key: cord-0914828-8wlx0i9v authors: Codd, Amy S; Hanna, Stephanie J; Compeer, Ewoud B; Richter, Felix C; Pring, Eleanor J; Gea-Mallorquí, Ester; Borsa, Mariana; Moon, Owen R; Scourfield, D Oliver; Gallimore, Awen M; Milicic, Anita title: Neutrophilia, lymphopenia and myeloid dysfunction: A living review of the quantitative changes to innate and adaptive immune cells which define COVID-19 pathology date: 2021-07-15 journal: Oxf Open Immunol DOI: 10.1093/oxfimm/iqab016 sha: ced5ba7c8a7c9a030e0c838f956a869a696cf13d doc_id: 914828 cord_uid: 8wlx0i9v Destabilisation of balanced immune cell numbers and frequencies is a common feature of viral infections. This occurs due to, and further enhances, viral immune evasion and survival. Since the discovery of the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), which manifests in coronavirus disease 2019 (COVID-19), a great number of studies have described the association between this virus and pathologically increased or decreased immune cell counts. In this review, we consider the absolute and relative changes to innate and adaptive immune cell numbers, in COVID-19. In severe disease particularly, neutrophils are increased, which can lead to inflammation and tissue damage. Dysregulation of other granulocytes, basophils, and eosinophils represent an unusual COVID-19 phenomenon. Contrastingly, the impact on the different types of monocytes leans more strongly to an altered phenotype, e.g. HLA-DR expression, rather than numerical changes. However, it is the adaptive immune response which bears the most profound impact of SARS-CoV-2 infection. T cell lymphopenia correlates with increased risk of ICU admission and death; therefore, this parameter is particularly important for clinical decision making. Mild and severe disease differ in the rate of immune cell counts returning to normal levels post disease. Tracking the recovery trajectories of various immune cell counts may also have implications for long-term COVID-19 monitoring. This review represents a snapshot of our current knowledge, showing that much has been achieved in a short period of time. Alterations in counts of distinct immune cells represent an accessible metric to inform patient care decision or predict disease outcomes. Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Since the outbreak, it has become apparent that there is a broad spectrum of clinical symptoms in people infected with SARS-CoV-2: from no obvious symptoms in around 40% of infected individuals (Oran and Topol, 2020) to a need for intensive-care unit (ICU) hospitalisation and use of ventilators in most severely affected patients . Additionally, the exacerbated immune response contributes to "acute respiratory distress syndrome" (ARDS), which is a prominent feature of severe COVID-19 Sun et al., 2020) . Many studies have reported singular and additive epidemiological and clinical risk factors associated with increased COVID-19 severity and mortality, including age and gender, and pre-existing conditions such as obesity, diabetes, hypertension and cardiovascular disease (Codo et al., 2020; Cummings et al., 2020; . Ethnicity is a complex etiological feature when considering the impact of COVID-19. In addition to some of the abovementioned comorbidities, ethnicity impacts socio-economic status, access to healthcare and occupational hazard. Ethnicity was inconsistently reported in the early stages of the pandemic , although the weight of evidence recently identified a significantly increased risk of COVID-19 infection in Black and Asian, compared to White individuals . Data from the USA shows that the mortality rate from COVID-19 is also higher in Black compared to white ethnic groups (Tai et al., 2021) . In this review we summarise the current knowledge of the changes observed in absolute counts and phenotypic frequencies of immune cells in SARS-CoV-2-infected individuals. We try to understand the nature of the immune response that leads to recovery over severe disease and how treatments can help to promote an immune response that leads to recovery. For hospitalised COVID-19 patients, rapid measures to guide stratification of care and resources are crucial, due to the burden of the pandemic on healthcare systems. A single centre evaluation of 'core' (full blood counts, urea, electrolytes, liver function and C-reactive protein) versus 'extended' (D-dimer, ferritin, high-sensitivity troponin I, lactate dehydrogenase, procalcitonin) clinical tests found the latter did not add sufficient cost-benefit prognostic value (Ponsford et al., 2021) . As a routine readout, cell counts provide a valuable overview of the main cell types involved in the immune response to COVID-19. Cell counts are informative even in the absence of mechanistic information explaining increased or decreased numbers, and thus could be used to guide clinical decision making and signpost more in-depth, descriptive research such as multi-dimensional phenotyping and biomarker identification. As yet there is no clear trend in overall white blood cell (WBC) count in individuals infected with SARS-CoV-2; a systematic review found increased WBC in 24.26% and decreased WBC in 10.55% in 20,662 hospitalized COVID-19 patients . However, metaanalysis of 45 studies covering such patients showed a trend between increased WBC count and disease severity (Moutchia et al., 2020) . This corroborates earlier observations of elevated WBC counts in severe disease and deceased COVID-19 patients (G. Qin et al., 2020; Ruan et al., 2020; and ICU admission (Elshazli et al., 2020) . Recent studies might explain the lack of a strong correlation between WBC count and disease severity, as an increase in the neutrophil to lymphocyte ratio (NLR), characterised by reduced lymphocytes and elevated neutrophils, has been found in patients with normal WBC counts upon hospital admission (S. Lombardi et al., 2020) . These non-convergent data demonstrate a variety of immune cell perturbations across the spectrum of COVID-19 severity, necessitating the examination of specific immune cell subsets. Neutrophils are the most prominent innate cells in the response to a viral infection and have been widely reported to be increased in the blood in COVID-19 patients (Elshazli et al., 2020; Qin et al., 2020) . Neutrophil infiltration of pulmonary capillaries has also been described, along with increased expression of neutrophil associated chemokines in lung epithelial cells (Barnes et al., 2020; Blanco-Melo et al., 2020 ). Neutrophilia appears to be selfpropelled by initial viral evasion of immune detection, leading to poor viral clearance, resulting in inflammation and cytokine storm (Borges et al., 2020; Cavalcante-Silva et al., 2021; Chau et al., 2021; Reusch et al., 2021) . There are limited data providing greater detail about neutrophil subsets in COVID-19, although Kuri-Cervantes et al. and Wilk et al. describe characteristics of neutrophils which may suggest impaired maturation (reduced CD15 expression and expression of developmental markers specifically in ARDS patients, respectively) Wilk et al., 2020) . Atypical neutrophil phenotypes have been associated with excessive Neutrophil Extracellular Trap (NET) production, which can cause hyperinflammation and tissue damage (Borges et al., 2020; Provine et al., 2021) . Increased numbers of peripheral neutrophils correlate with COVID-19 disease severity and poor outcome (Y. Elshazli et al., 2020; Qin et al., 2020; Wauters et al., 2021; Wei et al., 2020) . The contribution of dysfunctional neutrophil anti-viral responses to COVID-19 pathology is discussed in further detail in the innate immune response focused article of our living review series (Rodrigues et al., 2020) . The relationship between neutrophil and lymphocyte counts (neutrophil to lymphocyte ratio, NLR) represents a more powerful prognostic measure, both in COVID-19 and other acute disease settings (Curbelo et al., 2017; Liu et al., 2016; Qin et al., 2020) and is discussed in detail in the adaptive immunity section of this review. Basophils and eosinophils, which play a greater role in other innate immune functions, such as allergic and anti-microbial responses, are nevertheless also impacted in COVID-19. The relatively limited data indicates depletion of basophils occurs in the blood in showing some associations with severe disease (Laing et al., 2020; Mann et al., 2020; Mazzoni et al., 2020; Qin et al., 2020; Rodriguez et al., 2020) . It has been suggested however, that decreased basophil counts in the blood could be attributed to migration to the lungs (Carsana et al., 2020; Laing et al., 2020) . While the cause of basophil depletion is currently unknown, a basophil count of 25/µl in the blood may represent a threshold predictive of survival in ventilated patients (Renner et al., 2020) . The directionality of eosinophil perturbation in COVID-19 is less clear; several studies report decreased or unchanged eosinophil numbers in the blood Laing et al., 2020; Mazzoni et al., 2020; Qin et al., 2020; Rodriguez et al., 2020; . Contrastingly however, Lucas et al. describe a sustained increase in peripheral eosinophils in severe disease, along with increased IL-5, which is a contributing signalling factor for eosinopoiesis in the bone marrow (Lucas et al., 2020; Roufosse, 2018) . Increased IL-5 has been reported in other studies, however without eosinophil counts (C. . The limited data available on lung eosinophil infiltration are also conflicted (Barton et al., 2020; Kim et al., 2020; , although it is not clear if a distinction was made for the detection of migratory or lung-resident eosinophils. Whilst stratifying patients by peripheral blood eosinophil counts (> or < than 0.02x10 9 /L) has revealed differences in severity and mortality Xie et al., 2021) , overall, this suggests a need for more in-depth investigation of IL-5 associated eosinophil responses in the blood and potentially in the lungs. In terms of absolute numbers, studies reported unchanged Lombardi et al., 2020; Mann et al., 2020; Mazzoni et al., 2020) , increased (G. Hadjadj et al., 2020; Lucas et al., 2020; Wei et al., 2020; Wen et al., 2020) or decreased frequencies of the monocytic cell lineage in peripheral blood during COVID-19 (Boumaza et al., 2021; Qin et al., 2020; Sánchez-Cerrillo et al., 2020; Wilk et al., 2020) . Some of this discrepancy is likely due to the use of different markers and nomenclature in the studies: reporting global increases or decreases in monocytes is unlikely to capture their significance in the context of COVID-19 symptoms or clinical course, due to phenotypically and functionally distinct monocyte subsets. However, some trends have emerged. In peripheral blood, classical monocytes (M1; CD14 + CD16 -) were within the reference range in early disease (Lombardi et al., 2020) and remained stable in severe or moderate disease (Lucas et al., 2020) . Sánchez-Cerrillo et al. found that the frequency (rather than absolute count) of classical monocytes was reduced in the periphery, and enriched, although relatively infrequent, in bronchoscopy samples of patients with severe COVID-19 (Sánchez-Cerrillo et al., 2020) . Reduction in peripheral blood non-classical (M2; CD14 -CD16 + ) monocytes has been reported in severe and moderate disease and in ARDS compared to controls, although similarly this might be associated with a migration to the lung, due to the observed enrichment in bronchoscopy samples (Carsetti et al., 2020; Lucas et al., 2020; Sánchez-Cerrillo et al., 2020) . The most prominently reported monocytic perturbation is the expansion of intermediate (Mµ; CD14 + CD16 + ) monocytes in peripheral blood (Carsetti et al., 2020; Laing et al., 2020; Mann et al., 2020; Payen et al., 2020) . The significance of these changes in monocyte numbers is not yet clear. IL-6 production by intermediate monocytes in COVID-19 has been described in association with cytokine storm and severe disease (Y. , and in general, increased IL-6 levels correlate with disease severity Wan et al., 2020) . However, since an increase in intermediate monocytes in blood has been reported in both mild and severe disease, it is likely that other immune cells contribute to IL-6 production (Carsetti et al., 2020; Laing et al., 2020; Mann et al., 2020; Sánchez-Cerrillo et al., 2020; . The ability of SARS-CoV-2 to infect monocytes has been demonstrated in vitro, using primary monocytes and monocyte cell lines (Boumaza et al., 2021; Codo et al., 2020) , resulting in pro-and anti-inflammatory cytokine production, including IFN α, β, and λ, and TNF, IL-1β, IL-6, IL-10 and TGF-β (Boumaza et al., 2021; Codo et al., 2020) . Cytokine and chemokine production by monocytes in COVID-19 has recently been reviewed by our consortium (Rodrigues et al., 2020) . Some studies have also identified upregulation and coexpression of M1 and M2 markers on monocytes (Matic et al., 2020) , which further complicates efforts to understand the contribution of functional monocyte subsets in COVID-19. Boumaza et al. found that polarised monocyte cell lines (M1 or M2) showed no difference in the propensity for in vitro SARS-CoV-2 infection, however infection led to the general upregulation of M2 markers (Boumaza et al., 2021) . Further work is therefore required to establish a clear picture of the contributing role of the various monocyte subsets in COVID- Overall dendritic cell (DC) numbers are reduced in the peripheral blood in COVID-19 (Kuri-Cervantes et al., 2020; Laing et al., 2020; Lee et al., 2020; Liao et al., 2020; Wilk et al., 2020) . In humans, DCs are generally divided into conventional DC subsets (cDCs), specialised for antigen presentation, and plasmacytoid DC (pDCs) which primarily produce type 1 IFN and are important for anti-viral response (O'Keeffe et al., 2015) . In COVID-19, pDCs appear preferentially depleted (Hadjadj et al., 2020; Mann et al., 2020; . Lower pDC numbers in the periphery are reported to correlate with severe disease (Laing et al., 2020; Sanja et al., 2020; Wilk et al., 2020) . Additionally, Sanchez-Cerillo et al. did not detect pDCs in bronchiolar samples, although it is unclear if this was due to impaired migration or depletion in the periphery (Sánchez-Cerrillo et al., 2020) . In conclusion, the available data on the innate response to SARS-CoV-2 infection indicate an association between the efficacy of the early granulocyte (eosinophil, basophil and neutrophil) response and disease severity. It has been suggested that SARS-CoV-2 can evade immune sensing and inhibit signalling Rodrigues et al., 2020; Taefehshokr et al., 2020; Vabret et al., 2020; Zhang et al., 2021) , resulting in impaired activation of the innate response. Quantifying monocytes in COVID-19 is particularly complex as the classical, nonclassical and intermediate subtypes function at various stages of the immune response, including its resolution. Conclusions thus far are largely reliant on data pertaining to peripheral blood monocytes, which overlook, and could underestimate, the impact of monocyte migration to the lungs. While not reviewed here, it has also been suggested that altered expression of certain markers, e.g. HLA-DR (decreased), on monocytes and other cell types, is characteristic in COVID-19 (Boumaza et al., 2021; Giamarellos-Bourboulis et al., 2020; Kuri-Cervantes et al., 2020; Laing et al., 2020; Lombardi et al., 2020; Lucas et al., 2020; Matic et al., 2020; Wilk et al., 2020) . Laing et al. propose a COVID-19 innate and adaptive immune signature which is largely in agreement with the quantitative data discussed here: depletion of pDCs and basophils correlate with COVID-19 severity, while alterations in the proportion of different monocyte lineages demarcate COVID-19 from other respiratory infections (Laing et al., 2020) . Lymphopenia and the adaptive immune response to SARS-CoV-2 Reduction in lymphocytes, known as lymphopenia, is a common, although not exclusive, characteristic of COVID-19. Lymphopenia also occurs in infections with Ebola virus, respiratory syncytial virus (RSV) -which most commonly affects children, SARS-CoV-1, Middle Eastern Respiratory Virus (MERS) and Human Immunodeficiency Virus (HIV). It has also been reported for some strains of Influenza (Cunha et al., 2009; Leroy et al., 2001; Rivera and Messaoudi, 2016; Saleeby et al., 2008; Welliver, 2003) . In COVID-19, lymphopenia most prominently affects T cells, as discussed below; quantitative changes to NK cells, and, less commonly, B cells, have also been reported (Carsetti et al., 2020; Kuri-Cervantes et al., 2020; Laing et al., 2020; Lombardi et al., 2020; Mann et al., 2020; Qin et al., 2020; Schub et al., 2020; Xu et al., 2020) . NK cells NK cells are innate lymphocytes important in early viral infection control through direct killing of infected cells, by lysis or antibody directed cellular cytotoxicity (ADCC), and production of cytokines such as IFNγ (Frank and Paust, 2020; van Eeden et al., 2020) . NK cell depletion in peripheral blood has been reported as a part of COVID-19 associated lymphopenia (Lombardi et al., 2020) , although it has been noted that this is not as extensive as T cell lymphopenia (Laing et al., 2020) . Maucourant et al. further found that the absolute counts of both CD56 dim (cytotoxic and IFNγ + ) and CD56 bright (cytokine producing and IFNγ + ) NK cells were reduced (Maucourant et al., 2020) . This reduction positively correlates with COVID-19 severity, ICU admission and increased hospital stay (Carsetti et al., 2020; Jeannet et al., 2020; Mann et al., 2020; Odak et al., 2020; Qin et al., 2020; Schub et al., 2020; Xu et al., 2020; . Carsetti et al. and Odak et al. observed that an increase in NK cells was associated with asymptomatic infection, and mild COVID-19 patients had NK cell numbers comparable to healthy controls, respectively (Carsetti et al., 2020; Odak et al., 2020) . In addition to reduced NK cell numbers, Mazzoni et al. found that IL-6 levels negatively correlate with NK cell cytotoxic capacity (granzyme production) (Mazzoni et al., 2020) , suggesting a mechanism for poor infection control. Preservation of NK cell numbers and function in the periphery is therefore an important factor in efficient COVID-19 infection control. The mechanisms by which NK cells could control SARS-CoV-2 infection have recently been discussed by our consortium (Rodrigues et al., 2020) . It is not yet clear how the NK cell depletion occurs; RNAseq studies suggest enrichment of NK cells in the lungs (Chua et al., 2020; Liao et al., 2020; , although in the absence of quantitative data, it is not clear whether this is due to migration from the periphery or in situ expansion of lungresident NK cells. Robust antibody production has been widely detected in COVID-19 patients (A. T. Nielsen et al., 2020; W. Tan et al., 2020; Wu et al., 2020; . Accordingly, the percentage of antibody producing plasmablasts has been reported to increase to 10-31% of circulating B cells Varnaitė et al., 2020) . This can temporarily boost the WBC count, although the concurrent depletion of many other immune cell types, including CD19 + B cells (Carsetti et al., 2020; Mann et al., 2020; Schub et al., 2020; Xu et al., 2020) , seems to have largely masked this effect. It should also be noted that patients with severe COVID-19 can have high numbers of plasmablasts, suggesting that counts of these cells cannot be considered diagnostic without a qualitative analysis of the antibodies produced (Carsetti et al., 2020; Kuri-Cervantes et al., 2020; Mathew et al., 2020) . In line with this, higher amounts of IgM and IgG antibodies targeting SARS-CoV-2 nucleocapsid and spike proteins were associated with disease severity and poor clinical parameters W. Tan et al., 2020) . The best-documented change in immune cells numbers in COVID-19 is in T cells, with 40-80% of patients showing T cell lymphopenia on admission (Fei et al., 2020; Lombardi et al., 2020) , and a growing number of studies demonstrating a correlation between lymphopenia and severe COVID-19 (Fei et al., 2020; Lombardi et al., 2020; Qin et al., 2020; . Wang et al. found that T cell numbers are progressively reduced as the disease severity increases (J. . Peripheral T cell loss is associated with an increase in apoptotic cells and changes in the CXCR3 signalling pathway, suggesting that both T cell apoptosis and their migration towards inflamed tissue might contribute to peripheral lymphopenia (Adamo et al., 2020) . Reduced lymphocyte count upon admission was found to not only increase disease severity but also mortality Fei et al., 2020; . In particular, T cell counts <800/μl identify patients at risk of ICU admission and death (G. Diao et al., 2020; Xu et al., 2020) . Patients with lymphopenia also have an increased incidence of multi-organ injury, indicated by worse lung CT scores, reduced respiratory function and elevated indicators of hepatic injury (Fei et al., 2020) . Depletion of both CD4 + and CD8 + T cells has been reported in COVID-19 patients, in addition to phenotypic alterations in specific T cell subsets, which have recently been extensively reviewed by our consortium (Hanna et al., 2020) . While some studies report preferential depletion of CD8 + T cells Varnaitė et al., 2020) , there is currently a lack of consensus as to the prognostic significance of reduced CD4 + and CD8 + T cell counts. Some studies have reported that reduction of each of these subsets is associated with COVID-19 severity and the need for ICU care (G. Diao et al., 2020; Schub et al., 2020; Xu et al., 2020) . However, in other reports, while both populations are decreased, only a reduction in one T cell subtype, CD4 + (Lombardi et al., 2020; Qin et al., 2020; or CD8 + (J. is associated with disease severity or mortality. antigens and viruses (Lamichhane et al., 2020; Provine et al., 2021; Ussher et al., 2014; van Wilgenburg et al., 2016) . MAIT cells are depleted in COVID-19 patients (Parrot et al., 2020) and this decrease is associated with disease severity (Flament et al., 2021; Jouan et al., 2020) . Flament et al. further suggest CD8 + MAIT cells were most prominently reduced, however, in contrast, Parrot et al. found that double negative (DN) MAIT were decreased to a greater degree than CD8 + MAIT (Flament et al., 2021; Parrot et al., 2020) . Invariant natural killer T (iNKT) cells are another innate T cell type, with cytotoxic, cytokine producing and immunoregulatory roles, restricted by the recognition of lipid antigens (Matsuda et al., 2008) . Functional roles for iNKT cells have been described in the response to both chronic (HIV, Hepatitis) and acute (Influenza) viral infection (Juno et al., 2012) , prompting their investigation in COVID-19. iNKT cells are found to be depleted in severe COVID-19 disease (Jouan et al., 2020; Kuri-Cervantes et al., 2020) . There is little information as to the direct cause of T cell depletion in COVID-19. Reduction in CD4 + and CD8 + T cells also occurs in SARS-CoV-1 and MERS-CoV infections (Channappanavar et al., 2014) , however only the latter has been shown to infect T cells and directly cause apoptosis (Chu et al., 2016) . Thus far there are no peer reviewed data demonstrating SARS-CoV-2 infection of T cells. A study which used immortalised T cell lines to suggest cytotoxic SARS-CoV-2 infection of T cells has since been retracted (X. . Two studies have identified potential mechanisms by which T cells could be infected by SARS-CoV-2 (Pontelli et al., 2020; . Pontelli et al. show that PBMCs, including CD4 + and CD8 + T cells, are susceptible to SARS-CoV-2 infection in vitro, as assessed by immunostaining of viral antigens and viral replication (dsRNA) (Pontelli et al., 2020) . The infection correlated with the expression of apoptotic markers, which may suggest a mechanism for lymphopenia in severe COVID-19 patients (Pontelli et al., 2020) Indirect factors are also likely to contribute to the T cell number reduction in COVID-19. Inflammation, particularly in severe patients, is well described (G. Qin et al., 2020; Rodriguez et al., 2020; and levels of cytokines TNF, IL-6 and IL-10, and biochemical markers of inflammation negatively correlate with counts of CD4 + and CD8 + T cells . Increased TNF inflammation was observed post-mortem in secondary lymphoid organs and positively correlated with a loss in Bcl-6 + follicular helper T cells (Kaneko et al., 2020) . T cell numbers are also indirectly impacted by depletion and functional impairment of other immune cells. While more profound reduction in pDCs compared to cDCs is reported in COVID-19, cDCs from acute patients failed to respond to a maturation cocktail and did not simulate T cell proliferation (R. . Lack of cDC-mediated antigen presentation and co-stimulatory signals could therefore underlie low T cell counts. Depletion of NK cells negatively impacts signalling for DC maturation; additionally, NK cell cytokine production and co-stimulatory marker expression plays a direct role in T cell differentiation (Schuster et al., 2016) . Therefore, SARS-CoV-2 impairment of T cell responses involves multi-faceted disruption of the immune response. Furthermore, since enumeration of CD4 + and CD8 + T cell subsets in COVID-19 patients is not consistently accompanied by further in-depth phenotypic information, it is not clear whether reduced numbers of specific populations are due to conversions between particular phenotypes in the respective subset, or outright loss of cells (Chen and John Wherry, 2020; Hanna et al., 2020) . Reduced numbers of CD4 + and CD8 + T cells in COVID-19 patients may be due to failed activation/anergy, or hyperactivation, followed by apoptosis, each of which could contribute to severe COVID-19 (Bouadma et al., 2020) . Neutrophils and the neutrophil to lymphocyte ratio As mentioned above, the decline in lymphocyte numbers combined with an increased neutrophil count, or NLR, has proven to be a strong predictor of disease severity and outcome Meta-analysis has shown that the pooled risk ratio (RR) for mortality in COVID-19 patients with elevated NLR was 2.75 (Simadibrata et al., 2021) . Rodriguez et al. additionally found that the NLR decreased with recovery . The NLR also correlates with other immunological features of COVID-19. High NLR and anti-SARS-CoV-2 IgG antibody levels positively correlate with disease severity and negatively correlate with numbers of CD4 + T cells . Depletion and functional impairment of NK cells may also exacerbate the NLR: IL-6-, IL-8 and IL-10 induced upregulation of the NKG2A inhibitory NK cell receptor inhibits the production of IFNγ, which controls neutrophil accumulation in the lung (Antonioli et al., 2020) . IL-6, which is widely reported to be increased in COVID-19 (Elshazli et al., 2020; Mazzoni et al., 2020) , also directly regulates neutrophil trafficking (Fielding et al., 2008) . cases (Mann et al., 2020; Rodriguez et al., 2020) . As neutrophilia is associated with COVID-19 severity, it is crucial to collect more longitudinal data to aid in-hospital treatment allocation and patient stratification. NK cell counts increased over the second to third week of observation in patients who had a favourable outcome, although higher NK cells at baseline were most commonly observed in mild cases (Carsetti et al., 2020) . DC numbers also increased between the symptom onset and recovery . In individuals who experienced mild COVID-19, frequencies of monocytes and pDCs were comparable to healthy controls a median of 35 days after symptom onset; however, comparing frequencies may overlook a persistent overall reduction in absolute cell counts (Rodda et al., 2021) . Inflammatory classical macrophages persist in the early recovery stage (ERS; <7 days since negative PCR test) , however, phenotypic rather than quantitative changes appear to be more relevant over the clinical course with regard to the different subsets of monocytes. Mann et al. report an increase in proliferating Ki67 + monocytes prior to ICU admission, suggesting that monocyte expansion contributes to disease exacerbation, potentially identifying a therapeutic target (Mann et al., 2020) . Payen et al. note that HLA-DR expression by monocytes was below the threshold defined for immunosuppression throughout the monitoring period, only recovering >24 days after initial symptoms. HLA-DR expression also positively correlated with absolute numbers of CD4 + and CD8 + T cells (Payen et al., 2020) . Immune modulating treatment is also likely to contribute to the improved numbers of innate immune cells and, by reducing innate immune associated inflammation, clinical improvement observed in COVID-19 patients. However, the timing of treatment is crucial to avoid inhibition of the adaptive immune response, which is instrumental in resolving SARS-COV-2 infection (Robinson and Morand, 2021) . For example, glucocorticoids induce neutrophil apoptosis but also inhibit DC function and T cell development (Coutinho and Chapman, 2011) . Corticosteroid therapy has been administered to almost half of hospitalised COVID-19 patients (22%-44.9%) Fadel et al., 2020; Wen et al., 2020) . However, the rapidly evolving nature of the pandemic largely precludes in-depth longitudinal study of the effects of these treatments on quantitative immune cell recovery. The RECOVERY consortium concluded that dexamethasone and tocilizumab (an anti-IL-6 receptor antibody which increases IL-6 levels (Lucas et al., 2020) ), alone, or in combination, reduced mortality and hospital stay. However, this trial did not collect data pertaining to laboratory measures and therefore the effects on immune cell subsets cannot be ascertained (RECOVERY Collaborative Group, 2021; The RECOVERY Collaborative Group, 2021) . Additionally, a number of studies describe the improvement of innate cells, such as reduction in neutrophils and increase in non-classical and intermediate monocytes and DCs, in the absence of immunomodulatory treatment (Payen et al., 2020; Rodriguez et al., 2020) . While the data are still relatively limited, short term (<30 days) monitoring of T cell counts in convalescent individuals suggests cell number recovery occurs on a trajectory dictated by disease severity. In two studies, T cell numbers began to recover in most mild patients within 10-15 days, while the duration of T cell recovery in moderate and severe patients lasted 5-20 days (Mann et al., 2020; Rodriguez et al., 2020) . Another study found that in severe patients, T cells recovered to numbers comparable to mild cases at 16 days after disease onset, although this was after a further depletion on days 4-6 (J. . From the lowest 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 point at 11-14 days, CD4 + and CD8 + T cell recovery in a cohort of ICU patients reached significance 19-23 days after ICU admission, although it was not clear if this timepoint took place under continued critical care (Payen et al., 2020) . T cell frequencies remained lower than in healthy controls in both early and late recovery stages, although these data were not stratified based on disease severity prior to the recovery stage . However, impairment of polyfunctional responses, persistent activation and delayed quantitative recovery of IFNγ + CD8 + T cells has also been identified Payen et al., 2020; Yang et al., 2020) . It is not unexpected that recovery of T cell numbers takes longer in patients with more drastically depleted cell counts. Nevertheless, these trajectory data, along with the prognostic cell counts discussed in the previous sections, could be useful to predict the duration of patient's critical care requirements. More information is also warranted on the recovery of T cells and other immune cell numbers, in comparison to healthy controls, to determine if this has any impact on the so-called 'Long COVID' syndrome of persistent, erratic symptoms which seem to occur irrespective of initial disease severity, and is currently largely characterised based on symptoms, without interrogation of the underlying physiology (Mandal et al., 2020) . At the time of writing, two studies which have examined the recovery of adaptive immune cells up to 1, 3 and 8 months, report antigen specific responses (Dan et al., 2021; Rodda et al., 2021) . Dan et al. report that 40-50% of their cohort had detectable SARS-Cov-2-specific CD4 + and CD8 + T cell responses at >6 months, although these were not enumerated (Dan et al., 2021) . Higher numbers of functional, Spike-specific memory B and CD4 + T cells were detected in response to in vitro stimulation at one and three months in recovered individuals (Rodda et al., 2021) . Memory B cell increased, while memory CD4 + T cell remained similar (measured as counts or frequencies), from one to three months (Dan et al., 2021; Rodda et al., 2021) . Although lacking reinfection data, these studies encouragingly identify the capacity for reactivation of adaptive immunity against SARS-CoV-2, inviting further studies to determine if this response is protective. There are many factors not yet well understood in SARS-CoV-2 disease. Limitations of the available data include variation in study sizes, unevenly distributed or absent ethnicity data. In longitudinal studies, it is difficult to control for different numbers of data points per patient and the real-time nature of studies precludes uniform data collection timepoints. There is also an uneven distribution of data obtained from peripheral blood compared to BALF/ lung tissue, leading to a lack of understanding of cell migration over the disease course, and the potential for under-reporting of an effective lung-resident immune response. Relative cell recovery from these different types of samples may also confound attempts to correlate cell numbers with clinical parameters. Impairment of the immune response also goes beyond a simple depletion or expansion of immune cells. These metrics do however inform decision making in the clinical setting based on patient stratification, particularly in the case of T cell lymphopenia, where counts can be used to stratify patients according to their need of intensive care facilities (G. Diao et al., 2020; Xu et al., 2020) . Further analysis is required to determine at-risk lower limit cut-offs for NK cells, and more studies are required to reach a consensus upper limit of neutrophil counts and NLR. The prognostic value of other innate immune cells requires wider exploration of signalling, cytokine production and migration to the infection site, to determine the relationship between timing and efficacy in fighting SARS-CoV-2 infection. In particular, monocyte differentiation and phenotype proportions appear to correlate with disease severity, although this may prove ineffective as a prognostic indicator due to the complexity of monocyte characterisation. Due to the global impact of the SARS-CoV-2 pandemic, it is also important to consider the application of such metrics across the 'normal ranges' for diverse populations; for example, many of the studies reviewed here have a bias of sex or age range, due to the disproportionate impact of COVID-19 in some populations. Furthermore, it has been hypothesised that the worse survival rates reported in Black, Asian and minority ethnic backgrounds compared to white populations may be partly attributed to "underlying genetic factors" (Patel et al., 2020) . However, most research papers, including many of the studies outlined here, have not specified the ethnicity of patients, or have analysed a relatively homogenous population, further limiting our understanding of why some people are at higher risk of adverse outcomes from COVID-19 infection . 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 Box 1-What is the consensus?  Increased WBC counts occur in almost a quarter of COVID-19 patients and are associated with severe disease. However, WBC within physiological reference ranges may 'mask' a combination of extreme increases and decreases in the counts of different immune cells. Therefore, WBC count alone should not be used to stratify patients.  An increase in neutrophils in the blood occurs in most COVID-19 patients. Both increased neutrophils and an increased NLR positively correlate with disease severity. Therapeutic modulation of neutrophil activation signalling is being tested in clinical trials, if approved this may provide a more targeted approach to reducing inflammation compared to corticosteroids which must be administered with appropriate timing so as not to inhibit the adaptive immune response. Increased cell numbers can indicate effective activation and proliferation; however, in case of innate cells this is often delayed and, as a result, pathologically excessive in COVID-19. Decreased cell numbers e.g. T cells, can occur due to a failure to provide the required co-stimulation or activation followed by apoptosis; these opposing responses could dictate a poor or effective immune response, respectively. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 Data Availability Statement All data are contained within the manuscript. This review was facilitated by weekly releases of the Oxford-Cardiff COVID-19 Literature Consortium journal club-a database of reviewed articles and journals will be made available on request. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 https://mc.manuscriptcentral.com/oxfimm Manuscripts submitted to Oxford Open Immunology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 https://mc.manuscriptcentral.com/oxfimm Manuscripts submitted to Oxford Open Immunology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 https://mc.manuscriptcentral.com/oxfimm Manuscripts submitted to Oxford Open Immunology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 A: Altered immune cells in the peripheral blood. Top left, severe disease: Neutrophils, inflammatory intermediate monocytes (Mµ) and the neutrophil to lymphocyte ratio (NLR) are increased in severe COVID-19. Increased plasmablasts have been reported in both severe and mild disease. Bottom left: Many cells of the innate and adaptive immune system are depleted in severe COVID-19: Non-classical monocytes (M2), plasmacytoid dendritic cells (pDC) and basophils. Lymphopenia is widely reported in COVID-19, largely due to depletion of T cells; reductions in CD4+ and/ or CD8+ T cells (CD4, CD8) have been reported and innate lymphocytes and unconventional T cells are also decreased: NK cells, NKT cells, γδ T cells and MAIT cells. Centre, unchanged: some studies suggest eosinophils are unchanged in COVID-19 disease but may increase in the course of recovery. Classical monocytes (M1) are largely unchanged in COVID-19, however changes in expression of certain phenotypic proteins are associated with severe disease. Top right, mild COVID-19/recovery: Increasing basophils and eosinophils are also associated with recovery or mild COVID-19, along with higher numbers of conventional dendritic cells (cDC), NK cells and T cells which are indicative of an effective anti-viral immune response contributing to a milder form of COVID-19 and/or recovery. Bottom 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 left: decreased neutrophils and recovery of lymphocyte numbers, resulting in a reduction of the NLR, are associated with recovery from COVID-19. B: Altered immune cells in the lungs. Top left: Severe disease is largely characterised by inflammation in the lungs in association with increased neutrophils, M1 and Mµ monocytes. NK cells, cDCs and M2 monocytes are also increased, whereas basophils are decreased. Despite decreases in the blood, pDCs are rarely detected in the lung in severe COVID-19. In mild disease or recovery, neutrophils return to normal ranges. Increased T cells, in particular CD8+ T cells, occur as part of the recovery from COVID-19, although some studies have reported delayed recovery trajectories based on disease severity. DCs also increase with recovery. This figure was created using Servier Medical Art templates, which are licensed under a Creative Commons Attribution 3.0 Unported License; https://smart.servier.com. 190x275mm ( 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 Lymphopenia-induced T cell proliferation is a hallmark of severe NKG2A and COVID-19: another brick in the wall Usefulness of the neutrophil-to-lymphocyte ratio in predicting short-and longterm mortality in breast cancer patients Targeting potential drivers of COVID-19: Neutrophil extracellular traps COVID-19 Autopsies Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19 COVID-19 and Neutrophils: The Relationship between Hyperinflammation and Neutrophil Extracellular Traps Immune Alterations in a Patient with SARS-CoV-2-Related Acute Respiratory Distress Syndrome Monocytes and macrophages, targets of SARS-CoV-2: the clue for Covid-19 immunoparalysis Pulmonary post-mortem findings in a series of COVID-19 cases from northern Italy: a two-centre descriptive study Different Innate and Adaptive Immune Responses to SARS-CoV-2 Infection of Asymptomatic, Mild, and Severe Cases Neutrophils and COVID-19: The road so far T cell-mediated immune response to respiratory coronaviruses The Longitudinal Immune Response to Coronavirus Disease 2019: Chasing the Cytokine Storm Clinical and immunological features of severe and moderate coronavirus disease 2019 The Novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Directly Decimates Human Spleens and Lymph Nodes T cell responses in patients with COVID-19 Function of HAb18G/CD147 in invasion of host cells by severe acute respiratory syndrome coronavirus Middle East Respiratory Syndrome Coronavirus Efficiently Infects Human Primary T Lymphocytes and Activates the Extrinsic and Intrinsic Apoptosis Pathways COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis Elevated Glucose Levels Favor SARS-CoV-2 Infection and Monocyte Response through a HIF-1α/Glycolysis-Dependent Axis The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights Epidemiology, clinical course, and outcomes of critically ill adults with COVID-19 Diagnostic Importance of Relative Lymphopenia as a Marker of Swine Influenza (H1N1) in Adults Inflammation biomarkers in blood as mortality predictors in community-acquired pneumonia admitted patients: Importance of comparison with neutrophil count percentage or neutrophil-lymphocyte ratio Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection Reduction and Functional Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19). Front Immunol 11 Clinical Features of 85 Fatal Cases of COVID-19 from Wuhan. A Retrospective Observational Study Immunosuppression by activated human neutrophils. Dependence on the myeloperoxidase system Diagnostic and prognostic value of hematological and immunological markers in COVID-19 infection: A meta-analysis of 6320 patients Early Short Course Corticosteroids in Hospitalized Patients with COVID-19 Single-cell RNA-seq and V(D)J profiling of immune cells in COVID-19 patients Reduction of lymphocyte count at early stage elevates severity and death risk of COVID-19 patients: a hospital-based case-cohort study IL-6 regulates neutrophil trafficking during acute inflammation via STAT3 Outcome of SARS-CoV-2 infection is linked to MAIT cell activation and cytotoxicity Dynamic Natural Killer Cell and T Cell Responses to Influenza Infection Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure Tocilizumab in COVID-19: some clarity amid controversy Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients SARS-CoV-2 ORF9b Antagonizes Type I and III Interferons by Targeting Multiple Components of the RIG-I/MDA-5-MAVS, TLR3-TRIF, and cGAS-STING Signaling Pathways Pre-treatment neutrophil-to-lymphocyte ratio is associated with neutrophil and T-cell infiltration and predicts clinical outcome in patients with glioblastoma The Oxford-Cardiff COVID-19 Literature Consortium A systematic review of antibody mediated immunity to coronaviruses: kinetics, correlates of protection, and association with severity Clinical features of patients infected with 2019 novel coronavirus in Wuhan Significance of neutrophil-tolymphocyte ratio, platelet-to-lymphocyte ratio for predicting clinical outcomes in Severe COVID-19 is associated with deep and sustained multifaceted cellular immunosuppression SARS-CoV-2 proteome microarray for global profiling of COVID-19 specific IgG and IgM responses Phenotypical and functional alteration of unconventional T cells in severe COVID-19 patients Invariant NKT Cells: Regulation and Function during Viral Infection Loss of Bcl-6-Expressing T Follicular Helper Cells and Germinal Centers in COVID-19 Enhanced eosinophilic inflammation associated with antibody and complement-mediated pneumonic insults in severe COVID-19 Comprehensive mapping of immune perturbations associated with severe COVID-19 Neutrophil-to-lymphocyte ratio and lymphocyte-to-C-reactive protein ratio in patients with severe coronavirus disease 2019 (COVID-19): A metaanalysis A dynamic COVID-19 immune signature includes associations with poor prognosis Type I interferons are important co-stimulatory signals during T cell receptor mediated human MAIT cell activation The Incubation Period of Coronavirus Disease 2019 (COVID-19) From Publicly Reported Confirmed Cases: Estimation and Application Immunophenotyping of COVID-19 and influenza highlights the role of type I interferons in development of severe COVID-19 Early immune responses accompanying human asymptomatic Ebola infections Elevated Exhaustion Levels of NK and CD8+ T Cells as Indicators for Progression and Prognosis of COVID-19 Disease Clinical characteristics of 225 patients with COVID-19 in a tertiary Hospital near Wuhan Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19 Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 Prognostic Significance of Neutrophil-to-Lymphocyte Ratio in Patients with Sepsis: A Prospective Observational Study Blood neutrophil to lymphocyte ratio as a predictor of hypertension Neutrophil-to-lymphocyte ratio as an independent risk factor for mortality in hospitalized patients with COVID-19 Early Phases of COVID-19 Are Characterized by a Reduction in Lymphocyte Populations and the Presence of Longitudinal analyses reveal immunological misfiring in severe COVID-19 Long-COVID': a cross-sectional study of persisting symptoms, biomarker and imaging abnormalities following hospitalisation for COVID-19 Longitudinal immune profiling reveals key myeloid signatures associated with COVID-19 Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications SARS-CoV-2 infection induces mixed M1/M2 phenotype in circulating monocytes and alterations in both dendritic cell and monocyte subsets CD1d-restricted iNKT cells, the "Swiss-Army knife" of the immune system Natural killer cell immunotypes related to COVID-19 disease severity Impaired immune cell cytotoxicity in severe COVID-19 is IL-6 dependent The platelet-to-lymphocyte ratio, superior to the neutrophil-to-lymphocyte ratio, correlates with hepatitis C virus infection Clinical laboratory parameters associated with severe or critical novel coronavirus disease 2019 (COVID-19): A systematic review and meta-analysis Human B Cell Clonal Expansion and Convergent Antibody Responses to SARS-CoV-2 Reappearance of effector T cells is associated with recovery from COVID-19 Human dendritic cell subsets and function in health and disease Prevalence of Asymptomatic SARS-CoV-2 Infection The impact of ethnicity on clinical outcomes in COVID-19: A systematic review MAIT cell activation and dynamics associated with COVID-19 disease severity Ethnicity and covid-19 A Longitudinal Study of Immune Cells in Severe COVID-19 Patients Examining the utility of extended laboratory panel testing in the emergency department for risk stratification of patients with COVID-19: a singlecentre retrospective service evaluation MAIT cell activation augments adenovirus vector vaccine immunogenicity Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial T cell anergy in COVID-19 reflects virus persistence and poor outcomes Neutrophils in COVID-19 Molecular mechanisms of Ebola pathogenesis Divergent effects of acute versus chronic glucocorticoids in COVID-19 Functional SARS-CoV-2-Specific Immune Memory Persists after Mild COVID-19 The Oxford-Cardiff COVID19 Literature Consortium, 2020. Innate immunology in COVID-19 -A LIVING REVIEW PART II: Dysregulated inflammation drives immunopathology Systems-Level Immunomonitoring from Acute to Recovery Phase of Severe COVID-19 Targeting the Interleukin-5 Pathway for Treatment of Eosinophilic Conditions Other than Asthma. Front Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China Risk Factors for Severe Respiratory Syncytial Virus Disease in Children With Cancer: The Importance of Lymphopenia and Young Age COVID-19 severity associates with pulmonary redistribution of CD1c + DCs and inflammatory transitional and nonclassical monocytes High levels of SARS-CoV-2 specific T-cells with restricted functionality in severe course of COVID-19 Natural Regulators": NK Cells as Modulators of T Neutrophil-to-lymphocyte ratio on admission to predict the severity and mortality of COVID-19 patients: A metaanalysis Immune dysfunction following COVID-19, especially in severe patients Immunological and inflammatory profiles in mild and severe cases of COVID-19 Clinical characteristics of hospitalized patients with SARS-CoV-2 infection: A single arm meta-analysis Ethnicity and clinical outcomes in COVID-19: A systematic review and meta-analysis Covid-19: Perspectives on Innate Immune Evasion The Disproportionate Impact of COVID-19 on Racial and Ethnic Minorities in the United States Lymphopenia predicts disease severity of COVID-19: a descriptive and predictive study Viral Kinetics and Antibody Responses in Patients with Clinical Characteristics of 20,662 Patients with COVID-19 in mainland China: A Systemic Review and Meta-analysis Prognostic Role of Neutrophil-to-Lymphocyte Ratio in Solid Tumors: A Systematic Review and Meta-Analysis Dexamethasone in Hospitalized Patients with Covid-19 Pulmonary Pathology of Early-Phase 2019 Novel Coronavirus (COVID-19) Pneumonia in Two Patients With Lung Cancer Characteristics of Peripheral Blood Cells in COVID-19 Patients Revealed by a Retrospective Cohort Study CD161++ CD8+ T cells, including the MAIT cell subset, are specifically activated by IL-12+IL-18 in a TCR-independent manner Immunology of COVID-19: current state of the science Natural Killer Cell Dysfunction and Its Role in COVID-19 MAIT cells are activated during human viral infections Expansion of SARS-CoV-2-Specific Antibody-Secreting Cells and Generation of Neutralizing Antibodies in Hospitalized COVID-19 Patients Characteristics of lymphocyte subsets and cytokines in peripheral blood of 123 hospitalized patients with 2019 novel coronavirus pneumonia (NCP) Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China Characteristics of Peripheral Lymphocyte Subset Alteration Characteristics of lymphocyte subsets and their predicting values for the severity of CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells SARS-CoV-2 infects T lymphocytes through its spike protein-mediated membrane fusion Discriminating mild from critical COVID-19 by innate and adaptive immune single-cell profiling of bronchoalveolar lavages Dysregulation of the immune response affects the outcome of critical COVID-19 patients Respiratory syncytial virus and other respiratory viruses Immune cell profiling of COVID-19 patients in the recovery stage by single-cell sequencing A single-cell atlas of the peripheral immune response in patients with severe COVID-19 Neutralizing antibody responses to SARS-CoV-2 in a COVID-19 recovered patient cohort and their implications. medRxiv 2020.03.30 The role of peripheral blood eosinophil counts in COVID-19 patients Prognostic value of baseline neutrophil-to-lymphocyte ratio in outcome of immune checkpoint inhibitors Suppressed T cell-mediated immunity in patients with COVID-19: A clinical retrospective study in Wuhan Impaired T cell functions along with elevated activated Tregs at the early stage of asymptomatic SARS-CoV-2 infection Immune Phenotyping Based on the Neutrophil-to-Lymphocyte Ratio and IgG Level Predicts Disease Severity and Outcome for Patients With COVID-19 Analysis of clinical characteristics and laboratory findings of 95 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a retrospective analysis Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan The ORF8 protein of SARS-CoV-2 mediates immune evasion through down-regulating MHC-Ι Antibody Responses to SARS-CoV-2 in Patients With Novel Coronavirus Disease Rational targeting of immunosuppressive neutrophils in cancer Plateletto-lymphocyte ratio (PLR) and neutrophil-to-lymphocyte ratio (NLR) are associated with chronic hepatitis B virus (HBV) infection Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients Functional exhaustion of antiviral lymphocytes in COVID-19 patients Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study Acute SARS-CoV-2 Infection Impairs Dendritic Cell and T Cell Responses Pathogenic T cells and inflammatory monocytes incite inflammatory storm in severe COVID-19 patients