Abstract
Clinical symptoms of COVID-19 include fever, cough, and fatigue which may progress to acute respiratory distress syndrome (ARDS). The main hematological laboratory findings associated with the severe form of disease are represented by lymphopenia and eosinopenia which mostly occur in the elderly population characterized by cardiovascular comorbidities and immunosenescence. Besides, increased levels of D-dimer, procalcitonin, and C reactive protein (CRP) seem to be powerful prognostic biomarkers helping to predict the onset of coagulopathy. The host immune response to SARS-CoV-2 can lead to an aberrant inflammatory response or “cytokine storm” which contributes to the severity of illness. At immunological level, patients affected by a severe form of COVID-19 show poor clinical trajectories characterized by differential “immunotypes” for which T cell response seems to play a critical role in understanding pathogenic mechanisms of disease. Also, patients with mild to severe COVID-19 displayed macrophage activation syndrome (MAS), very low human leukocyte antigen D related (HLA-DR) expression with a parallel reduction of CD04+ lymphocytes, CD19 lymphocytes, and natural killer (NK) cells. Corticosteroids resulted the best therapy for the immune dysregulation whereas repurposing of tocilizumab (IL-6 receptor antagonist) appears to have mixed results in patients with COVID-19. Besides, anticoagulative therapy was associated with reduced in-hospital mortality and need of intubation among COVID-19 patients. Furthermore, the beneficial use of intravenous immunoglobulin (IVIG) and passive immunotherapy with convalescent plasma needs to be validated in large controlled clinical trials. In this review, we summarize the main hematological parameters with a prognostic value in COVID-19 and the basis of immunological reactivity during COVID-19, with a focus on ongoing clinical trials evaluating immune targets as possible therapeutic strategies.
Keywords: COVID-19, laboratory indexes, immune response, drug therapy
1. INTRODUCTION
The betacoronavirus 2 (SARS-CoV-2) induces the coronavirus disease 2019 (COVID-19) [1]. This pneumonia outbreak induced a rapid pandemic [2]. A predominant mechanism of cell entry exploited by SARS-CoV-2 is represented by the binding of its spike protein to the angiotensin-converting enzyme 2 (ACE2) receptors, mostly present on pulmonary epithelial cells but also on endothelial cells, lymphocytes and other cell types. [3]. COVID-19 shows a highly heterogeneous clinical presentation ranging from asymptomatic or mild disease (more than 80% of all cases) to respiratory failure requiring mechanical ventilation and multiple organ dysfunction syndromes or failure (MODS) [4,5]. The severe form of COVID-19 occurs especially in the elderly population (65 years and older) probably owing to the higher rate of pre-existing cardiovascular comorbidities as well as increased frailty and immunosenescence [6,7]. Remarkably, dysregulated innate and adaptive immune responses triggered by SARS-CoV-2 infection, such as hyperinflammation (cytokine storm), viral escape from innate sensing (particularly the interferon I pathway, IFN-I), hyperactivation of myeloid cells, and lymphopenia characterized by T cell and NK cell dysfunction seem to trigger precise immunological pathways which may contribute to COVID-19-induced systemic damages contributing to disease severity and death [8]. Besides, COVID-19 shows a significant impact on the hemostatic balance, and a pro-coagulant status may aid clinicians to identify patients with poor prognosis at an early stage of disease [9]. Therefore, the host immune response plays a key role in COVID-19 pathogenesis and seems to be different from other coronavirus infections owing to specific epigenetic-sensitive mechanisms unveiled in severe patients upon interactions between SARS-CoV-2 and host cells [10]. Now, COVID-19 is being clearly considered a hematologic disease [11], but the exact mechanistic role of both innate and adaptive immunity against SARS-CoV-2 still needs to be clarified to help to find new targets. Here, we discuss the main hematological parameters and mechanisms underlying the immunological reactivity during COVID-19 with a focus on current therapeutic strategies based on immune targets.
2. HEMATOLOGICAL RESPONSE DURING COVID-19
2.1. Laboratory findings and prognosis
COVID-19 is prevalently a respiratory infection with a prominent impact on the hematopoietic system and hemostasis underlying several cardiovascular complications [6,7]. Now, COVID-19 poses a great challenge for hematologists which should start novel therapeutic approaches going from targeted anti-inflammatory drugs to anticoagulation [11]. The dynamic changes of lymphocyte subsets and cytokineprofiles of patients affected by COVID-19 and their correlation with disease severity remain unclear. In this section, we summarize the main hematological biomarkers useful to screen, evaluate, and make a prognosis for patients affected by COVID-19 (Table 1 ).
Table 1.
Early infection | |||
---|---|---|---|
Parameter | Values (in adults) | ||
Peripheral blood leukocyte count | Normal (or slightly reduced) | 4.5 to 11.0 × 109/L | |
Lymphocyte count | Normal (or slightly reduced) | 1.3 to 4.5 109/L | |
Routine laboratory tests | Normal | ||
Hyperinflammation phase | |||
Peripheral blood leukocyte count | Reduced | <4.5 × 109/L | |
Lymphocyte count | Severely reduced | <1500 cells/mm3 | |
Eosinophil count | Reduced | <450-550 cells/ mm3 | |
Neutrophil count | Increased | >10,000/mm3 | |
Platelet count | Reduced | <150,000/mm3 | |
Inflammatory mediators | Increased levels of pro-inflammatory mediators, such as IL-6, CRP, ferritin, and PCT | IL-6 > 40 pg/ml; CRP ≥ 10 mg/L; Serum ferritin >300 ng/mL; PCT ≥ 0.5 ng/mL | |
Coagulative state | Increased DD | ≥0.5 mg/L |
Abbreviations: CRP: c reactive protein; DD: d-dimer; G-CSF: granulocyte colony stimulating factor; IL: interleukin; IP10: interferon-γ inducible protein 10; LDH: lactate dehydrogenase; MCP-1: monocyte chemoattractant protein-1; PCT: procalcitonin, PT: prothrombin time; TNF: tumor necrosis factor
Generally, the incubation period (1-14 days) as well as the early phase of disease are characterized by non-specific symptoms, including fever and cough. Besides, at hematological level these patients show a normal (or slightly reduced) count of peripheral blood leukocytes (4.5 to 11.0 × 109/L) and lymphocytes (1.3 to 4.5 109/L) as well as normal routine laboratory tests [[12], [13], [14]]. Otherwise, during advanced stages of COVID-19, patients showed an aberrant immune response represented by a modest increase in number of circulating neutrophils (>10,000/mm3) and a sustained decrease in number of T lymphocytes (<1500 cells/mm3), mainly CD04+ and CD08+ T cells, with respect to mild cases [[12], [13], [14]]. A retrospective study has demonstrated that a higher reduction in number of regulatory T cells (Tregs), CD04+ T cells and memory cells were features of a strong imbalance of immune system in severe cases [13].
In the early phase of infection, viral clearance mediated by type-I IFN is a key signaling to prevent further viral replication, T cell exhaustion, and cytokine storm. In most of infected patients, except in those with a severe form of disease, a robust T cell activation was observed with a dominance of CD04+ T responses (“broadly Th1”) over CD08+ [15]. Besides, recovery seems to be correlated with generation of T cell memory as suggested by the missing formation of effector and central memory T cells in severe with respect to mild form of disease [16]. Remarkably, a low number of CD04+ and CD08+ T and Tregs cells might result in exacerbated inflammatory responses leading to a hyperinflammatory immune response (cytokine storm) and worsen damaged tissue [17,18].
Sars-CoV-2 infection was characterized by a severe lymphopenia, with a drastic reduction of both CD04+ and CD08+ T cells, in moderate and severe COVID-19 cases with respect to mild symptoms [8,13]. The cause of peripheral T cell loss in moderate to severe COVID-19 remains elusive; however, beyond the established detrimental effects of the inflammatory cytokine milieu, a direct attachment of lymphocytes to the vascular wall, known as "endothelialitis" or "endothelitis", may be one of the causes leading to lymphopenia [7]. The cytokine storm can be exploited to monitor the COVID-19 course and at laboratory level is detectable by traditional inflammatory biomarkers, including high levels of interleukin 6 (IL-6) (IL-6 > 40 pg/ml), plasma C-reactive protein (CRP) (CRP ≥ 10 mg/L), procalcitonin (PCT) (PCT ≥ 0.5 ng/mL), and ferritin (serum ferritin>300 ng/mL) which were strongly associated with higher risk of developing ARDS [14,[19], [20]]. In particular, increased IL-6 levels have been associated with increased risk of death [21] and a gradual increase during hospitalization has been reported in non-survivors [9]. Besides, high serum CRP, procalcitonin and ferritin levels are emerged as poor prognostic factors [9]. A complex interplay between coagulative hemostasis and inflammation can trigger a coagulopathy in COVID-19, as feature of hospitalized patients affected by a severe form of disease (about 50%) [22]. Sars-CoV-2-induced coagulation disorders seem to mimic other systemic coagulopathies which are often present in severe infections, including disseminated intravascular coagulation (DIC) or thrombotic microangiopathy. In fact, standard coagulation parameters, including elevated levels of D-Dimer (DD) (DD ≥ 0.5 mg/L), are often found in patients with poor prognosis [14]. Despite the exact pathophysiological mechanisms need to be further investigated, assessing the risk of thrombosis in COVID-19 patients is critical to improve the clinical management in terms of anticoagulation therapy [[23], [24], [25], [26]].
2.2. Impact of common COVID-19 complications on immune response
Obesity is an independent risk and prognostic factor for severity of COVID-19 [27]. Indeed, by affecting insulin resistance and beta-cell function, obesity can limit the ability to evoke an appropriate metabolic response during activation of immune system. At immunological level, obesity is strongly linked to a chronic pro-inflammatory state by increasing IL-6, CRP, and adipokine levels [28]. Furthermore, obesity impacts both on tissue leukocyte expression and tissue regulatory (M2) phenotypic cells which are replaced by inflammatory macrophage subsets [28]. Thus, obesity could lead to a dysregulated immune response and increased viral shedding with relevant consequences on the clinical outcomes in COVID-19 patients. . Remarkably, obesity can also enhance pro-thrombotic DIC and risk of venous thromboembolism in COVID-19 [28]. To date, treatment with low molecular weight heparin can be used to limit coagulopathy in COVID-19 patients [29]. Deep vein thrombosis occurs in the upper limbs of patients requiring continuous positive airway pressure ventilator which often can compress the superficial or deep vessels of the upper limbs, thus favoring a thrombotic damage into microcirculation [30]. The mechanistic link between SARS-CoV-2 infection and coagulopathy is not clear; however, a direct attack to endothelial cells which express high levels of ACE2 receptor may be cause of abnormal coagulative state and sepsis [31,32]. Despite prevalence of chronic obstructive pulmonary disease (COPD) is low in COVID-19, a significant severity and mortality rate is seen in these patients [33]. Specific factors, such as dysregulated local and systemic inflammation, impaired host immunity, with the predominance of CD08+ and CD04+ cells, and structural injury may potentially contribute to such risk [34]. Besides, obese patients with COPD are at increased risk of developing severe COVID-19 owing to higher levels of ACE2 expressed in bronchial epithelial samples with respect to non obese controls [35].
3. IMMUNE REACTIVITY DURING COVID-19
Immune response of patients affected by sepsis are generally classified into three clinical groups including the “macrophage-activation syndrome” (MAS) [36], “sepsis-induced immunoparalysis” (low levels of the human leukocyte antigen D related, HLA-DR, on CD14 monocytes) [37], and an “intermediate functional state” without a clear dysregulation. Since IL-6 can inhibit HLA-DR expression [38], we could hypothesize that IL-6 over-production may be correlated with a lower HLA-DR expression in COVID-19 supporting the ongoing clinical investigations about the putative efficacy of anti-inflammatory drugs [39,40]. Moreover, the NK group 2 member A (NKG2A) receptor which can inhibit both NK cytokine secretion and cytotoxicity resulted overexpressed on both CD08+ T and NK cells of patients affected by COVID-19 vs healthy controls [41]. Since NKG2A overexpression can also lead to CD08+ T cells and NK exhaustion (a state of functional unresponsiveness) [41], SARS-CoV-2 may use this strategy to manipulate the host immune response against the viral pathogen. Monalizumab is an inhibiting antibody against NKG2A which can restore the function of CD08+ T and NK cells in cancers, thus may be a putative useful drug to treat patients affected by COVID-19 [41]. The B cell response is critical for the virus clearance and is the major responsible for the memory response preventing reinfection. SARS-CoV-2 can trigger a robust B cell response, as demonstrated by the rapid virus-specific IgM, IgG and IgA, and neutralizing IgG antibodies in the days following infection [8]. In most patients affected by COVID-19, the seroconversion occurs between 7 and 14 days after the onset of symptoms, and antibody titers may persist for a long period after clearance of the virus (36-50 days) but it is not clear whether and how antibody responses may contribute to disease [8]. From evidence, the cumulative IgM and IgG seroprevalence was 44% and 56%, respectively, on day 7 after the onset of symptoms and reached over 95% on day 20 and day 16, respectively. Besides, IgM and IgG levels appeared to remain above the cutoff value on day 28 [42]; and the long-term coexistence of IgG with SARS-CoV-2 raises the question of whether patients with antibodies are still at risk for re-infection [43]. Thus, one of the major challenges is to understand the mechanistic bases by which SARS-CoV-2 can overcame the presence of specific IgG antibodies for such a long time [44].
Innate immunity seems to play a pivotal role in SARS-CoV-2 clearance [45]. One strategy to pursue is to use drugs that mimic viral RNA in order to contribute to clearance of SARS-CoV-2. For example, combination of vaccine and innate immune stimulators may be more effective in the rapid clearance of SARS-CoV-2 [46]. Importantly, FDA has recently approved two mRNA-based vaccines (Pfizer/BioNTech and MODERNA) for their distribution in U.S. which demostrated to prevent severe COVID-19. These vaccines are based on mRNA technology which, instead of inoculating the antigen towards which an immune response is to be induced, inoculates the genetic sequence with instructions for producing the antigen (Spike protein)(https://www.who.int/). Both the increased proinflammatory responses and the reduction of antiviral cytokines in elderly people (the so called immunosenescence) may explain why disease severity and fatality are higher in aged COVID-19 patients [6,7]. In fact, the effective CD08+ T cell response might influence the severity of disease whereas variation in IL-10 levels might contribute to the relatively mild pneumonia symptoms in children affected by COVID-19 [31,32]. All patients affected by severe forms of COVID-19 display exaggerated inflammatory responses (immune dysregulation or MAS) which are characterized by pro-inflammatory cytokines. In particular, IL-6 can drive the immune dysregulation whereas IL-1β can guide MAS. At hematological level, immune dysregulation is featured by a monocyte-related cytokine overexpression and lymphopenia, mainly regarding CD04+ T and subsequently B cell count, CD08+ T and natural killer (NK) cell reduction leading to a higher risk of developing secondary bacterial infection [31,32]. Thus, the adaptive immune response might not be successfully triggered by immune system. Despite molecular mechanisms underlying lymphopenia are still unknown, it may be possible that SARS-Cov-2 can use a further strategy to directly infect circulating T cells and promote the cell death without taking advantage from ACE2 receptor which is absent in circulating lymphocytes [31,32].
SARS-CoV-2-specific memory T cells can persist in peripheral circulation providing a protective immunity [47]. Therefore, T cells studies may elucidate pathways for diagnostics of COVID-19 and to understand the cellular and molecular ways underlying the high clinical heterogeneity of disease.
4. INSIGHTS ON IMMUNE-BASED THERAPIES FOR COVID-19
During the first months of pandemic, the therapeutic arms seemed to be specific FDA-approved interleukin receptor antibodies mainly including tocilizumab and anakinra prescribed for systemic inflammatory response; however, later companies that produced tocilizumab stopped their randomized controlled studies in COVID-19 due to failure [48]. Thus, we are left with mainly some third degree observational studies to support the relevance of IL-6 in treating COVID-19 (Table 2 ). And, neither tocilizumab nor anakinra should be used in COVID-19 unless in an approved trial. Additional targets include IL-1 receptor [49], TNF-α [50] and interferon (IFN). On the other hand, multiple clinical trial groups around the world launched high-quality randomized clinical trials of corticosteroids for severe COVID-19 [51]. The UK-based RECOVERY trial has enrolled 6425 patients (2104 randomized to receive dexamethasone and 4321 randomized to receive usual care) and treatment with dexamethasone (6 mg/d for 10 days) was able to reduce mortality in patients receiving mechanical ventilation and supplemental oxygen as compared to usual care alone [52]. Besides, the REMAP-CAP trial randomized 403 patients with severe COVID-19 and receiving respiratory or cardiovascular organ support) to 1 of 3 open-label groups: fixed low-dose hydrocortisone, shock-dependent hydrocortisone, or no hydrocortisone. As a results, fixed-dose hydrocortisone and shock-dependent hydrocortisone were both likely superior to no hydrocortisone, but data were insufficient to confirm a single optimal regimen [53]. Moreover, controlled clinical trials are investigating the therapeutic efficacy of a humanized anti-granulocyte colony stimulating factors (GM-CSF) IgG1 monoclonal antibody TJ003234 (Table 2). Janus kinase (JAK1/JAK2) inhibition and the use of I IFN (β1α and β2α) are under consideration for severe patients affected by COVID-19. Type I IFN signals through JAK-STAT pathway and upregulated IFN-sensitive genes would eliminate infected cells. Other approaches are coming from pooled plasma of healthy donors, and intravenous immunoglobulin (IVIG) which represent one of the most widely diffused immunotherapies in many inflammatory diseases [54]. Some preliminary studies indicate that IVIG may be useful also during COVID-19 [55,56]; however, it is needed a randomized clinical trial with a large number of patients to verify the effectiveness of this therapy. Other therapeutic options would be to use convalescent plasma, intravenous hyperimmune globulin or neutralizing monoclonal antibodies (Table 3 ). Clinically, it is mandatory to validate the neutralizing activity of plasma or antibody preparations before their use in severe patients affected by COVID-19, mostly for convalescent plasma. Although there is a variable, temporal window, seroconversion time for IgM and IgG following onset of the disease was day 12 and 14, respectively, with 95% of infected patients who displayed IgM and 80% for IgG [57]. Preliminary reports suggested that plasma therapy may lead to improvement in around 35% of treated patients [58,59]. However, we still need to standardize the methodological steps necessary to make convalescent plasma treatments safe and readily available for treating COVID-19 patients [60]. In fact, patients affected by severe COVID-19 received convalescent plasma at a median of 21.5 days after first detection of viral shedding [61]. Results indicated that convalescent plasma treatment can discontinue SARS-CoV-2 shedding but cannot reduce mortality in critically end-stage patients, thus suggesting the need to start treatment earlier.
Table 2.
Mechanism of drug action | Trial | n Patients/status |
---|---|---|
IL-1 receptor antagonist Anakinra |
Phase 3 (NCT04330638) | 342 participants/Active Not Recruiting |
N/A (NCT04318366) | 1000 participants/Recruiting | |
EudraCT Number: 2020-001825-29 | Ongoing | |
EudraCT Number: 2020-001734-36 | Ongoing | |
IL-6 neutralization Siltuximab |
Phase 3 (NCT04330638) | 342 participants/Active Not Recruiting |
EudraCT Number: 2020-001500-41 | Ongoing | |
EudraCT Number: 2020-001413-20 | Ongoing | |
IL-6 receptor antagonist Sarilumab |
Phase 2 (NCT04315298) | 400 participants/Completed |
Phase 2 (NCT04321993) | 1000 participants/Recruiting | |
Phase 2/3 (NCT04327388) | 300 participants/Completed | |
EudraCT Number: 2020-001634-36 | Ongoing | |
EudraCT Number: 2020-001162-12 | Ongoing | |
Tocilizumab | Phase 2 (NCT04317092) | 400 participants/Active Not Recruiting |
Phase 3 (NCT04324073) | 239 participants/Active not recruiting | |
Phase 3 (NCT04320615) | 330 participants/Completed | |
Phase 3 (NCT04330638) | 342 participants/Active Not Recruiting | |
EudraCT Number: 2020-001386-37 | Ongoing | |
EudraCT Number: 2020-001375-32 | Ongoing | |
EudraCT Number: 2020-001770-30 | Ongoing | |
TNF inhibitor Baricitinib |
Phase 2 (NCT04330638) | 1000 participants/Active Not Recruiting |
Phase 3(NCT04330638) | 200 participants/Active Not Recruiting | |
EudraCT Number: 2020-001367-88 | Ongoing | |
EudraCT Number: 2020-001321-31 | Ongoing | |
Inhibition of viral replication by inducing type I IFN- stimulated genes IFNβ1α |
Phase 1(NCT04330638) |
328 participants/Active Not Recruiting |
Recombinant human IFN α1β | Phase 3 (NCT04315948) | 3100 participants/Recruiting |
Recombinant human IFN α2β | Phase 3 (NCT04320238) | 2944 participants/Recruiting |
Inhibition of complement activation by binding to terminal complement protein C5 Eculizumab |
Phase 1 (NCT04288713) | N/A/Available |
EudraCT Number: 2020-001246-18 | Ongoing | |
Regulation of innate response to inflammatory stimuli and injury by binding to danger-associated molecular patterns and Siglec G/10 CD24F |
Phase 3 (NCT04317040) | 230 participants/Active Not Recruiting |
GM-CSF neutralization Anti-GM-CSF Monoclonal AB |
Phase 2 (NCT04341116) | 144 participants/Recruiting |
GM-CSF neutralization, inhibition of viral infection of alveolar cells and virus clearance NKG2D-ACE2 CAR-NK Cells |
Phase 1/2 (NCT04341116) | 90 participants/Recruiting |
Abbreviations: ACE2: angiotensin-converting enzyme 2; CAR: chimeric antigen receptor; GM-CSF: granulocyte-macrophage colony-stimulating factor; N/A: not applicable; NK: natural killer; NKG2D: NK group 2 member D.
Table 3.
Treatment | Aim | Dose | Trial | n Patients/status |
---|---|---|---|---|
IVIG | Suppression of innate and adaptive inflammatory responses | 0.5g/kg/d for 5 days | Phase 3 (NCT04261426) | 80 participants/Not yet recruiting |
0.5 g/Kg for 4 days | Phase 3 (NCT04350580) | 138 participants/Recruiting | ||
0.5g/kg/d for 3 days | Phase 4 (NCT04411667) | 40 participants/ Active Not Recruiting | ||
Polyvalent IVIG in combination with the standard management | EudraCT Number: 2020-001768-27 | 39 participants/ Ongoing | ||
High dose IVIG plus SMT | EudraCT Number: 2020-001696-32 | 100 participants/Ongoing | ||
Immunoglobulin from cured patients | Viral inactivation | 0.2g/kg, ivdrip, once a day, for 3 days | Phase 1 (NCT04411667)) | 10 participants/Active Not Recruiting |
Human coronavirus immune plasma | Viral inactivation | 1 unit; ∼200-250 mL collected by pheresis | Phase 2 (NCT04323800) | 150 participants/ Recruiting |
Hyperimmune plasma | Viral inactivation | Administration of hyperimmune plasma at day 1 and based on clinical response on day 3 and 5 | Phase 1 (NCT04321421) | 49 participants/Completed |
Administration of two units of 300 mL | Phase 2 (NCT04392414) | 60 participants/Completed | ||
Plasma hyperimmune plus standard therapy | Phase 2/3 (NCT04385043) | 400 participants/Recruiting | ||
Anti-SARS-CoV-2 inactivated convalescent plasma | Viral inactivation | N/A | Phase 1 (NCT04292340) | 15 participants/Recruiting |
Day 0, Day 1: Transfusion of 200mL of ABO -Rh compatible inactivated convalescent plasma | Phase 2 (NCT04385186) | 60 participants/Not yet recruiting | ||
N/A | EudraCT Number: 2020-001310-38 | Ongoing | ||
Convalescent plasma | Viral inactivation | 1-2 units of ABO matched donor plasma at neutralization antibody titer >1:64 | Phase 1 (NCT04333251) | 115 participants/Not yet recruiting |
N/A | Phase 1 (NCT04327349) | 30 participants/Enrolling by invitation | ||
Phase 1/2 (NCT04366245) | 72 participants/Recruiting | |||
Day 1: CP-COVID19, 250 milliliters. Day 2: CP-COVID19, 250 milliliters. | Phase 2 (NCT04332380) | 10 participants/Completed | ||
4 units of convalescent plasma | Phase 2 (NCT04429854) | 423 participants/Recruiting | ||
200 cc of convalescent plasma | Phase 2 (NCT04393727) | 126 participants/Recruiting | ||
Day 1: CP-COVID19, 250 milliliters. Day 2: CP-COVID19, 250 milliliters. Hydroxychloroquine: 400 milligrams each 12 hours for 10 days |
Phase 2/3 (NCT04332835) | 80 participants/Not yet recruiting |
Abbreviations: IVIG: intravenous immunoglobulin; N/A: not applicable; SMT: standard medical treatment.
5. ANTOCOAGULATIVE THERAPY IN COVID-19
Hemostatic and inflammatory modifications are common in severe manifestations of COVID-19, and both the bedridden status and critical illness may constitute a prothrombotic milieu predisposing to venous and arterial thrombosis [62]. Hypercoagulative responses may result from specific interactions between host defense mechanisms and the coagulation system [63]. Often, D-dimer and factor VIII and von Willebrand factor are elevated and accompanied by complement activation, platelet activation, increase in fibrinogen, and neutrophil extracellular traps via epigenetic mechanisms [10,64,65]. Besides, some studies have also found antiphospholipid antibodies in patients with COVID-19 and heparin resistance [66,67]. Anticoagulation is associated with reduced in-hospital mortality and need of intubation among COVID-19 patients. However, the optimal antithrombotic regimen for patients with COVID-19, especially those with severe disease, remains uncertain and is currently an area of active clinical interest. A single-city, retrospective study enrolled 4400 adults admitted to New York City hospitals with COVID-19 of which 45% received prophylactic anticoagulation, 21% received therapeutic anticoagulation, and the remainder received none [68]. Overall, 24% of the patients died in the hospital. After multivariable adjustment, prophylactic and therapeutic treatment were each associated with about a 50% reduction in mortality, relative to no anticoagulation. Besides, thromboembolic disease was found in 42% of autopsies related to patients who did not received therapeutic anticoagulation [68]. The recently launched FREEDOM COVID-19 trial (Phase 4, NCT04512079) is evaluating prophylactic and full-dose enoxaparin as well as apixaban in 3600 hospitalized COVID-19 patients. However, no study results have been posted yet. Besides, the optimal type, dose, and duration of intermediate versus prophylactic doses of anticoagulative drugs (enoxaparin, eparin, enoxaparin/lovenox) is being tested in the ongoing randomized IMPROVE trial (Phase 4, NCT04367831) [69].
6. CONCLUDING REMARKS
COVID-19 cannot be considered a disease limited to lungs or cardiovascular system, rather it is a systemic infection with a relevant impact on the hematopoietic and coagulative systems. Lymphopenia shows a higher prognostic value together with inflammatory indices, including CRP and IL-6 which are useful to diagnose the cytokine storm and then stratify patients with poor disease progression. Coagulation abnormalities, such as elevated D-Dimer, PT and fibrin degradation products are particularly associated with disease worsening. The evaluation of individual immune response is currently part of the clinical evaluation; however, there are many questions about the immunity-based protection which remains unsolved owing to the clinical heterogeneity (from asymptomatic to mild symptoms), the association between presence of delayed/weak antibody responses and severe outcomes, and the long-time coexistence between the virus and its specific IgG. Now, it is clear that SARS-CoV-2 infection can both impair and hyperactivate the immune system [70], and patients affected by a severe form of COVID-19 seem to be characterized by differential “immunotypes” associated with poor clinical trajectories [71]. A timely administration of treatments may reduce viral load and avoid hyperactivation of pro-inflammatory pathways. Two mRNA-based vaccines have been recently approved as preventive strategy in order to teach our innate immune system as fight Sars-CoV-2. Hyper-inflammation is caused by a cytokine storm which results from an exaggerated immune response to the presence of SARS-CoV-2. This is considered one of the most powerful biomarkers of poor prognosis in patients affected by COVID-19. To date, no specific treatment has been proven to be effective; however, corticosteroids resulted the best therapy for the immune dysregulation. Many ongoing clinical trials are investigating new off-label therapeutic possibilities to reduce mortality and admission to mechanical ventilation rates, including the optimal antithrombotic regimen. Promising results were obtained by using hyperimmune plasma from patients recovered from the disease as well as repurposing of corticosteroids. More large clinical studies are required to assess the combination therapy really effective and safe in treatment of patients with moderate to severe form of COVID-19 in the second wave of pandemics [72,73].
Declaration of Competing Interest
The authors have no conflicts of interest to declare.
Funding Sources
The authors have no funding to declare.
Author's contribution statement
CN, GB, CC wrote the manuscript. MF, CL, and MR supervised the final version. All 6 the authors have read and approved the manuscript.
References
- 1.Zhu N., Zhang D., Wang W., Li X., Yang B., Song J. A novel coronavirus from patients with pneumonia in China. N Engl J Med. 2020;382:727–733. doi: 10.1056/NEJMoa2001017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Zhou P., Yang X.L., Wang X.G., Hu B., Zhang L., Zhang W. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273. doi: 10.1038/s41586-020-2012-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lu R., Zhao X., Li J., Niu P., Yang B., Wu H. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395:565–574. doi: 10.1016/S0140-6736(20)30251-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chen N., Zhou M., Dong X., Qu J., Gong F., Han Y. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;15(395):507–513. doi: 10.1016/S0140-6736(20)30211-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Huang C., Wang Y., Li X., Ren L., Zhao J., Hu Y. Clinical features of patients infected with 2019 novel coronavirus in Wuhan. China. Lancet. 2020;395:497–506. doi: 10.1016/S0140-6736(20)30183-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Napoli C., Tritto I., Mansueto G., Coscioni E., Ambrosio G. Immunosenescence exacerbates the COVID-19. Arch Gerontol Geriatr. 2020;90:104174. doi: 10.1016/j.archger.2020.104174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Napoli C., Tritto I., Benincasa G., Mansueto G., Ambrosio G. Cardiovascular involvement during COVID-19 and clinical implications in elderly patients. A review. Ann Med Surg (Lond). 2020;57:236–243. doi: 10.1016/j.amsu.2020.07.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Vabret N., Britton G.J., Gruber C., Hegde S., Kim J., Kuksin M. Immunology of COVID-19: current state of the science. Immunity. 2020;52:910–941. doi: 10.1016/j.immuni.2020.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhou F., Yu T., Du R., Fan G., Liu Y., Liu Z. Clinical course and risk factors for mortality of adult in patients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062. doi: 10.1016/S0140-6736(20)30566-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Crimi E., Benincasa G., Figueroa-Marrero N., Galdiero M., Napoli C. Epigenetic susceptibility to severe respiratory viral infections and its therapeutic implications: a narrative review. Br J Anaesth. 2020;125:1002–1017. doi: 10.1016/j.bja.2020.06.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Debuc B., Smadja D.M. Is COVID-19 a new hematologic disease? Stem Cell Rev Rep. 2020:1–5. doi: 10.1007/s12015-020-09987-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhou Y., Fu B., Zheng X., Wang D., Zhao C., Sun R. Pathogenic T cells and inflammatory monocytes incite inflammatory storm in severe COVID-19 patients. Natl Sci Rev. 2020 doi: 10.1093/nsr/nwaa041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Qin C., Zhou L., Hu Z., Zhang S., Yang S., Tao Y. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin Infect Dis. 2020 doi: 10.1093/cid/ciaa248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Terpos E., Ntanasis-Stathopoulos I., Elalamy I., Kastritis E., Sergentanis T.N., Politou M. Hematological findings and complications of COVID-19. Am J Hematol. 2020;95:834–847. doi: 10.1002/ajh.25829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Altmann D.M., Boyton R.J. SARS-CoV-2 T cell immunity: specificity, function, durability, and role in protection. Sci Immunol. 2020;5 doi: 10.1126/sciimmunol.abd6160. eabd6160. [DOI] [PubMed] [Google Scholar]
- 16.Odak I., Barros-Martins J., Bošnjak B., Stahl K., David S., Wiesner O. Reappearance of effector T cells is associated with recovery from COVID-19. EBioMedicine. 2020;57:102885. doi: 10.1016/j.ebiom.2020.102885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Moore J.B., June C.H. Cytokine release syndrome in severe COVID-19. Science. 2020;1(368):473–474. doi: 10.1126/science.abb8925. [DOI] [PubMed] [Google Scholar]
- 18.Ranger A., Haji R., Kaczmarski R., Danga A. Interleukin-6 blockade treatment for COVID-19 associated cytokine release syndrome in a patient with poorly controlled chronic myeloid leukaemia. Br J Haematol. 2020 doi: 10.1111/bjh.16901. doi: 10.1111/bjh.16901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bo Zhou J.S., Yadan W., Xiancang M. Utility of ferritin, procalcitonin, and c-reactive protein in severe patients with 2019 novel coronavirus disease. Research Square. 2020 doi: 10.21203/rs.3.rs-18079/v1. [DOI] [Google Scholar]
- 20.Luo X., Zhou W., Yan X., Guo T., Wang B., Xia H. Prognostic value of c-reactive protein in patients with COVID-19. Clin Infect Dis. 2020 doi: 10.1093/cid/ciaa641. ciaa641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wu C., Chen X., Cai Y., Xia J., Zhou X., Xu S. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in wuhan. China. JAMA Intern Med. 2020;180:1–11. doi: 10.1001/jamainternmed.2020.0994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Miesbach W., Makris M. COVID-19: Coagulopathy, risk of thrombosis, and the rationale for anticoagulation. Clin Appl Thromb Hemost. 2020;26 doi: 10.1177/1076029620938149. 1076029620938149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Liao D., Zhou F., Luo L., Xu M., Wang H., Xia J. Haematological characteristics and risk factors in the classification and prognosis evaluation of COVID-19: a retrospective cohort study. Lancet Haematol. 2020 doi: 10.1016/S2352-3026(20)30217-9. 2352-3026(20)30217-30219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Marietta M., Ageno W., Artoni A., De Candia E., Gresele P., Marchetti M. COVID-19 and haemostasis: a position paper from Italian Society on Thrombosis and Haemostasis (SISET) Blood Transfus. 2020;18:167–169. doi: 10.2450/2020.0083-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Han H., Yang L., Liu R., Liu F., Wu K.L., Li J. Prominent changes in blood coagulation of patients with SARS-CoV-2 infection. Clin Chem Lab Med. 2020;25(58):1116–1120. doi: 10.1515/cclm-2020-0188. [DOI] [PubMed] [Google Scholar]
- 26.Thachil J., Tang N., Gando S., Falanga A., Cattaneo M., Levi M. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J Thromb Haemost. 2020;18:1023–1026. doi: 10.1111/jth.14810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tamara A., Tahapary D.L. Obesity as a predictor for a poor prognosis of COVID-19: a systematic review. Diabetes Metab Syndr. 2020;14:655–659. doi: 10.1016/j.dsx.2020.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Simonnet A., Chetboun M., Poissy J., Raverdy V., Noulette J., Duhamel A. High prevalence of obesity in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) requiring invasive mechanical ventilation. Obesity. 2020;28:1195–1199. doi: 10.1002/oby.22831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kollias A., Kyriakoulis K.G., Dimakakos E., Poulakou G., Stergiou G.S., Syrigos K. Thromboembolic risk and anticoagulant therapy in COVID-19 patients: emerging evidence and call for action. Br J Haematol. 2020;189:846–847. doi: 10.1111/bjh.16727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bozzani A., Arici V., Franciscone M.M., Danesino V., Cascina A., Ticozzelli G. SARSCoV-2 infection and the upper limbs deep vein thrombosis risk. Ann Vasc Surg. 2020 doi: 10.1016/j.avsg.2020.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Li H., Liu L., Zhang D., Xu J., Dai H., Tang N. SARS-CoV-2 and viral sepsis: observations and hypotheses. Lancet. 2020;395:1517–1520. doi: 10.1016/S0140-6736(20)30920-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Li H., Chen K., Liu M., Xu H., Xu Q. The profile of peripheral blood lymphocyte subsets and serum cytokines in children with 2019 novel coronavirus pneumonia. J Infect. 2020;81:115–120. doi: 10.1016/j.jinf.2020.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Alqahtani J.S., Oyelade T., Aldhahir A.M., Alghamdi S.M., Almehmadi M., Alqahtani A.S. Prevalence, severity and mortality associated with COPD and smoking in patients with COVID-19: a rapid systematic review and meta-analysis. PLoS One. 2020;15:e0233147. doi: 10.1371/journal.pone.0233147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rovina N., Koutsoukou A., Koulouris N.G. Inflammation and immune response in COPD: where do we stand? Mediators Inflamm. 2013;2013:413735. doi: 10.1155/2013/413735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Higham A., Singh D. Increased ACE2 expression in bronchial epithelium of COPD patients who are overweight. Obesity (Silver Spring) 2020;28:1586–1589. doi: 10.1002/oby.22907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kyriazopoulou E., Leventogiannis K., Norrby-Teglund A., Dimopoulos G., Pantazi A., Orfanos S.E. Macrophage activation-like syndrome: an immunological entity associated with rapid progression to death in sepsis. BMC Med. 2017;15:172. doi: 10.1186/s12916-017-0930-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lukaszewicz A.C., Grienay M., Resche-Rigon M., Pirracchio R., Faivre V., Boval B. Monocytic HLA-DR expression in intensive care patients: interest for prognosis and secondary infection prediction. Crit Care Med. 2009;37:2746–2752. doi: 10.1097/CCM.0b013e3181ab858a. [DOI] [PubMed] [Google Scholar]
- 38.Ohno Y., Kitamura H., Takahashi N., Ohtake J., Kaneumi S., Sumida K. IL-6 down-regulates HLA class II expression and IL-12 production of human dendritic cells to impair activation of antigen-specific CD4(+) T cells. Cancer Immunol Immunother. 2016;65:193–204. doi: 10.1007/s00262-015-1791-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fu B., Xu X., Wei H. Why tocilizumab could be an effective treatment for severe COVID-19? J Transl Med. 2020;18:164. doi: 10.1186/s12967-020-02339-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Evangelos J., Giamarellos-Bourboulis E.J., Netea M.G., Rovina N., Akinosoglou K., Antoniadou A. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe. 2020;27:992–1000. doi: 10.1016/j.chom.2020.04.009. e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yaqinuddin A., Kashir J. Innate immunity in COVID-19 patients mediated by NKG2A receptors, and potential treatment using monalizumab, chloroquine, and antiviral agents. Med Hypotheses. 2020;140:109777. doi: 10.1016/j.mehy.2020.109777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Guan W.J., Ni Z.Y., Hu Y., Liang W.H., Ou C.Q., He J.X. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708–1720. doi: 10.1056/NEJMoa2002032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Xu X., Sun J., Nie S., Li H., Kong Y., Liang M. Seroprevalence of immunoglobulin M and G antibodies against SARS-CoV-2 in China. Nat Med. 2020;26:1193–1195. doi: 10.1038/s41591-020-0949-6. [DOI] [PubMed] [Google Scholar]
- 44.Fafi-Kremer S., Fauvelle C., Felmlee D.J., Zeisel M.B., Lepiller Q., Fofana I. Neutralizing antibodies and pathogenesis of hepatitis C virus infection. Viruses. 2012;4:2016–2030. doi: 10.3390/v4102016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Thevarajan I., Nguyen Tho, Koutsakos M., Druce J., Caly L., van de Sandt Ce. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26:453–455. doi: 10.1038/s41591-020-0819-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Florindo H.F., Kleiner R., Vaskovich-Koubi D., Acúrcio R.C., Carreira B., Yeini E. Immune-mediated approaches against COVID-19. Nat Nanotechnol. 2020;15:630–645. doi: 10.1038/s41565-020-0732-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Melgaço J.G., Azamor T., Ano Bom A.P.D. Protective immunity after COVID-19 has been questioned: what can we do without SARS-CoV-2-IgG detection? Cell Immunol. 2020;353:104114. doi: 10.1016/j.cellimm.2020.104114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Tsai A., Diawara O., Nahass R.G., Brunetti L. Impact of tocilizumab administration on mortality in severe COVID-19. Sci Rep. 2020;10:19131. doi: 10.1038/s41598-020-76187-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cavalli G., De Luca G., Campochiaro C., Della-Torre E., Ripa M., Canetti D. Interleukin-1 blockade with high-dose anakinra in patients with COVID-19, acute respiratory distress syndrome, and hyperinflammation: a retrospective cohort study. The Lancet Rheumatology. 2020:e325–e331. doi: 10.1016/S2665-9913(20)30127-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Richardson P., Griffin I., Tucker C., Smith D., Oechsle O., Phelan A. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet. 2020;395:30–31. doi: 10.1016/S0140-6736(20)30304-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sterne L., Marshall J., Diaz J., Villar J., Murthy S., Slutsky A. PROSPERO; 2020. A prospective meta-analysis of randomized trials of corticosteroid therapy for COVID-19. CRD42020197242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.RECOVERY Collaborative Group, Horby P., Lim W.S., Emberson J.R., Mafham M., Bell J.L., Linsell L. Dexamethasone in hospitalized patients with covid-19-preliminary report. N Engl J Med. 2020 doi: 10.1056/NEJMoa2021436. NEJMoa2021436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.The Writing Committee for the REMAP-CAP Investigators Effect of hydrocortisone on mortality and organ support in patients with severe COVID-19: the REMAP-CAP COVID-19 corticosteroid domain randomized clinical trial. JAMA. 2020;324:1317–1329. doi: 10.1001/jama.2020.17022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Perez E.E., Orange J.S., Bonilla F., Chinen J., Chinn I.K., Dorsey M. Update on the use of immunoglobulin in human disease: a review of evidence. J Allergy Clin Immunol. 2017;139:1–46. doi: 10.1016/j.jaci.2016.09.023. [DOI] [PubMed] [Google Scholar]
- 55.Shao Z., Feng Y., Zhong L., Xie Q., Lei M., Liu Z. Clinical efficacy of intravenous immunoglobulin therapy in critical ill patients with COVID-19: a multicenter retrospective cohort study. Clin Transl Immunology. 2020;9:e1192. doi: 10.1002/cti2.1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Xie Y., Cao S., Li Q., Chen E., Dong H., Zhang W. Effect of regular intravenous immunoglobulin therapy on prognosis of severe pneumonia in patients with COVID-19. J Infect. 2020;81:318–356. doi: 10.1016/j.jinf.2020.03.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhao J., Yuan Q., Wang H., Liu W., Liao X., Su Y., Wang X., Yuan J., Li T., Li J., Qian S., Hong C., Wang F., Liu Y., Wang Z., He Q., Li Z., He B., Zhang T., Fu Y., Ge S., Liu L., Zhang J., Xia N., Zhang Z. Antibody Responses to SARS-CoV-2 in Patients With Novel Coronavirus Disease 2019. Clin Infect Dis. 2020;71:2027–2034. doi: 10.1093/cid/ciaa344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Shen C., Wang Z., Zhao F., Yang Y., Li J., Yuan J. Treatment of 5 critically ill patients with COVID-19 with convalescent plasma. JAMA. 2020;323:1582–1589. doi: 10.1001/jama.2020.4783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Duan K., Liu B., Li C., Zhang H., Yu T., Qu J. Effectiveness of convalescent plasmatherapy in severe COVID-19 patients. Proc Natl Acad Sci U S A. 2020;117:9490–9496. doi: 10.1073/pnas.2004168117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Sullivan H.C., Roback J.D. Convalescent plasma: therapeutic hope or hopeless strategy in the SARS-CoV-2 Pandemic. Transfus Med Rev. 2020 doi: 10.1016/j.tmrv.2020.04.00161. S0887-7963(20)30025-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zeng Q.L., Yu Z.J., Gou J.J., Li G.M., Ma S.H., Zhang G.F. Effect of convalescent plasma therapy on viral shedding and survival in COVID-19 Patients. J Infect Dis. 2020;222:38–43. doi: 10.1093/infdis/jiaa228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ali A., Omore I., Asare L., Gabani M., Riaz M. Seated-immobility thromboembolism syndrome complicating coronavirus disease 2019 outbreak quarantine. Chest. 2020;158(A1614) doi: 10.1016/j.chest.2020.08.1453. [DOI] [Google Scholar]
- 63.Iba T., Levy J.H., Levi M., Connors J.M., Thachil J. Coagulopathy of coronavirus disease 2019. Crit Care Med. 2020;48:1358–1364. doi: 10.1097/CCM.0000000000004458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Escher R., Breakey N., Lämmle B. Severe COVID-19 infection associated with endothelial activation. Thromb Res. 2020;190:62. doi: 10.1016/j.thromres.2020.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Magro C., Mulvey J.J., Berlin D., Nuovo G., Salvatore S., Harp J. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: A report of five cases. Transl Res. 2020;220:1–13. doi: 10.1016/j.trsl.2020.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Helms J., Tacquard C., Severac F., Leonard-Lorant I., Ohana M., Delabranche X. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46:1089–1098. doi: 10.1007/s00134-020-06062-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.White D., MacDonald S., Bull T., Hayman M., de Monteverde-Robb R., Sapsford D. Heparin resistance in COVID-19 patients in the intensive care unit. J Thromb Thrombolysis. 2020;50:287–291. doi: 10.1007/s11239-020-02145-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nadkarni G.N., Lala A., Bagiella E., Chang H.L., Moreno P.R., Pujadas E. Anticoagulation, bleeding, mortality, and pathology in hospitalized patients with COVID-19. J Am Coll Cardiol. 2020;76:1815–1826. doi: 10.1016/j.jacc.2020.08.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Levi M., Thachil J., Iba T., Levy J.H. Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol. 2020;7:e438–e440. doi: 10.1016/S2352-3026(20)30145-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Liu J., Li S., Liu J., Liang B., Wang X., Wang H. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine. 2020;55:102763. doi: 10.1016/j.ebiom.2020.102763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Mathew D., Giles J.R., Baxter A.E., Oldridge D.A., Greenplate A.R., Wu J.E. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020 doi: 10.1126/science.abc8511. eabc8511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Donatelli F., Miceli A., Cirri S., Coscioni E., Napoli C. COVID-19 and the second wave during autumn: preventive strategies in cardiac and thoracic surgery divisions. Eur Surg. 2020:1–3. doi: 10.1007/s10353-020-00682-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Miceli A., Donatelli F., Glauber M., Napoli C. Urgent need for careful holistic assessment post-coronavirus disease 2019 (COVID-19) hospitalization: Crisis after crisis? . 2020. J Thorac Cardiovasc Surg. 2020:S0022–5223(20)32907–X.. doi: 10.1016/j.jtcvs.2020.10.081. [DOI] [PMC free article] [PubMed] [Google Scholar]