ArticlePDF Available

Anti-Rheumatic Drugs May Ameliorate the Clinical Course and Outcome of COVID-19 In Rheumatoid Arthritis Patients

Authors:

Abstract

Current data demonstrated that in patients with coronavirus disease-19 (COVID-19), there is a dysregulation of the immune system during the severe form of the disease. This dysregulation is expressed with an uncontrolled release of pro-inflammatory cytokines such as interleukin-1 (IL-1), IL-6, IL-17, tumour necrosis factor alpha (TNFa) and chemokines, associated with increased serum ferritin levels and other acute phase reactants. On the other side, these cytokines play a pivotal role in autoimmune rheumatic diseases (ARD), mostly in rheumatoid arthritis (RA) and the spondyloarthropathies. Patients affected with ARD represent a particular vulnerable group, considering that they may be in an immunocompromised status due to their ailment and its treatment on one side, but on the other side, they may be protected from their immunosuppressive therapy. To this end, we present five patients with RA treated with conventional synthetic (cs) disease-modifying anti-rheumatic drugs (DMARDs) and biologic (b) DMARDs who were affected from COVID-19 and we will try to give answers to the above hypothesis.
M
E
D
I
T
E
R
R
A
N
E
A
N
J
O
U
R
N
A
L
O
F
R
H
E
U
M
A
T
O
L
O
G
Y
E-ISSN: 2529-198X
MEDITERRANEAN JOURNAL OF RHEUMATOLOGY
Anti-Rheumatic Drugs May Ameliorate the Clinical Course
and Outcome of COVID-19 In Rheumatoid Arthritis Patients
Eleftherios Pelechas, Vassiliki Drossou,
Paraskevi V. Voulgari, Alexandros A. Drosos
Mediterr J Rheumatol 2022;33(1):68-74
March 2022 | Volume 33 | Issue 1
MEDITERRANEAN JOURNAL
OF RHEUMATOLOGY
33
1
2022
68
This work is licensed
under a Creative Commons
Attribution 4.0
International License.
The pandemic of coronavirus disease 19 (COVID-19)
caused by the severe acute respiratory syndrome corona-
virus-2 (SARS-CoV-2) is a matter of concern worldwide.
Environmental factors
such as smoking,
obesity, cardio-respira-
tory diseases, diabetes
mellitus and age may
increase the severity of
COVID-19.1,2 Current
data demonstrated an
acute dysregulation of
the immune system
during the severe form
of the disease. The
above is demonstrated
by the clinical picture and laboratory ndings, suggesting
a severe inammatory response of the innate immunity, af-
fecting mainly the respiratory system with the recruitment
in the lung parenchyma of macrophages, monocytes,
and lymphocytes, followed by the thrombotic diathesis
and multiorgan failure. Beside the clinical manifestations,
the immune response is expressed with the activation
of macrophages and lymphocytes generating a plethora
of cytokines like interleukin (IL)-1, IL-6, IL-17, tumour
necrosis factor a (TNFa), and various chemokines. As a
consequence, high levels of acute phase reactants are
produced, such as C-reactive protein (CRP), hyperfer-
ritinaemia and hyper-brinogenaemia. Afterwards, a
dysregulation of the adaptive immunity with reduction
of lymphocytes mostly CD4+ and CD8+ T-cells takes
place.3,4
Keywords: COVID-19, rheumatoid arthritis, hydroxychloroquine, tocilizumab, csDMARDs, bDMARDs
©Pelechas E, Drossou V, Voulgari PVV, Drosos AA.
ABSTRACT
Current data demonstrated that in patients with coronavirus disease-19 (COVID-19), there is a
dysregulation of the immune system during the severe form of the disease. This dysregulation is
expressed with an uncontrolled release of pro-inammatory cytokines such as interleukin-1 (IL-
1), IL-6, IL-17, tumour necrosis factor alpha (TNFa) and chemokines, associated with increased
serum ferritin levels and other acute phase reactants. On the other side, these cytokines play a
pivotal role in autoimmune rheumatic diseases (ARD), mostly in rheumatoid arthritis (RA) and the
spondyloarthropathies. Patients aected with ARD represent a particular vulnerable group, considering
that they may be in an immunocompromised status due to their ailment and its treatment on one
side, but on the other side, they may be protected from their immunosuppressive therapy. To this
end, we present ve patients with RA treated with conventional synthetic (cs) disease-modifying anti-
rheumatic drugs (DMARDs) and biologic (b) DMARDs who were aected from COVID-19 and we will
try to give answers to the above hypothesis.
Mediterr J Rheumatol 2022;33(1):68-74
https://doi.org/10.31138/mjr.33.1.68
Article Submitted: 7 Mar 2021; Revised Form: 11 Oct 2021; Article Accepted: 20 Oct 2021; Available Online: 31 Mar 2022
Cite this article as: Pelechas E, Drossou V, Voulgari PVV, Drosos AA. Anti-Rheumatic Drugs May Ameliorate the Clinical Course and Outcome
of COVID-19 In Rheumatoid Arthritis Patients. Mediterr J Rheumatol 2022;33(1):68-74.
Corresponding Author:
Alexandros A. Drosos, MD, FACR
Professor of Medicine/Rheumatology
Rheumatology Clinic, Department of
Internal Medicine
Medical School, University of Ioannina
Ioannina 45110, Greece
Tel.: +30 2651 007 503
Fax: +30 2651 007 054
E-mail: adrosos@uoi.gr
Web: www.rheumatology.gr
Anti-Rheumatic Drugs May Ameliorate the Clinical Course and Outcome of
COVID-19 In Rheumatoid Arthritis Patients
Eleftherios Pelechas , Vassiliki Drossou , Paraskevi V. Voulgari , Alexandros A. Drosos
Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
CASE-BASED REVIEW
69
On the other hand, autoimmune rheumatic diseases
(ARD) are also characterised by a dysregulation of the
immune system in which several pro-inammatory cy-
tokines such as IL-1, IL-6, IL-17, and TNFa which are
involved in the pathogenesis of rheumatoid arthritis (RA),
the spondyloarthropathies (SpA), and inammatory bow-
el disease (IBM).5 In the last two decades the introduction
of biological (b) disease-modifying anti-rheumatic drugs
(DMARDs) targeting cytokines, T and B cells, and with
the use of conventional synthetic (cs) DMARDS, has
revolutionized the treatment of the above diseases.6,7
The questions which arise here are: what is the impact of
CΟVID-19 inection in patients with ARD “already treated
with cs and bDMARDs”? Does their immunocompro-
mised status due to the disease itself and its treatment put
their life in danger? Are they protected against COVID-19
infection due to long use of cs and bDMARDs?.8 For this
reason, we present ve patients with RA, treated with cs
and bDMARDs, who were infected with COVID-19, and
discuss the relevant literature trying to answer the above
questions. An informed consent has been obtained from
all patients.
CASE PRESENTATION 1
A 42-year-old woman was diagnosed 6 years earlier with
RA on the basis of clinical and laboratory ndings. More
specically, bilateral symmetrical polyarthritis of the small
joints of the hands, positive anti-citrullinated protein anti-
bodies (ACPA), positive IgM rheumatoid factor (IgM-RF),
as well as high CRP and erythrocyte sedimentation rate
(ESR). She was treated with hydroxychloroquine (HCQ)
and methotrexate (MTX), plus 10mg of prednisone. She
responded very well to the above treatment regimen
since last year that developed a disease are. Since then,
adalimumab (ADA), an anti-TNFa inhibitor, was added.
This treatment resulted in complete clinical and laborato-
ry remission until recently. Indeed, on September 2, 2020
she experienced low-grade fever (37.7 °C), myalgias,
arthralgias, and malaise. Clinical examination revealed
no frank arthritis and the laboratory tests showed only
a high CRP (14mg/dl - normal values <6) and high ESR
(48mm/h). Chest x-ray was normal. Serological tests for
viruses and bacteria, as well as blood and urine cultures
were negative. However, the test for SARS-CoV-2 was
positive. At that time, she was receiving HCQ (200mg/
day), MTX (15mg/week), prednisone (2mg/day) and
ADA (40mg/14 days subcutaneously) (sc). The last ADA
injection was administered a week before the appear-
ance of fever. She was not a smoker. Past medical and
family history were unremarkable. We advised her to
discontinue MTX and ADA, and to stay in isolation at
home according to the national, European and American
recommendations.9-11 Ten days later, her symptoms
gradually subsided, and the symptoms resolved com-
pletely after 3 weeks. The repeated test for SARS-CoV-2
was negative. One month later, she was symptom-free,
a new test for SARS-CoV-2 was also negative, and she
started receiving her immunosuppressive therapy.
CASE PRESENTATION 2
A 50-year-old man was diagnosed as having RA 4 years
ago, on the basis of symmetrical polyarthritis aecting the
wrists, elbows and the knees bilaterally, and with positive
IgM-RF and high ESR and CRP. He was treated with MTX
20mg/week, plus prednisone 10mg/day. He responded
well, but a year later he relapsed, thus tocilizumab (TCZ),
an IL-6 receptor antagonist was initiated (162mg/week
sc). He responded very well to this treatment regimen,
but on September 24, 2020, he complained of arthral-
gias, myalgias, sore throat, and fever up to 38°C. Clinical
examination revealed no frank arthritis, chest x-ray was
normal, and laboratory tests showed white blood count
of 3.9/109L with normal dierential, high acute phase
reactants and a SARS-CoV-2 test positive. The rest of
the laboratory tests were negative or within normal limits.
Past medical and family history were not signicant. He
was not a smoker. At this time, he was receiving MTX
15mg/week, prednisone 2,5 mg/day and TCZ 162mg/
week. The last TCZ injection was administered three
days before the initiation of his symptoms. MTX and TCZ
were discontinued, and he stayed in isolation at home.9-
11 A week later, he felt well, without sore throat and fever,
and after 2 weeks, he had complete resolution of the
SARS-CoV-2 symptoms. The repeated test for SARS-
CoV-2 was negative and ve weeks later a new test for
SARS-CoV-2 was also negative so he started receiving
the above-mentioned treatment (MTX and TCZ).
CASE PRESENTATION 3
A 62-year-old female with seronegative RA diagnosed 5
years earlier, was in clinical remission since 2018, receiving
HCQ 200mg/day plus Etanercept (ETN) 50mg/week sc.
She presented on 18th November 2020 with arthralgias,
muscle aches, weakness, sore throat, altered sense of
smell, and fever (38.5 °C). She visited our clinic, where af-
ter a detailed clinical and laboratory investigation, she was
diagnosed as having SARS-CoV-2 infection. She had high
ESR and CRP, as well as high ferritin levels (805mcg/L,
normal values <35). She remained isolated at home re-
ceiving HCQ 200mg/day and azithromycin (AZT) 500mg/
day, while ETN was discontinued. She was not receiving
other drugs and she was not a smoker. Seven days later,
she felt well without fever, sore throat and myalgias, but the
olfactory dysfunction was still present. After three weeks
she was free of symptoms. The patient started receiving
ETN after a new SARS-CoV-2 negative test.
CASE PRESENTATION 4
A 55-year-old female with seropositive RA since 2016
receiving MTX 20mg/week and golimumab (GLM) 50mg/
ANTI-RHEUMATIC DRUGS IN COVID PATIENTS
MEDITERRANEAN JOURNAL
OF RHEUMATOLOGY
33
1
2022
70
MEDITERRANEAN JOURNAL
OF RHEUMATOLOGY
33
1
2022
month sc presented on 22nd November 2020 to us com-
plaining of arthralgias, myalgias, sore throat, and loss of
taste and smell. Clinical evaluation revealed no arthritis,
while the laboratory tests showed high ESR and CRP,
and positive SARS-CoV-2 test. She had no other co-
morbidities and was not a smoker. She stayed at home
receiving some analgesics, while MTX and GLM were
discontinued. Two weeks later she felt better, but the loss
of taste and smell had no improvement. After one month
she was free of symptoms with some improvement of
taste and smell. The SARS-CoV-2 test was negative,
and she started receiving MTX and GLM.
CASE PRESENTATION 5
A 38-year-old female with a 12-year history of seroneg-
ative RA presented on 15th December 2020 with severe
arthralgias and myalgias, as well as fever up to 38.5°C
lasting for 3 days. She was on treatment with HCQ
200mg/day and prednisone 2,5mg/day because she
delivered a healthy baby 6 months earlier. Past medical
and family history were unremarkable. Clinical examina-
tion revealed no frank arthritis and she tested positive for
SARS-CoV-2. The rest of the laboratory tests showed
only high CRP. She continued receiving her drugs plus
AZT 500mg/day and she stayed at home. After 10 days
she felt better without fever, myalgias and arthralgias.
One month later, the test for SARS-CoV-2 was negative.
DISCUSSION
In Table 1 we present the clinical and laboratory ndings
of those ve RA patients who were infected with SARS-
CoV-2. All infected patients presented systemic man-
ifestations and mild upper respiratory symptoms from
the nose and throat, while the laboratory investigation
revealed only high acute phase reactants, mostly CRP
and ESR, while one patient had also high ferritin levels. In
addition, one patient had low white blood cells. None of
these patients was hospitalized. Cs and bDMARDs were
discontinued with the exception of HCQ and prednisone.
The outcome of those patients was very good, lasting for
4-5 weeks until complete recovery.
Treatment decisions and therapeutic strategies in critically
ill patients of SARS-CoV-2 infection were initially limited.
Much of the therapy for severe Covid-19 are empirical
and depend mostly on clinical judgement.12 However,
up to date, dierent therapeutic protocols are applied
using anti-viral drugs, steroids (mostly dexamethasone),
supportive treatments, and several anti-rheumatic
drugs, such as HCQ, cytokine inhibitors, and others.13,14
Regarding the patients with ARD infected from SARS-
Table 1. Clinical and laboratory ndings of rheumatoid arthritis patients infected from SARS-CoV-2.
Patients Systemic
manifestations
Ear, nose,
and throat
symptoms
Respiratory
symptoms
Laboratory
tests Treatment Outcome
N:1
Arthralgias,
myalgias, malaise,
fever
None None High ESR
High CRP
HCQ
200mg/day,
prednisone
2.5mg
Good
N:2 Arthralgias,
myalgias, fever Sore throat None
High ESR
High CRP
Low WBC
Prednisone
2.5mg Good
N:3 Arthralgias, myalgia,
weakness, fever
Sore throat,
olfactory
dysfunction
None
High ESR
High CRP
High Ferritin
HCQ 200mg/
day AZT 500
mg/day
Good
N:4 Arthralgias, myalgias
Sore throat,
Loss of taste
and smell
None High ESR
High CRP None Good
N:5 Arthralgias,
Myalgias, fever None None High CRP
HCQ
200mg/day,
prednisone
2.5mg/d,
AZT 500mg/d
Good
SARS-CoV-2: Severe acute respiratory syndrome coronavirus-2; ESR: Erythrocyte sedimentation rate; CRP: C-reactive
protein; WBC: white blood cells; HCQ: hydroxychloroquine; AZT: Azithromycin.
71
TITLE
CoV-2, a study from Gianfrancesco et al. showed that
patients receiving prednisone >10 mg/day had a higher
probability to be hospitalised, while in those receiving
anti-TNFa therapy, the risk of hospitalisation was low.15
Haberman et al. showed that antimalarial therapy was
not associated with the likelihood of hospitalization in
patients infected from SARS-CoV-2. On the other hand,
bDMARDs and targeted synthetic (ts) DMARDs reduced
the odds of hospitalization.16 In a subsequent study,
the same investigators showed that among patients
with ARDs and SARS-CoV-2 infection, the outcome
was worse in those receiving corticosteroids, but none
in those receiving anti-cytokine therapies.17 HCQ has
been used in rheumatology for many years, especially for
the treatment of RA and systemic lupus erythematosus
(SLE). One questions that arises at this point is, how
could HCQ help patients suering from SARS-CoV-2
infection. The answer is simple: we do not know yet,
but what we do know is that HCQ inhibits or impairs
the presentation process of an antigen, or of a microor-
ganism to macrophages due to inhibition of toll-like re-
ceptors (TLRs). Another action of HCQ is that of helping
macrophages to destroy the ‘invader’ in the cytoplasm
through the action of lysosomes and blocks the major
histocompatibility complex (MHC), which is responsible
for the presentation of the ‘invader’ from macrophages
to T cells. Therefore, T cell activation is inhibited (Figure
1). Could the above mechanisms of HCQ block the virus
entry, and prevent its multiplication and spreading? We
do not know, yet.13,14 However, in vitro studies showed
that HCQ blocks viral replication by inhibiting the entry
of SARS-CoV-2 by interacting with glycosylation of
angiotensin converting enzyme-2 (ACE-2) receptor and
its binding with the spike protein.18 This remains to be
documented and there are some trials that are trying
to support it.19-21 Furthermore, high HCQ blood levels
are associated with reduced thrombotic events in SLE
patients,22 which is also a clinical manifestation of SARS-
ANTI-RHEUMATIC DRUGS IN COVID PATIENTS
Figure 1. Antigen-processing onto macrophage/APC and the action of hydroxychloroquine.
An antigen-presenting cell, takes up the pathogen (SARS-CoV-2) end engulfs it with phagocytosis (1). Lysosomes that
contain acids create an acidic environment and fuse with the phagocytosed “invader” creating the phagolysosome which
has an acidic environment too (2). Because of the acidic environment, the content of the phagolysosome breaks down
leaving various particles that will be processed as antigens (3). In the meantime, within the endoplasmic reticulum the ri-
bosomes are synthesizing MHC and allow the formation of an endosome which passes also through the Golgi apparatus
to form a new endosome. The viral breakdown particles will then fuse with the new endosome and the particles will bind
onto the groove of the MHC (4). Finally, the antigen presenting cell will express it on its self-surface (5). Hydroxychloro-
quine, accumulates in lysosomes, raising the pH establishing a non-acidic environment and inhibits the lysosomal activity
and, in this way, it prevents the MHC presentation to the surface of the antigen-presenting cell.
MEDITERRANEAN JOURNAL
OF RHEUMATOLOGY
33
1
2022
72
MEDITERRANEAN JOURNAL
OF RHEUMATOLOGY
33
1
2022
CoV-2 patients. On the other hand, a study by Koning et
al. found no protection for SARS-CoV-2 infection or the
development of the severe form of the disease with the
use of HCQ in patients with SLE.23 Recent data regarding
the eect of HCQ in Covid-19 did not show a clear clini-
cal benet, while a systematic review and meta-analysis
demonstrated that HCQ use was not associated with
any benet or harm regarding Covid-19 morbidity.24
IL-6, IL-1, and TNFa are the pivotal cytokines responsible
for the clinical deterioration of patients suering from
SARS-CoV-2 infection. TCZ is an IL-6 receptor antago-
nist which is used in rheumatology for patients suering
from RA, juvenile idiopathic arthritis and temporal arteritis.
There are several reports, using TCZ for critically ill pa-
tients infected with SARS-CoV-2, showing encouraging
results.25-30 On March 3 2020, TCZ has been approved
in China for use in patients with the SARS-CoV-2 infec-
tion. Indeed, TCZ maintains a crucial role in critically ill
patients with acute progressive SARS-CoV-2. More
than 200 cases of SARS-CoV-2 treated with TCZ have
been published mostly from Chinese, Italian and Spanish
investigators.31,32 In almost all cases, TCZ was found to
be eective, with an acceptable safety prole. The clinical
improvement has been associated with normalization
of CRP and other acute phase reactants.26,28 In a few
studies pulmonary improvement has been also noted by
improvement of imaging ndings.30 On the other hand,
other studies, showed that high IL-6 was associated
with severe phenotype disease of SARS-CoV-2.25 This is
probably due to the fact that IL-6 levels may increase a
few days after TCZ administration.33
Anakinra, is an IL-1 receptor antagonist which is used
in RA, Still’s disease, cryopyrin-associated periodic syn-
drome (CAPS), and in patients with gout. Canakinumab,
is a monoclonal antibody against IL-1 and it is used in
CAPS and gout patients. Anakinra, showed reduction
of CRP and progressive improvement of respiratory
parameters in critically ill patients when administered in-
travenously and in high doses.34 Recently, Moutsopoulos
HM described a patient suering from CAPS who
received Canakinumab 10 days before of being tested
positive for SARS-CoV-2. He reported that the patient
had a mild disease course and a week later the test for
SARS-CoV-2 was negative.35 TNFa inhibitors are used
in the last two decades for the treatment of RA, SpAs,
and other inammatory arthritides. It has been shown
that the use of TNFa inhibitors in SARS-CoV-2 patients
may improve disease outcome and prevent organ dam-
age.36 The question that we addressed at the beginning,
regarding the impact of SARS-CoV-2 infection in ARD
patients, is still a matter of discussion due to limited data
so far. However, single reports, as above, case series
and observational studies published recently indicated
that in patients with ARD and SARS-CoV-2 infection the
disease is expressed with mild clinical symptoms without
severe consequences probably due to their concomitant
use of cs and bDMARDs.15-17,37-44
On the other hand, the Covid-19 Global Rheumatology
Alliance (GRA) has published a series of papers dealing
with the impact of COVID-19 in ARD patients.45 The
results suggest that as in the general population, age
and comorbidities are to blame as the risk factors for
poor outcome in ARD patients infected by SARS-CoV-2.
Furthermore, glucocorticoids are also associated with an
unfavourable outcome in these patients. Finally, Stangfeld
et al. who examined the casualties in a large amount of
patients with ARDs, found that moderate or high disease
activity, and specic drugs (sulfasalazine, azathioprine,
cyclosporine-A, mycophenolate mofetil, tacrolimus,
cyclophosphamide, and the use of rituximab) were as-
sociated with increased odds of COVID-19 deaths.46
However, all the above apply to a mixed population of
ARD patients (SLE, scleroderma, RA and others) who
may have many dierent systemic manifestations and
treatments. The Janus-kinase (JAK) inhibitors tofacitinib
(TOFA) and Baricitinib (BARI), have been proposed as
potential therapeutic agents for SARS-CoV-2 infection.
Especially BARI may block the viral entry into the cells
by inhibiting the members of the numb-associated
kinase (NAK) family, such as adaptor associated protein-
kinase-1 (AAKI) and cyclin-G associated kinase (GAK),
which are involved in the viral endocytosis.47 Currently,
both drugs are used to treat RA and other inammatory
arthritides in a dose of 5mg twice daily for TOFA and
2mg twice daily for BARI. It has been shown that BARI
is able to inhibit eectively AAKI and GAK with the above
approved dose for RA. Moreover, BARI as a selective
inhibitor of JAK 1 and 2 and TOFA as an inhibitor of JAK1
and 3 are also able to inhibit the inammatory response of
SARS-CoV-2, especially IL-1 and INF-γ. Several studies
indicated that JAK inhibitors may have some benecial
eect in the treatment of COVID-19.48,49 However, recent
studies evaluating the impact of the COVID-19 on mor-
bidity and mortality in patients with ARDs indicated that
the use of JAK inhibitors had a negative eect.50,51
In the present report we present ve patients with RA
in clinical remission without systemic manifestations and
comorbidities who were treated with cs and bDMARDs
that achieved a good outcome. Indeed, our RA pa-
tients during the SARS-CoV-2 infection were receiving
csDMARDs (HCQ and MTX), while three of them were
receiving also TNFa inhibitors, and one patient was on
treatment with an IL-6 receptor antagonist. All had mild
clinical manifestations and very good outcome with com-
plete resolution of their symptoms, without hospitalisa-
tion and no additional therapy. Another factor which may
have inuenced the clinical manifestations of our patients
is that none of them had other comorbidities, like diabe-
tes mellitus, coronary disease, hypertension, and other
diseases which may play crucial role in the deterioration
73
TITLE
of the clinical course and outcome of SARS-CoV-2. All
patients, except one, restarted their immunosuppressive
therapy for RA after a negative test for Covid-19, which
is in line with the international guidelines. Alternatively,
the immunosuppressive therapy should be restarted
after a two-week, symptom free period of the patient.52,53
Thus, we can hypothesise that anti-rheumatic drugs
may ameliorate the clinical picture and disease course of
COVID-19 in RA patients. However, further studies with
a large number of patients are needed in order to conrm
the above benecial eects of the anti-rheumatic drugs.
ACKNOWLEDGEMENT
The authors would like to especially thank Ms Areti Fili for
her excellent secretarial support.
CONFLICT OF INTEREST
The authors declare no conict of interest.
REFERENCES
1. Zhou F, Y T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk
factors for mortality of adult inpatients with COVID-19 in Wuhan,
China: a retrospective cohort study. Lancet 2020;395(10229):1054-
62.
2. Wang T, Du Z, Zhu F, Cao Z, An Y, Gao Y, et al. Comorbidities
and multi-organ injuries in the treatment of COVID-19. Lancet
2020;395(10228)e52.
3. Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, et al. Dysregulation
of immune response in patients with coronavirus 2019 (COVID-19)
in Wuhan, China. Clin Infect Dis 2020;71(15):762-8.
4. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson
JJ, et al. COVID-19: consider cytokine storm syndromes and
immunosuppression. Lancet 2020;395(10229):1033-4.
5. Davison A, Diamond B. Autoimmune diseases. N Engl J Med
2001;345(5):340-50.
6. Rosman Z, Shoenfeld Y, Zandman-Goddard G. Biologic therapy
for autoimmune diseases: an update. BMC Med 2013;11:88.
7. Burmester GR, Feist E, Dorner T. Emerging cell and cytokine tar-
gets in rheumatoid arthritis. Nat Rev Rheumatol 2014;10(2):77-88.
8. Misra DP, Agarwal V, Gasparyan AY, Zimba O. Rheumatologists’
perspective on coronavirus disease 19 (COVID-19) and potential
therapeutic targets. Clin Rheumatol 2020;39(7):2055-62.
9. Recommendations regarding COVID 19 infection in Rheumatic
patients in Greece – The Greek Rheumatology Society and
Professional Association of Rheumatologists – ERE-EPERE.
Mediterr J Rheumatol 2020;31(1):6-7.
10. European League against Rheumatism. EULAR.org/eular_guid-
ance_for patients_covid19_outbreak.cfm. Accessed on 05 January
2020.
11. Mikuls TR, Johnson SR, Fraenkel L, Arasaratnam RJ, Baden LR,
Bermas BL, et al. American College of Rheumatology Guidance
for the management of Rheumatic Disease in Adult patients
during the COVID-19 Pandemic: Version 1. Arthritis Rheumatol
2020;72:1241-51.
12. Bhatraju PK, Ghassemieh BJ, Nichols M, Kim R, Jerome KR, Nalla
AK, et al. Covid-19 in critically ill patients in the Seattle region –
case series. N Engl J Med 2020;382(21):2012-22.
13. Ferro F, Elefante E, Baldini C, Bartoloni E, Puxeddu I, Talarico R,
et al. COVID-19: the new challenge for rheumatologists. Clin Exp
Rheumatol 2020;38(2):175-80.
14. Pelechas E, Drossou V, Voulgari PV, Drosos AA. Anti-rheumatic
drugs for the ght against the novel coronavirus infection (SARS-
CoV-2): What is the evidence? Mediterr J Rheumatol 2020;31(Suppl
2):259-67.
15. Gianfrancesco M, Hyrich KL, Al-adely S, Carmona L, Danil MI,
Gossec L, et al. Characteristics associated with hospitalization
for COVID-19 in people with rheumatic disease: data from the
COVID-19 global rheumatology alliance physician-reported regis-
try. Ann Rheum Dis 2020;79(7):859-66.
16. Haberman R, Axelrad J. Covid-19 in immune-mediated inam-
matory diseases. Case series from New York. N Engl J Med
2020:NEJMc2009567.
17. Haberman RH, Castillo R, Chn A, Yan D, Ramirez D, Sekar V, et
al. COVID-19 in patients with inammatory arthritis: a prospective
study on the eects of comorbidities and disease-modifying
antirheumatic drugs on clinical outcomes. Arthritis Rheumatol
2020;72(12):1981-9.
18. Gautret P, Lagier JC, Parola P, Hoang VT, Meddeb L, Maihe M,
et al. Hydroxychloroquine and azithromycin as a treatment of
COVID-19: results of an open-label non-randomized clinical trial.
Int J Antimicrob Agents 2020;56(1):105949.
19. Yao X, Ye F, Zhang M, Cui C, Huang B, Niu P, et al. In vitro antiviral
activity and projection of optimized dosing design of hydroxychlo-
roquine for the treatment of severe acute respiratory syndrome
coronavirus 2 (SARS-CoV-2). Clin Infect Dis 2020:ciaa237.
20. Virdis A, Tani C, Duranti E, Vagnani S, Carli L, Kuhl AA, et al. Early
treatment with hydroxychloroquine prevents the development
of endothelial dysfunction in a murine model of systemic lupus
erythematosus. Arthritis Res Ther 2015;17:277.
21. Xue H Liu Y, Luo P, Liu X, Qiu L, Liu D, et al. Hydroxychloroquine
treatment in COVID-19: a descriptive observational analysis
of 30 cases from a single center in Wuhan, China. J Med Virol
2020;92:2523-7.
22. Petri M, Konig MF, Li J, Goldman DW. Higher hydroxychloroquine
blood levels are associated with reduced thrombosis risk in
systemic lupus erythematosus. Arthritis Rheumatol 2021;73:997-
1004.
23. Konig MF, Kim AH, Scheetz MH, Graef ER, Liew JW, Simard J,
et al. Baseline use of hydroxychloroquine in systemic lupus ery-
thematosus does not preclude SARS-CoV-2 infection and severe
COVID-19. Ann Rheum Dis 2020;79(10):1386-8.
24. Putman M, Chock YPE, Tam H, Kim AHJ, Sattui E, Berenbaum F, et
al. Antirheumatic Disease therapies for the treatment of COVID-19:
a systematic review and meta-analysis. Arthritis Rheumatol
2021;73(1):36-47.
25. Fu B, Xu X, Wei H. Why tocilizumab could be an eective treatment
for severe COVID-19? J Transl Med 2020;18(1):164.
26. Xu X, Han M, Li T, Sun W, Wang D, Fu B, et al. Eective treatment
of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci
USA 2020;117(20):10970-5.
27. Luo P, Liu Y, Qiu L, Liu X, Liu D, Li J. Tocilizumab treatment in
COVID-19: a single center experience. J Med Virol 2020;92(7):814-
8.
28. Di Giambenedetto S, Ciccullo A, Borghetti A, Gambassi G, Landi F,
Visconti E, et al. O-label use of tocilizumab in patients with SARS-
CoV-2 infection. J Med Virol 2020;92(10):1787-8.
29. Piva S, Filippini M, Turla F, Cattaneo S, Margola A, de Fulviis S, et
al. Clinical presentation and initial management critically ill patients
with severe acute respiratory syndrome coronavirus 2 (SARS-
CoV-2) infection in Brescia, Italy. J Crit Care 2020;58:29-33.
30. Cellina M, Orsi M, Bombaci F, Sala M, Marino P, Oliva G. Favorable
changes of CT ndings in a patient with COVID-19 pneumonia after
treatment with tocilizumab. Diagn Interv Imaging 2020;1010(5):323-
4.
31. Sciascia S, Apra F, Baa A, Baldovino S, Boaro D, Boero R, et al.
Pilot prospective open, single-arm multicenter study on o-label
use of tocilizumab in patients with severe COVID-19. Clin Exp
Rheumatol 2020;38(3):529-32.
32. Campins L, Boixeda R, Perez-Cordon L, Aranega R, Lopera C,
Force L. Early tocilizumab treatment could improve survival among
COVID-19 patients. Clin Exp Rheumatol 2020;38(3):578.
33. Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T, et
al. Mechanisms and pathologic signicances in increase in serum in-
ANTI-RHEUMATIC DRUGS IN COVID PATIENTS
MEDITERRANEAN JOURNAL
OF RHEUMATOLOGY
33
1
2022
74
MEDITERRANEAN JOURNAL
OF RHEUMATOLOGY
33
1
2022
terleukin-6 (IL-6) and soluble IL-6 receptor after administration of an
anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid
arthritis and Castleman disease. Blood 2008;112(10):3959-64.
34. Cavalli G, De Luca G, Campochiaro C, Della-Torre E, Ripa M,
Canetti D, et al. Interleukin-1 blockade with high-dose anakinra in
patients with COVID-19, acute respiratory distress syndrome, and
hyperinammation: a retrospective cohort study. Lancet Rheumatol
2020;2(6):e325-e331.
35. Moutsopoulos HM. Anti-inammatory therapy may ameliorate the
clinical picture of COVID-19. Ann Rheum Dis 2020 Sep;79(9):1253-
4.
36. Roux CH, Brocq O, Gerald F, Pradier C, Bailly L. Clinical impact
of COVID-19 on a French population of spondyloarthritis patients.
Clin Rheumatol 2020;39(11):3185-3187. Doi: 10.1007/s10067-
020-05413-9.
37. Mihai C, Dobrota R, Schroder M, Garaiman A, Jordan S, Becker
MO. COVID-19 in a patient with systemic sclerosis treated with
tocilizumab for SSc-ILD, Ann Rheum Dis 2020;79:668-9.
38. Mathian A, Mahevas M, Rohmer J, Roumier M, Cohen-Aubart F,
Amador-Borrero B, et al. Clinical course of coronavirus disease
2019 (COVID-19) in a series of 17 patients with systemic lupus er-
ythematosus under long-term treatment with hydroxychloroquine.
Ann Rheum Dis 2020;79(6):837-9.
39. Han Y, Jiang M, Xia D, He L, Lv X, Liao X, et al. COVID-19 in a
patient with long-term use of glucocorticoids. A study of a familial
cluster. Clin Immunol 2020;214:108413.
40. Monti S, Balduzzi S, Delvino P, Bellis E, Quadrelli VS, Montecucco
C. Clinical course of COVID-19 in a series of patients with chronic
arthritis treated with immunosuppressive targeted therapies. Ann
Rheum Dis 2020;79(5):667-8.
41. Song J, Kang S, Choi SW, Seo KW, Lee S, So MW, et al. Coronavirus
disase 19 (COVID-19) complicated with pneumonia in a patient
with rheumatoid arthritis receiving conventional disease-modifying
antireumatic drugs. Rheumatol Int 2020;40(6):991-5.
42. Duret PM, Sebbag E, Mallick A, Gravier S, Spielmann L, Messer
L. Recovery from COVID-19 in a patient with spondyloarthritis
treated with TNF-alpha inhibitor etanercept. Ann Rheum Dis
2020;79(9):1251-2.
43. Emmi G, Bettiol A, Mattioli I, Silvestri E, di Scla G, Urban ML, et al.
SARS-CoV-2 infection among patients with systemic autoimmune
diseases. Autoimmun Rev 2020;19(7):102575.
44. Migkos MP, Kaltsonoudis E, Pelechas E, Drossou V, Karagianni
PG, Kavvadias A, et al. Use of conventional synthetic and biologic
disease-modifying anti-rheumatic drugs in patients with rheumatic
diseases contracting COVID-19: a single-center experience.
Rheumatol Int 2021 Mar 3;1-7.
45. Gianfrancesco M, Yazdany J, Robinson PC. Epidemiology and out-
comes of novel coronavirus 2019 in patients with immune-mediated
inammatory diseases. Curr Opin Rheumatol 2020;32(5):434-40.
46. Strangfeld A, Schäfer M, Gianfrancesco MA, Lawson-Tovey S,
Liew JW, Ljung L, et al. Factors associated with COVID-19-
related death in people with rheumatic diseases: results from
the COVID-19 Global Rheumatology Alliance physician-reported
registry. Ann Rheum Dis 2021 Jul;80(7):930-42.
47. Favalli EG, Ingegnoli F, de Lucia O, Cincinelli G, Cimaz R, Caporali
R. COVID-19 infection and rheumatoid arthritis: Faraway, so close!
Autoimmun Rev 2020;19(5):102523.
48. Richardson P, Grin I, Tucker C, Smith D, Oechsle O, Phelan A, et
al. Baricitinib as potential treatment for 2019-nCoV acute respira-
tory disease. Lancet 2020;395(10223):e30-e31.
49. Aitken T, Chin KL, Liew D, Ofori-Asenso R. Rethinking pandemic
preparation: global health security index (GHSI) is predictive
of COVID-19 burden, but in the opposite direction. J Infect
2020;81(2):318-56.
50. Bower H, Frisell T, di Giuseppe D, Delcoigne B, Ahlenius GM,
Baecklund E, et al. Impact of the COVID-19 pandemic on morbid-
ity and mortality in patients with inammatory joint diseases an in
the general population: a nationwide Swedish cohort study. Ann
Rheum Dis. 2021;80(8):1086-93.
51. Sparks JA, Wallace ZS, Seet AM, Gianfrancesco MA, Izadi Z,
Hyrich KL, et al. Associations of baseline use of biologic or targeted
synthetic DMARDs with COVID-19 severity in rheumatoid arthritis:
Results from the COVID-19 global rheumatology alliance physician
registry. Ann Rheum Dis 2021;80(9):1137-46.
52. Mikuls TR, Johnson SR, Fraenkel L, Arasaratnam RJ, Baden LR,
Bermas BL, et al. American College of Rheumatology Guidance
for the Management of Rheumatic Disease in Adult Patients
During the COVID-19 Pandemic: Version 1. Arthritis Rheumatol
2020;72(8):1241-51.
53. Barlow-Pay F, Htut TW, Khezrian M, Myint PK. Systematic review of
immunosuppressant guidelines in the COVID-19 pandemic. Ther
Adv Drug Saf 2021;12:2042098620985687.
... Pelechas E et al. reported five patients with RA under treatment with conventional and biological DMARDs which have SARS-CoV-2 infection. 13 The authors hypothesise that anti-rheumatic drugs may ameliorate the clinical picture and disease course of COVID-19 in RA patients. The efficacy of various DMARDs with different mechanism of action in the treatment of SARS-CoV-2 infection may explain with the fact that immune system dysregulation and release of higher amounts of cytokines are responsible in its pathogenesis. ...
Article
Full-text available
SARS-CoV-2 infection is a pandemic that affects predominantly upper airways and lungs. It may lead to reactivation of known inflammatory rheumatic diseases and/or initiation of various granulomatous disorders. Necrotising sarcoid granulomatosis (NSG) is a rare condition that can be confused with malignancy, granulomatosis with polyangiitis, and sarcoidosis. Herein we reported the development of NSG following a SARS-CoV-2 infection which mimicked granulomatosis with polyangiitis.
... Furthermore, some studies with these two immunossupressors found protection against more severe stages of COVID-19 as seen more recently by other authors. 43,56,59,76 Rituximab and other anti-CD20 antibodies that cause B-lymphocyte depletion, impairing humoral immunity, were associated, in LES and MS patients, with an aggravated clinical picture as seen by other authors. 77,78 As for IL-6 inhibitors, only in one study, they were associated with an increased number of hospitalisations in RA patients. ...
Article
Full-text available
The purpose of this work was to analyse published data on rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and multiple sclerosis (MS) and SARS-CoV-2 infection: susceptibility, post-infection autoimmune disease (AD) exacerbation, immunosuppressive therapies and long COVID. Supported by PICO strategy, two independent reviewers conducted the research in the PubMed/Medline database from January 2020 to June 2022 and included 16 articles on RA, 25 on MS and 12 on SLE. The quality assessment of the studies was performed using criteria from the National Institute of Health. Patients with RA or SLE had increased susceptibility to contracting SARS-CoV-2. It was higher in RA and increased with the patients’ comorbidities. For MS, susceptibility to SARS-CoV-2 was similar to the general population. Post-infection AD exacerbation occurred in AR, SLE and MS with an increased number of hospitalisations and deaths. Regarding therapies, in RA the use of glucocorticoids (GC) was associated with a worsening of the infection. A more severe clinical picture was associated with anti-CD20 in SLE and with anti-CD20 and methylprednisolone in MS. Considering long COVID, RA and SLE patients had a higher risk of complications opposite to MS patients. There was a higher susceptibility to SARS-CoV-2 infection in rheumatological diseases AR and SLE, exacerbated by age and comorbidities. For RA and MS, GC aggravated the infection and for SLE and MS anti-CD20 antibodies use. In all AD there was exacerbation and worsening of the clinical picture translated in long COVID, the latter with MS exception.
Article
Unlabelled: Coronavirus infections have been responsible since 2019 for respiratory illnesses with varying severity worldwide. Worst outcomes from coronavirus (COVID-19) have been reported in older patients andthose with comorbidities like rheumatic diseases. Some drugs used for treating rheumatic diseases are used in patients with COVID-19. Based on the limited data, rheumatic diseases do not seem to affect the disease course of COVID-19. We aimed to analyze the course of COVID-19 infections in patients with rheumatic diseases. Methods: A self-reported questionnaire was distributed online and to patients admitted with respiratory involvement. Data included demographic information, clinical presentation, severity, comorbidities, and laboratory parameters. Cases were matched by age, sex, the month of admission, and COVID-19 respiratory injury for patients with rheumatic diseases and patients without rheumatic diseases. Results: Twenty-two patients (4.4%) had rheumatic diseases before the COVID-19 infection. There were no differences in the use of treatment for COVID-19 infections in previous or present therapy or comorbidities. We found no significant difference in the duration of COVID-19 symptoms before admission, duration of hospital stay, or chest Xray Brixia score between the two groups. The lymphocyte count was lower in the patient group, while lactate dehydrogenase, ferritin, and D-dimer concentrations were higher compared to the control group. Thrombotic events were similar in rate. Conclusion: The poorer outcome from COVID-19 infections in patients with rheumatic diseases is related to older age and the presence of comorbidities rather than the rheumatic disease type or its treatment.
Article
Full-text available
Objective To investigate baseline use of biologic or targeted synthetic (b/ts) disease-modifying antirheumatic drugs (DMARDs) and COVID-19 outcomes in rheumatoid arthritis (RA). Methods We analysed the COVID-19 Global Rheumatology Alliance physician registry (from 24 March 2020 to 12 April 2021). We investigated b/tsDMARD use for RA at the clinical onset of COVID-19 (baseline): abatacept (ABA), rituximab (RTX), Janus kinase inhibitors (JAKi), interleukin 6 inhibitors (IL-6i) or tumour necrosis factor inhibitors (TNFi, reference group). The ordinal COVID-19 severity outcome was (1) no hospitalisation, (2) hospitalisation without oxygen, (3) hospitalisation with oxygen/ventilation or (4) death. We used ordinal logistic regression to estimate the OR (odds of being one level higher on the ordinal outcome) for each drug class compared with TNFi, adjusting for potential baseline confounders. Results Of 2869 people with RA (mean age 56.7 years, 80.8% female) on b/tsDMARD at the onset of COVID-19, there were 237 on ABA, 364 on RTX, 317 on IL-6i, 563 on JAKi and 1388 on TNFi. Overall, 613 (21%) were hospitalised and 157 (5.5%) died. RTX (OR 4.15, 95% CI 3.16 to 5.44) and JAKi (OR 2.06, 95% CI 1.60 to 2.65) were each associated with worse COVID-19 severity compared with TNFi. There were no associations between ABA or IL6i and COVID-19 severity. Conclusions People with RA treated with RTX or JAKi had worse COVID-19 severity than those on TNFi. The strong association of RTX and JAKi use with poor COVID-19 outcomes highlights prioritisation of risk mitigation strategies for these people.
Article
Full-text available
To examine whether patients with inflammatory arthritis (IA) treated with conventional synthetic (cs) disease-modifying anti-rheumatic drugs (DMARDs) and/or biologic (b) DMARDs, could be affected from SARS-CoV-2 infection and to explore the COVID-19 disease course and outcome in this population. This is a prospective observational study. During the period February–December 2020, 443 patients with IA who were followed-up in the outpatient arthritis clinic were investigated. All patients were receiving cs and/or bDMARDs. During follow-up, the clinical, laboratory findings, comorbidities and drug side effects were all recorded and the treatment was adjusted or changed according to clinical manifestations and patient’s needs. There were 251 patients with rheumatoid arthritis (RA), 101 with psoriatic arthritis (PsA) and 91 with ankylosing spondylitis (AS). We identified 32 patients who contracted COVID-19 (17 RA, 8 PsA, 7 AS). All were in remission and all drugs were discontinued. They presented mild COVID-19 symptoms, expressed mainly with systemic manifestations and sore throat, while six presented olfactory dysfunction and gastrointestinal disturbances, and all of them had a favorable disease course. However, three patients were admitted to the hospital, two of them with respiratory symptoms and pneumonia and were treated appropriately with excellent clinical response and outcome. Patients with IA treated with cs and/or bDMARDs have almost the same disease course with the general population when contract COVID-19.
Article
Full-text available
Objectives To estimate absolute and relative risks for all-cause mortality and for severe COVID-19 in inflammatory joint diseases (IJDs) and with antirheumatic therapies. Methods Through Swedish nationwide multiregister linkages, we selected all adult patients with rheumatoid arthritis (RA, n=53 455 in March 2020), other IJDs (here: spondyloarthropathies, psoriatic arthritis and juvenile idiopathic arthritis, n=57 112), their antirheumatic drug use, and individually matched population referents. We compared annual all-cause mortality March–September 2015 through 2020 within and across cohorts, and assessed absolute and relative risks for hospitalisation, admission to intensive care and death due to COVID-19 March–September 2020, using Cox regression. Results During March–September 2020, the absolute all-cause mortality in RA and in other IJDs was higher than 2015–2019, but relative risks versus the general population (around 2 and 1.5) remained similar during 2020 compared with 2015–2019. Among patients with IJD, the risks of hospitalisation (0.5% vs 0.3% in their population referents), admission to intensive care (0.04% vs 0.03%) and death (0.10% vs 0.07%) due to COVID-19 were low. Antirheumatic drugs were not associated with increased risk of serious COVID-19 outcomes, although for certain drugs, precision was limited. Conclusions Risks of severe COVID-19-related outcomes were increased among patients with IJDs, but risk increases were also seen for non-COVID-19 morbidity. Overall absolute and excess risks are low and the level of risk increases are largely proportionate to those in the general population, and explained by comorbidities. With possible exceptions, antirheumatic drugs do not have a major impact on these risks.
Article
Full-text available
Aims Individuals taking immunosuppressants are at increased susceptibility to viral infections in general. However, due to the novel nature of the COVID-19, there is a lack of evidence about the specific risks of the disease in this patient group. This systematic review aims to summarize the current international clinical guidelines to highlight areas where research is needed through critical appraisal of the evidence base of these guidelines. Methods We conducted a systematic review of clinical practice guidelines about the usage of immunosuppressants during the COVID-19 pandemic. Electronic databases including MEDLINE and the websites of relevant professional bodies were searched for English language guidelines that were published or updated between March 2020 and May 2020 in this area. We assessed the quality and consistency of guidelines. The evidence base underpinning these guidelines was critically appraised using GRADE criteria. Results Twenty-three guidelines were included. Most guidelines ( n = 15, 65.2%) informed and updated evidence based on expert opinion. The methodological quality of the guidelines varied, ranging from ‘very low’ to ‘moderate’. Guidelines consistently recommended that high-risk patients, including those who are taking high doses of steroids for more than a month, or a combination of two or more immunosuppressants, should be shielding during the outbreak. Most guidelines stated that steroids usage should not be stopped abruptly and advised on individualized risk–benefit analysis considering the risk of the effect of COVID-19 infection and the relapse of the autoimmune condition in patients. Discussion Clinical practice guidelines on taking immunosuppressants during the COVID-19 outbreak vary in quality. The level of evidence informing the available guidelines was generally low. Given the novel nature of COVID-19, the guidelines draw on existing knowledge and data, refer to the use of immunosuppressants and risks of serious infections of other aetiologies and have extrapolated these to form their evidence base.
Article
Full-text available
Objectives To determine factors associated with COVID-19-related death in people with rheumatic diseases. Methods Physician-reported registry of adults with rheumatic disease and confirmed or presumptive COVID-19 (from 24 March to 1 July 2020). The primary outcome was COVID-19-related death. Age, sex, smoking status, comorbidities, rheumatic disease diagnosis, disease activity and medications were included as covariates in multivariable logistic regression models. Analyses were further stratified according to rheumatic disease category. Results Of 3729 patients (mean age 57 years, 68% female), 390 (10.5%) died. Independent factors associated with COVID-19-related death were age (66–75 years: OR 3.00, 95% CI 2.13 to 4.22; >75 years: 6.18, 4.47 to 8.53; both vs ≤65 years), male sex (1.46, 1.11 to 1.91), hypertension combined with cardiovascular disease (1.89, 1.31 to 2.73), chronic lung disease (1.68, 1.26 to 2.25) and prednisolone-equivalent dosage >10 mg/day (1.69, 1.18 to 2.41; vs no glucocorticoid intake). Moderate/high disease activity (vs remission/low disease activity) was associated with higher odds of death (1.87, 1.27 to 2.77). Rituximab (4.04, 2.32 to 7.03), sulfasalazine (3.60, 1.66 to 7.78), immunosuppressants (azathioprine, cyclophosphamide, ciclosporin, mycophenolate or tacrolimus: 2.22, 1.43 to 3.46) and not receiving any disease-modifying anti-rheumatic drug (DMARD) (2.11, 1.48 to 3.01) were associated with higher odds of death, compared with methotrexate monotherapy. Other synthetic/biological DMARDs were not associated with COVID-19-related death. Conclusion Among people with rheumatic disease, COVID-19-related death was associated with known general factors (older age, male sex and specific comorbidities) and disease-specific factors (disease activity and specific medications). The association with moderate/high disease activity highlights the importance of adequate disease control with DMARDs, preferably without increasing glucocorticoid dosages. Caution may be required with rituximab, sulfasalazine and some immunosuppressants.
Article
Full-text available
Objective Hydroxychloroquine (HCQ) has a primary role in the prophylaxis and treatment of systemic lupus erythematosus (SLE) and may be protective against thrombosis in SLE. Optimal weight‐based dosing of HCQ is unknown. This study was undertaken to examine the usefulness of HCQ blood monitoring in predicting thrombosis risk in a longitudinal SLE cohort. Methods HCQ levels were serially quantified from EDTA whole blood by liquid chromatography–tandem mass spectrometry. The mean HCQ blood levels calculated prior to thrombosis or until the last visit were compared using t‐tests between patients with and those without thrombosis. Pooled logistic regression was used to analyze the association between rates of thrombosis and HCQ blood level. Rate ratios (RRs) and 95% confidence intervals (95% CIs) were calculated. Results In 739 patients with SLE, thrombosis occurred in 38 patients (5.1%). The mean ± SD HCQ blood level was lower in patients who developed thrombosis versus those who did not develop thrombosis (720 ± 489 ng/ml versus 935 ± 580 ng/ml; P = 0.025). Thrombosis rates were reduced by 13% for every 200‐ng/ml increase in the most recent HCQ blood level (RR 0.87 [95% CI 0.78–0.98], P = 0.025) and by 13% for mean HCQ blood level (RR 0.87 [95% CI 0.76–1.00], P = 0.056). Thrombotic events were reduced by 69% in patients with mean HCQ blood levels ≥1,068 ng/ml versus those with levels <648 ng/ml (RR 0.31 [95% CI 0.11–0.86], P = 0.024). This remained significant after adjustment for confounders (RR 0.34 [95% CI 0.12–0.94], P = 0.037). Conclusion Low HCQ blood levels are associated with thrombotic events in SLE. Longitudinal measurement of HCQ levels may allow for personalized HCQ dosing strategies. Recommendations for empirical dose reduction may reduce or eliminate the benefits of HCQ in this high‐risk population.
Article
Full-text available
SARS-CoV-2 is a positive-sense single-stranded RNA virus that causes the COVID-19 infection. Spike proteins are the most important proteins found on its capsule using the host's ACE2 receptors to invade respiratory cells. The natural course of the COVID-19 infection is variable, from asymptomatic to severe and potentially fatal. A small percentage of the severely infected patients will end up in an intensive care unit for ventilatory support. Elderly male patients with pre-existing medical conditions and smokers are at a disproportionate high risk to develop severe complications. Studies have shown that deaths occur due to a dysregulated immune system that overreacts, producing a plethora of cytokines, leading to the so-called "cytokine storm" phenomenon. In this direction, many drugs that are used in the everyday practice of Rheumatologists have been used. Indeed, pro-inflammatory cytokines such as the IL-1 and IL-6 have been shown to be the pivotal cytokines expressed, and anti-cytokine treatment has been tried so far with various results. In addition, hydroxychloroquine, an antimalarial drug, has been shown to reduce COVID-19 symptoms. Other drugs have also been used, such as intravenous pulses of immunoglobulins, and colchicine. Robust clinical trials are needed in order to find the suitable treatment. Current data indicate that hydroxychloroquine and cytokine targeting therapies may prove helpful in the fight of SARS-CoV-2 in appropriately selected patients.
Article
Full-text available
Objective Antirheumatic disease therapies have been used to treat coronavirus disease 2019 (COVID‐19) and its complications. We conducted a systematic review and meta‐analysis to describe the current evidence. Methods A search of published and preprint databases in all languages was performed. Included studies described ≥1 relevant clinical outcome for ≥5 patients who were infected with severe acute respiratory syndrome coronavirus 2 and were treated with antirheumatic disease therapy between January 1, 2019 and May 29, 2020. Pairs of reviewers screened articles, extracted data, and assessed risk of bias. A meta‐analysis of effect sizes using random‐effects models was performed when possible. Results The search identified 3,935 articles, of which 45 were included (4 randomized controlled trials, 29 cohort studies, and 12 case series). All studies evaluated hospitalized patients, and 29 of the 45 studies had been published in a peer‐reviewed journal. In a meta‐analysis of 3 cohort studies with a low risk of bias, hydroxychloroquine use was not significantly associated with mortality (pooled hazard ratio [HR] 1.41 [95% confidence interval (95% CI) 0.83, 2.42]). In a meta‐analysis of 2 cohort studies with some concerns/higher risk of bias, anakinra use was associated with lower mortality (pooled HR 0.25 [95% CI 0.12, 0.52]). Evidence was inconclusive with regard to other antirheumatic disease therapies, and the majority of other studies had a high risk of bias. Conclusion In this systematic review and meta‐analysis, hydroxychloroquine use was not associated with benefit or harm regarding COVID‐19 mortality. The evidence supporting the effect of other antirheumatic disease therapies in COVID‐19 is currently inconclusive.