Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int J Infect Dis ; 125: 233-240, 2022 Oct 31.
Article in English | MEDLINE | ID: covidwho-2131128

ABSTRACT

OBJECTIVES: Our aim was to compare outcomes of hospitalized adults with severe COVID-19 and cytokine storm treated with tocilizumab or baricitinib. METHODS: A prospective, investigational, real-world study was performed from April 2020 to April 2021 at our center. COVID-19 severity was classified by World Health Organization criteria, and cytokine storm was documented along predefined criteria. Eligible patients were enrolled at diagnosis if they fulfilled a priori inclusion criteria and received standard-of-care plus tocilizumab or baricitinib for >48 hours. Patients were followed per protocol for 28 days post-diagnosis. The primary outcome was all-cause mortality; secondary outcomes were invasive mechanical ventilation and major infectious complications. RESULTS: Of 463 patients, 102/463 (22.1%) received tocilizumab, and 361/463 (77.9%) baricitinib. Baseline characteristics were balanced. At 28 days, there was no difference in all-cause mortality (22/102, 21.6% vs 64/361, 17.7%; P-value = 0.38). Requirement for invasive mechanical ventilation was more frequent after tocilizumab (52/102, 50.9% vs 96/361, 26.6%; P <0.01), rate of major infectious complications was similar (32/102, 31.4% vs 96/361, 26.6%; P-value = 0.34). In logistic regression, the immunomodulatory drug was not retained as a predictor of all-cause mortality. Kaplan-Meier analysis revealed statistically similar survival distributions. CONCLUSION: All-cause mortality was similar between adults treated with baricitinib or tocilizumab for severe COVID-19 with cytokine storm.

2.
Front Immunol ; 13: 907125, 2022.
Article in English | MEDLINE | ID: covidwho-2123412

ABSTRACT

Common variable immunodeficiency (CVID) patients have markedly decreased immune response to vaccinations. In this study we evaluated humoral and T cell-mediated responses against severe acute respiratory syndrome coronavirus-2 (SARS-Cov-2) with additional flow cytometric changes in CVID patients receiving booster vaccination with BNT162b2 after two ChAdOx1 nCoV-19. The BNT162b2 vaccine raised the anti-spike protein S immunoglobulin G over the cut-off value from 70% to 83% in CVID, anti-neutralizing antibody had been raised over a cut-off value from 70% to 80% but levels after boosting were significantly less in both tests than in healthy controls (*p=0.02; **p=0.009 respectively). Anti-SARS-CoV-2 immunoglobulin A became less positive in CVID after boosting, but the difference was not significant. The cumulative interferon-γ positive T cell response by ELISpot was over the cut-off value in 53% of the tested individuals and raised to 83% after boosting. This and flow cytometric control of cumulative CD4+ and CD8+ virus-specific T cell absolute counts in CVID were also statistically not different from healthy individuals after boosting. Additional flow cytometric measures for CD45+ lymphocytes, CD3+, and CD19+ cells have not shown significant differences from controls except for lower CD4+T cell counts at both time points (**p=0.003; **p=0.002), in parallel CD4+ virus-specific T-cell ratio was significantly lower in CVID patients at the first time point (*p: 0.03). After boosting, in more than 33% of both CVID patients and also in their healthy controls we detected a decrease in absolute CD45+, CD3+, CD3+CD4+, and CD3+CD8+, CD19+, and CD16+56+ cell counts. CD16+CD56+ cell counts were significantly lower compared to controls before and after boosting (*p=0.02, *p=0.02). CVID patients receiving immunosuppressive therapy throughout the previous year or autologous stem cell transplantation two years before vaccination had worse responses in anti-spike, anti-neutralizing antibody, CD3+CD4+T, CD19+ B, and natural killer cell counts than the whole CVID group. Vaccinations had few side effects. Based on these data, CVID patients receiving booster vaccination with BNT162b2 after two ChadOx1 can effectively elevate the levels of protection against COVID-19 infection, but the duration of the immune response together with COVID-19 morbidity data needs further investigation among these patients.


Subject(s)
COVID-19 , Common Variable Immunodeficiency , Hematopoietic Stem Cell Transplantation , Adaptor Proteins, Signal Transducing , Antigens, CD19 , BNT162 Vaccine , ChAdOx1 nCoV-19 , Humans , Immunoglobulin G , SARS-CoV-2 , T-Lymphocytes , Transplantation, Autologous
3.
EJIFCC ; 33(2):187-193, 2022.
Article in English | EuropePMC | ID: covidwho-2092871

ABSTRACT

Patients suffering from malignant diseases have a high risk of developing severe or critical forms of COVID-19 (Coronavirus Disease 2019). Chronic lymphocytic leukaemia (CLL) is characterized by dysregulated adaptive and innate immune responses, because both T and B cells, the function of phagocytes and the activity of the complement system may be affected. Severe SARS-CoV-2 infection also influences the immunological functions mainly via causing the depletion of CD4+ and CD8+ T cells. We present the cases of two patients, whose de novo CLL were observed during severe COVID-19 pneumonia. A 43-year-old man with IDDM (Insulin dependent diabetes mellitus) was sent to hospital in February 2021. He had a bilateral severe COVID-19 pneumonia. There was a suspected sign of malignancy on a thoracic vertebra in his chest CT, and haematological consultation was requested. In parallel, a 53-year-old man was hospitalized in March of 2021 because of severe COVID-19 pneumonia. CLL was suspected based on his haematology test results (WBC: 123 G/L, lymphocytes: 91%, haemoglobin: 107 g/L). Flow cytometric analysis revealed CLL in both cases. Based on the result of the molecular genetic tests, the first patient had a good prognosis in Rai 0 stage, while the other patient suffered from Rai I stage with a worse prognosis. Both patients recovered from bilateral COVID-19 pneumonia without the need for intensive care unit treatment. The follow-up of these CLL patients that manifested during symptomatic COVID-19 disease further enriched our knowledge on such clinical conditions where the immune system is dysfunctional due to different simultaneous causes.

4.
International journal of infectious diseases : IJID : official publication of the International Society for Infectious Diseases ; 2022.
Article in English | EuropePMC | ID: covidwho-2092649

ABSTRACT

Objectives Our aim was to compare outcomes of hospitalized adults with severe COVID-19 and cytokine storm, treated with tocilizumab or baricitinib. Methods A prospective, investigational, real-world study was performed between April 2020–April 2021 at our centre. COVID-19 severity was classified by World Health Organization criteria, cytokine storm was documented along pre-defined criteria. Eligible patients were enrolled at diagnosis if they fulfilled a priori inclusion criteria and received standard-of-care plus tocilizumab or baricitinib for >48 hours. Patients were followed per protocol for 28 days post-diagnosis. Primary outcome was all-cause mortality, secondary outcomes were invasive mechanical ventilation and major infectious complications. Results From 463 patients, 102/463 (22.1%) received tocilizumab, 361/463 (77.9%) baricitinib. Baseline characteristics were balanced. At 28 days, there was no difference in all-cause mortality (22/102, 21.6% vs. 64/361, 17.7%;p=0.38). Requirement for invasive mechanical ventilation was more frequent after tocilizumab (52/102, 50.9% vs. 96/361, 26.6%;p<0.01), rate of major infectious complications was similar (32/102, 31.4% vs. 96/361, 26.6%;p=0.34). In logistic regression, immunomodulatory drug was not retained as a predictor of all-cause mortality. Kaplan–Meier analysis revealed statistically similar survival distributions. Conclusions All-cause mortality was similar between adults treated with baricitinib or tocilizumab for severe COVID-19 with cytokine storm.

5.
Frontiers in immunology ; 13, 2022.
Article in English | EuropePMC | ID: covidwho-1918657

ABSTRACT

Common variable immunodeficiency (CVID) patients have markedly decreased immune response to vaccinations. In this study we evaluated humoral and T cell-mediated responses against severe acute respiratory syndrome coronavirus-2 (SARS-Cov-2) with additional flow cytometric changes in CVID patients receiving booster vaccination with BNT162b2 after two ChAdOx1 nCoV-19. The BNT162b2 vaccine raised the anti-spike protein S immunoglobulin G over the cut-off value from 70% to 83% in CVID, anti-neutralizing antibody had been raised over a cut-off value from 70% to 80% but levels after boosting were significantly less in both tests than in healthy controls (*p=0.02;**p=0.009 respectively). Anti-SARS-CoV-2 immunoglobulin A became less positive in CVID after boosting, but the difference was not significant. The cumulative interferon-γ positive T cell response by ELISpot was over the cut-off value in 53% of the tested individuals and raised to 83% after boosting. This and flow cytometric control of cumulative CD4+ and CD8+ virus-specific T cell absolute counts in CVID were also statistically not different from healthy individuals after boosting. Additional flow cytometric measures for CD45+ lymphocytes, CD3+, and CD19+ cells have not shown significant differences from controls except for lower CD4+T cell counts at both time points (**p=0.003;**p=0.002), in parallel CD4+ virus-specific T-cell ratio was significantly lower in CVID patients at the first time point (*p: 0.03). After boosting, in more than 33% of both CVID patients and also in their healthy controls we detected a decrease in absolute CD45+, CD3+, CD3+CD4+, and CD3+CD8+, CD19+, and CD16+56+ cell counts. CD16+CD56+ cell counts were significantly lower compared to controls before and after boosting (*p=0.02, *p=0.02). CVID patients receiving immunosuppressive therapy throughout the previous year or autologous stem cell transplantation two years before vaccination had worse responses in anti-spike, anti-neutralizing antibody, CD3+CD4+T, CD19+ B, and natural killer cell counts than the whole CVID group. Vaccinations had few side effects. Based on these data, CVID patients receiving booster vaccination with BNT162b2 after two ChadOx1 can effectively elevate the levels of protection against COVID-19 infection, but the duration of the immune response together with COVID-19 morbidity data needs further investigation among these patients.

6.
Orv Hetil ; 163(20): 774-787, 2022 May 15.
Article in English | MEDLINE | ID: covidwho-1896505

ABSTRACT

Coronavirus disease 2019 (COVID-19) displays tremendous inter-individual variability, ranging from asymptomatic infections to life-threatening illness. Although more studies are needed, a picture has begun to emerge that variability in the immune system components is a main contributor to the heterogeneous disease courses. Here, we provide a concept for the interaction of the innate and adaptive immune systems with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to link the observations that have been made during the first two years of the pandemic. Inborn errors of, and autoantibodies directed against, type I interferons, dysregulated myeloid response, hyperinflammation, lymphopenia, lymphocyte impairment, and heterogeneous adaptive immunity to SARS-CoV-2 are discussed, as well as their impact in the course of COVID-19. In addition, we will also review part of the key findings that have helped define and delineate some of the essential attributes of SARS-CoV-2-specific humoral and cell-mediated immune memory. Orv Hetil. 2022; 163(20): 774-787.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Pandemics
7.
Vaccines (Basel) ; 10(4)2022 Mar 30.
Article in English | MEDLINE | ID: covidwho-1822474

ABSTRACT

In the present study, antibody and T cell-mediated immune responses elicited by BBIBP-CorV and BNT162b2 vaccines were compared 6 months after the two-dose immunization of healthy individuals. Additionally, antibody and T cell responses after the third dose of BBIBP-CorV or BNT162b2 were compared using a homologous or heterologous vaccination strategy. The third dose was consistently administered 6 months after the second dose. Six months following the two-dose vaccination, the cumulative IFNγ-positive T cell response was almost identical in participants immunized with either two doses of BNT162b2 or BBIBP-CorV vaccines; however, significant differences were revealed regarding humoral immunity: the two-dose BNT162b2 vaccine maintained a significantly higher antireceptor-binding domain (RBD) IgG, anti-spike (S1/S2) IgG, and IgA antibody levels. The BNT162b2 + BNT162b2 + BBIBP-CorV vaccine series elicited significantly lower anti-RBD IgG and anti-S1/S2 IgG levels than three doses of BNT162b2, while the anti-S IgA level was equally negligible in both groups. Importantly, the cumulative IFNγ-positive T cell response was highly similar in both groups. Surprisingly, the BBIBP-CorV + BBIBP-CorV + BNT162b2 vaccination series provided a much higher cumulative IFNγ-positive T cell response than that elicited by three doses of BNT162b2; moreover, the levels of anti-RBD IgG and anti-S IgA were almost identical. Only the mean anti-S1/S2 IgG levels were higher after receiving three mRNA vaccines. Based on these data, we can conclude that administering a third dose of BNT162b2 after two doses of BBIBP-CorV is an effective strategy to significantly enhance both humoral and T cell-mediated immune response, and its effectiveness is comparable to that of three BNT162b2 vaccines.

8.
Orv Hetil ; 162(52): 2071-2078, 2021 12 26.
Article in Hungarian | MEDLINE | ID: covidwho-1599870

ABSTRACT

Összefoglaló. Bár a SARS-CoV-2-pandémia próbára tette a diagnosztikus kapacitásokat, számos hasznos tapasztalattal is szolgált, melyek alacsonyabb mintaszám mellett nem lettek volna levonhatók. Míg korábban a PCR-vizsgálatok jellemzoen diagnosztikus, illetve kvantitatív követési célokat szolgáltak, a járvány során többségbe kerültek a szuro- és (kezdetben) a felszabadító vizsgálatok. Jól követheto volt, hogy a tesztek piacra juttatásának eroltetett üteme sokszor nem tette lehetové a teljesen kiforrott koncepciók létrehozását. Tekintettel arra, hogy a molekuláris diagnosztika során nem teljes vírusgenomokat, hanem célszakaszokat mutatunk ki, amelyek aránya a fertozés egyes szakaszaiban nem feltétlenül állandó, egyre valószínubb, hogy nem azonos célgének a legmegfelelobbek diagnosztikus, szuro- és felszabadító vizsgálatokhoz. A nagy mennyiségu, aspecifikusan végzett vizsgálat még kiváló fajlagosság mellett is a pozitív prediktív érték csökkenéséhez vezethet, amennyiben a fertozés tényleges prevalenciája a vizsgálati csoportban alacsony. Munkánkban megkíséreljük irodalmi és saját adatok felhasználásával összefoglalni az elmúlt két év fontosabb diagnosztikus tapasztalatait a teljesség igénye nélkül. Orv Hetil. 2021; 162(52): 2071-2078. Summary. Although the SARS-CoV-2 pandemic has been a great challenge for the diagnostic capacities, it also proved to be a unique source of experience. While previously PCR tests had overwhelmingly been used for targeted diagnostic and quantitative follow-up testing, screening and (initially) release tests became far more prevalent during the pandemic. It was well to be seen that the forced pace of bringing tests to market often gave way to not fully mature concepts. The PCR method is based on the detection of sequences, the proportions of which are likely to alter throughout the course of the disease. It is becoming increasingly clear that different target genes might be the best suitable for diagnostic, screening and release testing. Even with specific assays, an unprecedentedly high number of tests might result in the inflation of the positive predictive value, when the true prevalence of the infection remains very low among the tested individuals. Here we try to summarize some of the potentially most relevant diagnostic conclusions of the pandemic so far according to our own data and the literature. Orv Hetil. 2021; 162(52): 2071-2078.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Pandemics , Pathology, Molecular
9.
Int J Infect Dis ; 115: 8-16, 2022 Feb.
Article in English | MEDLINE | ID: covidwho-1536601

ABSTRACT

OBJECTIVES: Angiotensin-converting enzyme 2 (ACE2) represents the primary receptor for SARS-CoV-2 to enter endothelial cells. Here we investigated circulating ACE2 activity to predict the severity and mortality of COVID-19. METHODS: Serum ACE2 activity was measured in COVID-19 (110 critically ill and 66 severely ill subjects at hospital admission and 106 follow-up samples) and in 32 non-COVID-19 severe sepsis patients. Associations between ACE2, inflammation-dependent biomarkers, pre-existing comorbidities, and clinical outcomes were studied. RESULTS: Initial ACE2 activity was significantly higher in critically ill COVID-19 patients (54.4 [36.7-90.8] mU/L) than in severe COVID-19 (34.5 [25.2-48.7] mU/L; P<0.0001) and non-COVID-19 sepsis patients (40.9 [21.4-65.7] mU/L; P=0.0260) regardless of comorbidities. Circulating ACE2 activity correlated with inflammatory biomarkers and was further elevated during the hospital stay in critically ill patients. Based on ROC-curve analysis and logistic regression test, baseline ACE2 independently indicated the severity of COVID-19 with an AUC value of 0.701 (95% CI [0.621-0.781], P<0.0001). Furthermore, non-survivors showed higher serum ACE2 activity vs. survivors at hospital admission (P<0.0001). Finally, high ACE2 activity (≥45.4 mU/L) predicted a higher risk (65 vs. 37%) for 30-day mortality (Log-Rank P<0.0001). CONCLUSIONS: Serum ACE2 activity correlates with COVID-19 severity and predicts mortality.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Angiotensin-Converting Enzyme 2/blood , COVID-19/diagnosis , COVID-19/mortality , Endothelial Cells , Humans , Severity of Illness Index
10.
Acta Microbiol Immunol Hung ; 2021 Nov 19.
Article in English | MEDLINE | ID: covidwho-1526708

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (HSCT) and coronavirus disease 2019 (COVID-19) infection can both lead to severe cytokine release syndrome (sCRS) resulting in critical illness and death. In this single institution, preliminary comparative case-series study we compared clinical and laboratory co-variates as well as response to tocilizumab (TCZ)-based therapy of 15 allogeneic-HSCT- and 17 COVID-19-associated sCRS patients. Reaction to a TCZ plus posttransplant cyclophosphamide (PTCY) consolidation therapy in the allogeneic-HSCT-associated sCRS group yielded significantly inferior long-term outcome as compared to TCZ-based therapy in the COVID-19-associated group (P = 0.003). We report that a TCZ followed by consolidation therapy with a Janus kinase/signal transducer and activator of transcription (JAK/STAT) inhibitor given to 4 out of 8 critically ill COVID-19 patients resulted in their complete recovery. Non-selective JAK/STAT inhibitors influencing the action of several cytokines exhibit a broader effect than TCZ alone in calming down sCRS. Serum levels of cytokines and chemokines show similar changes in allogeneic-HSCT- and COVID-19-associated sCRS with marked elevation of interleukin-6 (IL-6), regulated upon activation normal T-cell expressed and secreted (RANTES), monocyte chemoattractant protein-1 (MCP-1) and interferon γ-induced protein 10 kDa (IP-10) levels. In addition, levels of IL-5, IL-10, IL-15 were also elevated in allogeneic-HSCT-associated sCRS. Our multi-cytokine expression data indicate that the pathophysiology of allogeneic-HSCT and COVID-19-associated sCRS are similar therefore the same clinical grading system and TCZ-based treatment approaches can be applied. TCZ with JAK/STAT inhibitor consolidation therapy might be highly effective in COVID-19 sCRS patients.

11.
Geroscience ; 43(5): 2321-2331, 2021 10.
Article in English | MEDLINE | ID: covidwho-1460452

ABSTRACT

In the present study, humoral and T cell-mediated immune responses elicited by BBIBP-CorV (inactivated virus) and BNT162b2 (mRNA-based) vaccines against SARS-CoV-2 virus were compared. Convalescent volunteers were also investigated to evaluate adaptive immunity induced by live virus. Although both vaccines induced antibody- and T cell-mediated immune responses, our analysis revealed significant quantitative and qualitative differences between the two types of challenges. The BBIBP-CorV vaccine elicited antireceptor-binding domain (RBD) IgG, as well as anti-spike protein (S) IgG and IgA antibodies in healthy individuals, the levels of which were much lower than after BNT162b2 vaccination but still higher than in the convalescent patients. The cumulative IFNγ-positive T cell response, however, was only twofold higher in participants injected with BNT162b2 compared to those who were primed and boosted with BBIBP-CorV vaccine. Moreover, the inactivated virus vaccine induced T cell response that targets not only the S but also the nucleocapsid (N) and membrane (M) proteins, whereas the mRNA vaccine was able to elicit a much narrower response that targets the S protein epitopes only. Thus, the pattern of BBIBP-CorV-induced T cell response in virus-naive participants was similar to the cell-mediated anti-SARS-CoV-2 response observed in convalescent patients. Based on these data, we can conclude that the BBIBP-CorV inactivated virus vaccine is immunologically effective. However, the duration of BBIBP-CorV-induced integrated, antibody, and T cell-mediated, immune responses needs further investigation.


Subject(s)
COVID-19 , Vaccines , BNT162 Vaccine , COVID-19 Vaccines , Humans , SARS-CoV-2 , T-Lymphocytes
13.
Acta Microbiol Immunol Hung ; 2021 Aug 06.
Article in English | MEDLINE | ID: covidwho-1348397

ABSTRACT

Large randomized clinical trials in severe Coronavirus Disease 2019 (COVID-19) patients have proven efficacy of intravenous tocilizumab. Our aim was to describe the laboratory parameters predicting in-hospital mortality of patients with tocilizumab administration in COVID-19 associated cytokine release syndrome (CRS).We evaluated high-dose (8 mg/kg) intravenous tocilizumab administration in severe and critically ill COVID-19 adult patients fulfilling predefined strict CRS criteria. A single-centre, prospective, observational cohort study was carried out among consecutive adult (≥18 years of age) in-patients with COVID-19 between April 1 and December 31, 2020. The primary endpoint was 28-day all-cause mortality. The changes in laboratory parameters from baseline on day 7 and 14 after administration of tocilizumab were analysed.In total, 1801 patients were admitted to our centre during the study period. One hundred and six patients received tocilizumab, and among them 62 (58.5%) required intensive care unit admittance while 25 (23.6%) deceased. At day 7 after tocilizumab administration, inflammatory markers (CRP, IL-6, ferritin) and lactate dehydrogenase (LDH) values were significantly lower among survivors. Subsequently, at day 14, differences of IL-6 and LDH levels has become more pronounced between subgroups. Restoration of absolute lymphocyte count (ALC) by day 7 and 14 was insufficient among patients who died.In our cohort, administration of high-dose tocilizumab for COVID-19 patients with CRS demonstrated clinical and sustained biochemical parameter improvement in 76.4%. In this patient population high and increasing LDH, IL-6, and low ALC levels had a predictive role for mortality.

14.
Orv Hetil ; 161(27): 1111-1121, 2020 07.
Article in Hungarian | MEDLINE | ID: covidwho-822860

ABSTRACT

INTRODUCTION: At present, neither specific curative treatment nor vaccines for novel coronavirus 2019 (COVID-19) are available. There is an urgent need to look for alternative strategies for COVID-19 treatment especially in the case of severe and/or critically ill patients with cytokine release syndrome (CRS). AIM: Convalescent plasma proved to increase survival rates in other severe viral infections. Therefore, convalescent plasma could be a promising treatment option for severe COVID-19 patients. METHOD: In our article, we present the first two critically ill Hungarian patients with COVID-19 infection treated with convalescent fresh frozen plasma. RESULTS: At the time of plasma therapy both patients were on mechanical ventilation and received antiviral agents and a full scale of supportive care. Each patient received 3 × 200 mL of convalescent plasma of recently recovered donors with sufficient novel anti-coronavirus IgG titers. Subsequent to convalescent plasma infusion, oxygenization improved and inflammatory markers decreased in both individuals. As compared to pretransfusion, lymphocyte counts increased and interleukin-6 level lessened. Both patients were weaned from mechanical ventilation within 2 weeks of treatment. No severe adverse effects were observed. CONCLUSIONS: Our experience indicates that convalescent plasma therapy is well tolerated and could potentially improve clinical outcomes. Optimal dose and timing as well as precise assessment of clinical benefit of convalescent plasma therapy will need further investigation in larger, well-controlled trials. This is the first report of the successful use of convalescent plasma in the treatment of critically ill patients with COVID-19 infection in Hungary. Orv Hetil. 2020; 161(27): 1111-1121.


Subject(s)
Coronavirus Infections/therapy , Pneumonia, Viral/therapy , COVID-19 , Critical Illness , Humans , Hungary , Immunization, Passive , Pandemics , Treatment Outcome , COVID-19 Serotherapy
15.
Orv Hetil ; 161(26): 1070-1077, 2020 06.
Article in Hungarian | MEDLINE | ID: covidwho-736545

ABSTRACT

During the past few months, a pandemic originating from China named new coronavirus disease (COVID-19) has shown how vulnerable the world is. To date, no medication supported by randomized clinical trials has been approved for the treatment of COVID-19. At the time of writing of this paper, severe acute respiratory syndrome caused by coronavirus-2 (SARS-CoV-2) has been responsible - according to modest estimations - for around 4 million of infections and 300 000 deaths. Unveiling details of patomechanism, in fatal cases the role of immune dysregulation, namely cytokine release syndrome (CRS) has been discovered. Based on the current knowledge, interleukin-6 (IL6) plays a pivotal role in COVID-19 associated CRS. Case reports and result of small case series suggest efficacy of an IL6 inhibitor monoclonal antibody (tocilizumab) in treating CRS. Authors describe a case and review recent knowledge on the treatment of COVID-19. To our knowledge, the first case of severe COVID-19-associated cytokine storm syndrome - treated succesfully with IL6 monoclocal antibody at a Hungarian department of infectology - is presented here. Orv Hetil. 2020; 161(26): 1070-1077.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/drug therapy , Pneumonia, Viral/drug therapy , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/epidemiology , Cytokine Release Syndrome/virology , Humans , Hungary/epidemiology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/epidemiology , Severity of Illness Index , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL