13
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Prognosis of patients with sickle cell disease and COVID-19: a French experience

      discussion

      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

          France is the country with the highest prevalence of sickle cell disease in Europe, with more than 26 000 patients diagnosed with the condition in 2018. Most of these patients are of sub-Saharan African origin. 1 Patients with sickle cell disease are thought to be at increased risk of COVID-19 complications. Aside from specific COVID-19-related morbidities, infections in patients with sickle cell disease 2 can provoke painful vaso-occlusive crisis and life-threatening acute chest syndrome. Thus, COVID-19 could be devastating for regions such as Africa or India, where an estimated 8–12 million patients with sickle cell disease live, or in the USA and Brazil, with more than 100 000 patients in each country. 3 Nevertheless, there are currently no data on the outcomes of patients with sickle cell disease and COVID-19. On March 13, 2020, at an early stage of the COVID-19 pandemic in France, we invited all practitioners involved in the management of patients with sickle cell disease to report on all inpatients with sickle cell disease and confirmed COVID-19 by RNA detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from nasal swabs. An email was sent to paediatricians, internists, and haematologists involved in sickle cell disease managment in France by our national consortia —MCGRE (Filière de santé maladies constitutionnelles rares du globule rouge et de l'érythropoïèse) and Laboratory of Excellence GR-Ex network. We prospectively collected data on outcomes in patients with sickle cell disease infected with COVID-19 using a standardised form. We compared the prevalence of intensive care unit (ICU) admission for inpatients with sickle cell disease by age range to that of COVID-19-positive inpatients in France during the same period. 4 Data were collected between March 13, 2020, and April 16, 2020. 83 inpatients with sickle cell disease infected by SARS-CoV-2 from 24 centres were enrolled (table 1 ). The median age was 33·5 years (range 19–68) for the 66 (80%) adults and 12 years (0·3–17) for the 17 (20%) children (defined as patients <18 years). 48 (58%) of 83 patients had a past medical history of acute chest syndrome, with a median of 2 episodes (range 1–10); 38 (46%) were being treated with hydroxyurea at admission (30 [51%] of 59 patients in the SS/Sβ0 subpopulation). Vaso-occlusive crisis was associated with COVID-19 in 44 (54%) of 81 inpatients and acute chest syndrome was associated with COVID-19 in 23 (28%) of 82 inpatients (table 1). Table 1 Patient characteristics by age range All patients (n=83) Patients aged 0–14 years (n=12) Patients aged 15–44 years (n=56) Patients aged 45–64 years (n=14) Patients aged 65–74 years (n=1) Age 30 (0·3–68) .. .. .. .. Sex Male 38 (46) 6 (50) 22 (39) 9 (64) 1 (100) Female 45 (54) 6 (50) 34 (61) 5 (36) 0 Haemoglobin genotype SS/Sβ° 71 (86) 11 (92) 48 (86) 12 (86) 0 SC 8 (10) 0 5 (9) 2 (14) 1 (100) Sβ+ 4 (5) 1 (8) 3 (5) 0 0 Hydroxyurea treatment at admission 38 (46) 4 (33) 28 (50) 6 (43) 0 Hydroxyurea dose (mg/kg/day) 17·9 (8·8–30·2) 18·8 (18·6–23·3) 18·2 (11·8–30·2) 13·7 (8·8–16·5) .. Weight (kg) 68 (5–110) 32 (5–49) 71·8 (41–110) 71·5 (59–95) 85 Vaso-occlusive crisis 44/81* (54) 6 (50) 34 (61) 4/12* (33) 0 Acute chest syndrome 23/82* (28) 2 (17) 17 (30) 4/13* (31) 0 Transfusion† 31 (37) 4 (33) 18 (32) 8 (57) 1 (100) Length of hospital stay (days) ‡ 8 (2–37) 4 (2–10) 7 (2–35) 10 (4–37) 22 Mechanical ventilation in the intensive care unit§ 9/17 (53) 0 3/7 (43) 5/7 (71) 1 (100) Data are n (%), n/N (%), or median (range). Percentages do not always equal 100% because of rounding. Ethnicity data were not collected in line with usual practice in France. * Data for vaso-occlusive crisis were not available for two patients and acute chest syndrome not available for one patient. † Simple transfusion or exchange transfusion (manual or automated) during the hospital stay. ‡ Hospitalisation was completed for 80 (96%) of 83 patients and is ongoing at the date of the notification for the other three. § 17 patients were admitted to the intensive care unit. 17 (20%) of 83 patients were admitted to the ICU. Nine (53%) required mechanical ventilation, including two patients treated with extracorporeal membrane oxygenation. Two patients died in the ICU with COVID-19 pneumopathy: two men with the SC haemoglobin genotype. Five (63%) of the 8 patients with the SC genotype were admitted to the ICU, compared with 12 (17 %) of 71 patients with the SS/Sβ0 genotype (p=0·0099 by Fisher's exact test). Among patients 40 years or older, 5 (31%) of 16 with the SS/Sβ0 genotype (median age 48·5 years, range 40–64) were admitted to the ICU versus five (100%) of five patients with the SC genotype (median age 50 years, 40–68; p=0·012). 15 (88%) of 17 patients with sickle cell disease admitted to the ICU were transfused with a median of 4 bags (range 2–7) of packed red blood cells per patient. Only 3 (20%) of 15 were transfused before ICU admission (1, 2, and 28 days before), and the other 12 were transfused after a median time of 1·5 days (range 0–9) after ICU admission. Two patients were treated with automated exchanges, 6 with simple transfusions, and 7 with exchange transfusions. One man with the SC genotype died 3 days after admission from a pulmonary embolism without transfusion, and a woman with the SS genotype was not transfused; instead she was treated with high-flow oxygen and then recovered. Of note, seven patients were directly admitted to the ICU; the median time to ICU transfer after hospital admission was 2 days (range 0–9). Among patients with the SS/Sβ0 genotype, three (25%) of 12 received a transfusion before ICU admission, which was not different from the proportion of transfusions received throughout the stay for the 22 (37%) of 59 patients with the SS/Sβ0 genotype not requiring ICU admission. Treatment with hydroxyurea at admission was similar in both groups (six [50%] of 12 patients with the SS/Sβ0 genotype admitted to the ICU vs 30 [51%] of 59 patients with the SS/Sβ0 genotype not admitted to the ICU, median dose 16·7 mg/kg [range 8·8–22·7] vs 17·9 mg/kg [8·9–30·2]). Although these data are relatively few, they do not support an effect of transfusions or hydroxyurea for preventing ICU admission for the management of COVID-19 in patients with sickle cell disease. The prevalence of ICU admission was significantly different between patients with sickle cell disease younger than 45 years and those 45 years or older; 9 (13%) of 68 patients with a median age of 28 years (range 0·3–44) versus eight (53%) of 15 patients with a median age of 54 years (45–68), p=0.0017. Compared with all other inpatients who tested positive for COVID-19 with the same age range, a biphasic trend was observed: a lower risk of ICU admission for young adults (15–44 years) with sickle cell disease than those without the condition (13% vs 24% admitted to ICU; odds ratio (OR) 0·44, 95% CI 0·16–0·99; p=0·039) and a higher but nonsignificant risk for older inpatients (45–64 years) with sickle cell disease (50% vs 36%; OR 1·76, 0·53–5·89; p=0·28, table 2 ). However, these data should be interpreted with caution because of a lack of statistical power to detect differences. A further limitation of this comparison is that the reasons for hospital admission in the patients without sickle cell disease with COVID-19 could be different (eg, respiratory complaints) compared with patients with sickle cell disease, in part because of sickle cell disease-related complications (eg, vaso-occlusive crisis). Nevertheless, this bias should affect age groups similarly. Moreover, we confirmed that 30 (71%) of 42 patients with sickle cell disease in this study had findings of COVID-19 pneumonia on chest CT scans. Table 2 ICU admission in patients with sickle cell disease and COVID-19 Inpatients with sickle cell disease (n=83) Hospitalised French population (n=17 745) * p value † ICU admission Deaths ICU admission* Deaths‡ Age range (years) All patients 17 (20) 2 (2) 6075 (34) 2891/42 212 (7) .. 0–14 2/12 (17) 0 32/110 (29) 1/592 (<1) 0·72 15–44 7/56 (13) 0 514/2112 (24) 105/7524 (1) 0·039 45–64 7/14 (50) 2/14 (14) 3049/8422 (36) 1016/19 689 (5) 0·28 65–74 1/1 (100) 0 2480/7101 (35) 1769/14 405 (12) .. Data are n (%) or n/N (%). * French general population younger than 75 years hospitalised with confirmed COVID-19 during the peak of the pandemic (April 7, 2020). 4 † Comparison of ICU admission prevalence by age range between inpatients with sickle cell disease and the French general population hospitalised with confirmed COVID-19 (Fisher's exact test). ‡ Death prevalence by age range among all confirmed inpatients with COVID-19 younger than 75 years from March 1, 2020, to April 14, 2020, in France. 4 These results suggest that COVID-19, even if potentially severe, does not seem to carry an increased risk of morbidity or mortality in patients with sickle cell disease, as most patients worldwide have the SS/Sβ0 genotype and are younger than 45 years. Our findings also suggest that vaso-occlusive crisis can complicate COVID-19 infection, occuring in around half of inpatients with sickle cell disease. The hypothesis of a protective effect against COVID-19 in patients with the SS/Sβ0 variant should be explored. Patients with the SS genotype have been shown to have high plasma interferon-α concentrations and their neutrophils showed a clear type I interferon signature. 5 SARS-CoV-2 does not seem to trigger substantial interferon responses ex vivo, which could explain increased viral replication. 6 However, older patients with sickle cell disease should be considered vulnerable to SARS-CoV-2 and should follow guidelines from their respective country to prevent being exposed to it. These patients should also be closely monitored if they become hospitalised becasue of COVID-19.

          Related collections

          Most cited references4

          • Record: found
          • Abstract: found
          • Article: not found

          Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19

          Abstract Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging coronavirus that has resulted in nearly 1,000,000 laboratory-confirmed cases including over 50,000 deaths. Although SARS-CoV-2 and SARS-CoV share a number of common clinical manifestations, SARS-CoV-2 appears to be highly efficient in person-to-person transmission and frequently cause asymptomatic infections. However, the underlying mechanism that confers these viral characteristics on high transmissibility and asymptomatic infection remain incompletely understood. Methods We comprehensively investigated the replication, cell tropism, and immune activation profile of SARS-CoV-2 infection in human lung tissues with SARS-CoV included as a comparison. Results SARS-CoV-2 infected and replicated in human lung tissues more efficiently than that of SARS-CoV. Within the 48-hour interval, SARS-CoV-2 generated 3.20 folds more infectious virus particles than that of SARS-CoV from the infected lung tissues (P<0.024). SARS-CoV-2 and SARS-CoV were similar in cell tropism, with both targeting types I and II pneumocytes, and alveolar macrophages. Importantly, despite the more efficient virus replication, SARS-CoV-2 did not significantly induce types I, II, or III interferons in the infected human lung tissues. In addition, while SARS-CoV infection upregulated the expression of 11 out of 13 (84.62%) representative pro-inflammatory cytokines/chemokines, SARS-CoV-2 infection only upregulated 5 of these 13 (38.46%) key inflammatory mediators despite replicating more efficiently. Conclusions Our study provided the first quantitative data on the comparative replication capacity and immune activation profile of SARS-CoV-2 and SARS-CoV infection in human lung tissues. Our results provided important insights on the pathogenesis, high transmissibility, and asymptomatic infection of SARS-CoV-2.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Pandemic influenza A (H1N1) virus infections in children with sickle cell disease.

              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              The proteome of neutrophils in sickle cell disease reveals an unexpected activation of the interferon alpha signaling pathway

              Polymorphonuclear neutrophils (PMN) are key actors in the pathophysiology of sickle cell disease (SCD), but signaling pathways underlying their activation and sustained inflammation are not well documented. We thus investigated the protein profile of neutrophils from SCD patients (SS genotype) using a proteomic approach. Unexpectedly, SCD neutrophils exhibit a high expression of interferon signaling proteins (ISP) belonging to the type 1 interferon (IFN-1) response pathway. We also showed that SCD patients at steady state displayed a higher level of plasmatic IFNα. Overall, we reported a dramatic high-level expression of ISP in neutrophils from SS patients suggesting an abnormal activation that could be important in developing new anti-inflammatory therapies. SCD is a hemoglobinopathy leading to major red blood cell (RBC) dysfunction, but other cell types (vascular endothelium, leukocytes, platelets) 1-3 also represent key actors in the pathophysiology of the disease. Important studies have highlighted the role of PMN, both during the vaso-occlusive crisis (VOC) and the associated long-term morbidity and mortality. 4 In SCD, patients have an increased leukocyte count at steady state, and exhibit neutrophil activation, rendering them more susceptible to inflammatory stimuli. 5 Moreover recent data have demonstrated the presence of different sub-phenotypes of PMN especially in cancer and inflammation 6,7 as well as in a preclinical model of SCD. 8 Despite these advances, signaling pathways underlying sustained inflammation in SCD remain elusive. In addition, a fine understanding of PMN activation profile is necessary to better decipher the inflammatory paradigm in SCD and develop tailored therapies. In the present study, we investigated for the first time the proteomic profile of PMN in SCD at basal state by a label-free global proteomic approach. We performed a proteomic comparative study of purified neutrophils from four SS patients (SS1-4) at basal state and four AA blood type healthy donors (AA1-4). All patients included in this study were homozygous (SS genotype), aged 2 to 18 years (mean age 9.7 years), and without any associated co-morbidity. They were free from any infections and exhibited C-reactive protein level 0.3) were selected, their LFQ values were z score transformed and analyzed by Euclidian clustering. Clusters 1 and 2 correspond to proteins upregulated and downregulated in SS neutrophils, respectively. (D) Cluster analysis of differentially expressed proteins with the “cellular response to type I interferon” GOBP annotation. (E) Characterization of neutrophils from SS patients by flow cytometry analysis, typical result: over-expression of the Fc fragment of IgG/CD64 and under-expression of L-selectin/CD62L. Data are presented as mean ± standard devaition (SD). Mann-Whitney test was used to compare SS and AA; *P 1.3 or <0.7. Sixty-eight proteins were overexpressed (cluster 1, Figure 1C), and 33 proteins were down-regulated in the SS group compared to the AA group (cluster 2, Figure 1C). Prior to further investigating a new biological pathway, we aimed to confirm the already known surface markers of sickle cell neutrophils. Indeed, several studies in mouse models or in patients have shown an activated and aged phenotype of PMN in SCD. 8,10 By using our proteomics data, we found two proteins described as markers of activation (Fc fragment of IgG, high affinity Ia, receptor/CD64) and ageing (L-Selectin/CD62L) of neutrophils respectively overexpressed (5.8-fold) and underexpressed (0.6-fold) in SS patients compared to the AA group (Online Supplementary Table S1 and Online Supplementary Proteomic File). These data were confirmed by cytometry analysis of freshly isolated neutrophils from five SS patients and five controls (Figure 1E). We therefore concluded that the proteomic analysis could be a relevant tool for exploring neutrophil abnormalities in SCD. A Fisher exact test performed using proteins down-regulated in SS neutrophils did not evidence any common biological pathway (data not shown). In contrast, analyses of the upregulated proteins revealed a major involvement of the type1 interferon (IFN-I) response (Online Supplementary Table S1 and Online Supplementary Proteomic File). Importantly, major ISP including IFIT1, IFIT2, IFIT3, ISG15, ISG20, GBP2, IFI35, MX1 and MX2 were increased 3- to 84-fold in the proteome of SS neutrophils compared to the one of AA neutrophils (Figure 1D). Moreover, we found a significant overexpression of STAT1 and STAT2 in the neutrophil proteome of the SS group consistent with an activation of the IFN-related JAK/STAT signaling pathways. In order to confirm the proteomic data, we assessed the overexpression of the main ISP using Western blotting experiments of purified neutrophils from 10 other SS patients at steady state and 10 other AA controls. In agreement with the proteomic data, we found a significant increase of ISP expression including MX1, ISG15 and IFIT1 as well as the STAT1 and STAT2 proteins, in neutrophil lysates from SS patients compared to controls (Figure 2A). The nuclear translocation of STAT1 and STAT2 are activated by JAK and TYK2-mediated phosphorylation of the Y701 and Y689, respectively, that stimulates the IFN-1 responses. 11,12 We showed that both Y701 of STAT1 and Y689 of STAT2 were highly phosphorylated in SS compared to AA neutrophils (Figure 2A). These findings confirmed the strong activation of the IFN-1 signaling pathway in SS neutrophils via the JAK/STAT1/2 pathway. In order to investigate further and to assess whether the IFN-I response was due to either IFNα or IFNb, we measured the level of both cytokines in the plasma of 34 healthy AA donors and 37 SS patients at steady state, using the novel digital-ELISA technology. Interestingly, we found a significantly increased level of IFNα in the plasma from half of the SS patients compared to AA controls (Mann-Whitney test, P<0.001), even though no difference was observed for IFNb (Figure 2B). Although the specific role of the different types of IFN-1 is not fully understood, it appears that IFNα, in contrast to IFNb is mainly involved in autoimmune and auto-inflammatory diseases. 13 It is noteworthy that 20 SS patients exhibited an increase of IFNα from 10- to 1,000-fold compared to healthy individuals although 17 of the 37 plasma samples had normal levels of IFNα. Clinical and biological investigations of these 37 SS patients did not show any correlation between the plasmatic level of IFNα and biologic markers including leucocyte, neutrophils, reticulocytes and platelets counts, hemoglobin (Hb) level, Hb haplotypes or age (Online Supplementary Table S2), and plasmatic cytokine concentration (including CX3CL-1, Rantes, MCP-1, MCP-3, TNFα, IL1b, IL10, IL18, and IL6). Since it is known that neutrophil extracellular trap formation (NETosis) plays a role in the pathogenesis of SCD, we next investigated the NETosis by measuring the neutrophil elastase and nucleosome, in the plasma from 28 patients investigated for IFNα and IFNb. As expected, we found a significant high level of both markers of NETosis (Figure 2C) in SS patients compared to AA controls, but no correlation with the IFNα level (data not shown). Finally, we analyzed the clinical data from the patients, and found no significant difference between the “high IFN” and “low IFN” group of patients and the number of acute events (including number of VOC per year, acute chest syndrome, stroke, cerebral vasculopathy, acute splenic sequestration nor splenectomy). It is noteworthy that no patient has been treated with hydroxycarbamide and none of them has followed a transfusion program. Moreover, it is interesting to note that of the four SS plasma samples used for the neutrophil proteomic analysis one had low IFNα level, while the other three exhibited 7- to 60-fold increased levels compared to controls, although all four neutrophil samples expressed high level of ISP. Therefore, it is highly probable that plasma IFNα has a transient secretion while the downstream activation of the signaling pathway is persistent. Altogether, our data indicated that SS patients may have inappropriate transient high IFNα secretions (i.e., outside of any acute and infectious events), responsible for the activation of the IFN-1 signaling pathway in neutrophils. Although the mechanism of this activation remains to be elucidated, some recent data described a clear relationship between INF-1 responses and red blood cell alloimmunization in murine models. 14,15 Since alloimmunization represents a detrimental issue in SCD, our data highlight the importance of testing the link between ISP and alloimmunization in SS patients. In conclusion, we showed for the first time by quantitative proteomic analyses of purified neutrophils a particular immune and inflammatory signature in SCD. Our findings provide evidence of a dysfunction of the IFNα signaling pathway that could play an important role in the pathogenesis of SCD. Future studies using a cohort of patients are needed to determine the relationship between IFNα activation and clinical complications and to establish if ISP may represent therapeutic targets to decrease inflammation in SCD. Supplementary Material Disclosures and Contributions Supplementary Appendix
                Bookmark

                Author and article information

                Contributors
                Journal
                Lancet Haematol
                Lancet Haematol
                The Lancet. Haematology
                Elsevier Ltd.
                2352-3026
                18 June 2020
                18 June 2020
                Affiliations
                [a ]French Sickle Cell Referral Centre, Department of Internal Medicine, Georges Pompidou European Hospital, AP-HP, 75015 Paris, France
                [b ]French Sickle Cell Referral Centre, Mondor Hospital, AP-HP, Creteil, France
                [c ]Department of Paediatrics, Hôpital Armand Trousseau, APHP, Paris, France
                [d ]French Sickle Cell Referral Centre, Necker-Enfants malades Hospital, AP-HP, Paris, France
                [e ]French Sickle Cell Referral Centre, Bicètre Hospital, AP-HP, Paris, France
                [f ]Internal Medicine Department, Tenon Hospital, APHP, Paris, France
                [g ]Internal medicine Department, Centre Hospitalier de Versailles, Versailles, France
                [h ]Department of Paediatrics, Hôpital du Sud Francilien, Corbeil-Essonnes, France
                Article
                S2352-3026(20)30204-0
                10.1016/S2352-3026(20)30204-0
                7302791
                32563282
                0a3c2348-803d-4c05-b255-2654ab7a53aa
                © 2020 Elsevier Ltd. All rights reserved.

                Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID-19. The COVID-19 resource centre is hosted on Elsevier Connect, the company's public news and information website. Elsevier hereby grants permission to make all its COVID-19-related research that is available on the COVID-19 resource centre - including this research content - immediately available in PubMed Central and other publicly funded repositories, such as the WHO COVID database with rights for unrestricted research re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for free by Elsevier for as long as the COVID-19 resource centre remains active.

                History
                Categories
                Article

                Comments

                Comment on this article