8
views
0
recommends
+1 Recommend
3 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found

      Care for patients with rheumatic diseases during COVID‐19 pandemic: A position statement from APLAR

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          1 INTRODUCTION The outbreak of coronavirus disease 2019 (COVID‐19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) was first reported in China in December 2019. This disease now affects the whole world. Patients with rheumatic diseases are at higher risk of respiratory infections including influenza and pneumococcal pneumonia, which is attributed to the underlying disease, comorbidities and immunosuppressive therapy, 1 but to date we lack good information about the virus SARS‐CoV‐2. Nonetheless, immunosuppressive treatments are essential to control disease activity and prevent functional deterioration in these patients. Rheumatologists need to be vigilant in preventing rheumatic disease patients from contracting the disease during this pandemic, especially patients with chronic lung problems (eg scleroderma with lung fibrosis) and chronic kidney disease (eg lupus nephritis) and those on high‐dose glucocorticoids and immunosuppressants (Appendix 1). In the desperate search to find effective treatments for COVID‐19, drugs largely used by rheumatologists have entered the spotlight, including the caution against use of non‐steroidal anti‐inflammatory drugs (NSAIDs), the potential of antimalarials and biologic disease‐modifying anti‐rheumatic drugs (bDMARDs), for example anti‐interleukin‐6 (IL‐6) and targeted synthetic DMARDS (tsDMARDs) Janus‐activated kinase (JAK) inhibitors to manage cytokine storm syndrome (CSS)/cytokine release syndrome associated with COVID‐19. Here, we try to provide guidance regarding clinical decision‐making both for patients with COVID‐19 and those with rheumatic diseases, and strategies to mitigate further harm to these patients. 2 METHODS An Asia‐Pacific League Against Rheumatism (APLAR) COVID‐19 task force comprising rheumatologists from 9 Asia‐Pacific countries was convened on 31 March, 2020. A set of guidance statements was developed and refined based on best available evidence up to 26 April, 2020 and expert opinion. Given the overall limited nature of the data, a systematic review was not performed. The final guidance statements integrate both the task force members' assessment of the evidence quality and the ratio of risk and benefit from the treatment or action. We assert that the key guiding principle should be to “first do no harm,” especially given the unknown efficacy of proposed DMARDs and biologics and their established potential harms. This guidance document has been reviewed and endorsed by the APLAR executive committee and the APLAR scientific committee chairpersons. 3 HOW CAN WE MINIMIZE THE RISK OF RHEUMATIC DISEASE PATIENTS FROM EXPOSURE TO COVID‐19? In the absence of a vaccine or a therapeutic agent, a “mitigation approach”, including “social distancing”, frequent hand washing and quarantining strategies are the primary interventions to hamper the spread of infection. 2 Another approach, known as “suppression strategies” includes strict lockdown measures – social distancing in entire populations, the closure of schools and community spaces, aggressive case finding and contact tracing, have succeeded in maintaining low case counts of COVID‐19. During this extraordinary time, the rheumatology community faces unprecedented challenges as care could be postponed/delayed or handled through virtual care to minimize contact exposure and to practice social distancing. Comorbid conditions are common in patients with COVID‐19. 3 Smoking can cause an increase in the release of IL‐6 in bronchial epithelial cells, 4 and upregulate angiotensin‐converting enzyme‐2 (ACE2) receptors, the known receptor for SARS‐CoV. 5 This is particularly relevant as some of the Asia‐Pacific countries, for example China, has a high male smoking rate. 6 Globally the quality of evaluation, monitoring and treatment of comorbidities in rheumatic disease patients is variable with considerable scope for improvement. 7 Rheumatologists should be vigilant in assessing and managing comorbidities not only to improve morbidity and mortality, but hopefully to minimize risk of COVID‐19 in rheumatic disease patients. 4 NON‐STEROIDAL ANTI‐INFLAMMATORY DRUGS In patients with acute respiratory tract infections, short‐term use of NSAIDs are associated with increased risk of cardiovascular events and nephrotoxicity, 8 , 9 , 10 higher rates of complications, and delays in the prescription of effective antibiotic treatment. 11 Despite the lack of evidence relating specifically to people with COVID‐19, regular NSAID use should not be recommended as the first line option for managing the symptoms of COVID‐19. 12 Nonetheless, arthritis patients taking NSAIDs for symptomatic relief should continue their treatment as needed. 5 USE OF IMMUNOSUPPRESSANTS AND RISK OF COVID‐19 INFECTION Epidemiologic studies have identified advanced age, male gender and presence of comorbidities (hypertension, obesity, diabetes, coronary heart disease, chronic obstructive lung disease and chronic kidney disease) as poor prognostic factors for COVID‐19. 13 Despite the lack of data on the true prevalence and risk of COVID‐19 in rheumatic disease patients, immunosuppressed status (the use of chemotherapy or conditions requiring immunosuppressive treatment) was not reported to be a risk factor and risk for adverse outcome. One patient with systemic sclerosis‐associated interstitial lung disease (SSC‐ILD) on tocilizumab and 7 patients on bDMARDs or tsDMARDs who developed COVID‐19 recovered uneventfully. 14 , 15 , 16 Nonetheless, at least 2 patients on rituximab 17 developed respiratory failure and 1 of them died despite treatment with tocilizumab. 18 In order to gather real‐world data to inform treatment strategies and better characterize individuals at increased risk of infection, the COVID‐19 Global Rheumatology Alliance has successfully developed online portals and case report forms to enable healthcare providers around the world to enter information on individuals with rheumatic disease who have been diagnosed with COVID‐19, with clinical data of the first 110 patients published. 19 For now, patients with stable rheumatic diseases should continue their treatment. In case of infection (including COVID‐19), treatment of infection gains precedence and immunosuppressive treatment may be de‐escalated or temporarily withheld in consultation with the treating rheumatologist (Appendix 1). 5.1 Glucocorticoid therapy Acute lung injury and acute respiratory distress syndrome (ARDS) are partly caused by host immune responses. Severe COVID‐19‐associated pneumonia patients may exhibit features of systemic hyper‐inflammation or CSS. COVID‐19 infection with CSS typically occurs in subjects with ARDS and historically, non‐survival in ARDS was linked to sustained IL‐6 and IL‐1 elevation. 20 Corticosteroids suppress lung inflammation but also inhibit immune responses and pathogen clearance. The effectiveness of adjunctive glucocorticoid therapy in the management of COVID‐19 infected patients remains controversial. 21 , 22 Until results from ongoing randomized‐controlled trials are available, the World Health Organization (WHO) has advised against routine use of systemic corticosteroids for treatment of viral pneumonia outside of clinical trials unless they were indicated for other reasons (eg septic shock) (Appendix 2). In rheumatic disease patients on long‐term steroids, it is very important to remind them not to stop their prednisone even if they develop symptoms suggestive of COVID‐19 (Appendix 1). For patients with active rheumatic disease, after excluding concurrent active infection, the prednisone dose could be increased carefully according to the severity of the organ manifestation, in spite of the risk of COVID‐19. 5.2 Conventional synthetic disease‐modifying anti‐rheumatic drugs Preclinical and limited clinical data suggested that hydroxychloroquine (HCQ) and chloroquine (CLQ) have antiviral activities against SARS‐CoV‐2. 23 , 24 , 25 In contrast, a small but randomized study from China in patients with mild to moderate COVID‐19 treated with HCQ or placebo found no difference in recovery rates, 26 and French investigators failed to confirm antiviral activity or clinical benefit of the HCQ and azithromycin combination in 11 hospitalized patients with severe COVID‐19. 27 In a French series of 17 systemic lupus erythematosus (SLE) patients with COVID‐19 on long‐term HCQ, 11 (65%) and 5 (29%) developed respiratory failure and ARDS respectively despite having blood HCQ concentrations within the therapeutic range for the management of SLE. 28 Whether blood HCQ concentrations may be effective for the antiviral activity against SARS‐CoV‐2 remained uncertain. Nonetheless, data from this study suggest that HCQ may not be able to prevent severe COVID‐19 in these patients. The US Food and Drug Administration (FDA) cautioned against use of HCQ or CLQ for COVID‐19 outside of the hospital setting or a clinical trial due to risk of heart rhythm problems (Appendix 2). The APLAR task force agreed there are insufficient clinical data to recommend either for or against HCQ or CLQ for COVID‐19, and clinicians should monitor patients for adverse effects, especially prolonged QTc interval. Health authorities should ensure adequate supply of these drugs since the HCQ shortage not only will limit availability to patients with COVID‐19 if efficacy is truly established but also represents a real risk to patients with rheumatic diseases. On the other hand, rheumatologists should remind patients to continue HCQ and not to taper the dosage even if they develop symptoms suggestive of COVID‐19 and reassurance should be given that this drug should not increase their risk of infection. 5.3 Biologic disease‐modifying anti‐rheumatic drugs Once hospitalized, for some patients with COVID‐19, death can occur within a few days, many with ARDS, and some with multi‐organ dysfunction syndrome. 14 In those critically ill patients, there are both clinical signs and symptoms, as well as laboratory abnormalities, that suggest a CSS is occurring in response to the viral infection. According to data from the Chinese cohorts, patients with severe disease and requiring intensive care often show leucopenia, lymphopenia, significantly higher levels of C‐reactive protein (CRP), IL‐6, IL‐10, and tumor necrosis factor‐α (TNF‐α). 29 In this setting, biologic drugs selectively blocking inflammatory cytokines, such as TNF‐α inhibitors, anti‐IL‐6, anti‐IL‐1 and JAK inhibitors are currently employed in the treatment of severe cases of COVID‐19 in an experimental manner or undergoing clinical trials (Appendix 2). Tocilizumab, has been shown effective in treating CSS, a common complication of chimeric antigen receptor‐T cell therapy used for treating refractory acute lymphoblastic leukemia 30 and may be effective in Chinese COVID‐19 patients with severe and critical disease. 31 Anti‐IL‐6R antibody is currently included in the treatment recommendation for Chinese COVID‐19 patients (Appendix 2). These concepts have led to interests in JAK inhibitors, for example baricitinib, as potential treatments for CSS complicated with severe COVID‐19. ACE2 is a cell‐surface protein widely existing on cells in the heart, kidney, blood vessels, especially alveolar epithelial cells. SARS‐CoV‐2 was believed to invade and enter lung cells through ACE2‐mediated endocytosis. One of the known regulators of endocytosis is the AP2‐associated protein kinase 1 (AAK1). AAK1 inhibitors can interrupt the passage of the virus into cells and can be helpful in preventing virus infections. Baricitinib, apart from being a JAK inhibitor, is also an AAK1 inhibitor. Baricitinib was thought to be a possible candidate for treatment of COVID‐19, considering its relative safety and high affinity. 32 On the other hand, JAK–STAT (signal transducer and activator of transcription) signal blocking by baricitinib produces an impairment of interferon‐mediated antiviral response, with a potential facilitating effect on the evolution of SARS‐CoV‐2 infection, and therefore may not be a suitable treatment. 33 While we are waiting for the results from the control trials to resolve this controversy, rheumatologists should be particularly cautious of serious infectious events on the use of this agent, in particular viral infection, for example herpes zoster. 6 CONCLUSIONS Rheumatologists worldwide are trying new strategies to optimize care for rheumatic disease patients during this unprecedented COVID‐19 pandemic. Concerted efforts from healthcare providers in different healthcare systems are required to continue clinical assessments and ensure adequate supply of immunosuppressive therapy. Worsening of rheumatic disease may induce a systemic inflammatory state which may represent an adjunctive risk factor for major susceptibility to viral infection. On the other hand, rheumatologists are cautiously enthusiastic that a variety of immune‐modulating drugs and targeted cytokine inhibitors available for rheumatic disease patients may also benefit patients as prophylaxis for COVID‐19 or with COVID‐19‐induced CSS. Because of insufficient data, APLAR could not recommend any specific treatments for patients with COVID‐19. Nevertheless, rheumatologists/immunologists are expert in the use of these agents and we should be to the forefront in advising around their application, noting risks and benefits are not yet clear and should not be taken for granted in COVID‐19. We emphasize the ongoing importance of critical review of emerging literature to inform current and future treatment decisions. International registries have been created to collect data on rheumatic patients with COVID‐19. Ultimately, time and these registries will tell what the right decision is regarding maintaining current therapy for patients with rheumatic diseases. The APLAR task force will respond quickly and efficiently to place the evidence base behind our recommendations and update them should new findings emerge from clinical trials.

          Related collections

          Most cited references31

          • Record: found
          • Abstract: found
          • Article: found

          Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus–Infected Pneumonia in Wuhan, China

          In December 2019, novel coronavirus (2019-nCoV)-infected pneumonia (NCIP) occurred in Wuhan, China. The number of cases has increased rapidly but information on the clinical characteristics of affected patients is limited.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro

            Dear Editor, In December 2019, a novel pneumonia caused by a previously unknown pathogen emerged in Wuhan, a city of 11 million people in central China. The initial cases were linked to exposures in a seafood market in Wuhan. 1 As of January 27, 2020, the Chinese authorities reported 2835 confirmed cases in mainland China, including 81 deaths. Additionally, 19 confirmed cases were identified in Hong Kong, Macao and Taiwan, and 39 imported cases were identified in Thailand, Japan, South Korea, United States, Vietnam, Singapore, Nepal, France, Australia and Canada. The pathogen was soon identified as a novel coronavirus (2019-nCoV), which is closely related to sever acute respiratory syndrome CoV (SARS-CoV). 2 Currently, there is no specific treatment against the new virus. Therefore, identifying effective antiviral agents to combat the disease is urgently needed. An efficient approach to drug discovery is to test whether the existing antiviral drugs are effective in treating related viral infections. The 2019-nCoV belongs to Betacoronavirus which also contains SARS-CoV and Middle East respiratory syndrome CoV (MERS-CoV). Several drugs, such as ribavirin, interferon, lopinavir-ritonavir, corticosteroids, have been used in patients with SARS or MERS, although the efficacy of some drugs remains controversial. 3 In this study, we evaluated the antiviral efficiency of five FAD-approved drugs including ribavirin, penciclovir, nitazoxanide, nafamostat, chloroquine and two well-known broad-spectrum antiviral drugs remdesivir (GS-5734) and favipiravir (T-705) against a clinical isolate of 2019-nCoV in vitro. Standard assays were carried out to measure the effects of these compounds on the cytotoxicity, virus yield and infection rates of 2019-nCoVs. Firstly, the cytotoxicity of the candidate compounds in Vero E6 cells (ATCC-1586) was determined by the CCK8 assay. Then, Vero E6 cells were infected with nCoV-2019BetaCoV/Wuhan/WIV04/2019 2 at a multiplicity of infection (MOI) of 0.05 in the presence of varying concentrations of the test drugs. DMSO was used in the controls. Efficacies were evaluated by quantification of viral copy numbers in the cell supernatant via quantitative real-time RT-PCR (qRT-PCR) and confirmed with visualization of virus nucleoprotein (NP) expression through immunofluorescence microscopy at 48 h post infection (p.i.) (cytopathic effect was not obvious at this time point of infection). Among the seven tested drugs, high concentrations of three nucleoside analogs including ribavirin (half-maximal effective concentration (EC50) = 109.50 μM, half-cytotoxic concentration (CC50) > 400 μM, selectivity index (SI) > 3.65), penciclovir (EC50 = 95.96 μM, CC50 > 400 μM, SI > 4.17) and favipiravir (EC50 = 61.88 μM, CC50 > 400 μM, SI > 6.46) were required to reduce the viral infection (Fig. 1a and Supplementary information, Fig. S1). However, favipiravir has been shown to be 100% effective in protecting mice against Ebola virus challenge, although its EC50 value in Vero E6 cells was as high as 67 μM, 4 suggesting further in vivo studies are recommended to evaluate this antiviral nucleoside. Nafamostat, a potent inhibitor of MERS-CoV, which prevents membrane fusion, was inhibitive against the 2019-nCoV infection (EC50 = 22.50 μM, CC50 > 100 μM, SI > 4.44). Nitazoxanide, a commercial antiprotozoal agent with an antiviral potential against a broad range of viruses including human and animal coronaviruses, inhibited the 2019-nCoV at a low-micromolar concentration (EC50 = 2.12 μM; CC50 > 35.53 μM; SI > 16.76). Further in vivo evaluation of this drug against 2019-nCoV infection is recommended. Notably, two compounds remdesivir (EC50 = 0.77 μM; CC50 > 100 μM; SI > 129.87) and chloroquine (EC50 = 1.13 μM; CC50 > 100 μM, SI > 88.50) potently blocked virus infection at low-micromolar concentration and showed high SI (Fig. 1a, b). Fig. 1 The antiviral activities of the test drugs against 2019-nCoV in vitro. a Vero E6 cells were infected with 2019-nCoV at an MOI of 0.05 in the treatment of different doses of the indicated antivirals for 48 h. The viral yield in the cell supernatant was then quantified by qRT-PCR. Cytotoxicity of these drugs to Vero E6 cells was measured by CCK-8 assays. The left and right Y-axis of the graphs represent mean % inhibition of virus yield and cytotoxicity of the drugs, respectively. The experiments were done in triplicates. b Immunofluorescence microscopy of virus infection upon treatment of remdesivir and chloroquine. Virus infection and drug treatment were performed as mentioned above. At 48 h p.i., the infected cells were fixed, and then probed with rabbit sera against the NP of a bat SARS-related CoV 2 as the primary antibody and Alexa 488-labeled goat anti-rabbit IgG (1:500; Abcam) as the secondary antibody, respectively. The nuclei were stained with Hoechst dye. Bars, 100 μm. c and d Time-of-addition experiment of remdesivir and chloroquine. For “Full-time” treatment, Vero E6 cells were pre-treated with the drugs for 1 h, and virus was then added to allow attachment for 2 h. Afterwards, the virus–drug mixture was removed, and the cells were cultured with drug-containing medium until the end of the experiment. For “Entry” treatment, the drugs were added to the cells for 1 h before viral attachment, and at 2 h p.i., the virus–drug mixture was replaced with fresh culture medium and maintained till the end of the experiment. For “Post-entry” experiment, drugs were added at 2 h p.i., and maintained until the end of the experiment. For all the experimental groups, cells were infected with 2019-nCoV at an MOI of 0.05, and virus yield in the infected cell supernatants was quantified by qRT-PCR c and NP expression in infected cells was analyzed by Western blot d at 14 h p.i. Remdesivir has been recently recognized as a promising antiviral drug against a wide array of RNA viruses (including SARS/MERS-CoV 5 ) infection in cultured cells, mice and nonhuman primate (NHP) models. It is currently under clinical development for the treatment of Ebola virus infection. 6 Remdesivir is an adenosine analogue, which incorporates into nascent viral RNA chains and results in pre-mature termination. 7 Our time-of-addition assay showed remdesivir functioned at a stage post virus entry (Fig. 1c, d), which is in agreement with its putative anti-viral mechanism as a nucleotide analogue. Warren et al. showed that in NHP model, intravenous administration of 10 mg/kg dose of remdesivir resulted in concomitant persistent levels of its active form in the blood (10 μM) and conferred 100% protection against Ebola virus infection. 7 Our data showed that EC90 value of remdesivir against 2019-nCoV in Vero E6 cells was 1.76 μM, suggesting its working concentration is likely to be achieved in NHP. Our preliminary data (Supplementary information, Fig. S2) showed that remdesivir also inhibited virus infection efficiently in a human cell line (human liver cancer Huh-7 cells), which is sensitive to 2019-nCoV. 2 Chloroquine, a widely-used anti-malarial and autoimmune disease drug, has recently been reported as a potential broad-spectrum antiviral drug. 8,9 Chloroquine is known to block virus infection by increasing endosomal pH required for virus/cell fusion, as well as interfering with the glycosylation of cellular receptors of SARS-CoV. 10 Our time-of-addition assay demonstrated that chloroquine functioned at both entry, and at post-entry stages of the 2019-nCoV infection in Vero E6 cells (Fig. 1c, d). Besides its antiviral activity, chloroquine has an immune-modulating activity, which may synergistically enhance its antiviral effect in vivo. Chloroquine is widely distributed in the whole body, including lung, after oral administration. The EC90 value of chloroquine against the 2019-nCoV in Vero E6 cells was 6.90 μM, which can be clinically achievable as demonstrated in the plasma of rheumatoid arthritis patients who received 500 mg administration. 11 Chloroquine is a cheap and a safe drug that has been used for more than 70 years and, therefore, it is potentially clinically applicable against the 2019-nCoV. Our findings reveal that remdesivir and chloroquine are highly effective in the control of 2019-nCoV infection in vitro. Since these compounds have been used in human patients with a safety track record and shown to be effective against various ailments, we suggest that they should be assessed in human patients suffering from the novel coronavirus disease. Supplementary information Supplementary information, Materials and Figures
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Clinical and immunologic features in severe and moderate Coronavirus Disease 2019

              Journal of Clinical Investigation
                Bookmark

                Author and article information

                Contributors
                haqsyedatiqul@gmail.com
                Journal
                Int J Rheum Dis
                Int J Rheum Dis
                10.1111/(ISSN)1756-185X
                APL
                International Journal of Rheumatic Diseases
                John Wiley and Sons Inc. (Hoboken )
                1756-1841
                1756-185X
                27 May 2020
                : 10.1111/1756-185X.13863
                Affiliations
                [ 1 ] Department of Medicine and Therapeutics The Chinese University of Hong Kong Hong Kong China
                [ 2 ] The First Department of Internal Medicine University of Occupational and Environmental Health Kitakyushu Japan
                [ 3 ] Apollo Indraprastha Hospitals New Delhi India
                [ 4 ] Division of Allergy, Immunology and Rheumatology Department of Internal Medicine School of Medicine College of Medicine Taipei Medical University Taipei Taiwan
                [ 5 ] Allergy, Arthritis & Rheumatism Clinic Farrer Park Hospital Singapore City Singapore
                [ 6 ] Yong Loo Lin School of Medicine National University of Singapore Singapore City Singapore
                [ 7 ] Deptartment of Rheumatology BSMMU Dhaka Bangladesh
                [ 8 ] Department of Rheumatology Key Laboratory of Rheumatology and Clinical Immunology Ministry of Education Peking Union Medical College & Chinese Academy of Medical Science Peking Union Medical College Hospital National Clinical Research Center for Dermatologic and Immunologic Diseases Ministry of Science & Technology Beijing China
                [ 9 ] Makati Medical Center Makati Philippines
                [ 10 ] Department of Internal Medicine Seoul National University Hospital Seoul South Korea
                [ 11 ] Laboratory for Autoimmune Diseases RIKEN Center for Integrative Medical Sciences Yokohama Japan
                [ 12 ] BSM Medical University Dhaka Bangladesh
                Author notes
                [*] [* ] Correspondence

                Syed Atiqul Haq, BSM Medical University, Dhaka, Bangladesh.

                Email: haqsyedatiqul@ 123456gmail.com

                Author information
                https://orcid.org/0000-0001-6410-8852
                https://orcid.org/0000-0002-0807-7139
                https://orcid.org/0000-0001-6685-4170
                https://orcid.org/0000-0002-5384-3437
                https://orcid.org/0000-0003-4154-7283
                Article
                APL13863
                10.1111/1756-185X.13863
                7283887
                32462761
                bf286212-af0f-4245-8006-38326f7876d7
                © 2020 Asia Pacific League of Associations for Rheumatology and John Wiley & Sons Australia, Ltd

                This article is being made freely available through PubMed Central as part of the COVID-19 public health emergency response. It can be used for unrestricted research re-use and analysis in any form or by any means with acknowledgement of the original source, for the duration of the public health emergency.

                History
                : 10 April 2020
                : 03 May 2020
                : 03 May 2020
                Page count
                Figures: 0, Tables: 0, Pages: 6, Words: 9864
                Categories
                Expert Comments
                Expert Comments
                Custom metadata
                2.0
                corrected-proof
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.8.4 mode:remove_FC converted:10.06.2020

                Rheumatology
                Rheumatology

                Comments

                Comment on this article