Buscar en
Revista Colombiana de Reumatología (English Edition)
Toda la web
Inicio Revista Colombiana de Reumatología (English Edition) Infections and systemic sclerosis: an emerging challenge
Journal Information
Vol. 27. Issue S1.
Systemic Sclerosis
Pages 62-84 (April 2020)
Share
Share
Download PDF
More article options
Visits
8477
Vol. 27. Issue S1.
Systemic Sclerosis
Pages 62-84 (April 2020)
Review Article – Meta-analysis
Full text access
Infections and systemic sclerosis: an emerging challenge
Infecciones y esclerosis sistémica: un desafío emergente
Visits
8477
Julián E. Barahona-Correaa,b, Alejandro De la Hoza, María José Lópeza, Javier Garzóna,c, Yannick Allanoree, Gerardo Quintana-Lópezb,d,
Corresponding author
gquintanal@unal.edu.co

Corresponding author.
a Division of Infectious Diseases, Hospital Universitario San Ignacio, Bogota, Colombia
b Grupo Reumavance, Division of Rheumatology, Hospital Universitario Fundación Santa Fé de Bogotá, Bogota, Colombia
c Department of Internal Medicine, Pontificia Universidad Javeriana, Bogota, Colombia
d Rheumatology Unit, School of Medicine, Universidad Nacional de Colombia, Bogota, Colombia
e Service de Rhumatologie, centre de référence Maladies auto-immunes systémiques rares, INSERM U1016, Institut Cochin, Hôpital Cochin, Paris, France
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (1)
Tables (4)
Table 1. Potential infectious complications/etiologies by immunosuppressant in patients with Systemic sclerosis.
Table 2. Screening recommendations and available prophylactic therapies proposed for autoimmune diseases.
Table 3. Potential prophylaxis to be considered in patients with SSC.
Table 4. Major recommendations for vaccination in patients with autoimmune inflammatory rheumatic diseases and patients with altered immunocompetence.
Show moreShow less
Abstract

Mounting evidence has shown non-systemic sclerosis (SSc) related complications as a rising cause of hospital admission and mortality, out of which infections are among the top-five causes. Patients with SSc are at an increased risk of infection due to several features of the treatment options and to the disease itself. For instance, lung involvement is associated with a higher frequency of respiratory infections, whereas the presence of digital ulcers or calcinosis may result in skin and soft tissue infections, and even osteomyelitis. On the other hand, the growing trend towards immunomodulation and immunosuppression in patients with autoimmune diseases will place SSc patients at a higher risk of infectious complications, including opportunistic infections. A low suspicion threshold and an increasing awareness among treating specialists, particularly rheumatologists, are warranted for prevention, early diagnosis and management of infectious complications. Nonetheless, data on risk management strategies in SSc, such as vaccination and antimicrobial prophylaxis, are scarce. A narrative non-systematic review was performed to provide an update of infectious complications in patients with SSc.

Keywords:
Systemic scleroderma
Infection
Communicable diseases
Antibiotic prophylaxis
Vaccination
Risk assessment
Resumen

La evidencia creciente muestra que las complicaciones no asociadas a la esclerosis sistémica (ES) son una causa cada vez más frecuente de hospitalización y mortalidad, dentro de las cuales las infecciones se encuentran entre las primeras cinco causas. Los pacientes con ES presentan un riesgo elevado de infección asociado a las opciones terapéuticas y por la enfermedad misma. Por ejemplo, el compromiso pulmonar se asocia a una mayor frecuencia de infecciones respiratorias, mientras que la presencia de úlceras digitales o calcinosis pueden resultar en infecciones de piel y tejidos blandos, e incluso osteomielitis. Por otro lado, la tendencia creciente hacia la inmunomodulación y la inmunosupresión como tratamiento de las enfermedades autoinmunes, pondrá a estos pacientes en un mayor riesgo de infecciones, incluidas las infecciones oportunistas. Son necesarios un umbral bajo de sospecha y un alto nivel de alerta entre las especialidades tratantes, particularmente los reumatólogos, para la prevención, el diagnóstico temprano y el manejo de las complicaciones infecciosas. Sin embargo, la información respecto a estrategias de gestión de riesgo en ES, como la vacunación o la profilaxis antibiótica, es escasa. Se realizó una revisión narrativa no sistemática que presenta una actualización sobre las complicaciones infecciosas en pacientes con ES.

Palabras clave:
Esclerodermia sistémica
Infección
Enfermedades transmisibles
Profilaxis antibiótica
Vacunación
Medición de riesgo
Full Text
Introduction

Systemic sclerosis (SSc), also called scleroderma, is an immune-mediated chronic connective tissue disease characterized by a disproportionate deposition of collagen in the skin and internal organs; fibrosis and vasculopathy are its main features.1,2 Two main subsets, limited and diffuse, are described according to the extension of cutaneous involvement.3 SSc presents the highest standardized mortality ratio (SMR, i.e., risk of death as compared with the general population) among rheumatic diseases.4 As a systemic disease, a myriad of signs and symptoms may be present, particularly renal, cardiopulmonary, gastrointestinal, and musculoskeletal. Thus, its management is an increasing challenge for rheumatologists and related specialties.

With regard to infectious diseases, patients with SSc are at an increased risk of infection due to the disease itself, mainly respiratory infections due to interstitial lung disease (ILD)5 and skin/soft tissue infections due to the presence of digital ulcers (DU) or calcinosis, that can progress to osteomyelitis.6,7

As well, the growing trend towards immunomodulation and immunosuppression will place SSc patients at a higher risk of infections, as increasingly observed in other autoimmune diseases,8 including opportunistic infections (OIs). Medications such as cyclophosphamide (CYC), mycophenolate mofetil (MMF), methotrexate (MTX), azathioprine (AZA), glucocorticoids, and biological therapy (i.e., rituximab [RTX]) are recommended for the management of disease manifestations.9–12 Several novel immunologic targets are under research with promising results.13 In addition, the intravenous route of application of prostanoids and some immunosuppressants may generate an increased risk of infection. Further, the prolonged use of proton pump inhibitors (PPI) and the use of antibiotic courses for microbial overgrowth are interventions to be considered as risk factors for infection.

Hereby, we offer an updated review of infectious diseases in patients with SSc, including their epidemiology, clinical manifestations and risk-management strategies.

Methods

A narrative non-systematic review was performed. We completed a literature search in 4 databases (Cochrane Database, Medline, Embase and LILACS [which includes literature from Latin America]). Articles in English, Spanish, French, Italian, Portuguese and German were included. The search strategy included the following terms: “Systemic scleroderma”, “Infection”, “Infectious Disease Medicine”, “Communicable Diseases”. Gray literature was obtained using these terms in Google and a ‘snowball’ strategy was applied to retrieve useful literature from the revised articles’ references.

Infections and SSc: epidemiology

Recent advances in the treatment of SSc have influenced the causes of death and in-hospital admission. A shift from renal crisis to cardiovascular (e.g., congestive heart failure [CHF]), pulmonary (i.e., ILD and pulmonary artery hypertension [PAH]), neoplastic and infectious causes as main drivers of death and hospitalization has been observed.5,14–26 A meta-analysis that included data until 2006 found a SMR of 3.53, which has not changed significantly over the last 40 years.14 Rubio-Rivas et al. reported a lower SMR (2.72) using a similar approach; the authors estimated a cumulative survival from onset (first Raynaud’s symptom) at 87% and 74% at 5 and 10 years, respectively, whereas from diagnosis the survival was 75% and 62%, respectively. Further, they described a trend towards improvement in mortality between the studies performed prior and after 1990, although statically significant only at 5-year survival from diagnosis (71% vs. 84%).27 Previously, the main driver of death in patients with SSc was renal crisis. The use of angiotensin-converting enzyme inhibitors led to a profound reduction from 42% of cases to less than 6% in the early 2000s.22 However, mounting evidence has shown that non-SSc-related causes for both death and admission are approaching 50%,14,15,17,20,22,23 out of which infections are becoming an important cause.

Infection as a cause of death

Two meta-analyses including data from 1960 to the first decade of the 2000s reported a 7% death frequency secondary to infection in SSc.14,19 Further, reports from the international European Scleroderma Trials and Research (EUSTAR) database, ranked infections among the top-five causes of death.15,24 A study using death certificates from France supported a similar frequency and reported infection as the cause of death in 11% of certificates. The authors reported a high mortality from lung infections and a five-fold higher rate of infectious deaths among patients with SSc compared with the general population24; similar evidence has been reported from several cohorts around the world.16,17,20,21,23,28

Some reports suggest a temporal shift in causes of death with infections being on the rise. Steen et al. reported that the proportion of infections as a cause of death among their cohort (1972–2001) ranged from 2% (1972–76) to 9% (1997–2001); although no statistical differences between periods were observed, a trend towards a higher frequency appears to be present.22 Further, two studies with a 10-year difference in the USA, based on the Healthcare Cost and Utilization Project-Nationwide Inpatient Sample (HCUP-NIS) databases and using a similar methodology, presented a significant rise in infections as a cause of death and admission26,29; Chung et al. (2002–2003) showed that infections were among the top-three causes of death, accounting for 13% of the events.26 Whereas Poudel et al., ten years later (2012–2013) reported that the most common diagnoses of deceased inpatients were infection/septicemia (32.7%), followed by pulmonary (20%) and cardiovascular (15.7%) involvement. Infection (other than opportunistic) had a high adjusted OR for mortality (3.36, CI 2.73–4.41).29

Infection as a cause of admission

Several cohorts have reported infections as a common cause of admission in patients with SSc, being ranked among the top three causes and as the most common non-SSc-related cause.17,18,21,26,29,30 Respiratory, skin/soft tissue, gastrointestinal, and urinary tract systems are the most common sites of infection causing hospital admission, although their frequencies differ by cohort. Infections have been associated with higher costs, longer length of stay and mortality.29

The occurrence of infections in SSc may be explained by an increasing use of immunosuppressive agents. Poudel et al. explored this hypothesis by comparing non-SSc-related hospitalizations against SSc-related hospitalizations with infection/septicemia, using a surrogate variable (a code for “chemotherapy” was available, which may represent the use of CYC or RTX, although it is not possible to ascertain). They found a significantly higher proportion of chemotherapy use in SSc hospitalizations with infection/septicemia, both in those who lived (9.2% vs. 4.1%, p < 0.0001) and those who died (17.4% vs. 6.3%, p = 0.0005). Those patients who died had higher proportions of PAH (43.3%), respiratory failure (72.4%), and CHF (38.4%), which may represent a more aggressive disease that required stronger immunosuppression and therefore, a higher risk of infection. Interestingly, the authors found that a higher income was associated with a higher risk of death. They proposed an access paradox, in which, among other causes, a higher income may represent a better access to immunosuppressive agents.29 Conversely, another study analyzing the immunosuppressive therapy hypothesis found no statistical association between respiratory infection and the use of immunosuppressants as a group.17 In this line, recent studies with Tocilizumab (TCZ)31,32 and Abatacept (ABA)33 do not support an increased risk of infections, as will be discussed later.

Infections and SSc: organ-specific involvement

Infectious complications in SSc can be attributed to disease- or drug-associated factors (Fig. 1). The former appears to be more frequent, mainly represented by soft tissue and pulmonary infections.

Fig. 1.

Infectious complications and risk factors in systemic sclerosis.

(0.19MB).
Skin and soft tissue infections

Skin involvement due to DU is one of the most frequent complications of SSc, and it can be associated with soft tissue infection in 19–54% of patients.6,7,34–36 The etiological mechanism of DU differs between ischemic ulcers, those related to articular deformities, and those associated with calcinosis.5 With regard to infectious complications, literature reports do not usually discriminate by mechanism, thus defining a clear risk difference is difficult.

Nonetheless, particularly in the presence of calcinosis, the development of cellulitis is a frequent complication.37,38 Progression to streptococcal pyomyositis,39 and cases of necrotizing fasciitis40 have also been reported.

Some studies have reported more infectious complications among the limited cutaneous SSc subset patients.41 DU infections can progress to osteomyelitis, with an incidence as high as 8%, which can increase to 17% in patients with active DU6,42–44 and 42% when those ulcers have any sign of infection.7 The most frequently isolated pathogens are Staphylococcus aureus, Pseudomonas aeruginosa and Escherichia coli, a pivotal information for the election of empiric antibiotic treatment.45 Furthermore, disease related disability is worsened by these infections, since 8–16% of patients require surgical amputation as part of their management.46 Patients who need surgical debridement, amputation or other acute intervention, will present an increased risk of wound infection, necrosis and slow wound healing, thus requiring a careful monitorization.47

Respiratory involvement

In some SSc series, the respiratory system is the most frequent site of infectious complications, accounting for up to 24% of the reported events.8 Diverse mechanisms have been proposed to explain this phenomenon, including ILD, which develops in almost 50–60 % of SSc patients, being the main cause of death in several series48,49; 40% of these deaths are attributed to lung infection.50 Due to pulmonary structural involvement, bacterial infections are frequent (e.g., pneumonia with or without necrosis, lung abscess, empyema, etc.).51 As patients with lung involvement present frequent flares, it is vital to rule out infectious complications as a cause of acute respiratory symptoms; early bronchoscopy may be necessary to identify infectious agents.52 Procalcitonin has showed some usefulness in this scenario, where a positive value can suggest an active bacterial infection, although the threshold is not well established.53,54

On the other side, esophageal dysmotility has been associated with aspiration pneumonia (OR 1.23, CI 1.05–10.55), major infection (OR 3.22, CI 1.01–10.51)30 and in-hospital death25; some authors have proposed that microaspiration may contribute to ILD.55,56 Pneumonia may be difficult to distinguish from ILD.52 Thus, Sehra et al.25 proposed that “strict aspiration precautions should be considered for SSc patients who are admitted to the hospital, including simple measures like elevation of the head of the bed, avoiding over-sedation, and swallowing evaluations to probably help decrease in-hospital mortality”.

Bacterial pneumonia and TB have also been reported in SSc. Two population-based studies in Taiwan (2000–2006 and 2001–2012) explored the risk of TB in SSc57 and rheumatic diseases,58 compared to age-matched healthy patients. SSc patients were found to have a significant 2.81-fold greater risk of overall TB infection (Incidence Rate Ratio [IRR] 2.81, CI 1.36–5.37) and 2.53-fold greater risk of pulmonary TB (IRR 2.53, CI 1.08–5.30); although non-significant, a trend towards a higher risk of extrapulmonary TB was reported (IRR 4.22, CI 0.27–17.57). SSc was found to be an independent risk factor for TB infection, with a hazard ratio (HR) of 2.99 (CI 1.58–5.63). Risk factors for TB in SSc were explored: age > 60 years and the presence of PAH were statistically significant in the multivariate analysis; interestingly, exposure to glucocorticoids or CYC were only significant in the univariate analysis.57 Lu et al. reported similar findings, although their methods differed, as the authors pooled together data from SSc and inflammatory myositis as a group.58 Moreover, among patients who received dexamethasone pulse therapy as part of their treatment, pulmonary TB was reported in 9% and extrapulmonary TB in 20%.59 Extrapulmonary TB has also been associated with the use of CYC, high doses of glucocorticoids and Tumor Necrosis Factor (TNF)-ɑ inhibitors.60,61 Some series have reported non-TB mycobacteria infections in 18% of patients with ILD.62

Lung involvement by other opportunistic microorganisms include infection by Pneumocystis jirovecii which, although seldom reported in SSc patients,63,64 has been associated with high dose glucocorticoids (i.e., ≥20 mg/day in this study) or lower glucocorticoids doses with concomitant use of CYC65 with a mortality close to 50%. Therefore, a low threshold of suspicion must be considered among rheumatologists and treating specialists to make the diagnosis in patients with highly immunosuppressive treatment who consult with progressive dyspnea and dry cough66; prophylaxis should be considered in high risk patients.67

Aspergillus fumigatus fungal ball has been found on large bullous spaces in patients with ILD, so it should be considered in patients with hemoptysis and lung masses.68 A case of pulmonary actinomycosis in a patient on infliximab was reported.69

Gastrointestinal involvement

Small intestinal bacterial overgrowth (SIBO) is a common gastrointestinal manifestation. Several antibiotics, such as amoxicillin, quinolones (e.g., ciprofloxacin), tetracyclines (e.g., doxycycline), metronidazole, minimally absorbable antibiotics (e.g., rifaximin) and trimethoprim/sulfamethoxazole (TMP/SMX) may be considered for SIBO treatment.70,71 Cyclical courses may be given monthly during 10–14 days. From an infectious diseases perspective, one should not forget that the regular use of antibiotics is linked with antibiotic associated diarrhea and a higher risk of Clostridioides difficile infection.72,73 Although clindamycin is often considered as the major driver of C. difficile infection, most of the mentioned antibiotics for the treatment of SIBO may be associated as well.73 A myriad of symptoms may be present and range from mild to severe systemic compromise. The main acute symptoms that should prompt active search are malaise, abdominal pain, diarrhea, nausea, vomiting, fever and leukocytosis.74 Moreover, the presence of gastrointestinal involvement has been related to a greater incidence of non-strongyloidiasis diarrhea (OR 2.28, CI 1.21–6.56).30

Interestingly, a high incidence of esophageal candidiasis was reported in the late 80 s in patients with SSc who needed PPIs and was linked by the authors to esophageal dysmotility, rather than to immunosuppressive treatment.75,76 Nonetheless, this phenomenon appears to be uncommon in recent literature. In fact, only one case was reported among the retrieved articles.30 Further, in our experience, esophageal candidiasis is seldom seen in clinical practice.

Helicobacter pylori prevalence in SSc patients is an interesting phenomenon, as it can be almost twice the one of immunocompetent population, although this prevalence is mainly driven by ELISA positivity.77 Its occurrence has been associated with the presence of disease activity and severity, and its detection and eradication have shown significant improvement in SSc symptoms and disease activity.78,79 Several eradication regimens are available and should be considered based on local epidemiology, patient preferences, risk factors and medical consideration.80

Another complication with the potential of infectious complications is the requirement of parenteral nutrition, mostly derived from gastrointestinal involvement, such as pseudo-obstruction, malabsorption, and malnutrition. It is considered as a safe intervention, although the incidence of bacteremia presents a variable rate (0.19–1 in 1000 catheter-days).81,82

Opportunistic infections and others

The advent of biological therapy and the use of both glucocorticoids and other immunosuppressants in rheumatology practice render patients on these therapies prone to OI, for whom a specific definition has been proposed.83 Currently, OIs in SSc are considered to be less frequent than in other rheumatic diseases, given that most of the available information is limited to case reports. Thus, recommendations for a prophylactic or diagnostic approach are difficult, but they should be considered in highly immunosuppressed patients.

Extrapolated data of OIs from other diseases in which similar therapies are used (e.g., rheumatic, malignancies, transplants, etc.) might be considered. This kind of infections have been described in patients with high immunosuppression requirements, especially with high glucocorticoids dose (i.e., ≥30 mg/day), CYC or biological therapy.

Several different OIs have been reported in SSc, such as P. jirovecii pneumonia (as previously described)63–65; disseminated candidiasis on high glucocorticoids doses (i.e., prednisolone at a dose of 40 mg/day, followed by methylprednisolone pulse therapy)84; cryptococcosis, with pulmonary and extrapulmonary involvement85,86; cerebral toxoplasmosis87; tenosynovitis, septic arthritis and skin abscesses caused by atypical mycobacteria88–90; and infections by Nocardia farcinica with subcutaneous abscesses or pulmonary infections.91,92

Similarly, viral reactivations have been reported in patients on high immunosuppressive therapy. Cytomegalovirus (CMV) reactivations present complications similar to those described in other high risk groups, including splenic rupture, pneumonitis, pancreatitis, esophagitis or duodenal perforation, mainly related with high dose glucocorticoids and CYC.93–96 Human Herpes Virus 6 (HHV6) asymptomatic reactivation has been detected in SSc patients, although it may represent an epiphenomenon linked to immunosuppression, without clinical relevance.97 There are also reports of a Herpes Simplex Virus (HSV) -1 necrotizing pleuritis98 and parvovirus B19 alveolitis associated with ILD.99 Herpes Zoster Virus (HZV) reactivations can happen in 5% of patients, mainly with cutaneous involvement, but complications such as Guillain-Barré syndrome, have been reported.100 Hepatitis B Virus (HBV) reactivation prevalence in rheumatoid arthritis has been described in 3–3.5 %, but there are no data of its prevalence in SSc patients. Nevertheless, when high immunosuppressive doses are considered as part of SSc treatment, patients with a high risk profile (i.e., health care workers, high risk sexual activity, hemodialysis patients, injectable drug users, multiple sexual partners, and household contacts of HBV-infected individuals) must be screened for chronic HVB infection, and prophylaxis must be offered if indicated.101,102 HBV reactivation must be suspected when an increase in liver enzymes is observed, mainly alanine aminotransferase, particularly in patients with evidence of past HBV infection (i.e., positive HBV core antibody).102 If suspected, serum HBV DNA should be assessed and consultation with a specialist (e.g., gastroenterology, infectious diseases) is advisable. A low threshold of suspicion must be considered: although hepatotoxicity from medications is often seen, HBV reactivation may explain this finding and must be ruled out in at-risk patients.

Data on Human Papilloma Virus (HPV) infection incidence in SSc are contrasting, but up to a five-fold increased incidence when compared with healthy age matched controls has been reported.103,104 Routinely gynecological screening must be considered based on national guidelines.

Immunosupressants and infections

Currently, treatment strategies are directed towards management for specific organ involvement and symptoms relief, with immunomodulation as a pivotal strategy for the management of skin, lung and articular involvement.9,10 In selected patients, transplantation proceedings, such as hematopoietic stem cell transplantation (HSCT), are also available with a subsequent risk of infection.105 Solid organ transplantation is also feasible and is particularly offered for patients with organ-specific involvement, including lung (i.e., ILD or PAH), heart, and kidney; even heart-lung transplant is possible in selected cases. Clinical and survival outcomes are comparable to those observed in patients without SSc, including infectious events,106–109 for which recent guidelines have been issued.110 Further, other frequent interventions, particularly intravenous medications, such as prostanoids may turn SSc patients more prone to infections.5Table 1 summarizes potential infectious complications/etiologies by immunosuppressant in patients with SSc.

Table 1.

Potential infectious complications/etiologies by immunosuppressant in patients with Systemic sclerosis.

Glucocorticoids (146,148) 
  • Bacteria: Gram-positive, Gram-negative, Listeria monocytogenes, Nocardia spp, TB, Non-TB- mycobacteria

  • Fungi: Candida spp., Aspergillus spp., Pneumocystis jirovecii, Cryptococcus, Histoplasma capsulatum

  • Virus: HSV, HZV, CMV, EBV, Influenza, Adenovirus

  • Parasites: Toxoplasma, Cryptosporidium spp, Strongyloides spp

 
Cyclophosphamide (5,112,114,118–127,184,185) 
  • Bacteria: Gram-positive, Gram-negative, mycobacteria

  • Fungi: P.jirovecii, C. albicans, invasive fungal disease

  • Virus: HSV, HZV, BK-virus and adenovirus associated hemorrhagic cystitis, JC-virus/PML, HBV reactivation

  • Increased risk of upper RTI, pneumonia, UTI, skin/soft tissue infection.

 
Methotrexate (102,125,126,144,145) 
  • Skin and soft tissue infections, RTI

  • Virus: HZV (debated)

 
Azathioprine (102,112,115,125,126,135–138) 
  • Virus: HZV, HSV, CMV, EBV

 
Mycophenolate mofetil (113,116,117,129–132) 
  • RTI, UTI and skin/soft tissue infection, pneumonia, HZV

 
Rituximab (102,125,153–155,157,158,160) 
  • Bacteria: Enterobacteria, Gram-positive.

  • Fungi: P. jirovecii, A. fumigatus, Candida spp.

  • Virus: HBV reactivation, HZV, CMV, JC virus/PM

 
Tocilizumab (31,102,125,126,162–165,168) 
  • Bacteria: S. aureus (including disseminated infections), E. coli, Streptococcus intermedius, Non-TB-mycobacteria

  • Virus: HZV

  • Necrotizing skin and soft tissue infections

  • UTI, RTI, nasopharyngitis

 
Abatacept (102,125,126,169–171,174) 
  • Virus: HSV

  • Upper RTI, pneumonia, UTI

  • Seldom seen: OIs (peritoneal tuberculosis, fungal esophagitis, fungal eye infection, fungal sinusitis, and Candida spp.)

 
TNF α inhibitors (69,177–180) 
  • Bacteria: Non-TB-mycobacteria

  • Virus: HZV, HBV reactivation

  • Fungi: endemic mycoses, CMV, P. jirovecii, Aspergillus spp.

  • RTIs, UTIs, gastrointestinal, skin/soft tissue infections

 

CMV, Cytomegalovirus; EBV, Epstein-Barr Virus; HBV, Hepatitis B Virus; HCV, Hepatitis C Virus; HSV, Herpes Simplex Virus; HZV, Herpes Zoster Virus; JC-virus, John Cunningham -virus; OI, Opportunistic Infection; PML, Progressive Multifocal Leukoencephalopathy; RTI, Respiratory Tract Infection; TB, Tuberculosis; TNF, Tumor Necrosis Factor; UTI, Urinary Tract Infection.

This table is based on references and author’s experience.

Synthetic immunosuppressantsCyclophosphamide

CYC is an alkylating agent that inhibits DNA replication, particularly in hematopoietic cells and lymphocytes due to their high proliferation rate, ultimately leading to their apoptosis.111 CYC appears as a first-line therapy for the treatment of ILD and is considered in severe skin involvement.9 This agent has been associated with respiratory infections, although the presence of lung structural changes that may account for an increased risk of infections should not be disregarded.5 Data on CYC and infections will be approached from two standpoints: regular- and high-dose, as infection risk is expected to differ. Although a recommended standard regular dose is lacking,10 a dose of 500−600 mg/m2 on a monthly basis is widely accepted, which will be discussed in this subheading. In contrast, high doses exceed this dosage and are mainly reported in Hematopoietic Stem Cell Transplant (HSCT) trials, a subject that will be addressed later.

Randomized controlled trials of CYC compared with a variety of interventions have shed light onto this subject. Ludici et al. reported UTIs in 6 patients (13.3%) and respiratory infections in 4 (8.8%).112 A recent metanalysis of CYC in connective tissue disease-associated ILD found no difference in pneumonia rates when compared either to placebo or MMF113; only two studies versus placebo were included: one study reported 6 out of 79 patients with pneumonia, as the only recorded infectious event,114 and, the other study reported no differences in infections between groups, although 13.6% (n = 3) of patients in the intervention group presented intercurrent upper respiratory infection115 with a more intense immunosuppressive therapy (i.e., glucocorticoids plus CYC followed by AZA). The comparison of CYC against MMF found similar results.113,116,117 In a meta-analysis of 18 studies until 2010 of CYC for lung involvement, Poormoghim et al reported only one discontinuation of CYC due to pneumonia and only one episode of HZV.118

Some other infectious complications have been reported. For instance, one of the frequent adverse events during CYC therapy is hematuria, which is usually considered secondary to CYC itself.118 However, BK-virus associated hemorrhagic cystitis in a patient with SSC treated with CYC has been reported, thus highlighting the need to rule out an infectious cause.119 Further, a case of hemorrhagic cystitis associated to adenovirus was described in a HSCT, probably secondary to CYC mobilization (considered as a high-dose).120

Although infections due to CYC appear to be rather infrequent, the experience in other rheumatic diseases should prompt awareness. Woytala et al reported infections in 37% of patients (n = 24) with different rheumatic diseases, including skin, upper and lower respiratory, and UTIs; few episodes of HZV (3%) were observed.121 Experience in systemic lupus erythematosus (SLE) has suggested an increased risk of infection from a variety of etiologies122–124; CYC has been considered as a low risk medication for progressive multifocal leukoencephalopathy (PML).125,126 An association with HZV appears to be present in rheumatoid arthritis (RA) on CYC.126 HBV reactivation has been reported in the literature and a shorter period to reactivation (8 weeks) compared to other immunosuppressants was identified.127

In conclusion, an increased risk of upper respiratory infections and probably UTIs appears to be associated with CYC. However, treatment interruption is not often needed.

Mycophenolate mofetil

MMF is a potent inhibitor of inosine monophosphate dehydrogenase (IMPDH), which is pivotal for the proliferation of T and B lymphocytes.111,128 MMF is recommended among the first line therapies for ILD,9 although its safety in SSc treatment appears as an objective in the research agenda.10

A systematic review on MMF safety in SSc reported 34 infectious events in 487 included patients, including respiratory (n = 11), skin (n = 4), and UTIs (n = 2).129 Boulos et al. reported only one discontinuation due to recurrent infections after 5 years on therapy in patients treated with MMF (n = 74).130 Gordon et al.131 compared MMF alone vs. MMF plus belimumab in 20 patients, with a 1:1 randomization. No differences in infection rates were observed. Naidu et al. reported a similar infection rate compared to placebo in Indian patients.132

A recent meta-analysis of CYC in ILD in SSc that included two studies,113,116,117 found no difference in pneumonia rates when comparing MMF with CYC.

Data from renal transplant and SLE patients have reported a decreased production of IgG and IgM, thus increasing infection risk and suppressing response to some vaccines.133

These findings support an acceptable safety profile of MMF from an infectious risk standpoint, and may support its election over CYC, although evidence from lupus nephritis appears to show no difference between both.134

Azathioprine

Azathioprine (AZA) is a cytotoxic agent that interferes with DNA and RNA cycles through the insertion of 6-tioguanine nucleotide.111 AZA is considered as a second line agent for maintenance therapy in ILD.9 Evidence of infections in SSc is scarce and its safety in these patients appears as an objective in the research agenda.10 Ludici et al. did not report infectious events in 24 patients on AZA,112 whereas Dheda et al. found one episode of TB in 8 patients assessed.115 Nonetheless, some evidence from the treatment of inflammatory bowel disease suggests a 3-fold increased risk of OIs with the use of AZA, particularly viral (i.e., HZV, HSV, CMV, Epstein-Barr Virus [EBV]),135 and a 7% frequency of infectious events.136–138 An association with HZV appears to be present in RA on AZA.126

Methotrexate

Methotrexate (MTX) is a folic acid analogue that inhibits dihydrofolate reductase activity, thus undermining lymphocyte proliferation and promoting an anti-inflammatory milieu.139 MTX should be considered for the management of skin and articular involvement in SSc.9,10

The evidence from randomized controlled trials in the management of SSc has shown infections as a rare event in this population, with doses up to 25 mg/week.140–143 Evidence from IBD treatment appears to support its safety.138 MTX has been associated with a mild increased risk of infection, particularly skin/soft tissue and respiratory tract infections, in patients with rheumatoid arthritis (RA).144,145 Although an increased risk of HZV has been suggested, a large national database-based study in the USA found no association.126

Glucocorticoids

Glucocorticoids influence the inflammatory response through several pathways. Specifically, by a reduction in the innate response and in the production of inflammatory cytokines, an inhibition of both classic and alternative pathways of the complement system, and a reduced capacity for antigen presentation, lymphocyte proliferation and differentiation; a shift towards a Th2 phenotype is generated.146 Low dose glucocorticoids have been recommended, mainly based on expert opinion, for the management of articular symptoms9 and some evidence for myositis is available, although its use is debated; however, a wide share of patients with SSc receive them as comedication with other immunosuppressants.147 Immunological alterations explain the increased risk of infectious events due to both intra- or extra-cellular microorganisms, including bacteria, fungi, virus, mycobacteria and parasites146; OIs, such as P. jirovecii pneumonia, TB and HZV are increasing major concerns.148 High-dose glucocorticoids have been associated to an increased risk of fatal infections in patients with autoimmune diseases.149,150

Biological immunosupressants as potential novel interventions for SScRituximab

Rituximab (RTX) is a monoclonal antibody that targets CD20 and depletes most of B-cells virtually sparing plasmatic cells, thus altering several immunologic pathways.13,151 Although not readily approved by regulatory agencies for the treatment of SSc, RTX is recommended by experts for the treatment of refractory ILD and articular involvement.9 Mounting evidence supports its efficacy, either alone or in combination, for these systems and even in skin involvement.152–155 Infectious disease risk appears to depend on the underlying diagnosis, as it may be as high as 50% in patients with lymphoma or below 5% in patients with RA or systemic vasculitis. Hypogammaglobulinemia is the main driver of infection in these patients, in whom the highest risk occurs during the first 3 months of therapy, although substantial risk is present thereafter.156 Neutropenia can also occur and severe neutropenia is reported in up to 30% of patients. Early neutropenia is rare, nonetheless, the so-called late-onset neutropenia is observed in up to 15% of patients 1–5 months after the end of therapy. Its mechanism is still unknown as is considered to be immune-mediated; it can persist for months, may resolve spontaneously, and in persistent cases, its response to granulocyte colony-stimulating factor therapy is poor or transient. Its impact on infection risk remains to be elucidated.157 Different microorganisms have been reported as etiologies of infectious events on RTX for both neoplastic and autoimmune diseases, particularly by Enterobacteriaceae. Further, OIs have been reported including P. jirovecii pneumonia, A. fumigatus, Candida spp., HZV, CMV and PML,158 although for the latter, RTX has been considered as a low risk medication.125 However, evidence is conflicting as data derived from RA did not revealed an increased risk of OIs nor viral reactivations, although the lack of long-term follow-up hampers to ascertain this risk.159 Regarding specific data on SSc, Thibaut et al. reported in a literature review that infectious events are frequent, but usually not severe.153 Melsens et al. did not report infectious complications in 17 patients with SSc.154 Noteworthy, interim results from the EUSTAR Network reported infections in 76 out of 248 (30%) patients with SSc using RTX.155

One of the major concerns of RTX, is its increased risk of reactivation of latent HBV infection, that is considered the highest among all the available biologics: a rate of up to 16.9%, although this information comes from patients on concomitant antineoplastic agents.160 This risk appears to be lower among rheumatic diseases (<10%), thus, close monitoring is advised.102

Tocilizumab

Tocilizumab is a humanized monoclonal antibody directed towards IL-6 receptor.161 TCZ is considered a promising molecule in the treatment of SSc and is recommended as an alternative in articular involvement.9,13 The FaSScinate phase 2 study explored its utility in SSc and reported that up to 56% of patients (n = 24) presented at least one infectious event. Noteworthy, this frequency did not differ from the placebo arm, although serious infectious events were more frequent in the TCZ arm (16% vs. 5%). One death due to lung infection was reported. The most frequent infections were nasopharyngitis (n = 5), UTI (n = 3) and HZV (n = 3).31 Data from the phase 3 trial did not report an increased risk of infection when compared to placebo.32 Noteworthy, Distler & Distler expressed a relevant concern on infectious disease risk: the authors of the study reported two events of proximal interphalangeal joint osteomyelitis in patients receiving TCZ, probably associated with digital ulcers. As TCZ inhibition of IL-6 has been shown to blunt PCR elevation and probably signs of infection, a low threshold of clinical suspicion is warranted162; from an infectious disease perspective, the follow-up of osteomyelitis under IL-6 inhibition may present a major challenge. Evidence from RA experience has suggested an increased risk of necrotizing skin infections, as well as S. aureus and non-TB mycobacteria disseminated infections.163–166 Neutropenia was observed in up to 30% of RA patients in pivotal studies, although less than 5% presented severe neutropenia. Its mechanism is still unknown, although a redistribution of the neutrophil compartment has been proposed. Its consequences on infection risk are unclear, although data from RA trials do not support an association with an increased risk; dose interruption is an effective risk mitigation strategy that yields a rapid recovery of neutrophil counts.167,168

Abatacept

Abatacept is a soluble fusion molecule formed by the extra-cellular domain of CTLA-4, which inhibits the activation of T lymphocytes through its union with CD80 and CD86, thus blocking the interaction with CD28 161. The efficacy of ABA in the management of skin and articular involvement in SSc has been suggested 13. Chakravarty et al. (n = 10) found no statistical differences in infection rate between the intervention and the control group; two events of upper respiratory infection were reported in the ABA arm.169 Similarly, a retrospective study (n = 11) in patients with articular involvement reported 3 infectious events: two episodes of bronchitis and one HSV infection.170 Results from a phase 2 trial in SSc suggested a comparable infection risk profile between ABA and placebo.32 Pooled-evidence from RA studies does not support an increased risk for infection compared to other therapies,171–173 although the long-term extension (5 years) report of the ACQUIRE study showed that 70% of patients experienced an infectious event during follow up, mainly mild or moderate.174

Belimumab

In a pilot study, Gordon et al compared MMF alone vs. MMF plus belimumab (BEL) in 20 patients, with a 1:1 randomization. Although no differences between the interventions in the assessed outcomes were observed, basic science evidence supports its potential use13 and the authors suggest the performance of a larger clinical trial.131 No differences in infection rates were observed, with 18 in the MMF/BEL vs. 16 in the MMF alone. The most frequent infections in the MMF/BEL were upper respiratory involvement (n = 5), UTI (n = 3) and HSV stomatitis (n = 2).

Tumor necrosis factor α inhibitors

Tumor Necrosis Factor (TNF) α inhibitors might play a role in the management of articular compromise, although they are not currently recommended by experts as a first line therapy.9,13,175 Data for etanercept, adalimumab and infliximab have been reported from small studies in patients with SSc.176,177 Regarding infectious disease risk, a low frequency of events has been reported: uncomplicated HZV (although this patient received concomitant prednisone and AZA),177 infected DU and one sepsis episode.178 Lam et al. reported no cases of OIs in 18 patients.179 Anecdotally, one case of pulmonary actinomycosis was reported on infliximab.69 Nonetheless, safety concerns based on data from other autoimmune/autoinflammatory diseases should be considered. Patients on TNF-α inhibitors may present a double risk of serious infections compared to placebo.180 Several infectious complications have been reported, namely respiratory infections, UTIs, gastrointestinal infections, skin/soft tissue infections, HZV, and HBV reactivation.180 OIs have been described, such as endemic mycoses, CMV, P. jirovecii pneumonia, Aspergillus spp., among others, in which atypical manifestations may be observed.180 As TB infection is a major concern on TNF-α inhibitors, active screening prior to biologic application must be performed. Non-TB-mycobacteria infections may be observed.180

Hematopoietic stem cell transplant and high dose CYC

Hematopoietic Stem Cell Transplant (HSCT) is an emerging therapeutic strategy in selected patients with SSC. The procedure is performed in three main phases: 1) mobilization of stem cells from the bone marrow to the peripheral blood using priming agents (i.e., high dose CYC), followed by the collection of the mobilized stem cells; 2) immunoablation or myeloablation through the administration of conditioning regimens (i.e., total body irradiation or alkylating agents); 3) infusion of autologous CD4+ stem cells, the so-called “transplantation”.181,182

Infectious complications are usual in the vast majority of transplantation procedures and three risk phases have been proposed. The first phase is known as ‘early pre-engraftment’, which occurs previous to the “transplantation” during the first 2–4 weeks, and is characterized by infections due to mucositis and neutropenia. The most frequent etiologies are bacterial (Gram-positive or Gram-negative), HSV and Candida spp. The second phase is known as ‘early post-engraftment’, and occurs from days 30–100 after the “transplantation”. The most frequent etiologies are bacterial, fungal (e.g., Aspergillus spp., P. jirovecii), and viral (e.g., CMV, EBV) infections. The last phase is known as ‘late post-engraftment’ and is characterized by infections due to encapsulated bacteria, Nocardia spp., Aspergillus spp., P. jirovecii, CMV, EBV, among others. Nonetheless, an increased incidence of respiratory virus infections and complications is present throughout all the phases.105 These periods are as applicable in patients with refractory systemic autoimmune diseases undergoing autologous stem cell transplantation. Szodoray et al. reported that immune reconstitution started with CD56 + NK cells around day 30, followed by CD8 + T cells around day 60 post-transplantation. The number of B cells and CD4 + T cells became normal within 150 days.183

The HSCT experience in SSc has illustrated the risk of infection. The SCOT trial compared CYC in a high dose regimen (i.e., initial intravenous dose of 500 mg/m2, followed by 11 monthly infusions of 750 mg/m2) with HSCT (which included a mobilization dose of CYC of 120 mg/Kg), and reported a similar infection rate among both groups. In the CYC arm, 84% of patients (n = 31) presented an infectious disease during follow-up, although only 9 patients presented a serious infection. The most frequent infections were UTIs (40%) and upper respiratory infections (21.6%); up to 10% of patients presented an infected DU. In contrast, in the HSCT group 97% of patients (n = 33) presented an infectious episode and serious infections appeared to be more frequent. The most frequent infections were HZV (38.2%), upper respiratory infection (26.5%) and pneumonia (26.5%); bacteremia was reported in 11.8% of patients. Varicella Zoster occurred in 12 patients in the HSCT compared to 1 in the CYC arm. Five cases of CMV reactivation were observed in the HSCT arm.184

The ASTIS trial compared CYC (i.e., 750 mg/m2 monthly for 12 months) with HSCT (i.e., mobilization dose of CYC of 200 mg/Kg for 4 days); viral infection or reactivation after randomization were recorded. Infections were infrequent among the CYC group (n = 77) with only one episode of HSV primary infection. In contrast, 27.8% of patients (n = 22) in the HSCT group presented an infectious disease. Reactivation of EBV (12.5%), HSV (22%) and recurrent CMV (18.7%) were the most frequent phenomena.185

The ASSIST trial compared CYC (i.e., 1000 mg/m2 monthly for 12 months) with HSCT (i.e., mobilization dose of CYC of 200 mg/Kg for 4 days). The authors reported only one episode of cellulitis on CYC and one episode of C. difficile positive stool sample in both arms, Micrococcus bacteremia and CMV reactivation were observed in the HSCT arm.186

Del Papa et al., in a retrospective study of HSCT in SSc, recorded eight cases of fever of unknown origin, five cases of fever with positive blood culture and three cases of pneumonia. All the observed infections resolved with adequate antibiotic treatment.182

Solid organ transplantation

Solid organ transplant recipients also have an increased risk of infectious complications, in close relation to the dose and type of immunosuppressive drugs used. Other factors related include surgical complications, prolonged hospitalization, other invasive procedures, and viral reactivations, mainly CMV, with increased risk of opportunistic infections, allograft injury and rejection.187,188

In any post-transplant patient, the risk of infection is a function of the net state of immunosuppression. As described in bone marrow transplant recipients, the risk of infection has been divided into three periods, the first is referred as “early period”, which includes infections within the first month post transplantation and is characterized by health care-related infections, donor derived infections (e.g., HSV, rabies, HIV, Trypanosoma cruzi) and recipient derived infections, usually related to previous colonization. The second or “intermediate period” includes infections during the period from 1 to 6 months after transplantation. It is characterized by the greatest risk of opportunistic infections, mainly intracellular microorganisms, including P. jirovecii, TB, Cryptococcus neoformans, CMV, EBV, Toxoplasma gondii, among others. The third or “late period” occurs after the sixth month post transplantation and is characterized by an increased incidence of community acquired infections, mainly pneumonia and UTIs. Infections by Aspergillus spp., Zygomycota, CMV infections (colitis and retinitis), and post-transplantation lymphoproliferative disorder, associated in most cases to EBV have been described.188

In solid organ transplant recipients, the most common site of infection is the allograft. In SSc patients, the most frequently transplanted organ is the lung. Bacterial infection is the most frequent infectious complication (35–66%) for recipients with unilateral or bilateral lung transplant. Bacterial infections presenting in the immediate post transplantation period usually show relation with pre-transplantation colonization, the surgical procedure itself and accompanying technical complications, intubation or prolonged hospitalization. Late onset bacterial infections are related to increased immunosuppression due to rejection, invasive diagnostic procedures and the development of bronchiolitis obliterans.189

Other therapies and their potential risk of infection

The intravenous route of some therapies for SSc management should raise awareness towards the potential for infectious complications. Prostanoids (e.g., epoprostenol, treprostinil) are recommended for the management of PAH, Raynaud’s phenomenon and DU.9,10 Cellulitis and sepsis have been described with their application. Among the most frequent sepsis etiologies are Staphylococcus epidermidis, Micrococcus spp., S. aureus, Pseudomonas spp., and Enterobacter spp.190,191 A higher infection rate associated with treprostinil compared to epoprostenol has been suggested.5,192,193

In patients with gastroesophageal reflux disease or gastrointestinal dysmotility, the use of high dose PPIs is recommended.9 Recently, the prolonged use of PPIs has been associated with a higher risk of C.difficile infection73,194,195 and community-acquired pneumonia, although evidence for the latter is conflicting.196–199

With regard to ILD management with anti-fibrotic therapy, current evidence supports its efficacy in SSc. Nintedanib is not associated with an increased risk of infection in SSc or other patient population.200–203 As observed with most tyrosine kinase inhibitors, a common side effect is diarrhea, which has not been associated with an infectious etiology. Although its cause is still unknown, receptor inhabitation and direct mucosal damage have been proposed.204,205 On the other hand, although direct evidence of pirfenidone in SSc is lacking, trials are ongoing; to date, pirfenidone has not been associated with an increased risk of infection in other patient population.206,207

Other therapeutic strategies addressing several domains of the disease have not been associated with a higher risk of infection in SSc. These include phosphodiesterase-5 inhibitors, calcium channel blockers, angiotensin-converting enzyme inhibitors, angiotensin receptor antagonist, endothelin receptor antagonists, and riociguat.10,208–213

Infection prevention

Risk management strategies, including prophylaxis and vaccination must be considered within the bundle of care. Tables 2 and 3 summarize screening recommendations, potential prophylaxis and available prophylactic therapies to be considered in patients with SSc. Several recommendations guidelines for the prevention of infection in patients receiving immunosuppressive treatments are available.157,168,214–223 However, there is a lack of information in the literature on infectious diseases prevention in patients with SSc, probably due to the low incidence of the disease. Thus, extrapolated data from other diseases, in which similar therapies are used (e.g., rheumatic, malignancies, transplants, etc.), might be considered. When considering vaccination and chemoprophylaxis for patients with SSc, the first thought a rheumatologist should have is: how immunosuppressed is my patient and what is the mechanism of immunosuppression? One shall not forget an increased risk of infections due to the disease itself. Vaccination, screening for latent or active infections, and several other measures for infection prevention should be performed prior to immunosuppressive agent application (215,223,224)

Table 2.

Screening recommendations and available prophylactic therapies proposed for autoimmune diseases.

Recommendation by infectious disease  Available therapies 
Tuberculosis (TB)Screening: TST or IGRA (based on local guidelines).TST > = 5 mm or IGRA (+):1) Rule out active TB (Chest X-rays, sputum smear and culture).2) ID/ TB specialist consult should be considered. 3) Initiate LTBI treatment TST < 5 mm or IGRA (-): 1) Routine screening is not recommended and should be based on suggestive symptoms or potential TB exposure.  Isoniazid 300 mg PO/day + Pyridoxine 50 mg PO/day for nine months (270 doses)Other therapies are available and should be considered based on the patient history and the medical concept.Should be given for at least one month prior to immunosuppression 
P. jirovecii pneumoniaaTo be considered in patients receiving CYC, GC or RTX.   TMP/SMX 800 mg/160 mg PO on alternate days or 400 mg/80 mg PO every daybIn case of adverse reaction, other therapies may be considered – ID specialist consult should be considered. 
Hepatitis B Virus (HBV)Screening should be considered in every patient prior to immunosuppressive therapy. Baseline HBV profile should include: antiHbs, HbsAg, and antiHbc. 1) antiHbs (-), HbsAg (-), antiHbc (-): vaccination should be initiated.2) antiHbs (-), HbsAg (+/-) antiHbc (+): Prophylaxis should be considered. Complete profile must be performed, including: HbeAg, antiHbeAg, ALT, serum HBV DNA. 3) antiHbs (+), HbAg (-), antiHbc (+/-): patient is protected (previous vaccination or resolved infection).  ID specialist consult must be considered. Several options are available, including lamivudine, tenofovir (TDF, TAF), entecavir.Major concern: patients receiving RTX. 
Herpes Zoster VirusSuggested by some authors for patients exposed to CYC 236. No consensus available. Patients must be individualized.  ID specialist consult must be considered. Valacyclovir? 
Herpes Simplex VirusNo consensus available. Suppressive therapy may be considered if recurrent episodes (> = 3/year) occur. Patients must be individualized.  ID specialist consult must be considered. Acyclovir: suppressive therapy dose. 
Helminths and amoebasAntiparasitic prophylaxis should be considered, particularly ifbiological therapy or GC are planned. Amebiasis prophylaxis in endemic regions has been proposed;repeated every year if immunosuppression persists or potential exposure (e.g., still living in endemic area).  Helminthic superinfection: ivermectin (200ug/Kg/day [i.e., one drop per Kg]), twodoses 15 days apart; or albendazole 400 mg/day, for 3 daysAmebiasis (endemic regions): secnidazole 2 g, single dose 

antiHbc, Hepatitis B virus total core antibody; antiHbeAg, Hepatitis B virus E antigen antibody; antiHbs, Hepatitis B virus surface antibody; CYC, Cyclophosphamide; GC, Glucocorticoids; HbeAg, Hepatitis B virus E antigen; HbsAG, Hepatitis B virus surface antigen; ID, Infectious diseases; IGRA, Interferon Gamma Release Assay; LTBI, Latent TB Infection; RTX, Rituximab; TAF, Tenofovir Alafenamide Fumarate; TDF, Tenofovir Disoproxil Fumarate; TMP/SMX, Trimethoprim/Sulfamethoxazole; TNF, Tumor Necrosis Factor; TST, Tuberculin Skin Test.

This table is based on references and author’s experience.

a

Based on expert recommendations, however, current data are contrasting. Lower TMP/SMX doses have been proposed.

b

Regimen proposed for ANCA-associated vasculitis.

Table 3.

Potential prophylaxis to be considered in patients with SSC.

CYCGlucocorticoids (GC)RTXOthersf
<15 mg  15−30 mg  >30 mg 
TB  Every patient should be screened. Treatment for LTBI positive patients must be administered at least one month prior to immunosuppression. ID specialist consultation advisable.
PCPa  Yb  Y?c  N? 
HBV  Prophylaxis must be considered in patients with HbsAg (+/-) and antiHbc (+). ID specialist consultation must be performed.
HZV 
HSV  Consider if recurrent (> = 3 per year). ID specialist consultation must be performed.
Parasitesd  Consider particularly when GCs or biologics are planned.
Otherse  No evidence. Patients must be individualized. ID specialist consultation must be performed.

antiHbc, HBV total core antibody; CYC, Cyclophosphamide; GC, Glucocorticoids; HbsAg, HBV surface antigen; HBV, Hepatitis B virus; HSV, Herpes Simplex Virus; HZV, Herpes Zoster Virus; ID, Infectious diseases; LTBI, Latent TB Infection; PCP, Pneumocystis jirovecii pneumonia; RTX, Rituximab; TB, Tuberculosis.

This table is based on references and author’s experience.

a

Based on expert recommendations, however current data are contrasting.

b

Additional risk factors: baseline lymphopenia, low CD4 count, CYC use, TNF α inhibitors or RTX use, initial GC use >60 mg.

c

Prophylaxis has been suggested for patients on RTX based on an apparent increased risk.

d

Refers to helminths and amoebas.

e

Including: Candida spp., Cryptococcus spp., Toxoplasma, Endemic mycoses (e.g., Histoplasmosis), Leishmania spp., etc.

f

Including: Methotrexate, Mycophenolate Mophetil, Azathioprine, Abatacept, Tocilizumab, Tumor Necrosis Factor α inhibitor, Belimumab.

Antibiotic prophylaxis: when to consider

As shown, the most frequently used immunosuppressants in SSc may increase the risk of infectious complications.5 Experience from other autoimmune diseases has suggested the protective effect of chemoprophylaxis whenever immunosuppression is necessary.67

A particular interest and evidence for P. jirovecii pneumonia are available, in which contrasting incidences are identified depending on the underlying disease, from as low as 0.1% in RA to as high as 12% in granulomatosis with polyangiitis.225 One shall consider that P. jirovecii pneumonia has a low rate among patients with rheumatic diseases, although its course tends to be severe and potentially fatal.226 For instance, recent guidelines for the management of ANCA-associated vasculitis “encourage the use of prophylaxis against P. jirovecii in all patients treated with CYC, where not contraindicated”.227,228 Mounting evidence supports the utility of prophylaxis in patients on glucocorticoids, particularly in those with high doses (≥ 30 mg/day), since a dose-dependent risk is observed.67,229 In addition, the use of RTX also appears to be associated with a higher risk of P. jirovecii pneumonia and a protective effect of prophylaxis has been reported.230 Winthrop & Baddley recently proposed a tailored prophylaxis schema for different autoimmune rheumatic diseases on glucocorticoids, in which SSc patients with ≥ 30 mg/day dose would benefit and those with doses below 15 mg/day would not; those patients who receive between 15 mg and 30 mg/day should be assessed on an individual risk basis, based on the presence of at least one of these factors: baseline lymphopenia, low CD4 count, CYC use, anti-TNF or RTX use, initial glucocorticoids dose ≥ 60 mg/day (Table 3). Nonetheless, these recommendations are based on expert opinion and the authors advocate for further studies67; some other authors believe that “more personalized risk assessments are needed to inform P. jirovecii pneumonia prophylaxis”, as data are contrasting.226 There are no studies in SSc showing these outcomes, probably due to the difficulty in gathering large cohorts of patients with this disease. However, there are reports of SSc patients on these therapies who develop opportunistic infections by Mycobacterium avium intracellulare and P. jirovecii.231

As the optimal TMP/SMX prophylactic dose in rheumatic diseases is unclear, an open RCT in Japan explored this subject. Patients were mainly exposed to glucocorticoids (>0.6 mg/Kg/day), and received a daily low dose TMP/SMX (80 mg/400 mg, or 40 mg/200 mg, or escalating dose from 8 mg/40 mg until 40 mg/200 mg was reached); no events of P. jirovecii pneumonia were observed in any group. The authors suggested a better tolerability and safety of 40 mg/200 mg doses.232 Recommended regimens for ANCA-associated vasculitis prophylaxis on CYC are: TMP/SMX 160 mg/800 mg on alternate days or 80 mg/400 mg every day. In case of adverse reaction or contraindication, pentamidine, atovaquone or dapsone are alternatives,227,228 however, only the latter is available in Colombia. In this scenario, an infectious diseases specialist consultation is advisable. As quality evidence is unavailable regarding doses, one might extrapolate recommended doses for human immunodeficiency virus (HIV), namely dapsone 100 mg/day or, in situations in which the recommended agents cannot be administered or are not tolerated, clindamycin plus primaquine.233 British guidelines on ANCA-associated vasculitis suggest the use of similar prophylaxis regimen in any patient with immunosuppression, including RTX.234 Some authors have suggested that one shall not rely solely on CD4-cell count for the guidance of P. jirovecii prophylaxis in autoimmune/inflammatory diseases.235

The incidence of HZV in patients under immunosuppression is an increasing challenge, however, the utility of prophylaxis among patients with autoimmune rheumatic diseases is unclear and safety data for vaccination are lacking. A recent study in patients receiving CYC for SLE or vasculitis explored the effect of anti-viral prophylaxis with valacyclovir. The authors reported an increased risk of HZV in patients with lymphopenia <500/μL at CYC initiation (HR 4.36, CI 0.51-37-31) and suggested that valacyclovir should be considered in these scenarios; none of the 19 patients receiving valacyclovir presented HZV. Interestingly, a higher risk of HZV has been found in SLE, which may suggest a different risk based on the underlying disease.236 Data are lacking for SSc.

As aforementioned, SSc patients are at an increased risk of TB (and probably non-TB-mycobacteria) infection due to the disease itself (particularly in the presence of pulmonary involvement and PAH) and to pharmacological immunosuppression.57,58 Glucocorticoids and TNF-α inhibitors are well-known risk factors for TB.237,238 Therefore, screening for TB infection using either a tuberculin skin test or an interferon gamma release assay is the basis of active disease prevention, and should be implemented in patients who will undergo immunosuppression.215,224 Whenever latent TB infection is detected, it must be treated according to guidelines.224,239 Expert recommendations for RA suggest to screen for pulmonary non-TB-mycobacteria prior to biologic therapy in patients with lung structural abnormalities (particularly bronchiectasis) or those with unexplained chronic cough. Although there is a higher risk of non-TB-mycobacteria in patients with ILD, such as SSc,62 data to support the need of prophylaxis are not available.238

Vaccines

Some preventable infectious diseases, such as influenza and pneumococcal pneumonia, may manifest more severely in patients with SSc. For instance, patients with ILD who become infected by influenza are at greater risk of respiratory failure and morbidity. Therefore, vaccination becomes an important strategy of care.240 However, safety and efficacy of vaccination in patients on immunosuppressive therapy and patients with autoimmunity has always concerned clinicians, as some studies have shown immunosuppressive therapy may hamper immunologic response to vaccination.241,242 These believes may explain a low vaccination rate against influenza in these patients, as described in a French cohort of patients with systemic autoimmune diseases on immunosuppressive therapy.243

Influenza

Recent research on influenza vaccination in patients with SSc has proven that different presentations of the influenza vaccine are not only safe in this population, but effective. A study by Sampaio-Barros et al. evaluating the outcomes of vaccination with H1N1 vaccine, found that there were no statistically significant differences in seroprotection (83.7 vs. 76.1%; P = 0.20) or seroconversion (76.1 vs.72.8%; P = 0.61) rates between SSc patients and healthy controls, despite receiving different immunosuppressive medications. A similar incidence of systemic side effects after vaccination was also observed (25 vs. 31.5%; P = 0.33).244 Another study by Setti et al., assessed the effectiveness of influenza AH3N2/AH1N1/B vaccine in individuals with SSc compared to a healthy control group. Although a higher effectiveness was observed in the control group, a good humoral response was achieved by SSc patients with no higher incidence of adverse effects or alterations in the course of the disease, proving its safety and efficacy.240

Pneumococcal pneumonia

Public health authorities recommend vaccination for Streptococcus pneumoniae (PCV13 or PPVP23) in immunocompromised adults245 and different studies have described a fair immune response to pneumococcal vaccine in patients with SSc. A study by Hesselstrand et al., comparing the immune response of patients with SSc with or without DMARDs (disease-modifying antirheumatic drugs) against a control group, receiving the PPVP23 or the PCV13 pneumococcal vaccine, found that the production of antibodies against pneumococcal serotypes in SSc patients without DMARDs was similar to that of the healthy control group. No differences were described between the PPVP23 and the PCV13 vaccine. However, patients on DMARDS had a lower antibody production.246 Other study by Mercado et al. described the immunogenicity of different pneumococcal antigens after vaccination with the PPVP23 in a group of 18 patients with SSC. They found that after vaccination, all patients had a two-fold rise in antibodies against the different antigens despite being treated with CYC. No differences were observed between limited SSc and diffuse SSc.247

Additional recommendations for immunization in patients with autoimmune inflammatory rheumatic diseases from the CDC,222 the Infectious Diseases Society of Colombia (ACIN),222 the Infectious Diseases Society of America (IDSA),222 the Colombian Association of Rheumatology (ASOREUMA)216 and the European League Against Rheumatism (EULAR),221 are summarized in Table 4. Novel interventions adapted to the South American context should be assessed for future recommendations on vaccination in patients with autoimmune diseases, such as vaccination for dengue virus, meningococcal disease and vaccination with the recently developed inactivated vaccine for HZV (Shingrix), which has shown to be more effective in preventing HZV and postherpetic neuralgia.248

Table 4.

Major recommendations for vaccination in patients with autoimmune inflammatory rheumatic diseases and patients with altered immunocompetence.

Recommendation  EULAR (2011)(221)  IDSA (2013)(222)  ACIN (2016)(223)  ASOREUMA (2016)(216)  Pérez-Díaz et al. (2016)(215)  CDC (2018)(222) 
  • Assess which vaccines were previously administered to the patient

 
  • Live vaccines must be administered 4wk before immunosuppression

 
     
  • All inactivated vaccines, according to age, should be administered >2 wk before immunosuppression

 
 
  • Vaccines should be administered if the inflammatory and autoimmune disease is stable

 
       
  • Live and live attenuated vaccines should not be administered to patients with high-grade immunosuppression

 
  • Vaccination should be administered before starting rituximab or after 6 months of ceasing therapy

 
         
  • Inactivated vaccines can be administered in patients on DMARDS and TNF-α blockers

 
     
  • Live vaccines can be administered one month after stopping high dose corticosteroids

 
           
  • Low dose corticosteroids do not contraindicate live vaccines

 
           
  • Live vaccines should be withheld for 3 months after Anti-TNFα or B-cell antibodies are stopped

 
           
  • Those who live in a household with immunocompromised patients should be vaccinated following the recommendations of the annually updated schedule, except for oral polio

 
     
  • Varicella vaccine should be administered to patients without evidence of varicella immunity with low-level immunosuppression or ≥4 wk prior to immunosuppression

 
     
  • Administer Zoster vaccine to patients aged ≥60 years prior to immunosuppressive therapy or with mild immunosuppressive therapy

 
   
  • Administer Zoster vaccine to patients ≥50 years with immunity for varicella and who will be on high grade immunosuppression.

 
     
  • Annual inactivated influenza vaccination should be administered

 
  • PPSV23 and PCV13 polysaccharide pneumococcal vaccination should be administered

 
  • Tetanus toxoid vaccination should be administered as in the general population 6mo after rituximab

 
 
  • Hepatitis A and B vaccinations are recommended in patients at risk of contracting those diseases.

 
   
  • Hepatitis B vaccine is strongly recommended

 
   
  • Vaccinate against HPV as recommended for general populations specially patients with SLE

 
   
  • Do not vaccinate with tuberculosis vaccine BCG

 
   
  • Oral polio vaccine is not recommended

 
       
  • Avoid yellow fever vaccine

 
 
High level immunosuppression:
  • Prednisone at doses ≥ 20 mg/day or equivalent for ≥ 14 days

  • Certain biologic immune modulators: TNF-α blockers or rituximab

  • After HSCT, high level immunosuppression is highly variable and depends on:

    • Type of transplant

    • Type of donor

    • Stem cell source

    • Complications (GVHD)

 
Low level immunosuppression:
  • Methotrexate at doses ≤0.4 mg/kg/week

  • Azathioprine ≤3 mg/kg/day

  • 6 mercaptopurine ≤1.5 mg/kg/day

  • Prednisone <20 mg/day or equivalent for less than 14 days

ACIN, Asociación Colombiana de Infectología (Colombian Association of Infectious Diseases); ASOREUMA, Asociación Colombiana de Reumatología (Colombian Association of Rheumatology); BCG, Bacillus Calmette-Guerin; CDC, Centers for Disease Control; DMARDS, Disease-modifying anti-rheumatic drugs; EULAR, European League Against Rheumatism; GVHD, Graft-vs-Host disease; Hib, Haemophilus influenza b; IDSA, Infectious Diseases Society of America; HSCT, Hematopoietic Stem Cell Transplantation; PCV, Pneumococcal conjugate vaccine; PPSV, Pneumococcal polysaccharide vaccine; SLE, Systemic lupus erythematosus; TNF-α, Tumor necrosis factor α.

Pre-treatment considerations

A baseline screening for different infectious diseases should be considered, as strong immunosuppression in patients with SSc is expected. Interpretation of screening tests has been discussed elegantly elsewhere.102,215,224 This screening should be based on local epidemiology, which should always include: TB, HIV, Hepatitis A Virus, HBV, Hepatitis C Virus, Toxoplasma, EBV, CMV, Varicella Zoster Virus, syphilis and a stool test. Some other entities to be considered are: Chagas (Trypanosoma cruzi) and Human T Lymphocyte Virus 1–2.215 Screening for histoplasmosis it not routinely recommended and should only be considered in endemic areas.215 Antiparasitic prophylaxis should be considered, particularly if biological therapy or glucocorticoids are planned. To prevent helminthic superinfection, the use of ivermectin (200ug/Kg/day [i.e., one drop per Kg]) in two doses 15 days apart, or albendazole 400 mg/day for 3 days is recommended.223 Amebiasis prophylaxis in endemic regions has been proposed using secnidazole 2 grams in a single dose, and repeating it every year if immunosuppression persists or if the patient is still living in an endemic area.215

Conclusion

Infections are a growing concern in the management of SSc, as evidence suggests them as a rising cause of admission and mortality. Most of the studies on this issue come from the USA and Europe, with few studies from Asia, which may not represent Latin-American populations, thus, local studies are warranted. Patients with SSc present a higher risk of infectious complications due to the structural damage of the disease and immunosuppressive treatment. Novel therapeutic agents have shown promising results; however, an increasing incidence of infections may be expected as has been observed in other rheumatic diseases. Data on the effects of antimicrobial prophylaxis and risk management strategies in SSc are lacking, although experience from other autoimmune diseases may shed light on this issue. An increasing awareness among treating specialties, particularly rheumatologists is warranted. The wide adoption of risk management strategies, including underused measures such as vaccination, is necessary.

Funding

The present work did not receive any funding.

Conflicts of interest

The authors declare there are no conflicts of interest relevant for the present manuscript.

Acknowledgements

We thank Dr. Diana Romero-Alvernia for her valuable comments on the manuscript.

References
[1]
F. Rongioletti, C. Ferreli, L. Atzori, U. Bottoni, G. Soda.
Scleroderma with an update about clinico-pathological correlation.
G Ital Dermatol Venereol., 153 (2018), pp. 208-215
[2]
L.P. Prada, P.X. Coral.
Esclerosis sistémica.
Reumatología Fundamentos de Medicina, eight edit, pp. 493-508
[3]
C.P. Denton, D. Khanna.
Systemic sclerosis.
Lancet, 390 (2017), pp. 1685-1699
[4]
C.C. Mok, C.L. Kwok, L.Y. Ho, P.T. Chan, S.F. Yip.
Life expectancy, standardized mortality ratios, and causes of death in six rheumatic diseases in Hong Kong, China.
Arthritis Rheum., 63 (2011), pp. 1182-1189
[5]
G. Bussone, A. Berezné, L. Mouthon, A. Berezne, L. Mouthon.
[Infectious complications of systemic sclerosis].
Presse Med, 38 (2009), pp. 291-302
[6]
Y. Allanore, C.P. Denton, T. Krieg, P. Cornelisse, D. Rosenberg, B. Schwierin, et al.
Clinical characteristics and predictors of gangrene in patients with systemic sclerosis and digital ulcers in the Digital Ulcer Outcome Registry: a prospective, observational cohort.
Ann Rheum Dis, 75 (2016), pp. 1736-1740
[7]
D. Giuggioli, A. Manfredi, M. Colaci, F. Lumetti, C. Ferri.
Osteomyelitis complicating scleroderma digital ulcers.
Clin Rheumatol, 32 (2013), pp. 623-627
[8]
F. Rahal, R. Chitouane, S. Slimani, S. Lefkir, N. Brahimi, A. Ladjouze-Rezig.
Infections during rheumatic diseases: A serie of 146 patients.
Ann Rheum Dis, 71 (2013),
[9]
A. Fernández-Codina, K.M. Walker, J.E. Pope.
Scleroderma algorithm group. treatment algorithms for systemic sclerosis according to experts.
Arthritis Rheumatol, 70 (2018), pp. 1820-1828
[10]
O. Kowal-Bielecka, J. Fransen, J. Avouac, M. Becker, A. Kulak, Y. Allanore, et al.
Update of EULAR recommendations for the treatment of systemic sclerosis.
Ann Rheum Dis., 76 (2017), pp. 1327-1339
[11]
V. Smith, C.A. Scirè, R. Talarico, P. Airo, T. Alexander, Y. Allanore, et al.
Systemic sclerosis: State of the art on clinical practice guidelines.
RMD Open., 4 (2018), pp. 1-9
[12]
R.E. Pellar, J.E. Pope.
Evidence-based management of systemic sclerosis: Navigating recommendations and guidelines.
Semin Arthritis Rheum., 46 (2017), pp. 767-774
[13]
C. Bruni, E. Praino, Y. Allanore, O. Distler, A. Gabrielli, F. Iannone, et al.
Use of biologics and other novel therapies for the treatment of systemic sclerosis.
Expert Rev Clin Immunol., 13 (2017), pp. 469-482
[14]
M. Elhai, C. Meune, J. Avouac, A. Kahan, Y. Allanore.
Trends in mortality in patients with systemic sclerosis over 40 years: A systematic review and meta-analysis of cohort studies.
Rheumatology., 51 (2012), pp. 1017-1026
[15]
A.J. Tyndall, B. Bannert, M. Vonk, P. Airò, F. Cozzi, P.E. Carreira, et al.
Causes and risk factors for death in systemic sclerosis: a study from the EULAR Scleroderma Trials and Research (EUSTAR) database.
Ann Rheum Dis., 69 (2010), pp. 1809-1815
[16]
S. Hu, Y. Hou, Q. Wang, M. Li, D. Xu, X. Zeng.
Prognostic profile of systemic sclerosis: analysis of the clinical EUSTAR cohort in China.
Arthritis Res Ther., 20 (2018), pp. 235
[17]
S. Netwijitpan, C. Foocharoen, A. Mahakkanukrauh, S. Suwannaroj, R. Nanagara, S. N, et al.
Indications for hospitalization and in-hospital mortality in Thai systemic sclerosis.
Clin Rheumatol., 32 (2013), pp. 361-367
[18]
M. Piga, L. Casula, S. Sanna, D. Perra, A. Floris, A. Antonelli, et al.
Population-based analysis of hospitalizations for patients with systemic sclerosis in a West-European region over the period 2001–2012.
Rheumatol Int., 36 (2016), pp. 73-81
[19]
A. Komócsi, A. Vorobcsuk, R. Faludi, T. Pintér, Z. Lenkey, G. Költo, et al.
The impact of cardiopulmonary manifestations on the mortality of SSC: A systematic review and meta-analysis of observational studies.
Rheumatology., 51 (2012), pp. 1027-1036
[20]
P.D. Sampaio-Barros, A.B. Bortoluzzo, R.G. Marangoni, L.F. Rocha, A.P.T. Del Rio, A.M. Samara, et al.
Survival, causes of death, and prognostic factors in systemic sclerosis: analysis of 947 Brazilian patients.
J Rheumatol., 39 (2012), pp. 1971-1978
[21]
S. Shenavandeh, R. Naseri.
Assessment of hospitalization and mortality of scleroderma in-patients: a thirteen-year study.
Reumatologia., 55 (2017), pp. 163-168
[22]
V.D. Steen, T.A. Medsger.
Changes in causes of death in systemic sclerosis, 1972–2002.
Ann Rheum Dis., 66 (2007), pp. 940-944
[23]
R.P.V. de Rezende, R.A. Gismondi, H.C. Maleh, E.M. de Miranda Coelho, C.S. Vieira, M.L.G. Rosa, et al.
Distinct mortality profile in systemic sclerosis: a death certificate study in Rio de Janeiro, Brazil (2006-2015) using a multiple causes of death analysis.
Clin Rheumatol., (2017), pp. 1-6
[24]
M. Elhai, C. Meune, M. Boubaya, J. Avouac, E. Hachulla, A. Balbir-Gurman, et al.
Mapping and predicting mortality from systemic sclerosis.
Ann Rheum Dis., 76 (2017), pp. 1897-1905
[25]
S.T. Sehra, A. Kelly, J.F. Baker, C.T. Derk.
Predictors of inpatient mortality in patients with systemic sclerosis: a case control study.
Clin Rheumatol., 35 (2016), pp. 1631-1635
[26]
L. Chung, E. Krishnan, E.F. Chakravarty.
Hospitalizations and mortality in systemic sclerosis: results from the Nationwide Inpatient Sample.
Rheumatology., 46 (2007), pp. 1808-1813
[27]
M. Rubio-Rivas, C. Royo, C.P. Simeón, X. Corbella, V. Fonollosa.
Mortality and survival in systemic sclerosis: systematic review and meta-analysis.
Semin Arthritis Rheum., 44 (2014), pp. 208-219
[28]
X. Li, Y.-Q. Qian, N. Liu, R. Mu, Y. Zuo, G.-C. Wang, et al.
Survival rate, causes of death, and risk factors in systemic sclerosis: a large cohort study.
Clin Rheumatol., 37 (2018), pp. 3051-3056
[29]
D. Ram Poudel, M. George, R. Dhital, P. Karmacharya, N. Sandorfi, C.T. Derk.
Mortality, length of stay and cost of hospitalization among patients with systemic sclerosis: results from the National Inpatient Sample.
Rheumatology., 57 (2018), pp. 1611-1622
[30]
C. Foocharoen, Y. Siriphannon, A. Mahakkanukrauh, S. Suwannaroj, R. Nanagara, C. F, et al.
Incidence rate and causes of infection in Thai systemic sclerosis patients.
Int J Rheum Dis., 15 (2012), pp. 277-283
[31]
D. Khanna, C.P. Denton, A. Jahreis, J.M. van Laar, T.M. Frech, M.E. Anderson, et al.
Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial.
Lancet., 387 (2016), pp. 2630-2640
[32]
D. Khanna, C. Lin, M. Kuwana, Y. Allanore, A. Batalov, I. Butrimiene, et al.
Efficacy and safety of tocilizumab for the treatment of systemic sclerosis: results from a phase 3 randomized controlled trial [abstract].
Arthritis Rheumatol., 70 (2018), pp. 898
[33]
D. Khanna, C. Spino, E. Bush, S. Johnson, L. Chung, J. Molitor, et al.
Abatacept Vs. placebo in early diffuse cutaneous systemic sclerosis—results of a phase 2 investigator initiated, double-blind, placebo-controlled, multicenter, randomized controlled trial study [abstract].
Arthritis Rheumatol, 70 (2018), pp. 900
[34]
L. Mouthon, P. Carpentier, A. Khau Van Kien, P. Clerson, H. Maillard, E. Hachulla, et al.
Impact of recurrent ischemic digital ulcers on hand disability in systemic sclerosis.
Ann Rheum Dis., 71 (2013),
[35]
S.I. Nihtyanova, G.M. Brough, C.M. Black, C.P. Denton.
Clinical burden of digital vasculopathy in limited and diffuse cutaneous systemic sclerosis.
Ann Rheum Dis., 67 (2008), pp. 120-123
[36]
M. Vonk, A. Voskuyl, M. Walravens, P. Paassen, A. Schuerwegh.
Burden of disease in dutch patients with systemic sclerosis and digital ulcers; data from thedata from the dutch national registry.
Arthritis Rheum., 63 (2011),
[37]
J. Bernardo Cofino, L. Trapiella Martinez.
Superinfected calcinosis cutis as a presentation of a limited form systemic sclerosis.
Reumatol Clin., 10 (2014), pp. 187-188
[38]
N. Tangri, B.M. Young.
Soft-tissue infection and underlying calcinosis of CREST syndrome.
CMAJ., 175 (2006), pp. 1059
[39]
R.L.J. Minor, S. Baum, K.S. Schulze-Delrieu.
Pyomyositis in a patient with progressive systemic sclerosis. Case report and review of the literature.
Arch Intern Med, 148 (1988), pp. 1453-1455
[40]
M.Y. Mok, S.Y. Wong, T.M. Chan, W.M. Tang, W.S. Wong, C.S. Lau.
Necrotizing fasciitis in rheumatic diseases.
Lupus., 15 (2006), pp. 380-383
[41]
F. Galluccio, G. Fiori, L. Amanzi, F. Braschi, I. Miniati, S. Guiducci, et al.
Digital ulcers (DU) in systemic sclerosis (SSc): staging, characteristics and sub-setting through observation of 1614 digital lesions.
Clin Exp Rheumatol., 28 (2010), pp. S180
[42]
I. Chiapparoli, F. Locatelli, P. Orsolini, R. Caporali, C. Montecucco, V. Codullo.
Osteomyelitis complicating digital ulcers in systemic sclerosis.
Ann Rheum Dis., 77 (2018), pp. 1519
[43]
Y. Braun-Moscovici, Z. Keidar, M. Braun, D. Markovits, K. Toledano, Y. Tavor, et al.
The prevalence of osteomyelitis in infected digital ulcers in systemic sclerosis patients.
Ann Rheum Dis., 76 (2017), pp. 910
[44]
L. Mouthon, P. Carpentier, A. Khau Van Kien, P. Clerson, H. Maillard, E. Hachulla, et al.
Evaluation of the impact of recurrent ischaemic digital ulcers on hand disability in patients with SSc (eclipse)-report of the cohort at the time of inclusion.
Rheumatology., 51 (2012), pp. ii21
[45]
D. Giuggioli, A. Manfredi, M. Colaci, F. Lumetti, M. Sebastiani, C. Ferri.
Scleroderma digital ulcers complicated by infection with fecal pathogens.
Arthritis Rheum., 63 (2011),
[46]
R. Cimino, A. Rotundo, C. Pintaudi, S. Giancotti, S. Mazzuca.
Current approach to digital ulcers in systemic sclerosis patients.
Ital J Med., 9 (2015), pp. 24
[47]
M. Tägil, J. Dieterich, P. Kopylov.
Wound healing after hand surgery in patients with systemic sclerosis--a retrospective analysis of 41 operations in 19 patients.
J Hand Surg Eur Vol., 32 (2007), pp. 316-319
[48]
A.H. Low, S.C. Ng, E. Tay, J. Thumboo.
Disease manifestations and pulmonary complications in Asian patients with systemic sclerosis.
Int J Rheum Dis., 13 (2010), pp. 189
[49]
W. Stevens, V. Thakkar, O. Moore, J. Byron, S. Proudman, J. Zochling, et al.
Lung disease is the major cause of death in the Australian SSc cohort study.
[50]
M. Tanaka, R. Koike, R. Sakai, K. Saito, S. Hirata, H. Nagasawa, et al.
Pulmonary infections following immunosuppressive treatments during hospitalization worsen the short-term vital prognosis for patients with connective tissue disease-associated interstitial pneumonia.
Mod Rheumatol., 25 (2015), pp. 609-614
[51]
M. Di Franco, B. Lucchino, M. Spaziante, C. Iannuccelli, G. Valesini, G. Iaiani.
Lung infections in systemic rheumatic disease: focus on opportunistic infections.
Int J Mol Sci., 18 (2017), pp. 293
[52]
H.W. Farber, R.W. Simms, R. Lafyatis.
Care of patients with scleroderma in the intensive care setting.
J Intensive Care Med., 25 (2010), pp. 247-258
[53]
Y. Shi, X. Rui.
Procalcitonin kinetics: a reliable tool for diagnosis and monitoring of the course of bacterial infection in critically ill patients with autoimmune diseases.
Intensive Care Med., 39 (2013), pp. 2233-2234
[54]
O. Sleglova, H. Dejmkova, J. Uhrova.
Usefulness of procalcitonin measurement in differentiating between activity of systemic autoimmune disease and bacterial infection.
Arthritis Rheum., 60 (2009), pp. 132
[55]
I. Marie, S. Dominique, H. Levesque, P. Ducrotté, P. Denis, M.F. Hellot, et al.
Esophageal involvement and pulmonary manifestations in systemic sclerosis.
[56]
X.J. Zhang, A. Bonner, M. Hudson, Canadian Scleroderma Research Group, M. Baron, J. Pope.
Association of gastroesophageal factors and worsening of forced vital capacity in systemic sclerosis.
J Rheumatol., 40 (2013), pp. 850-858
[57]
S.-M. Ou, W.-C. Fan, K.-T. Chou, K.-T. Cho, C.-M. Yeh, Su VY-F, et al.
Systemic sclerosis and the risk of tuberculosis.
J Rheumatol., 41 (2014), pp. 1662-1669
[58]
M.-C. Lu, C.-L. Lai, C.-C. Tsai, M. Koo, N.-S. Lai.
Increased risk of pulmonary and extra-pulmonary tuberculosis in patients with rheumatic diseases.
Int J Tuberc Lung Dis., 19 (2015), pp. 1500-1506
[59]
Q.M. Ahmad, I.H. Shah, Q. Nauman, F. Sameem, J. Sultan.
Increased incidence of tuberculosis in patients of systemic sclerosis on dexamethasone pulse therapy: a short communication from Kashmir.
Indian J Dermatol., 53 (2008), pp. 24-25
[60]
M. Valence, O. Decaux, M. Revest, P. Jégo, P. Tattevin.
Association of tuberculosis and deep venous thromboembolism in patients with autoimmune diseases.
Eur J Intern Med., 26 (2015), pp. e23-4
[61]
L. Escobedo, L. Minerva, A. Carmen, Z. Fabián, V. Karene, I. Amaia, et al.
Central nervous system tuberculosis in patients with systemic rheumatic disease that were receiving immunosuppresive therapy.
[62]
R.F. Mizutani, E.M.S. Lombardi, U. De Paula Santos, M. Terra-Filho.
Silica exposure, silicosis, autoimmune diseases, tuberculosis and nontuberculous pulmonary mycobacterial disease.
Eur Respir J., 48 (2016),
[63]
A. Shimizu, H. Oka, T. Matsuda, S. Ozaki.
(1→3)-Beta-D glucan is a diagnostic and negative prognostic marker for Pneumocystis carinii pneumonia in patients with connective tissue disease.
Clin Exp Rheumatol., 23 (2005), pp. 678-680
[64]
J. Ogawa, M. Harigai, K. Nagasaka, T. Nakamura, N. Miyasaka.
Prediction of and prophylaxis against Pneumocystis pneumonia in patients with connective tissue diseases undergoing medium- or high-dose corticosteroid therapy.
Mod Rheumatol., 15 (2005), pp. 91-96
[65]
C.A. Mecoli, D. Saylor, A.C. Gelber, L. Christopher-Stine.
Pneumocystis jiroveci pneumonia in rheumatic disease: a 20-year single-centre experience.
Clin Exp Rheumatol., 35 (2017), pp. 671-673
[66]
H.-C. Tsai, M.-J. Tsai, F.-C. Lin, S.-C. Chang.
Opportunistic pulmonary infection in patients with connective tissue disease: a single-center experience.
Chest., 140 (2011), pp. 756A
[67]
K.L. Winthrop, J.W. Baddley.
Pneumocystis and glucocorticoid use: to prophylax or not to prophylax (and when?); that is the question.
Ann Rheum Dis., 77 (2018), pp. 631-633
[68]
S. Nandi, A. Santra, L. Ghoshal, S. Kundu.
Interstitial lung disease in systemic scleroderma, complicated with bilateral pulmonary aspergilloma: an unusual association.
J Clin Diagnostic Res., 9 (2015),
[69]
I. Marie, L. Lahaxe, H. Levesque, P. Heliot.
Pulmonary actinomycosis in a patient with diffuse systemic sclerosis treated with infliximab.
QJM, 101 (2008), pp. 419-421
[70]
N. Pittman, S.M. Rawn, M. Wang, A. Masetto, K.A. Beattie, M. Larché.
Treatment of small intestinal bacterial overgrowth in systemic sclerosis: a systematic review.
Rheumatology., 57 (2018), pp. 1802-1811
[71]
A. Emmanuel.
Current management of the gastrointestinal complications of systemic sclerosis.
Nat Rev Gastroenterol Hepatol., 13 (2016), pp. 461-472
[72]
A. Rodríguez-Varón, O.M. Muñoz, J. Pulido-Arenas, S.B. Amado, M. Tobón-Trujillo.
Antibiotic-associated diarrhea: clinical characteristics and the presence of clostridium difficile.
Rev Gastroenterol México., 82 (2017), pp. 129-133
[73]
S.M. Gualtero, L.A. Abril, N. Camelo, S.D. Sanchez, F.A. Davila, G. Arias, et al.
[Characteristics of clostridium difficile infection in a high complexity hospital and report of the circulation of the NAP1/027 hypervirulent strain in Colombia].
Biomedica., 37 (2017), pp. 466-472
[74]
L. Alcalá Hernández, E. Reigadas Ramírez, E. Bouza Santiago.
Clostridium difficile infection.
Med Clin (Barc)., 148 (2017), pp. 456-463
[75]
L. Hendel, E. Svejgaard, I. Walsøe, M. Kieffer, A. Stenderup.
Esophageal candidosis in progressive systemic sclerosis: occurrence, significance, and treatment with fluconazole.
Scand J Gastroenterol., 23 (1988), pp. 1182-1186
[76]
A.J. Geirsson, A. Akesson, T. Gustafson, A. Elner, F.A. Wollheim.
Cineradiography identifies esophageal candidiasis in progressive systemic sclerosis.
Clin Exp Rheumatol., 7 (1989), pp. 43-46
[77]
W.C. Yong, S. Upala, A. Sanguankeo.
Helicobacter pylori infection in systemic sclerosis: a systematic review and meta-analysis of observational studies.
Clin Exp Rheumatol., 36 (2018), pp. 168-174
[78]
M. Radic, D. Bonacin, J. Morovic Vergles, J. Radic, V. Kovacic, E. Siegert, et al.
Eradication of Helicobacter pylori reduces disease severity and activity in systemic sclerosis patients.
Eur J Clin Invest., 44 (2014), pp. 64-65
[79]
C. Balaji, M. Bhuvanesh, C. Saranya, R. Ramesh, M. Saravanan, S. Rajeswari.
Helicobacter Pylori infection in systemic sclerosis and its association with upper gastrointestinal dysfunction.
Indian J Rheumatol., 12 (2017), pp. 204-208
[80]
A. Laserna, J.E. Barahona-Correa, L.-H. Alba.
Treatment of the Helicobacter pylori infection: a critical appraisal of the literature.
Univ Médica., 59 (2018), pp. 1-13
[81]
H. Endo, Y. Kitsunoki, T. Yamamoto, M. Kaburagi, S. Muraoka, H. Abe, et al.
Long-term home parenteral nutrition therapy in eleven patients with severe scleroderma bowel disease.
Int J Rheum Dis., 13 (2010), pp. 183
[82]
E. Harrison, A.L. Herrick, M. Dibb, J.T. McLaughlin, S. Lal, E. H, et al.
Long-term outcome of patients with systemic sclerosis requiring home parenteral nutrition.
Clin Nutr., 34 (2015), pp. 991-996
[83]
K.L. Winthrop, S.A. Novosad, J.W. Baddley, L. Calabrese, T. Chiller, P. Polgreen, et al.
Opportunistic infections and biologic therapies in immune-mediated inflammatory diseases: consensus recommendations for infection reporting during clinical trials and postmarketing surveillance.
Ann Rheum Dis., 74 (2015), pp. 2107-2116
[84]
S. Ito, T. Honma, M. Ueno, M. Kikuchi, T. Sato, M. Nakano, et al.
An autopsy case of progressive systemic sclerosis with disseminated Candida albicans embolism.
Japanese J Rheumatol., 5 (1994), pp. 363-369
[85]
K. Sripavatakul, C. Foocharoen.
Spontaneous pneumothorax from cryptococcal pneumonia in systemic sclerosis: a case report.
J Med Case Rep., 5 (2011), pp. 309
[86]
E. Ishikawa, K. Yoneda, K. Nakai, J. Moriue, I. Yokoi, A. Munehiro, et al.
Primary cutaneous cryptococcosis in a patient with CREST syndrome.
Eur J Dermatol., 22 (2012), pp. 421-422
[87]
C.R. Bamford.
Toxoplasmosis mimicking a brain abscess in an adult with treated scleroderma.
Neurology., 25 (1975), pp. 343-345
[88]
J.-C. Gerster, B. Duvoisin, J. Dudler, I.C. Berner.
Tenosynovitis of the hands caused by Mycobacterium kansasii in a patient with scleroderma.
J Rheumatol., 31 (2004), pp. 2523-2525
[89]
A.R. Cantwell Jr, J.W. Wilson.
Scleroderma with ulceration secondary to atypical mycobacteria.
Arch Dermatol., 94 (1966), pp. 663-664
[90]
V. Napier, P.A. Sarojini, J.T. Ramani Bai.
Atypical mycobacterial infection in a case of systemic sclerosis.
Indian J Dermatol Venereol Leprol., 59 (1993), pp. 33-34
[91]
C. Auzary, L. Mouthon, M. Soilleux, P. Cohen, P. Boiron, L. Guillevin, et al.
Localized subcutaneous Nocardia farcinica abscess in a woman with overlap syndrome between systemic scleroderma and polymyositis.
Ann Med Interne (Paris)., 150 (1999), pp. 582-584
[92]
S. Mehta, B. Cowan, A. Flynn.
Pulmonary nocardia farcinica infection in a patient with scleroderma-associated interstitial lung disease.
Am J Respir Crit Care Med, 191 (2015),
[93]
R. Amathieu, L. Tual, S. Rouaghe, J. Stirnemann, O. Fain, G. Dhonneur.
[Splenic rupture associated with CMV infection: case report and review].
Ann Fr Anesth Reanim., 26 (2007), pp. 674-676
[94]
M.A. Contreras, C. Salas, J.L. Andreu, J. Mulero.
Cytomegalovirus esophagitis as a treatable complication of systemic sclerosis.
J Clin Rheumatol., 7 (2001), pp. 384-387
[95]
T. Ozaki, H. Yamashita, S. Kaneko, H. Yorifuji, H. Takahashi, Y. Ueda, et al.
Cytomegalovirus disease of the upper gastrointestinal tract in patients with rheumatic diseases: a case series and literature review.
Clin Rheumatol., 32 (2013), pp. 1683-1690
[96]
D. Ciervo, E. Antonielli, S. Baroncelli, A. Mancini, G. Degl’Innocenti, A. Crociani, et al.
A rare case of cytomegalovirus pancreatitis in a woman with new onset systemic sclerosis.
Ital J Med., 9 (2015), pp. 23
[97]
F. Broccolo, F. Drago, G. Cassina, A. Fava, L. Fusetti, B. Matteoli, et al.
Selective reactivation of human herpesvirus 6 in patients with autoimmune connective tissue diseases.
J Med Virol., 85 (2013), pp. 1925-1934
[98]
D. Michailidou, J.V. Lettera, I. Forde, P.J. Cohen, A.J. Wolff, D.A. Kaufman.
Acute Necrotizing Herpetic Pleuritis in a Patient with Systemic Sclerosis and Immunosuppression: Report of a Novel Pulmonary Herpes Infection.
Am J Med., 129 (2016), pp. e25-27
[99]
A. Ghinoi, M.T. Mascia, D. Giuggioli, R. Magistro, G. Barbolini, C.M. Magro, et al.
Coexistence of non-specific and usual interstitial pneumonia in a patient with severe cystic scleroderma lung involvement and parvovirus B19 infection.
Clin Exp Rheumatol, 23 (2005), pp. 431-433
[100]
M. Yamaguchi, A. Chiba, T. Yanagawa, T. Mato, K. Hirai, T. Inoue, et al.
Guillain-Barré syndrome following herpes zoster in a patient with systemic sclerosis.
Mod Rheumatol., 11 (2001), pp. 251-254
[101]
S. Zingarelli, P. Airò, M. Frassi, C. Bazzani, M. Scarsi, M. Puoti.
Prophylaxis and therapy of HBV infection in 20 patients treated with disease modifying antirheumatic drugs or with biological agents for rheumatic diseases.
Reumatismo., 60 (2008), pp. 22-27
[102]
C. Koutsianas, K. Thomas, D. Vassilopoulos.
Hepatitis B Reactivation in Rheumatic Diseases: Screening and Prevention.
Rheum Dis Clin North Am., 43 (2017), pp. 133-149
[103]
M. Martin, C. Mougin, J.-L. Prétet, H. Gil, N. Meaux-Ruault, E. Puzenat, et al.
Screening of human papillomavirus infection in women with systemic sclerosis.
Clin Exp Rheumatol., 32 (2014),
[104]
C. Vacchi, M. Colaci, G. Cassone, C. Esposito, F. Lumetti, D. Giuggioli, et al.
Persistence of human papillomavirus in the cervix of women with systemic sclerosis.
Ann Rheum Dis., 76 (2017), pp. 1267
[105]
J.R. Wingard, J. Hsu, J.W. Hiemenz.
Hematopoietic stem cell transplantation: an overview of infection risks and epidemiology.
Infect Dis Clin North Am., 24 (2010), pp. 257-272
[106]
D. Bertrand, J. Dehay, J. Ott, R. Sberro, C. Brunelle, N. Kamar, et al.
Kidney transplantation in patients with systemic sclerosis: a nationwide multicentre study.
Transpl Int., 30 (2017), pp. 256-265
[107]
S.K. Gadre, O.A. Minai, X.-F. Wang, Q. Zhang, M. Budev, A.R. Tonelli.
Lung or heart-lung transplant in pulmonary arterial hypertension: what is the impact of systemic sclerosis?.
Exp Clin Transplant, 15 (2017), pp. 676-684
[108]
J.C. Lenaerts, J.L. Lenaerts, R. Westhovens, W. Droogne, A. Ciarka, J. Van Cleemput, et al.
Cardiac transplantation in systemic sclerosis: single-centre experience of three cases.
Rheumatology (Oxford)., 57 (2018), pp. 1120-1122
[109]
R. Jablonski, J. Dematte, S. Bhorade.
Lung transplantation in scleroderma: recent advances and lessons.
Curr Opin Rheumatol., 30 (2018), pp. 562-569
[110]
M. Green, E.A. Blumberg, L. Danziger-Isakov, S. Huprikar, C.N. Kotton, D. Kumar.
Foreword: 4th edition of the American Society of Transplantation Infectious Diseases Guidelines.
Clin Transplant., 33 (2019),
[111]
J.B. Flórez-Suárez.
Quintana-López G. Inmunosupresores.
Reumatología Fundamentos de Medicina, 8th ed., pp. 221-234
[112]
M. Iudici, G. Cuomo, S. Vettori, M. Bocchino, A. Sanduzzi Zamparelli, S. Cappabianca, et al.
Low-dose pulse cyclophosphamide in interstitial lung disease associated with systemic sclerosis (SSc-ILD): efficacy of maintenance immunosuppression in responders and non-responders.
Semin Arthritis Rheum., 44 (2015), pp. 437-444
[113]
H. Barnes, A.E. Holland, G.P. Westall, N.S. Goh, I.N. Glaspole.
Cyclophosphamide for connective tissue disease-associated interstitial lung disease.
Cochrane database Syst Rev., 1 (2018),
[114]
D.P. Tashkin, R. Elashoff, P.J. Clements, J. Goldin, M.D. Roth, D.E. Furst, et al.
Cyclophosphamide versus placebo in scleroderma lung disease.
N Engl J Med., 354 (2006), pp. 2655-2666
[115]
R.K. Hoyles, R.W. Ellis, J. Wellsbury, B. Lees, P. Newlands, N.S.L. Goh, et al.
A multicenter, prospective, randomized, double-blind, placebo-controlled trial of corticosteroids and intravenous cyclophosphamide followed by oral azathioprine for the treatment of pulmonary fibrosis in scleroderma.
Arthritis Rheum., 54 (2006), pp. 3962-3970
[116]
G. Zhang, T. Xu, H. Zhang, S. Ye, Q. Wang, L. Zhang, et al.
[Randomized control multi-center clinical study of mycophenolate mofetil and cyclophosphamide in the treatment of connective tissue disease related interstitial lung disease].
Zhonghua Yi Xue Za Zhi., 95 (2015), pp. 3641-3645
[117]
D.P. Tashkin, M.D. Roth, P.J. Clements, D.E. Furst, D. Khanna, E.C. Kleerup, et al.
Mycophenolate mofetil versus oral cyclophosphamide in scleroderma-related interstitial lung disease (SLS II): a randomised controlled, double-blind, parallel group trial.
Lancet Respir Med., 4 (2016), pp. 708-719
[118]
H. Poormoghim, M. Moradi Lakeh, M. Mohammadipour, F. Sodagari, N. Toofaninjed.
Cyclophosphamide for scleroderma lung disease: a systematic review and meta-analysis.
Rheumatol Int., 32 (2012), pp. 2431-2444
[119]
A. Belotti Masserini, P.L. Colombelli, S. Panza, G. Dognini, F. Cagnoni, C. Mastaglio, et al.
Bkvirus associated haemorrhagic cystitis in a scleroderma patient after cyclophosphamide therapy.
Clin Exp Rheumatol, 32 (2014),
[120]
H. Nakamura, T. Odani, S. Yasuda, A. Noguchi, Y. Fujieda, M. Kato, et al.
Autologous haematopoietic stem cell transplantation for Japanese patients with systemic sclerosis: long-term follow-up on a phase II trial and treatment-related fatal cardiomyopathy.
Mod Rheumatol., 28 (2018), pp. 879-884
[121]
P.J. Woytala, E. Morgiel, A. Luczak, K. Czesak-Woytala, P. Wiland, P.J. W, et al.
The safety of intravenous cyclophosphamide in the treatment of rheumatic diseases.
Adv Clin Exp Med., 25 (2016), pp. 479-484
[122]
G.S. Alarcón.
Relative risk and attributable risk needed to interpret “factors predictive of infection in cyclophosphamide-treated lupus patients”: comment on the article by Pryor et al.
[123]
B.D. Pryor, S.G. Bologna, L.E. Kahl.
Risk factors for serious infection during treatment with cyclophosphamide and high-dose corticosteroids for systemic lupus erythematosus.
Arthritis Rheum., 39 (1996), pp. 1475-1482
[124]
Y. Santamaría-Alza, J. Sánchez-Bautista, J.F. Fajardo-Rivero, C.L. Figueroa.
Invasive fungal infections in Colombian patients with systemic lupus erythematosus.
Lupus., 27 (2018), pp. 1116-1122
[125]
E.S. Molloy, C.M. Calabrese, L.H. Calabrese.
The risk of progressive multifocal leukoencephalopathy in the biologic era: prevention and management.
Rheum Dis Clin North Am., 43 (2017), pp. 95-109
[126]
E.F. Chakravarty.
Incidence and prevention of herpes zoster reactivation in patients with autoimmune diseases.
Rheum Dis Clin North Am., 43 (2017), pp. 111-121
[127]
N. Droz, L. Gilardin, P. Cacoub, F. Berenbaum, D. Wendling, B. Godeau, et al.
Kinetic profiles and management of hepatitis B virus reactivation in patients with immune-mediated inflammatory diseases.
Arthritis Care Res (Hoboken)., 65 (2013), pp. 1504-1514
[128]
T. Ueda, T. Sakagami, T. Kikuchi, T. Takada.
Mycophenolate mofetil as a therapeutic agent for interstitial lung diseases in systemic sclerosis.
Respir Investig., 56 (2018), pp. 14-20
[129]
M.A. Omair, A. Alahmadi, S.R. Johnson.
Safety and effectiveness of mycophenolate in systemic sclerosis. A systematic review.
[130]
D. Boulos, G.-S. Ngian, A. Rajadurai, K. Elford, W. Stevens, S. Proudman, et al.
Long-term efficacy and tolerability of mycophenolate mofetil therapy in diffuse scleroderma skin disease.
Int J Rheum Dis., 20 (2017), pp. 481-488
[131]
J.K. Gordon, V. Martyanov, J.M. Franks, E.J. Bernstein, J. Szymonifka, C. Magro, et al.
Belimumab for the treatment of early diffuse systemic sclerosis: results of a randomized, double-blind, placebo-controlled, pilot trial.
Arthritis Rheumatol., 70 (2018), pp. 308-316
[132]
G. Naidu, S. Sharma, V. Dhir, S. Dhooria, A. Sinha, A. Mb, et al.
A randomised controlled trial to compare the efficacy of oral mycophenolate mofetil with placebo in patients with systemic sclerosis related early interstitial lung disease.
Ann Rheum Dis., 77 (2018), pp. 751-752
[133]
S.Y. Patel, J. Carbone, S. Jolles.
The expanding field of secondary antibody deficiency: causes, diagnosis, and management.
Front Immunol., 10 (2019), pp. 33
[134]
D.J. Tunnicliffe, S.C. Palmer, L. Henderson, P. Masson, J.C. Craig, A. Tong, et al.
Immunosuppressive treatment for proliferative lupus nephritis.
Cochrane database Syst Rev., 6 (2018),
[135]
M. Toruner, E.V. Loftus, W.S. Harmsen, A.R. Zinsmeister, R. Orenstein, W.J. Sandborn, et al.
Risk factors for opportunistic infections in patients with inflammatory bowel disease.
Gastroenterology., 134 (2008), pp. 929-936
[136]
D.H. Present, S.J. Meltzer, M.P. Krumholz, A. Wolke, B.I. Korelitz.
6-Mercaptopurine in the management of inflammatory bowel disease: short- and long-term toxicity.
Ann Intern Med., 111 (1989), pp. 641-649
[137]
K.D. Glazier, A.L. Palance, L.H. Griffel, K.M. Das.
The ten-year single-center experience with 6-mercaptopurine in the treatment of inflammatory bowel disease.
J Clin Gastroenterol., 39 (2005), pp. 21-26
[138]
L.P. McLean, R.K. Cross.
Adverse events in IBD: to stop or continue immune suppressant and biologic treatment.
Expert Rev Gastroenterol Hepatol., 8 (2014), pp. 223-240
[139]
L.F. Restrepo, R. Giraldo, J. Londoño, C. Pinzón, A. Cortes, G. Ballesteros, et al.
Farmacogenética del metotrexato en artritis reumatoide.
Revisión sistemática. Rev Colomb Reumatol., 23 (2016), pp. 102-114
[140]
Su T-IK, D. Khanna, D.E. Furst, G. Danovitch, C. Burger, P. Maranian, et al.
Rapamycin versus methotrexate in early diffuse systemic sclerosis: results from a randomized, single-blind pilot study.
Arthritis Rheum., 60 (2009), pp. 3821-3830
[141]
M. Krishna Sumanth, V.K. Sharma, B.K. Khaitan, A. Kapoor, T. Tejasvi.
Evaluation of oral methotrexate in the treatment of systemic sclerosis.
Int J Dermatol., 46 (2007), pp. 218-223
[142]
J.E. Pope, N. Bellamy, J.R. Seibold, M. Baron, M. Ellman, S. Carette, et al.
A randomized, controlled trial of methotrexate versus placebo in early diffuse scleroderma.
[143]
F.H. Van den Hoogen, A.M. Boerbooms, A.J. Swaak, J.J. Rasker, H.J. van Lier, L.B. van de Putte.
Comparison of methotrexate with placebo in the treatment of systemic sclerosis: a 24 week randomized double-blind trial, followed by a 24 week observational trial.
Br J Rheumatol., 35 (1996), pp. 364-372
[144]
S. Bernatsky, M. Hudson, S. Suissa.
Anti-rheumatic drug use and risk of serious infections in rheumatoid arthritis.
Rheumatology., 46 (2007), pp. 1157-1160
[145]
A. McLean-Tooke, C. Aldridge, S. Waugh, G.P. Spickett, L. Kay.
Methotrexate, rheumatoid arthritis and infection risk: what is the evidence?.
Rheumatology., 48 (2009), pp. 867-871
[146]
G.G. Fareau, R. Vassilopoulou-Sellin.
Hypercortisolemia and infection.
Infect Dis Clin North Am., 21 (2007), pp. 639-657
[147]
M. Iudici.
What should clinicians know about the use of glucocorticoids in systemic sclerosis?.
Mod Rheumatol., 27 (2017), pp. 919-923
[148]
J. Youssef, S.A. Novosad, K.L. Winthrop.
Infection Risk and Safety of Corticosteroid Use.
Rheum Dis Clin North Am., 42 (2016), pp. 157-176
[149]
K. Migita, Y. Sasaki, N. Ishizuka, T. Arai, T. Kiyokawa, E. Suematsu, et al.
Glucocorticoid therapy and the risk of infection in patients with newly diagnosed autoimmune disease.
Medicine (Baltimore)., 92 (2013), pp. 285-293
[150]
K. Migita, T. Arai, N. Ishizuka, Y. Jiuchi, Y. Sasaki, Y. Izumi, et al.
Rates of serious intracellular infections in autoimmune disease patients receiving initial glucocorticoid therapy.
[151]
J.E. Barahona-Correa, M.A. Franco-Cortés, J. Ángel-Uribe, L.S. Rodríguez-Camacho.
Comparison of plasma cytokine levels before and after treatment with rituximab in patients with rheumatoid arthritis and systemic lupus erythematosus-associated polyautoimmunity.
Univ Médica., 59 (2018), pp. 1-16
[152]
P. Fraticelli, C. Fischetti, F. Salaffi, M. Carotti, M. Mattioli, G. Pomponio, et al.
Combination therapy with rituximab and mycophenolate mofetil in systemic sclerosis. A single-centre case series study.
Clin Exp Rheumatol, 36 (2018), pp. 142-145
[153]
M. Thiebaut, D. Launay, S. Rivière, T. Mahévas, S. Bellakhal, E. Hachulla, et al.
Efficacy and safety of rituximab in systemic sclerosis: French retrospective study and literature review.
Autoimmun Rev., 17 (2018), pp. 582-587
[154]
K. Melsens, E. Vandecasteele, E. Deschepper, V. Badot, D. Blockmans, G. Brusselle, et al.
Two years follow-up of an open-label pilot study of treatment with rituximab in patients with early diffuse cutaneous systemic sclerosis.
Acta Clin Belg., 73 (2018), pp. 119-125
[155]
M. Elhai, O. Distler, V. Smith, M. Matucci-Cerinic, J.J. Alegre-Sancho, M.-E. Truchetet, et al.
Rituximab in systemic sclerosis: safety and efficacy data from the eustar network.
Ann Rheum Dis., 77 (2018), pp. 122
[156]
E.A.A. Christou, G. Giardino, A. Worth, F. Ladomenou.
Risk factors predisposing to the development of hypogammaglobulinemia and infections post-Rituximab.
Int Rev Immunol., 36 (2017), pp. 352-359
[157]
M. Mikulska, S. Lanini, C. Gudiol, L. Drgona, G. Ippolito, M. Fernández-Ruiz, 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., 24 (2018), pp. S71-82
[158]
J.-J. Tudesq, G. Cartron, S. Rivière, D. Morquin, L. Iordache, A. Mahr, et al.
Clinical and microbiological characteristics of the infections in patients treated with rituximab for autoimmune and/or malignant hematological disorders.
Autoimmun Rev., 17 (2018), pp. 115-124
[159]
T. Kelesidis, G. Daikos, D. Boumpas, S. Tsiodras.
Does rituximab increase the incidence of infectious complications? A narrative review.
Int J Infect Dis., 15 (2011), pp. e2-16
[160]
R.P. Perrillo, R. Gish, Y.T. Falck-Ytter.
American Gastroenterological Association Institute technical review on prevention and treatment of hepatitis B virus reactivation during immunosuppressive drug therapy.
Gastroenterology., 148 (2015), pp. 221-244
[161]
E.A. Jauregui.
Agentes biológicos no anti-TNF y pequeñas moléculas.
Reumatología Fundamentos de Medicina, 8th ed., pp. 253-270
[162]
O. Distler, J.H.W. Distler.
Tocilizumab for systemic sclerosis: implications for future trials.
Lancet., 387 (2016), pp. 2580-2581
[163]
S. Manabe, H. Yanagi, H. Ozawa, A. Takagi.
Necrotising soft tissue infection without systemic toxicity in a patient with rheumatoid arthritis treated with tocilizumab.
[164]
M.T.T. Nguyen, J. Pødenphant, P. Ravn.
Three cases of severely disseminated Staphylococcus aureus infection in patients treated with tocilizumab.
[165]
D. Kobayashi, S. Ito, A. Hirata, A. Abe, A. Murasawa, I. Narita, et al.
Mycobacterium abscessus pulmonary infection under treatment with tocilizumab.
Intern Med., 54 (2015), pp. 1309-1313
[166]
T. Koike, M. Harigai, S. Inokuma, N. Ishiguro, J. Ryu, T. Takeuchi, et al.
Effectiveness and safety of tocilizumab: postmarketing surveillance of 7901 patients with rheumatoid arthritis in Japan.
J Rheumatol., 41 (2014), pp. 15-23
[167]
R.J. Moots, A. Sebba, W. Rigby, A. Ostor, B. Porter-Brown, F. Donaldson, et al.
Effect of tocilizumab on neutrophils in adult patients with rheumatoid arthritis: pooled analysis of data from phase 3 and 4 clinical trials.
Rheumatology (Oxford)., 56 (2017), pp. 541-549
[168]
K.L. Winthrop, X. Mariette, J.T. Silva, E. Benamu, L.H. Calabrese, A. Dumusc, 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, immunoglobuli.
Clin Microbiol Infect., 24 (2018), pp. S21-40
[169]
E.F. Chakravarty, V. Martyanov, D. Fiorentino, T.A. Wood, D.J. Haddon, J.A. Jarrell, et al.
Gene expression changes reflect clinical response in a placebo-controlled randomized trial of abatacept in patients with diffuse cutaneous systemic sclerosis.
Arthritis Res Ther., 17 (2015), pp. 159
[170]
M. Elhai, M. Meunier, M. Matucci-Cerinic, B. Maurer, G. Riemekasten, T. Leturcq, et al.
Outcomes of patients with systemic sclerosis-associated polyarthritis and myopathy treated with tocilizumab or abatacept: a EUSTAR observational study.
Ann Rheum Dis., 72 (2013), pp. 1217-1220
[171]
N.D. Tank, B.N. Karelia, B.N. Vegada.
Biological response modifiers in rheumatoid arthritis: systematic review and meta-analysis of safety.
J Pharmacol Pharmacother., 8 (2017), pp. 92-105
[172]
F. Montastruc, C. Renoux, M. Hudson, S. Dell’Aniello, T.A. Simon, S. Suissa.
Abatacept initiation in rheumatoid arthritis and the risk of serious infection: A population-based cohort study.
Semin Arthritis Rheum., (2019),
[173]
K.L. Grøn, E.V. Arkema, B. Glintborg, F. Mehnert, M. Østergaard, L. Dreyer, et al.
Risk of serious infections in patients with rheumatoid arthritis treated in routine care with abatacept, rituximab and tocilizumab in Denmark and Sweden.
Ann Rheum Dis., (2019),
[174]
M.C. Genovese, C. Pacheco-Tena, A. Covarrubias, G. Leon, E. Mysler, M. Keiserman, et al.
Longterm safety and efficacy of subcutaneous abatacept in patients with rheumatoid arthritis: 5-year results from a phase IIIb trial.
J Rheumatol., 45 (2018), pp. 1085-1092
[175]
J.H.W. Distler, S. Jordan, P. Airo, J.J. Alegre-Sancho, Y. Allanore, A. Balbir Gurman, et al.
Is there a role for TNFα antagonists in the treatment of SSc? EUSTAR expert consensus development using the Delphi technique.
Clin Exp Rheumatol., 29 (2011), pp. S40-5
[176]
G. Murdaca, F. Spanò, M. Contatore, A. Guastalla, F. Puppo.
Potential use of TNF-α inhibitors in systemic sclerosis.
Immunotherapy., 6 (2014), pp. 283-289
[177]
M.A. Omair, V. Phumethum, S.R. Johnson.
Long-term safety and effectiveness of tumour necrosis factor inhibitors in systemic sclerosis patients with inflammatory arthritis.
Clin Exp Rheumatol., 30 (2012), pp. S55-9
[178]
C.P. Denton, M. Engelhart, N. Tvede, H. Wilson, K. Khan, X. Shiwen, et al.
An open-label pilot study of infliximab therapy in diffuse cutaneous systemic sclerosis.
Ann Rheum Dis., 68 (2009), pp. 1433-1439
[179]
G.K. Lam, L.K. Hummers, A. Woods, F.M. Wigley.
Efficacy and safety of etanercept in the treatment of scleroderma- associated joint disease.
J Rheumatol., 34 (2007), pp. 1636-1637
[180]
R. Pereira, P. Lago, R. Faria, T. Torres.
Safety of anti-TNF therapies in immune-mediated inflammatory diseases: focus on infections and malignancy.
Drug Dev Res., 76 (2015), pp. 419-427
[181]
U.A. Walker, L.A. Saketkoo, O. Distler.
Haematopoietic stem cell transplantation in systemic sclerosis.
RMD open, 4 (2018),
[182]
N. Del Papa, F. Onida, E. Zaccara, G. Saporiti, W. Maglione, E. Tagliaferri, et al.
Autologous hematopoietic stem cell transplantation has better outcomes than conventional therapies in patients with rapidly progressive systemic sclerosis.
Bone Marrow Transplant., 52 (2017), pp. 53-58
[183]
P. Szodoray, L. Varoczy, G. Papp, S. Barath, B. Nakken, G. Szegedi, et al.
Immunological reconstitution after autologous stem cell transplantation in patients with refractory systemic autoimmune diseases.
Scand J Rheumatol., 41 (2012), pp. 110-115
[184]
K.M. Sullivan, E.A. Goldmuntz, L. Keyes-Elstein, P.A. McSweeney, A. Pinckney, B. Welch, et al.
Myeloablative autologous stem-cell transplantation for severe scleroderma.
N Engl J Med., 378 (2018), pp. 35-47
[185]
J.M. Van Laar, D. Farge, J.K. Sont, K. Naraghi, Z. Marjanovic, J. Larghero, et al.
Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial.
JAMA., 311 (2014), pp. 2490-2498
[186]
R.K. Burt, S.J. Shah, K. Dill, T. Grant, M. Gheorghiade, J. Schroeder, et al.
Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): an open-label, randomised phase 2 trial.
Lancet., 378 (2011), pp. 498-506
[187]
P.F. Halloran.
Immunosuppressive drugs for kidney transplantation.
N Engl J Med., 351 (2004), pp. 2715-2729
[188]
J.A. Fishman.
Infection in solid-organ transplant recipients.
N Engl J Med., 357 (2007), pp. 2601-2614
[189]
J. Gavalda, A. Roman, A. Pahissa.
Risks and Epidemiology of Infections after Lung or Heart-Lung Transplantation.
Transplant infections, third edit., pp. 114-137
[190]
D.B. Badesch, V.F. Tapson, M.D. McGoon, B.H. Brundage, L.J. Rubin, F.M. Wigley, et al.
Continuous intravenous epoprostenol for pulmonary hypertension due to the scleroderma spectrum of disease. A randomized, controlled trial.
Ann Intern Med., 132 (2000), pp. 425-434
[191]
Centers for Disease Control and Prevention (CDC).
Bloodstream infections among patients treated with intravenous epoprostenol or intravenous treprostinil for pulmonary arterial hypertension--seven sites, United States, 2003–2006.
MMWR Morb Mortal Wkly Rep, 56 (2007), pp. 170-172
[192]
N. Kitterman, A. Poms, D.P. Miller, S. Lombardi, H.W. Farber, R.J. Barst.
Bloodstream infections in patients with pulmonary arterial hypertension treated with intravenous prostanoids: insights from the REVEAL REGISTRY®.
Mayo Clin Proc., 87 (2012), pp. 825-834
[193]
F. López-Medrano, M. Fernández-Ruiz, M.J. Ruiz-Cano, E. Barrios, M. Vicente-Hernández, J.M. Aguado, et al.
High incidence of bloodstream infection due to gram-negative bacilli in patients with pulmonary hypertension receiving intravenous treprostinil.
Arch Bronconeumol., 48 (2012), pp. 443-447
[194]
M. Ogielska, P. Lanotte, C. Le Brun, A.S. Valentin, D. Garot, A.-C. Tellier, et al.
Emergence of community-acquired Clostridium difficile infection: the experience of a French hospital and review of the literature.
Int J Infect Dis., 37 (2015), pp. 36-41
[195]
C. Carvajal, C. Pacheco, F. Jaimes.
[Clinical and demographic profile and risk factors for Clostridium difficile infection].
Biomedica., 37 (2017), pp. 53-61
[196]
C.-S. Eom, C.Y. Jeon, J.-W. Lim, E.-G. Cho, S.M. Park, K.-S. Lee.
Use of acid-suppressive drugs and risk of pneumonia: a systematic review and meta-analysis.
CMAJ., 183 (2011), pp. 310-319
[197]
J. Johnstone, K. Nerenberg, Loeb M. Meta-analysis.
proton pump inhibitor use and the risk of community-acquired pneumonia.
Aliment Pharmacol Ther., 31 (2010), pp. 1165-1177
[198]
A.A. Lambert, J.O. Lam, J.J. Paik, C. Ugarte-Gil, M.B. Drummond, T.A. Crowell.
Risk of community-acquired pneumonia with outpatient proton-pump inhibitor therapy: a systematic review and meta-analysis.
[199]
K.B. Filion, D. Chateau, L.E. Targownik, A. Gershon, M. Durand, H. Tamim, et al.
Proton pump inhibitors and the risk of hospitalisation for community-acquired pneumonia: replicated cohort studies with meta-analysis.
[200]
O. Distler, K.B. Highland, M. Gahlemann, A. Azuma, A. Fischer, M.D. Mayes, et al.
Nintedanib for systemic sclerosis-associated interstitial lung disease.
N Engl J Med., 380 (2019), pp. 2518-2528
[201]
K.R. Flaherty, A.U. Wells, V. Cottin, A. Devaraj, S.L.F. Walsh, Y. Inoue, et al.
Nintedanib in progressive fibrosing interstitial lung diseases.
[202]
L. Richeldi, R.M. du Bois, G. Raghu, A. Azuma, K.K. Brown, U. Costabel, et al.
Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis.
N Engl J Med., 370 (2014), pp. 2071-2082
[203]
T. Ogura, H. Taniguchi, A. Azuma, Y. Inoue, Y. Kondoh, Y. Hasegawa, et al.
Safety and pharmacokinetics of nintedanib and pirfenidone in idiopathic pulmonary fibrosis.
Eur Respir J., 45 (2015), pp. 1382-1392
[204]
M. Kato, S. Sasaki, T. Nakamura, K. Kurokawa, T. Yamada, Y. Ochi, et al.
Gastrointestinal adverse effects of nintedanib and the associated risk factors in patients with idiopathic pulmonary fibrosis.
[205]
L. Lancaster, B. Crestani, P. Hernandez, Y. Inoue, D. Wachtlin, L. Loaiza, et al.
Safety and survival data in patients with idiopathic pulmonary fibrosis treated with nintedanib: pooled data from six clinical trials.
BMJ open Respir Res., 6 (2019),
[206]
T.E. King, W.Z. Bradford, S. Castro-Bernardini, E.A. Fagan, I. Glaspole, M.K. Glassberg, et al.
A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis.
N Engl J Med., 370 (2014), pp. 2083-2092
[207]
A. Fischer, J. Distler.
Progressive fibrosing interstitial lung disease associated with systemic autoimmune diseases.
Clin Rheumatol., (2019),
[208]
E. Hachulla, P.-Y. Hatron, P. Carpentier, C. Agard, E. Chatelus, P. Jego, et al.
Efficacy of sildenafil on ischaemic digital ulcer healing in systemic sclerosis: the placebo-controlled SEDUCE study.
Ann Rheum Dis., 75 (2016), pp. 1009-1015
[209]
A. Wei, Z. Gu, J. Li, X. Liu, X. Wu, Y. Han, et al.
Clinical adverse effects of endothelin receptor antagonists: insights from the meta-analysis of 4894 patients from 24 randomized double-blind placebo-controlled clinical trials.
J Am Heart Assoc., 5 (2016),
[210]
A.E. Thompson, B. Shea, V. Welch, D. Fenlon, J.E. Pope.
Calcium-channel blockers for Raynaud’s phenomenon in systemic sclerosis.
[211]
M. Roustit, S. Blaise, Y. Allanore, P.H. Carpentier, E. Caglayan, J.-L. Cracowski.
Phosphodiesterase-5 inhibitors for the treatment of secondary Raynaud’s phenomenon: systematic review and meta-analysis of randomised trials.
Ann Rheum Dis., 72 (2013), pp. 1696-1699
[212]
L.J. Rubin, N. Galiè, F. Grimminger, E. Grünig, M. Humbert, Z.-C. Jing, et al.
Riociguat for the treatment of pulmonary arterial hypertension: a long-term extension study (PATENT-2).
Eur Respir J., 45 (2015), pp. 1303-1313
[213]
H.-A. Ghofrani, N. Galiè, F. Grimminger, E. Grünig, M. Humbert, Z.-C. Jing, et al.
Riociguat for the treatment of pulmonary arterial hypertension.
N Engl J Med., 369 (2013), pp. 330-340
[214]
L. Drgona, C. Gudiol, S. Lanini, B. Salzberger, G. Ippolito, M. Mikulska.
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 or myeloid cells surface antigens [II]: CD22, CD30, CD33.
Clin Microbiol Infect., 24 (2018), pp. S83-94
[215]
C.E. Pérez-Díaz, E. Uribe-Pardo, O.-J. Calixto, ÁA. Faccini-Martínez, A.J. Rodriguez-Morales.
Infections with biological therapy: strategies for risk minimization in tropical and developing countries.
J Infect Dev Ctries., 10 (2016), pp. 1278-1285
[216]
E. Forero, M. Chalem, G. Vásquez, E. Jauregui, L.F. Medina, L.F. Pinto Peñaranda, et al.
Gestión de riesgo para la prescripción de terapias biológicas.
Rev Colomb Reumatol., 23 (2016), pp. 50-67
[217]
J. Aguilar-Company, M. Fernández-Ruiz, R. García-Campelo, A.C. Garrido-Castro, I. Ruiz-Camps.
ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Cell surface receptors and associated signaling pathways).
Clin Microbiol Infect., 24 (2018), pp. S41-52
[218]
G. Redelman-Sidi, O. Michielin, C. Cervera, C. Ribi, J.M. Aguado, M. Fernández-Ruiz, 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 (Immune checkpoint inhibitors, cell adhesion inhibitors, sphingosine-1-phosphate rece.
Clin Microbiol Infect., 24 (2018), pp. S95-107
[219]
M. Reinwald, J.T. Silva, N.J. Mueller, J. Fortún, C. Garzoni, J.W. de Fijter, 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 (Intracellular signaling pathways: tyrosine kinase and mTOR inhibitors).
Clin Microbiol Infect., 24 (2018), pp. S53-70
[220]
M. Fernández-Ruiz, Y. Meije, O. Manuel, H. Akan, J. Carratalà, J.M. Aguado, 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 (Introduction).
Clin Microbiol Infect., 24 (2018), pp. S2-9
[221]
S. Van Assen, N. Agmon-Levin, O. Elkayam, R. Cervera, M.F. Doran, M. Dougados, et al.
EULAR recommendations for vaccination in adult patients with autoimmune inflammatory rheumatic diseases.
Ann Rheum Dis., 70 (2011), pp. 414-422
[222]
D.K. Kim, L.E. Riley, P. Hunter.
Advisory committee on immunization practices recommended immunization schedule for adults aged 19 years or older - United States.
MMWR Morb Mortal Wkly Rep., 67 (2018), pp. 158-160
[223]
J.M. Gómez Muñoz, J.C. Gómez Rincón, A. Alí Munive, C.A. Cano Gutierrez, P.X. Coral Alvarado, W. Coronell Rodríguez, et al.
Guías para la inmunización del adolescente y adulto en Colombia. Documento de actualización, 2016.
Infectio, 20 (2016), pp. 192-210
[224]
E. Forero, M. Chalem, G. Vásquez, E. Jauregui, L.F. Medina, L.F.P. Penaranda, et al.
Risk management for prescribing biological therapies.
Rev Colomb Reumatol., 23 (2016), pp. 50-67
[225]
A. Roux, F. Gonzalez, M. Roux, M. Mehrad, J. Menotti, J.-R. Zahar, et al.
Update on pulmonary Pneumocystis jirovecii infection in non-HIV patients.
Med Mal Infect., 44 (2014), pp. 185-198
[226]
G. Schmajuk, K. Jafri, M. Evans, S. Shiboski, M. Gianfrancesco, Z. Izadi, et al.
Pneumocystis jirovecii pneumonia (PJP) prophylaxis patterns among patients with rheumatic diseases receiving high-risk immunosuppressant drugs.
Semin Arthritis Rheum., (2018),
[227]
M. Yates, R.A. Watts, I.M. Bajema, M.C. Cid, B. Crestani, T. Hauser, et al.
EULAR/ERA-EDTA recommendations for the management of ANCA-associated vasculitis.
Ann Rheum Dis., 75 (2016), pp. 1583-1594
[228]
M. Yates, R. Watts, I. Bajema, M. Cid, B. Crestani, T. Hauser, et al.
Validation of the EULAR/ERA-EDTA recommendations for the management of ANCA-associated vasculitis by disease content experts.
[229]
J.W. Park, J.R. Curtis, J. Moon, Y.W. Song, S. Kim, E.B. Lee, et al.
Prophylactic effect of trimethoprim-sulfamethoxazole for pneumocystis pneumonia in patients with rheumatic diseases exposed to prolonged high-dose glucocorticoids.
Ann Rheum Dis., 77 (2018), pp. 644-649
[230]
A. Kronbichler, J. Kerschbaum, S. Gopaluni, J. Tieu, F. Alberici, R.B. Jones, et al.
Trimethoprim-sulfamethoxazole prophylaxis prevents severe/life-threatening infections following rituximab in antineutrophil cytoplasm antibody-associated vasculitis.
Ann Rheum Dis., 77 (2018), pp. 1440-1447
[231]
P.J. Clements, J.G. Goldin, E.C. Kleerup, D.E. Furst, R.M. Elashoff, D.P. Tashkin, et al.
Regional differences in bronchoalveolar lavage and thoracic high-resolution computed tomography results in dyspneic patients with systemic sclerosis.
Arthritis Rheum., 50 (2004), pp. 1909-1917
[232]
M. Utsunomiya, H. Dobashi, T. Odani, K. Saito, N. Yokogawa, K. Nagasaka, et al.
Optimal regimens of sulfamethoxazole-trimethoprim for chemoprophylaxis of Pneumocystis pneumonia in patients with systemic rheumatic diseases: results from a non-blinded, randomized controlled trial.
Arthritis Res Ther., 19 (2017), pp. 7
[233]
Adolescents P on OI in H-I and A and. Guidelines for the prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: recommendations from the Centers for Disease Control and Prevention, the National Institutes of Health, and the HIV Medicine Association of the [Internet]. 2018 [cited 2018 Dec 24]. Available from: http://aidsinfo.nih.gov/contentfiles/lvguidelines/adult_oi.pdf.
[234]
E. Ntatsaki, D. Carruthers, K. Chakravarty, D. D’Cruz, L. Harper, D. Jayne, et al.
BSR and BHPR guideline for the management of adults with ANCA-associated vasculitis.
Rheumatology., 53 (2014), pp. 2306-2309
[235]
G. Baulier, N. Issa, F. Gabriel, I. Accoceberry, F. Camou, P. Duffau.
Guidelines for prophylaxis of Pneumocystis pneumonia cannot rely solely on CD4-cell count in autoimmune and inflammatory diseases.
Clin Exp Rheumatol., 36 (2018), pp. 490-493
[236]
C. Garnier, D. Ribes, D. Chauveau, A. Huart, G. Pugnet, D. Adoue, et al.
Zoster after cyclophosphamide for systemic lupus erythematosus or vasculitis: incidence, risk factors, and effect of antiviral prophylaxis.
J Rheumatol., 45 (2018), pp. 1541-1548
[237]
K.L. Winthrop, T. Chiller.
Preventing and treating biologic-associated opportunistic infections.
Nat Rev Rheumatol., 5 (2009), pp. 405-410
[238]
K.L. Winthrop, M. Iseman.
Bedfellows: mycobacteria and rheumatoid arthritis in the era of biologic therapy.
Nat Rev Rheumatol., 9 (2013), pp. 524-531
[239]
World Health Organization. Latent tuberculosis infection: updated and consolidated guidelines for programmatic management. [Internet]. 2018 [cited 2018 Dec 24]. Available from: https://www.who.int/tb/publications/2018/latent-tuberculosis-infection/en/.
[240]
M. Setti, D. Fenoglio, F. Ansaldi, G. Filaci, S. Bacilieri, L. Sticchi, et al.
Flu vaccination with a virosomal vaccine does not affect clinical course and immunological parameters in scleroderma patients.
[241]
L.B.S. Gelinck, A.E. van der Bijl, W.E.P. Beyer, L.G. Visser, T.W.J. Huizinga, R.A. van Hogezand, et al.
The effect of anti-tumour necrosis factor treatment on the antibody response to influenza vaccination.
Ann Rheum Dis., 67 (2007), pp. 713-716
[242]
O. Elkayam, A. Bashkin, M. Mandelboim, I. Litinsky, D. Comaheshter, D. Levartovsky, et al.
The effect of infliximab and timing of vaccination on the humoral response to influenza vaccination in patients with rheumatoid arthritis and ankylosing spondylitis.
Semin Arthritis Rheum., 39 (2010), pp. 442-447
[243]
F. Lanternier, C. Henegar, L. Mouthon, P. Blanche, L. Guillevin, O. Launay.
Low influenza-vaccination rate among adults receiving immunosuppressive therapy for systemic inflammatory disease.
Ann Rheum Dis., 67 (2008), pp. 1047
[244]
P.D. Sampaio-Barros, D.C.O. Andrade, L.C.P. Seguro, S.G. Pasoto, V.S.T. Viana, A.C.M. Ribeiro, et al.
Pandemic non-adjuvanted influenza A H1N1 vaccine in a cohort of patients with systemic sclerosis.
Rheumatology., 57 (2018), pp. 1721-1725
[245]
Centers for Disease Control and Prevention (CDC).
Use of 13-valent pneumococcal conjugate vaccine and 23-valent pneumococcal polysaccharide vaccine for adults with immunocompromising conditions: recommendations of the Advisory Committee on Immunization Practices (ACIP).
MMWR Morb Mortal Wkly Rep, 61 (2012), pp. 816-819
[246]
R. Hesselstrand, J. Nagel, T. Saxne, P. Geborek, L. Skattum, M.C. Kapetanovic.
Immunogenicity and safety of pneumococcal vaccination in patients with systemic sclerosis.
Rheumatology., 57 (2018), pp. 625-630
[247]
U. Mercado, H. Acosta, R. Diaz-Molina.
Antibody response to pneumococcal polysaccharide vaccine in systemic sclerosis.
J Rheumatol., 36 (2009), pp. 1549-1550
[248]
K.L. Dooling, A. Guo, M. Patel, G.M. Lee, K. Moore, E.A. Belongia, et al.
Recommendations of the advisory committee on immunization practices for use of herpes zoster vaccines.
MMWR Morb Mortal Wkly Rep., 67 (2018), pp. 103-108

Please cite this article as: Barahona-Correa JE, De la Hoz A, López MJ, Garzón J, Allanore Y, Quintana-López G. Infecciones y esclerosis sistémica: un desafío emergente.Rev Colomb Reumatol. 2020;27:62–84.

Article options
Tools