Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cytotherapy ; 25(6 Supplement):S239, 2023.
Article in English | EMBASE | ID: covidwho-20239698

ABSTRACT

Background & Aim: Immune checkpoint inhibitors (ICI) revolutionized solid tumor treatment, however, in many tumors only partial response is achieved. Allocetra-OTS has an immune modulating effect on macrophages and dendritic cells and showed an excellent safety profile in patients including patients with sepsis and Covid-19. Here we investigated the anti-tumoral effect of Allocetra-OTS cellular therapy, in peritoneal solid tumor animal models. Methods, Results & Conclusion(s): Allocetra-OTS is manufactured from enriched mononuclear fractions and induced to undergo early apoptosis. Balb/c mice were inoculated intraperitoneally (IP) with AB12 (mesothelioma) with pLenti-PGK-V5-Luc-Neo and treated with anti- CTLA4 with or without Allocetra-OTS. Mice were monitored daily for clinical score and weekly using IVIS (Fig.1). Kaplan-Meier log rank test was done for survival. For Allocetra-OTS preparation, enriched mononuclear fractions were collected by leukapheresis from healthy eligible human donors and induced to undergo early apoptosis. Anti- CTLA4 standalone therapy significantly improved survival (Fig.2) from mean 34+/-9 to 44.9 +/-20 days. However, OTS standalone therapy was non-inferior and improved survival to 52.3 +/-20 days. Anti-CTLA4 + Allocetra-OTS combination therapy, ameliorated survival to 86.7+/-20 days with complete cancer remission in 60-100% of mice. Similar anti- tumoral effects of Allocetra-OTS were seen in mesothelioma model in a combination therapy with either anti-PD1 or cisplatin and using anti-PD1 in ID8 ovary cancer model. Based on single cell analysis confirmed by flow cytometry and pathology, the mechanism of action seems to be related or at least associated with an increase in f/480high peritoneal macrophages and a decrease in recruited macrophages, and to f/480high infiltration of the tumor. However, further studies are needed to confirm these observations. During IP tumor progression, Allocetra-OTS as a standalone therapy or in combination with ICI, or cisplatin, significantly reduced tumor size and resulted in complete remission in up to 100% treated mice. Similar results were obtained in ID8 ovary cancer. Based on excellent safety profile in > 50 patients treated in prior clinical trials for sepsis and Covid-19, Phase I/II clinical trial of Allocetra-OTS plus chemotherapy has started and three patient already recruited. A second phase I/II clinical trial of Allocetra- OTS plus anti-PD1, as a second- and third-line therapy in various cancers, was initiated in Q1 2023. [Figure presented]Copyright © 2023 International Society for Cell & Gene Therapy

2.
Hla ; 101(4):364-365, 2023.
Article in English | EMBASE | ID: covidwho-2292141

ABSTRACT

We found a higher incidence of myocarditis in young males who had received Pfizer-BioNTech BNT162b2 vaccinations as compared with historical controls and unvaccinated individuals. The analyses focused on risk following the first and second vaccine in adults and adolescents, as well as risk in adults following the third (booster) vaccine. Males, mainly aged 12-30 years, were found to be at higher risk. However, the question remains what causes lead one specific young male, but not another, to develop post-vaccination myocarditis. The HLA molecule is known to play an important role in infectious and auto-inflammatory diseases. We hypothesized that differences in HLA alleles could lead to either protection or susceptibility to vaccination-induced myocarditis. On this basis, HLA typing was performed using next-generation sequencing technology for the HLA-A, -B, -C, -DRB1, -DQB1 and -DPB1 loci, in 21 wellcharacterized patients who developed myocarditis after the second Pfizer BNT162b2 vaccination. The HLA genotypes were compared with high-resolution HLA data of 272 healthy controls from the Hadassah Bone Marrow registry samples, who are representative of HLA frequencies in the Israeli population. Our findings demonstrated that in HLA class II, DRB1*14:01 (19.04% vs. 5.3%, Pcorr = 0.028, OR = 4.17), HLA-DQB1*05:03 (19.04% vs. 6.06%, Pcorr = 0.034, OR = 3.64) and DRB1*15:03 (7.14% vs. 0.0%, Pcorr = 0.003, OR = 41.76) were significantly associated with disease susceptibility. We further discovered susceptibility motifs in the HLA-DR peptidebinding grooves: His60 (Pcorr0.01, OR = 3.52) and Arg70 (Pcorr = 0.0047, OR = 3.43). Our findings suggest that immunogenetic fingerprints in HLA peptide-binding grooves may have changed the binding affinity of different peptides derived from the Pfizer-BioNTech BNT162b2 vaccination, and induced myocarditis.

3.
Autoimmunity, COVID-19, Post-COVID19 Syndrome and COVID-19 Vaccination ; : 427-436, 2022.
Article in English | Scopus | ID: covidwho-2272035

ABSTRACT

Postvaccination myocarditis was reported as early as 1957 in association with smallpox vaccination. Following Emergency Use Authorization of the Pfizer-BioNTech BNT162b2 vaccine by the FDA on December 11, 2020, approval was granted in Israel on December 20, 2020. A national vaccination campaign for novel coronavirus infection (COVID-19) using the Pfizer vaccine ensued in Israel. The vaccination campaign was based on a 2-dose regimen administered with a 21-day interval, and later a third (booster) vaccine was added. A unique nationwide active surveillance program with hospital reports of diagnoses of myocarditis and verification of vaccinations. Based on comparisons with historical (pre-COVID-19) incidence of myocarditis, observed-to-expected ratios were calculated. The results clearly demonstrated an association between vaccination and myocarditis, mainly in young males and mainly in the first week after the second vaccine. These studies are the only nationwide active surveillance studies performed to date and provide the most accurate estimates of myocarditis occurrence after the Pfizer-BioNTech BNT162b2 mRNA-based vaccination. © 2023 Elsevier Inc. All rights reserved.

4.
Autoimmunity, COVID-19, Post-COVID19 Syndrome and COVID-19 Vaccination ; : 647-672, 2022.
Article in English | Scopus | ID: covidwho-2263201

ABSTRACT

Treatment of COVID-19 can be categorized into prophylactic treatment, early-stage treatment, and late-stage treatment. Prophylactic treatment, as either pre or postexposure passive immunization with monoclonal antibodies, is currently limited to high-risk groups, with preexisting risk factors for severe disease and death in case of contracting COVID-19. Additional prophylactic treatment for hospitalized patients includes anticoagulation. In early treatment, when the infectious state is dominant, antiviral agents are used as well as passive immunization with monoclonal antibodies. Late-stage treatment in progressive and-inflammatory disease characterized by a cytokine storm and lung involvement in most severe/critical patients, includes corticosteroids, interluekin-6 inhibitors, and JAK inhibitors. Oxygen support is mandatory in severe patients and in patients with moderate to severe adult respiratory distress syndrome and refractory hypoxemia. Rescue procedures include protonation, alveolar recruitment maneuvers, neuromuscular blockade, pulmonary vasodilators, and extracorporeal membrane oxygenation. Additional potential treatments that have not been yet authorized are beyond the scope of this discussion. © 2023 Elsevier Inc. All rights reserved.

5.
Journal for ImmunoTherapy of Cancer ; 10(Supplement 2):A1432, 2022.
Article in English | EMBASE | ID: covidwho-2161960

ABSTRACT

Background Immune checkpoint inhibitors (ICI) revolutionized solid tumor treatment, however, in many tumors only partial response is achieved. Allocetra-OTS has an immune modulating effect on macrophages1 and showed an excellent safety profile in patients including patients with sepsis.2 Here we investigated the anti-tumoral effect of Allocetra-OTS cellular therapy, in solid tumor animal models. Methods Allocetra-OTS is manufactured from enriched mononuclear fractions and induced to undergo early apoptosis. In an immunecompetent model, Balb/c mice were inoculated intraperitoneally (IP) with AB12 (mesothelioma) with pLenti-PGK-V5-Luc-Neo and treated with anti-CTLA4 with or without Allocetra-OTS. Mice were monitored daily for clinical score and weekly using IVIS. Kaplan-Meier log rank test was done for survival. For Allocetra-OTS preparation, enriched mononuclear fractions were collected by leukapheresis from healthy eligible human donors and induced to undergo early apoptosis.To follow tumor growth in vivo, HeLa-CD19 cells were stably transduced with pLenti-PGK-V5-Luc-Neo. For CAR preparation, fresh mononuclear cells (MNC) were transfected with CD19-CAR plasmids. SCID-Bg mice were injected IP with human HeLa-CD19 or HeLa-CD19-luciferase cells, 10x10 allocetraOTS or vehicle, and 10x10 CD19-CAR T cells or mock T cells. Results In immune competent Balb/c mesothelioma model, anti-CTLA4 standalone therapy significantly improved survival from mean 34+/-9 to 44.9 +/-20 days (p<0.05). Similarly, Allocetra- OTS standalone therapy improved survival to 52.3 +/-20 days (p<0.02). However, anti-CTLA4 + Allocetra-OTS combination therapy, ameliorated survival to 86.7+/-20 days (p<0.0001) with complete cancer remission in 60-100% of mice (figure 1 & 2). Similar anti-tumoral effects of Allocetra- OTS were seen in mesothelioma model in a combination therapy with either anti-PD1 or cisplatin. In the CAR-T model, SCID-Bg mice were sacrificed or died from tumor progression in 30+/-5 days (range 27-37). CAR T cell therapy significantly improved survival to 55+/-11 days (p < 0.05 vs MOCK) but Alloctra-OTS further improved survival to 75+/-10 (p < 0.001) with 20-40% complete remission. Conclusions During IP tumor progression, Allocetra-OTS as a standalone therapy or in combination with ICI, cisplatin or CAR-T therapy, significantly reduced tumor size and resulted in complete remission in up to100% treated mice. Based on excellent safety profile in > 40 patients treated in prior clinical trials for sepsis and Covid-19, Phase I/II clinical trial of Allocetra-OTS plus chemotherapy has started and first patient already recruited. A second Phase I/II clinical trial of Allocetra-OTS plus anti-PD1, as a second- and third-line therapy in various cancers, is planned for Q4 2022.

6.
Annals of Oncology ; 33:S759, 2022.
Article in English | EMBASE | ID: covidwho-2041531

ABSTRACT

Background: ICI revolutionized solid tumor treatment, however, in many tumors only partial response is achieved. Here we investigated the anti-tumoral effect of Allocetra—OTS cellular therapy, in solid tumor models. Methods: Allocetra-OTS is manufactured from enriched mononuclear fractions and induced to undergo early apoptosis.Balb/c mice were inoculated in the peritoneal cavity with AB12 (mesothelioma) stably transduced with pLenti-PGK-V5-Luc-Neo for IVIS visualization and treated with anti-CTLA4, anti-PD1, or cisplatin, with or without Allocetra-OTS (also administered as monotherapy). Kaplan-Meier log rank test was done for survival. CAR T model was induced in SCID-Bg mice were inoculated intraperitoneally with human HeLa-CD19, followed by treatment with 10×109 cells of Allocetra-OTS or vehicle, and 1×107 CD19-CAR T cells or mock T cells. Results: Anti-CTLA4 therapy significantly improved survival from mean 34±9 to 44.9 ±20 days (p<0.05). However, Allocetra-OTS monotherapy improved survival to 52.3 ±20 days (p<0.02) and anti-CTLA4 + Allocetra-OTS combination therapy to 86.7±20 days (p<0.0001) with complete cancer remission in 60-100% of mice. Similar results were seen in combination therapy with either anti-PD1 or cisplatin. In the CAR-T model, SCID-Bg mice survived 30±5 days (range 27–37) and were sacrificed or died from tumor progression. Results were verified using IVIS of intraperitoneal HeLaCD19-Luc cells. CAR T cell therapy significantly improved survival to 55±11 days (p < 0.05 vs MOCK) but Alloctra-OTS further improved survival to 75±10 (p < 0.001) with 20-40% complete remission. Conclusions: During intraperitoneal tumor progression, Allocetra-OTS as monotherapy or in combination with ICI, cisplatin or CAR-T therapy, significantly reduced tumor size and resulted in complete remission in up to100% treated mice. Based on excellent safety profile in > 40 patients treated in prior clinical trials for sepsis and COVID-19, phase I/II clinical trial of Allocetra-OTS plus chemotherapy is planned for Q3 2022, and a second phase I/II clinical trial of Allocetra-OTS plus anti-PD1, as a second- and third-line therapy in various cancers, is planned for Q4 2022. Legal entity responsible for the study: The authors. Funding: Enlivex Therapeutics Ltd. Disclosure: D. Mevorach: Financial Interests, Personal, Royalties, Founder & CSO: Enlivex Therapeutics LTD. E. Yalon, E. Regev, C. Ankri, O. Hershkovitz, Y. Shabat, B. Reicher: Financial Interests, Personal, Full or part-time Employment: Enlivex Therapeutics LTD.

7.
Annals of the Rheumatic Diseases ; 81:962, 2022.
Article in English | EMBASE | ID: covidwho-2009063

ABSTRACT

Background: AIRD patients (pts) may be more susceptible to severe COVID19. Objectives: To determine the risk factors for severe COVID19 and the effect of vaccinations among AIRD pts followed at dedicated rheumatology clinics. Methods: At the onset of the pandemic, we established a national registry of AIRD pts, diagnosed with COVID19, based on voluntary reporting by the treating rheumatologist. 12 centers from Israel participated in the study. COVID19 was confrmed by a positive SARS CoV2 PCR. The indications for PCR testing were clinical symptoms or close contact with an infected person. Severe illness was defned by SpO2 <94% in room air, respiratory rate of >30 breaths/min, PaO2/FiO2 <300 mm Hg, or lung infltrates >50% on imaging. The registry included demographic data, AIRD diagnosis and duration, visceral involvement, co-morbidities, immunomodulatory treatment, date of diagnosis and severity of COVID19 disease, management, complications, duration of hospitalization, the dates of the mRNA vaccinations, lab results and outcome. We analyzed data from 1.3.2020 to 30.11.2021 Results: During the study period we experienced 4 outbreaks of COVID19 infection. Initially social distancing, followed by a lockdown were imposed. The low number of cases led to relaxation of the measures. Two more severe outbreaks followed, which triggered 2 new lockdowns. The 3rd outbreak ended almost 2 months after vaccination started (BNT162b2 mRNA COVID19 vaccine). From March 1st 2020 to April 30, 2021, 298 AIRD pts (70.8% females, mean (SD) age 53.3(15.3)) with confrmed COVID19 infection were included. 43.3%(129) had visceral involvement due to the AIRD. 58.7%(175 pts) were on conventional synthetic disease modifying drugs (csDMARDs), 44.6% (133) on biologic/targeted DMARDs and 40% (120) on prednisone. Almost 2/3 of pts had at least one comorbidity. In a multivariate logistic regression analysis age, AIRD with pulmonary involvement, diabetes and treatment with prednisone, mycophenolate mofetil or JAK inhibitors were associated with hospitalization. Older age, renal and vascular involvement due to the AIRD, and congestive heart failure were associated with higher mortality. The 4th outbreak occurred 6 months after the introduction of vaccines, with spreading of the delta variant: 110 AIRD pts with COVID19 were recorded. Demographic data, clinical AIRD's characteristics, immunomodulatory treatment and comorbidities were similar to the previous outbreaks. However, during the 4th outbreak, the proportion of pts with severe COVID19, the hospitalization and mortality rate were signifcantly lower as compared to the frst 3 outbreaks (15% vs 24%, 27% vs 53%, 6.7% vs 9.1%, respectively). Among COVID19 pts, 25% received a 3rd vaccine dose (booster), 56% contracted infection more than 5 months after the 2nd vaccine dose and 24% were unvaccinated. Most of the pts who received the booster contracted the disease within a week of vaccination. The odds ratio for hospitalization in vaccinated pts compared to unvaccinated was 0.11 (0.01-0.63 95% CI, p=0.041) in those vaccinated within the previous 1-5 months, and 0.38 (0.21-0.67 95% CI, p=0.001) in those vaccinated more than 6 months ago. 9 pts died, 5 were more than 6 months after the 2nd mRNA vaccine, 2 were unvaccinated and 1 patient received the booster on the same day of COVID19 diagnosis. Conclusion: Before the vaccination campaign, the hospitalization and mortality rate in our cohort were similar to the data reported by other registries. COVID19 tends to be more severe, with increased mortality in patients with active AIIRD and visceral involvement (pulmonary, cardiac, renal), advanced age and co-morbidities. The delta outbreak occured 6 months after the implementation of vaccinations and was associated with signifcantly lower hospitalization and mortality rates, despite the increased aggressiveness of the variant. Vaccination of AIIRD pts with 3 doses of mRNA vaccines protects from severe COVID19 disease, hospitalization, and death.

8.
Journal of Clinical Oncology ; 40(16), 2022.
Article in English | EMBASE | ID: covidwho-2005652

ABSTRACT

Background: Chimeric antigen receptor (CAR) T cells can activate an immune response to a cancer-specific antigen but is less effective in solid tumors. Immune check point inhibitors (ICI) revolutionized the treatment of solid tumors, however, in many tumors only partial response is achieved. Here we questioned the role of synergistic effect of Allocetra-OTS (cellular therapy for in-vivo reprogramming macrophages and dendritic cells, Enlivex Therap.) on solid tumor progression. Methods: To follow tumor growth in vivo, HeLa-CD19 cells were stably transduced with pLenti-PGK-V5-Luc-Neo. For CAR preparation, fresh mononuclear cells (MNC) were transfected with CD19-CAR plasmids. For the intraperitoneal solid tumor model, SCID-Bg mice were injected intraperitoneally (IP) with human HeLa- CD19 or HeLa-CD19-luciferase cells, 10×106 allocetra-OTS or vehicle, and 10×106 CD19-CAR T cells or mock T cells. In an immune-competent model, Balb/c mice were treated IP with AB12 (mesothelioma) with pLenti-PGK-V5-Luc-Neo and treated with anti-CTLA4 with or without Allocetra-OTS. Mice were monitored daily for clinical signs and peritoneal fluid accumulation and weekly for tumor growth. Kaplan-Meier log rank test was done for survival. Peritoneal cells were evaluated using single cell analysis and flow cytometry. Tumors were examined for bacterial presence by immunohistochemistry staining with antilipoteichoic acid (LTA) and antilipopolysaccharide (LPS). For allocetra-OTS preparation, enriched mononuclear fractions were collected by leukapheresis from healthy eligible human donors and induced to undergo early apoptosis. Results: SCID mice survived 30±5 days (range 27-37) and were sacrificed or died from solid tumor in the peritoneal cavity after accumulation of bloody peritoneal fluid and clinical deterioration. Results were verified using IVIS of intraperitoneal HeLaCD19- Luc cells. CAR T cell therapy significantly ameliorated survival to 55±11 days (p < 0.05 vs MOCK) but Alloctra-OTS further ameliorated survival to 75±10 (p < 0.001) with 20-40% complete remission. In AB12 model, anti CTLA4 therapy significantly ameliorated survival from 26±5 to 38 ±9 days (p < 0.05). However, Allocetra-OTS monotherapy ameliorated survival to 45 ±12 days (p < 0.02) and combinational therapy to 75±9 days (p < 0.0001) with complete remission in 60-75% of mice. Single cell analysis revealed that restoration of large peritoneal macrophages (LPM), were associated with antitumor activity. Conclusions: During intraperitoneal tumor progression, allocetra-OTS as monotherapy or combinational therapy with CAR or anti-CTLA4 significantly reduced tumor size and enable complete remission in up to 75% treated mice. Based on excellent safety profile in > 30 patients treated for sepsis and Covid19, human phase I/II of allocetra-OTS plus ICI, for peritoneal metastases, is planned for 2022.

9.
Cytotherapy ; 24(5):S121-S122, 2022.
Article in English | EMBASE | ID: covidwho-1996728

ABSTRACT

Background & Aim: COVID-19 has become pandemic, with mortality estimated between 1–4% )in alpha and delta variants) and complications among hospitalized patients resulting in up to 15–25% of inpatients being admitted to the intensive care unit (ICU). Two studies (one of 5 and and of 16 patients), were designed to determine the safety and efficacy of treatment with Allocetra-OTS for reprogramming macrophages and resolution of cytoikine storm in patients with severe/critical COVID-19. The two studies were approved by the Ministry of Health’s (MOH) Ethical Committee. Methods, Results & Conclusion: Methods: 2 clinical studies were done in three medical centres in Israel. Patient inclusion criteria was mainly severe or critical condition by NIH criteria. Further details are found in NCT04590053 and NCT04513470.18/21 had significant ARDS. An enriched MNC fraction was collected via leukapheresis from healthy, eligible human donors, prepared by Enlivex Ltd. and gamma irradiated. Apoptosis and viability of apoptotic cells were determined using Annexin V- and PI staining (Medical & Biological Laboratories, Nagoya, Aichi, Japan). Lack of proliferation was shown using CFSE and bead stimulation. Every patient recived one dose of 109 cells. Results. administration of Allocetra-OTS in 21 patients (11 severe and 10 critical with non ivasive oxygen support) with severe-to-critical Covid-19, of whom was safe with 5 unrelated SAEs. Allocetra-OTS was well tolerated when given in conjunction with standard therapy (remdesivir, enoxaparin, and dexamethasone) and showed early recovery;5.5 days in average till discharge. 2/21 patients were still hospitalized by (Figure Presented) day 28 (end of study).18 patients had mild-to severe ARDS and 16/18 (88.8%) completely recovered within few days. The cytokine storm was resolved in all discharged patients as shown by laboratory and 30 cytokine/chemokine measurements (Fig. 1 shows pro-inflammatory cytokines). Conclusion: Allocetra showed excellent safty profile and promising results regarding resolution of inflammation and respiratory failure, A double blind large comparative study in this population is now recruiting patients in 3 countries.

10.
Cytotherapy (Elsevier Inc.) ; 24(5):S121-S122, 2022.
Article in English | Academic Search Complete | ID: covidwho-1783863
11.
Circulation ; 144(25):E578-E578, 2021.
Article in English | Web of Science | ID: covidwho-1663212
12.
Annals of the Rheumatic Diseases ; 80(SUPPL 1):892, 2021.
Article in English | EMBASE | ID: covidwho-1358785

ABSTRACT

Background: The epidemiology of COVID19 among patients with AIIRD may be influenced by a dysregulated immune system, immunosuppressive therapies and behavioral patterns. Data regarding the epidemiology of COVID19 among patients with AIIRD is scarce. Objectives: To assess the pattern of COVID19 pandemic among patients with AIIRD compared to the general population in Israel Methods: At the beginning of the COVID-19 pandemic, we established a national registry of patients with AIIRD, diagnosed with COVID-19, based on voluntary reporting by the treating rheumatologist. All the members of the Israeli Society of Rheumatology were encouraged to participate and repeatedly reminded to report any new cases. Rheumatology centers from 11 hospitals from the Northern and Central part of Israel participated in this study. The registry included demographic data, AIIRD diagnosis and duration, systemic organ involvement, co-morbidities, treatment (conventional synthetic disease modifying drugs (csDMARDs), biologic/targeted (b/ts) DMARDs, corticosteroids use, dose and treatment duration, date of COVID19 diagnosis, severity of the viral disease and complications, duration of hospitalization, if required, treatment for COVID 19, laboratory results and outcome. The diagnosis of COVID 19 was made by a positive SARS CoV2 PCR. The indications for SARS CoV2 PCR testing in Israel comprise clinical symptoms or exposure to a confirmed close contact. Severe illness was defined by SpO2 <94% in room air, respiratory rate of ≥30 breaths/min, PaO2/FiO2 <300 mm Hg, or lung infiltrates ≥50% on chest imaging. The epidemiological data regarding the number of COVID19 confirmed patients, the number of severe cases and the rate of mortality among the general population per day and per week, were extracted from the data dashboard of the Israeli Ministry of Health. We analyzed data from 02.2020 to 15.01.2021. Results: During the study period we experienced 3 waves of COVID 19 pandemic. The governmental management of COVID19 spread, at the beginning of the pandemic, included inforcement of severe travel restrictions and social distancing, followed eventually by a preventive lockdown, in spite of the relatively low number of cases. Easing of the restrictions, lifting the travel ban, opening of the commerce and schools led to 2 much more severe waves, which triggered 2 new lockdowns. Up to January 2021, 549763 Israelis had confirmed COVID19, 30% of whom had severe disease, 0.84% died (30% of the patients with severe disease). We identified 190 AIIRD patients (mean(SD) age 52(18), 30% males) who had confirmed COVID19. The weekly incidence curve of patients with rheumatic diseases correlated with the curve of the general population (Figure 1). Sixty-one % of the patients with AIIRD received csDMARDs, 41% were on b/ tsDMARDs, 39% on chronic corticosteroids, 12% on ≥10mg prednisone. Forty-seven% of patients required hospitalization, 20% had severe COVID19. Sixteen patients (42% of patients with severe COVID19) (mean(SD), median age 64.7(15.4),67)) died (systemic sclerosis-4 patients, rheumatoid arthritis -6, systemic lupus erythematosus -2, antiphospholipid syndrome-2, granulomatous polyangiitis -1, polymyalgia rheumatica-1). The AIIRD was active in 56% of them, 50% received csDMARDs, none of them were on b/tsDMARDs, 31% received chronic prednisone≥10 mg. All patients who died had at least 2 comorbidities. Conclusion: The pattern of spread of COVID19 in AIIRD patients is similar to the general population despite repeated mass media alerts for enhanced social distancing for elderly and immune suppressed patients. The disease tends to be more severe with enhanced mortality, especially in those with active AIIRD disease and organ involvement (lungs, heart, renal), older age and co-morbidities. A reporting bias cannot be excluded.

13.
Arthritis & Rheumatology ; 72:2, 2020.
Article in English | Web of Science | ID: covidwho-1017536
SELECTION OF CITATIONS
SEARCH DETAIL