizumab Treatment for Multiple Sclerosis. Open Forum Infect Dis 2019;6:1–3 nonetheless, screening for HBV is mandatory before beginning of any immunosuppressive treatment.

The American Gastroenterological Association and the European Association for the Study of the Liver have issued guidelines regarding the management of HBV reactivation during immunosuppressive treatment. Antiviral prophylaxis is strongly recommended to high-risk patients defined as having anticipated incidence of reactivation higher than 10%. In MS and NMOSD, this population is comprised of patients presenting positive antiHBc receiving B cell depleting DMDs, alemtuzumab, or cladribine for more than 4 weeks. Patients receiving azathioprine, methotrexate, and prednisone at low doses for any duration are considered low risk for reactivation and are not required to receive prophylaxis. A less supported alternative is to screen HBV DNA every 3 months.3737 Reddy KR, Beavers KL, Hammond SP, Lim JK, Falck-Ytter YTAmerican Gastroenterological Association Institute. American Gastroenterological Association Institute guideline on the prevention and treatment of hepatitis B virus reactivation during immuno-suppressive drug therapy. Gastroenterology 2015;148(01):215-–219, quiz e16–e17,3838 Idilman R. The summarized of EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. Turk J Gastroenterol 2017;28(05):412–416

Hepatitis B reactivation is less clear during treatment with other DMDs. Clinical trials of oral DMDs were not uniform in screening HBV, and severe liver disease has not been reported.3939 Epstein DJ, Dunn J, Deresinski S. Infectious complications of multiple sclerosis therapies: Implications for screening, prophylaxis, and management. Open Forum Infect Dis 2018;5(08): ofy174 Nonetheless, screening is recommended, and monitoring may be considered individually.

HPV in MS and NMOSD treated patients

Human papillomavirus (HPV) viruses are responsible for the occurrence of cutaneous warts, 99% of cervical cancers, and most of anogenital cancers.4040 Nguyen ML, Flowers L. Cervical cancer screening in immunocompromised women. Obstet Gynecol Clin North Am 2013;40(02): 339–357 Multiple sclerosis and NMOSD treatments may compromise normal antiviral response to HPV. Fingolimod therapy, for instance, has been associated with treatment refractory warts.4141 Triplett J, Kermode AG, Corbett A, Reddel SW. Warts and all: Fingolimod and unusual HPV-associated lesions. Mult Scler 2019; 25(11):1547–1550

There are 2 vaccines currently available in Brazil for the prevention of HPV, the HPV 2 (HPV 16 and 18 serotypes) and HPV 4 (HPV 6, 11, 16, and 18 serotypes) vaccines. The national public health system (SUS) offers the HPV4 vaccine to boys and girls aged from 9 to 14 and to HIV, cancer, transplant, and immunosuppressed patients from 9 to 26 years of age free of charge. Nonetheless, the HPV4 vaccine has been approved for use in females between 9 and 45 and males between 9 and 26 years of age. As MS and NMOSD treated patients are more prone to developing HPV infection clinical manifestations, we recommend all male and female patients receive the HPV4 vaccine when possible.

Brazilian guidelines on cervical cancer screening suggest collecting cervical cytology yearly in all women over 25 years of age who are or have previously been sexually active. Periodic screening should be maintained until the age of 64.4040 Nguyen ML, Flowers L. Cervical cancer screening in immunocompromised women. Obstet Gynecol Clin North Am 2013;40(02): 339–357 Due to the potential increased risk of cervical cancer in MS and NMOSD treated patients, in agreement with other specialists, we suggest carrying yearly cervical cytology in conjunction with pelvic examination in all previously sexually active patients.4040 Nguyen ML, Flowers L. Cervical cancer screening in immunocompromised women. Obstet Gynecol Clin North Am 2013;40(02): 339–357 In addition, we recommend avoiding DMTs associated with impaired antiviral response, such as fingolimod, in case of cervical cancer diagnosis.

Tuberculosis screening

Latent tuberculosis should be screened annually in patients using MS DMDs, except for interferons and glatiramer.3939 Epstein DJ, Dunn J, Deresinski S. Infectious complications of multiple sclerosis therapies: Implications for screening, prophylaxis, and management. Open Forum Infect Dis 2018;5(08): ofy174,4242 Baldassari LE, Feng J, Macaron G, et al. Tuberculosis screening in multiple sclerosis: effect of disease-modifying therapies and lymphopenia on the prevalence of indeterminate TB screening results in the clinical setting. Mult Scler J Exp Transl Clin 2019;5 (03):2055217319875467,4343 Navas C, Torres-Duque CA, Munoz-Ceron J, et al. Diagnosis and treatment of latent tuberculosis in patients with multiple sclerosis, expert consensus. On behalf of the Colombian Association of Neurology, Committee of Multiple Sclerosis. Mult Scler J Exp Transl Clin 2018;4(01):2055217317752202 Interferon gamma release tests (IGRAs), such as QuantiFERON, are more accurate than tuberculosis skin tests and BCG vaccination does not interfere with them. A meticulous medical history should be taken aiming to identify previous contact with confirmed cases of tuberculosis. In addition, chest radiography or tomography should be performed to differentiate active from latent tuberculosis infection. The latter is treated with isoniazid 300 mg/d for 6 to 9 months or rifampicin for 4 months. We recommend at least a month of antituberculosis therapy before starting DMDs for patients with latent tuberculosis.

Varicella virus zoster infection in patients on fingolimod or high-dose corticosteroids

Fingolimod augments the risk of varicella zoster virus (VZV) reactivation. Although overall low, the risk associated with fingolimod is higher than with placebo (11 versus 6 per 1,000 patients/year). Hence, we recommend that patients without evidence of VZV immunity (i.., IgG negative) should receive 2 doses of the vaccine with a 4-week interval. The first dose of fingolimod should be scheduled for at least 1 month after the second dose. Moreover, in case of recent high-dose corticosteroid use for the treatment of a relapse, the VZV vaccine must be administered at least 30 days following corticosteroid interruption.4444 Mills EA, Mao-Draayer Y. Understanding Progressive Multifocal Leukoencephalopathy Risk in Multiple Sclerosis Patients Treated with Immunomodulatory Therapies: A Bird's Eye View. Front Immunol 2018;9:138

PML

Progressive multifocal leukoencephalopathy (PML) is a poliomavirus-associated disease caused by the John Cunningham virus (JCV) which often leads to permanent disability or death. Progressive multifocal leukoencephalopathy has been described in MS patients treated with natalizumab, fingolimod, dimethyl-fumarate, and ocrelizumab.4444 Mills EA, Mao-Draayer Y. Understanding Progressive Multifocal Leukoencephalopathy Risk in Multiple Sclerosis Patients Treated with Immunomodulatory Therapies: A Bird's Eye View. Front Immunol 2018;9:138 There are no reports of PML on MS or NMOSD patients treated with rituximab; however, cases of PML have been described in patients suffering from other diseases who were treated with the drug.4545 Van Assche G, Van Ranst M, Sciot R, et al. Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn's disease. N Engl J Med 2005;353(04):362–3684747 Berger JR, Malik V, Lacey S, Brunetta P, Lehane PB. Progressive multifocal leukoencephalopathy in rituximab-treated rheumatic diseases: a rare event. J Neurovirol 2018;24(03):323–331

Concern about the risk of developing PML is the main reason for discontinuing natalizumab.4848 Conway DS, Hersh CM, Harris HC, Hua LH. Duration of natalizumab therapy and reasons for discontinuation in a multiple sclerosis population. Mult Scler J Exp Transl Clin 2020;6(01): 2055217320902488 Risk factors for natalizumab-associated PML include prior immunosuppressant (IS) therapy, the presence of serum anti–JC virus (JCV) antibodies, and longer natalizumab therapy duration—especially beyond 2 years.4949 Foley 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;,5050 Ho PR, Koendgen H, Campbell N, Haddock B, Richman S, Chang I. Risk of natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: a retrospective analysis of data from four clinical studies. Lancet Neurol 2017;16 (11):925–933 Mindfulness of the risk factors is important in preventing the disease. We recommend monitoring at each medical appointment, according to the table below (▸Table 4). Strategies for minimizing PML risk include extended interval dosing of natalizumab (i.e., infusions every 6–8 weeks) or switching treatment to other high efficacy options, such as anti-CD20 monoclonal antibodies, alemtuzumab, or cladribine.5151 Ryerson LZ, Foley J, Chang I, et al. Risk of natalizumab-associated PML in patients with MS is reduced with extended interval dosing. Neurology 2019;93(15):e1452–e1462

Table 4
Progressive multifocal leukoencephalopathy, risk stratification on natalizumab-treated patients* * Adapted from Ho et al. (2017).52

Multiple sclerosis patients with previous natalizumab use may experience PML due to “carry-over” risk, even after being switched to other drugs. In fact, most of the patients who developed PML on fingolimod, dimethyl-fumarate, or ocrelizumab fit into that category. Nonetheless, “de novo” PML cases have been described for each of the aforementioned medications. “Carry-over” PML risk can be mitigated through prevention and surveillance. We suggest magnetic resonance imaging (MRI) screening every 3 to 4 months up to 12 months following the switch from natalizumab to any of those medications.5252 Wattjes MP, Rovira À, Miller D, et al; MAGNIMS study group. Evidence-based guidelines: MAGNIMS consensus guidelines on the use of MRI in multiple sclerosis–establishing disease prognosis and monitoring patients. Nat Rev Neurol 2015;11(10): 597–606. Doi: 10.1038/nrneurol.2015.157
https://doi.org/10.1038/nrneurol.2015.15...

Few cases of PML have also been reported in patients under treatment with alemtuzumab, cladribine, teriflunomide, and interferons. Nonetheless, all are associated with low risk of developing PML, as most of the reported cases had coexisting risk factors for the development of the disease.5353 Kartau M, Sipilä JO, Auvinen E, Palomäki M, Verkkoniemi-Ahola A. Progressive Multifocal Leukoencephalopathy: Current Insights. Degener Neurol Neuromuscul Dis 2019;9:109–121

Post exposure measures – measles and chickenpox

Immunocompromised patients can develop severe forms of measles and chickenpox. Therefore, postexposure measures for susceptible individuals are imperative and involve timely vaccination and immunoglobulin administration. A restricted group of patients will be eligible to receive postexposure vaccination, as live vaccines should be avoided in immunosuppressed patients.

To prevent severe forms of chickenpox, the VZV vaccine should be administered up to 5 days following significant exposure of non-immunosuppressed patients (e.g., patients under beta-interferons or glatiramer acetate). Varicella zoster virus immunoglobulin administration should ideally be performed up to 4 days following significant exposure of immunosuppressed patients; however, it is still feasible up to 10 days after exposure. It is available in the Brazilian public health system (SUS). Less supported alternatives include intravenous immunoglobulin (IVIg) (400 mg/kg one dose), oral acyclovir (20 mg/kg each dose, 4 times daily for 7 days, maximum daily dose of 3,200 mg), or valacyclovir (20 mg/kg each dose, 3 times daily for 7 days, maximum daily dose of 3,000 mg) administration. When chosen, acyclovir and valacyclovir should be used after 7 to 10 days of exposure.5454 Lachiewicz AM, Srinivas ML. Varicella-zoster virus post-exposure management and prophylaxis: A review. Prev Med Rep 2019; 16:101016

Severe measles prevention can be achieved through postexposure vaccine within 72 hours of exposure of non-immunosuppressed patients. Specific immunoglobulin is unavailable. Alternatively, IMIg (0.5 ml/kg up to 15 ml divided among multiple muscle groups) or IVIg (400 mg/kg one dose) should be administered up to 6 days following exposure of immunosuppressed patients.5555 SPSP. Spsp – nota informativa (julho de 2018): atualização sobre sarampo. 1‣13; 2020.

Prevention of yellow fever

Yellow fever is an endemic disease in Brazil without specific treatment. In 2017 and 2018, Brazil experienced one of the most severe yellow fever epidemics of all times with almost 3,000 infected patients and a 39.3% lethality rate.5656 Ministério da Saúde. Monitoramento do Período Sazonal da Febre Amarela Brasil – 2018/2019. informe n° 03 | 2018/2019. Since then, the number of seasonally infected patients has decreased, but the single dose vaccine is still recommended for patients living in or travelling to endemic areas.

The yellow fever vaccine (YFV) is highly effective, inducing neutralizing antibodies in 99% of recipients.5757 Domingo C, Niedrig M. Safety of 17D derived yellow fever vaccines. Expert Opin Drug Saf 2009;8(02):211–221 It is a live attenuated virus vaccine and should not be administered to young (< 6 months of age), immunosuppressed, pregnant, or breastfeeding patients. The vaccine seems to be safe for MS patients being treated with injectable medications but should not be administered to patients on other DMTs, despite its reported safety on natalizumab-treated patients.5858 Huttner A, Eperon G, Lascano AM, et al. Risk of MS relapse after yellow fever vaccination: A self-controlled case series. Neurol Neuroimmunol Neuroinflamm 2020;7(04):1–6 There is some controversy on whether the vaccine increases the chance of relapses in MS. Farez et al. reported a statistically significant increase in relapse rate following the YFV (rate ratio = 12.778; p < 0.001) in a 2011 study involving 7 MS patients living in endemic areas of Argentina.5959 Farez MF, Correale J. Yellow fever vaccination and increased relapse rate in travelers with multiple sclerosis. Arch Neurol 2011;68(10):1267–1271 However, in a recent study carried by Huttner et Al., there was no reported increase in relapse rate after the vaccination of 23 patients travelling to endemic areas.5858 Huttner A, Eperon G, Lascano AM, et al. Risk of MS relapse after yellow fever vaccination: A self-controlled case series. Neurol Neuroimmunol Neuroinflamm 2020;7(04):1–6 We recommend an individualized approach when indicating the YFV to patients living or travelling to endemic areas. Travelling should be discouraged, when possible. Patients on non-injectable DMTs may benefit from barrier methods, such as using insect repellent, placing nets on doors and windows, and wearing long-sleeved shirts and pants. If the infection risk is remarkably high, DMT interruption followed by vaccination may be indicated, if there are no records of recent MS inflammatory activity or previously aggressive disease. If it is decided to do so, the ideal conduct is to wait 3 to 6 months after cessation of immunosuppression prior to vaccination (patients who received B cell depleting DMDs should wait 6 months).2020 Ministério da Saúde Manual dos Centros de Referência para Imunobiológicos Especiais- 5a edição; 2019 In case of recent MS inflammatory activity of previously aggressive disease, we recommend avoiding the vaccine, considering the current available evidence.

In conclusion, the long-term management of MS and NMOSD has become feasible due to the increasing number of DMDs being developed and approved. However, disease activity and adequate control are inevitably tied to increased risk of developing infections. Infections may worsen the course of MS and NMOSD through heightened risk and the severity of relapses. Conversely, DMDs may tamper adequate immune response to infections, leading to possibly worse outcomes. Mitigating strategies of infectious risk must be assessed from time of diagnosis to DMDs initiation and follow-up.

Adequate infection screening and vaccination are primordial to reduce the risk of infection in MS/NMOSD patients. Neurologists should be mindful to screen for preventable and treatable infections such as tuberculosis, syphilis, hepatitis B and C, HIV, varicella, measles, rubella, and toxoplasmosis before starting DMDs.22 Fragoso YD, Elso FG, Carrá A Differential diagnosis of multiple sclerosis in Latin America. Mult Scler J Exp Transl Clin 2017;3(03): 2055217317714279,66 Buonomo AR, Zappulo E, Viceconte G, Scotto R, Borgia G, Gentile I. Risk of opportunistic infections in patients treated with alemtuzumab for multiple sclerosis. Expert Opin Drug Saf 2018;17(07): 709–717 When possible (i.e. patients with non-aggressive MS), immunizations should be offered prior to the initiation of DMDs. Disease-modifying drugs should be started at least 2 weeks after inactivated vaccines and at least 4 weeks after live-attenuated vaccines have been administered.1919 Sociedade Brasileira de Imunizações. Calendário de Vacinaçao Pacientes Especiais 2020-2021. 2020,2020 Ministério da Saúde Manual dos Centros de Referência para Imunobiológicos Especiais- 5a edição; 2019 NMOSD patients should not have immunosuppressive treatment delayed due to vaccination, as there is an increased risk of relapses following vaccines in non-immunosuppressed NMOSD patients.1818 Mealy MA, Cook LJ, Pache F, et al. Vaccines and the association with relapses in patients with neuromyelitis optica spectrum disorder. Mult Scler Relat Disord 2018;23:78–82

Infection risk monitoring is also necessary during patients' follow-up. Neurologists should reassess infection risk when switching DMDs and take each specific DMD recommendation into consideration. Infection prophylaxis is indicated for patients who present recurrent UTIs, or who are receiving alemtuzumab or eculizumab. When in doubt, infectious disease specialists or urologists should be consulted. Lymphocyte counts, HBV/HCV/HPV infection, and PML risk should also be checked routinely. Neurologists should be reminded that PML has been associated to DMDs other than natalizumab. In addition, PML carry-over risk should be monitored after suspension of natalizumab, as PML is a preventable fatal disease, and a high level of suspicion is necessary.

There are currently no national guidelines on the infectious risk management of patients with CNS demyelinating disorders. Through this review, we have aimed to provide information and practical recommendations to assist Brazilian neurologists with infectious diseases scenarios which may be experienced in clinical practice.

References

  • 1
    Leray E, Moreau T, Fromont A, Edan G. Epidemiology of multiple sclerosis. Rev Neurol (Paris) 2016;172(01):3–13
  • 2
    Fragoso YD, Elso FG, Carrá A Differential diagnosis of multiple sclerosis in Latin America. Mult Scler J Exp Transl Clin 2017;3(03): 2055217317714279
  • 3
    Solomon AJ. Diagnosis, Differential Diagnosis, and Misdiagnosis of Multiple Sclerosis. Continuum (Minneap Minn) 2019;25(03):611–635
  • 4
    Arvin AM, Wolinsky JS, Kappos L, et al. Varicella-zoster virus infections in patients treated with fingolimod: risk assessment and consensus recommendations for management. JAMA Neurol 2015;72(01):31–39
  • 5
    Winthrop KL, Mariette X, Silva JT, 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 (Soluble immune effector molecules [II]: agents targeting interleukins, immunoglobulins and complement factors). Clin Microbiol Infect 2018;24(Suppl 2):S21–S40
  • 6
    Buonomo AR, Zappulo E, Viceconte G, Scotto R, Borgia G, Gentile I. Risk of opportunistic infections in patients treated with alemtuzumab for multiple sclerosis. Expert Opin Drug Saf 2018;17(07): 709–717
  • 7
    Olberg HK, Eide GE, Cox RJ, et al. Antibody response to seasonal influenza vaccination in patients with multiple sclerosis receiving immunomodulatory therapy. Eur J Neurol 2018;25(03):527–534
  • 8
    Kim W, Kim SH, Huh SY, et al. Reduced antibody formation after influenza vaccination in patients with neuromyelitis optica spectrum disorder treated with rituximab. Eur J Neurol 2013;20(06): 975–980
  • 9
    Bar-Or A, Calkwood JC, Chognot C, et al. Effect of ocrelizumab on vaccine responses in patients with multiple sclerosis: The VELOCE study. Neurology 2020;95(14):e1999–e2008. Doi: 10.1212/WNL.0000000000010380
    » https://doi.org/10.1212/WNL.0000000000010380
  • 10
    Schwid SR, Decker MD, Lopez-Bresnahan MRebif-Influenza Vaccine Study Investigators. Immune response to influenza vaccine is maintained in patients with multiple sclerosis receiving interferon beta-1a. Neurology 2005;65(12):1964–1966
  • 11
    Bar-Or A, Freedman MS, Kremenchutzky M, et al. Teriflunomide effect on immune response to influenza vaccine in patients with multiple sclerosis. Neurology 2013;81(06):552–558
  • 12
    Olberg HK, Cox RJ, Nostbakken JK, Aarseth JH, Vedeler CA, Myhr KM. Immunotherapies influence the influenza vaccination response in multiple sclerosis patients: an explorative study. Mult Scler 2014;20(08):1074–1080
  • 13
    Metze C, Winkelmann A, Loebermann M, et al. Immunogenicity and predictors of response to a single dose trivalent seasonal influenza vaccine in multiple sclerosis patients receiving disease-modifying therapies. CNS Neurosci Ther 2019;25(02):245–254
  • 14
    Keshtkar-Jahromi M, Argani H, Rahnavardi M, et al. Antibody response to influenza immunization in kidney transplant recipients receiving either azathioprine or mycophenolate: a controlled trial. Am J Nephrol 2008;28(04):654–660
  • 15
    von Hehn C, Howard J, Liu S, et al. Immune response to vaccines is maintained in patients treated with dimethyl fumarate. Neurol Neuroimmunol Neuroinflamm 2017;5(01):e409
  • 16
    Lebrun C, Vukusic SFrench Group for Recommendations in Multiple Sclerosis (France4MS) and the Société Francophone de la Sclérose En Plaques (SFSEP) Immunization and multiple sclerosis: Recommendations from the French multiple sclerosis society. Mult Scler Relat Disord 2019;31:173–188
  • 17
    Farez MF, Correale J, Armstrong MJ, et al. Practice guideline update summary: Vaccine-preventable infections and immunization in multiple sclerosis: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology 2019;93(13):584–594
  • 18
    Mealy MA, Cook LJ, Pache F, et al. Vaccines and the association with relapses in patients with neuromyelitis optica spectrum disorder. Mult Scler Relat Disord 2018;23:78–82
  • 19
    Sociedade Brasileira de Imunizações. Calendário de Vacinaçao Pacientes Especiais 2020-2021. 2020
  • 20
    Ministério da Saúde Manual dos Centros de Referência para Imunobiológicos Especiais- 5a edição; 2019
  • 21
    Merck. Mavenclad - prescribing information; 2019
  • 22
    Armstrong C. IDSA releases recommendations on vaccinations in immunocompromised patients. Am Fam Physician 2014; 90:664–666
  • 23
    Berkhout A, Clark JE, Wen SC-H. In utero exposure to biologic disease-modifying anti-rheumatic drugs and effects to the infant: infectious complications, vaccine response, and safety of live vaccine administration. Expert Rev Vaccines 2019;18(05): 495–504
  • 24
    Chitnis T. Pediatric Central Nervous System Demyelinating Diseases. Continuum (Minneap Minn) 2019;25(03):793–814
  • 25
    Hughes AM, Ponsonby AL, Dear K, et al; Ausimmune Investigator Group. Childhood infections, vaccinations, and tonsillectomy and risk of first clinical diagnosis of CNS demyelination in the Ausimmune Study. Mult Scler Relat Disord 2020;42:102062
  • 26
    Suleiman L, Waubant E, Aaen G, et al; Network of Pediatric Multiple Sclerosis Centers. Early infectious exposures are not associated with increased risk of pediatric-onset multiple sclerosis. Mult Scler Relat Disord 2018;22:103–107. Doi: 10.1016/j.msard.2018.03.015.Early
    » https://doi.org/10.1016/j.msard.2018.03.015.Early
  • 27
    Di Pietrantonj C, Rivetti A, Marchione P, Debalini MG, Demicheli V. Vaccines for measles, mumps, rubella, and varicella in children. Cochrane Database Syst Rev 2020;2020:•••
  • 28
    Stowe J, Andrews N, Miller E. Do Vaccines Trigger Neurological Diseases? Epidemiological Evaluation of Vaccination and Neurological Diseases Using Examples of Multiple Sclerosis, Guillain-Barré Syndrome and Narcolepsy. CNS Drugs 2020;34(01): 1–8
  • 29
    Langer-Gould A, Qian L, Tartof SY, et al. Vaccines and the risk of multiple sclerosis and other central nervous system demyelinating diseases. JAMA Neurol 2014;71(12):1506–1513
  • 30
    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(02): 184–191
  • 31
    Barmettler S, Ong MS, Farmer JR, Choi H, Walter J. Association of Immunoglobulin Levels, Infectious Risk, and Mortality With Rituximab and Hypogammaglobulinemia. JAMA Netw Open 2018;1(07):e184169
  • 32
    Hersh CM, Cohen JA. Alemtuzumab for the treatment of relapsing-remitting multiple sclerosis. Immunotherapy 2014;6(03):249–259
  • 33
    Lycke J, et al. Lymphocyte subset dynamics following alemtuzumab administration in the CARE-MS I trial. in 89; 2012
  • 34
    Benamu E, Montoya JG. Infections associated with the use of eculizumab: recommendations for prevention and prophylaxis. Curr Opin Infect Dis 2016;29(04):319–329
  • 35
    Fisher H, Oluboyede Y, Chadwick T, et al. Continuous low-dose antibiotic prophylaxis for adults with repeated urinary tract infections (AnTIC): a randomised, open-label trial. Lancet Infect Dis 2018;18(09):957–968
  • 36
    Ciardi MR, et al. Reactivation of Hepatitis B Virus with Immune-Escape Mutations after Ocrelizumab Treatment for Multiple Sclerosis. Open Forum Infect Dis 2019;6:1–3
  • 37
    Reddy KR, Beavers KL, Hammond SP, Lim JK, Falck-Ytter YTAmerican Gastroenterological Association Institute. American Gastroenterological Association Institute guideline on the prevention and treatment of hepatitis B virus reactivation during immuno-suppressive drug therapy. Gastroenterology 2015;148(01):215-–219, quiz e16–e17
  • 38
    Idilman R. The summarized of EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. Turk J Gastroenterol 2017;28(05):412–416
  • 39
    Epstein DJ, Dunn J, Deresinski S. Infectious complications of multiple sclerosis therapies: Implications for screening, prophylaxis, and management. Open Forum Infect Dis 2018;5(08): ofy174
  • 40
    Nguyen ML, Flowers L. Cervical cancer screening in immunocompromised women. Obstet Gynecol Clin North Am 2013;40(02): 339–357
  • 41
    Triplett J, Kermode AG, Corbett A, Reddel SW. Warts and all: Fingolimod and unusual HPV-associated lesions. Mult Scler 2019; 25(11):1547–1550
  • 42
    Baldassari LE, Feng J, Macaron G, et al. Tuberculosis screening in multiple sclerosis: effect of disease-modifying therapies and lymphopenia on the prevalence of indeterminate TB screening results in the clinical setting. Mult Scler J Exp Transl Clin 2019;5 (03):2055217319875467
  • 43
    Navas C, Torres-Duque CA, Munoz-Ceron J, et al. Diagnosis and treatment of latent tuberculosis in patients with multiple sclerosis, expert consensus. On behalf of the Colombian Association of Neurology, Committee of Multiple Sclerosis. Mult Scler J Exp Transl Clin 2018;4(01):2055217317752202
  • 44
    Mills EA, Mao-Draayer Y. Understanding Progressive Multifocal Leukoencephalopathy Risk in Multiple Sclerosis Patients Treated with Immunomodulatory Therapies: A Bird's Eye View. Front Immunol 2018;9:138
  • 45
    Van Assche G, Van Ranst M, Sciot R, et al. Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn's disease. N Engl J Med 2005;353(04):362–368
  • 46
    Kim W, Kim HJ. Monoclonal Antibody Therapies for Multiple Sclerosis and Neuromyelitis Optica Spectrum Disorder. J Clin Neurol 2020;16(03):355–368
  • 47
    Berger JR, Malik V, Lacey S, Brunetta P, Lehane PB. Progressive multifocal leukoencephalopathy in rituximab-treated rheumatic diseases: a rare event. J Neurovirol 2018;24(03):323–331
  • 48
    Conway DS, Hersh CM, Harris HC, Hua LH. Duration of natalizumab therapy and reasons for discontinuation in a multiple sclerosis population. Mult Scler J Exp Transl Clin 2020;6(01): 2055217320902488
  • 49
    Foley 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;
  • 50
    Ho PR, Koendgen H, Campbell N, Haddock B, Richman S, Chang I. Risk of natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: a retrospective analysis of data from four clinical studies. Lancet Neurol 2017;16 (11):925–933
  • 51
    Ryerson LZ, Foley J, Chang I, et al. Risk of natalizumab-associated PML in patients with MS is reduced with extended interval dosing. Neurology 2019;93(15):e1452–e1462
  • 52
    Wattjes MP, Rovira À, Miller D, et al; MAGNIMS study group. Evidence-based guidelines: MAGNIMS consensus guidelines on the use of MRI in multiple sclerosis–establishing disease prognosis and monitoring patients. Nat Rev Neurol 2015;11(10): 597–606. Doi: 10.1038/nrneurol.2015.157
    » https://doi.org/10.1038/nrneurol.2015.157
  • 53
    Kartau M, Sipilä JO, Auvinen E, Palomäki M, Verkkoniemi-Ahola A. Progressive Multifocal Leukoencephalopathy: Current Insights. Degener Neurol Neuromuscul Dis 2019;9:109–121
  • 54
    Lachiewicz AM, Srinivas ML. Varicella-zoster virus post-exposure management and prophylaxis: A review. Prev Med Rep 2019; 16:101016
  • 55
    SPSP. Spsp – nota informativa (julho de 2018): atualização sobre sarampo. 1‣13; 2020.
  • 56
    Ministério da Saúde. Monitoramento do Período Sazonal da Febre Amarela Brasil – 2018/2019. informe n° 03 | 2018/2019.
  • 57
    Domingo C, Niedrig M. Safety of 17D derived yellow fever vaccines. Expert Opin Drug Saf 2009;8(02):211–221
  • 58
    Huttner A, Eperon G, Lascano AM, et al. Risk of MS relapse after yellow fever vaccination: A self-controlled case series. Neurol Neuroimmunol Neuroinflamm 2020;7(04):1–6
  • 59
    Farez MF, Correale J. Yellow fever vaccination and increased relapse rate in travelers with multiple sclerosis. Arch Neurol 2011;68(10):1267–1271

Publication Dates

  • Publication in this collection
    09 Jan 2023
  • Date of issue
    2022

History

  • Received
    07 June 2021
  • Accepted
    04 Oct 2021
Academia Brasileira de Neurologia - ABNEURO R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices Torre Norte, 04101-000 São Paulo SP Brazil, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revista.arquivos@abneuro.org