T cell responses in patients with - COVID-19 - Nature

Page created by Ricky Gibbs
 
CONTINUE READING
Progress
                                                                                                    typically occurs for only 2–4 days around

T cell responses in patients with                                                                   symptom onset and rapidly recovers15.
                                                                                                    By contrast, COVID-19-​associated
                                                                                                    lymphopenia may be more severe or
COVID-19                                                                                            persistent than in these other infections and
                                                                                                    seems to be more selective for T cell lineages.
                                                                                                    It is possible that the peripheral lymphopenia
Zeyu Chen and E. John Wherry
                                                                                                    observed in patients with COVID-19 reflects
Abstract | The role of T cells in the resolution or exacerbation of COVID-19, as well               recruitment of lymphocytes to the respiratory
                                                                                                    tract or adhesion to inflamed respiratory
as their potential to provide long-​t erm protection from reinfection with
                                                                                                    vascular endothelium. However, although
SARS-​CoV-2, remains debated. Nevertheless, recent studies have highlighted                         autopsy studies of patients’ lungs and
various aspects of T cell responses to SARS-​CoV-2 infection that are starting to                   single-​cell RNA sequencing (scRNA-​seq) of
enable some general concepts to emerge.                                                             bronchoalveolar lavage fluid do identify the
                                                                                                    presence of lymphocytes, the lymphocytic
                                                                                                    infiltration is not excessive5,16. In addition,
COVID-19 caused by SARS-​CoV-2                   observed in patients with severe disease,          a recent scRNA-​seq analysis of the upper
infection is a global pandemic, with more        relevant features of CD8+ versus CD4+ T cell       respiratory tract from patients with
than 15.8 million infections and more than       responses in patients who are hospitalized,        COVID-19 showed that there was a markedly
641,000 deaths as of 25 July 2020, according     features of T cell differentiation that may        decreased contribution of cytotoxic T
to the COVID-19 Map of the Johns Hopkins         be altered and current data on whether the         lymphocytes in patients with severe disease
Coronavirus Resource Center. SARS-​CoV-2         overall magnitude of the T cell response in        compared with those with moderate disease9.
infection can result in a range of clinical      patients with COVID-19 is insufficient or          In severe disease, lymphopenia may be
manifestations, from asymptomatic or             excessive and how these features may relate        associated with high levels of IL-6, IL-10 or
mild infection to severe COVID-19 that           to disease. Finally, we discuss emerging           tumour necrosis factor (TNF)6,10,14, potentially
requires hospitalization. Patients who           data on SARS-​CoV-2-​specific T cells              through a direct effect of these cytokines on
are hospitalized often progress to severe        found in patients who have recovered from          T cell populations17,18 and/or indirect effects
pneumonia and acute respiratory distress         COVID-19 and the implications for T cell           via other cell types, such as dendritic cells19
syndrome (ARDS)1–3. Although relatively          memory. In this rapidly emerging field,            and neutrophils20,21. Hyperactivation of T cells
little is known about the immunology             we summarize the most recent studies               or high levels of expression of pro-​apoptotic
of individuals who are asymptomatic or           that have addressed T cell responses in            molecules, such as FAS (also known as
individuals with mild disease who do not         COVID-19. Some of these are as yet only            CD95)8, TRAIL or caspase 3 (ref.11), could
require hospitalization, recent studies have     available on preprint servers and conclusions      also contribute to T cell depletion.
revealed important insights into the immune      from non-​peer-​reviewed data should be                 Thus, although the mechanisms of
responses of patients who are hospitalized.      treated with caution. A summary of the data        lymphopenia in COVID-19 remain
Similar to other respiratory viral infections,   sets that are published versus those available     incompletely understood, the reduction in
adaptive immune responses4–9, particularly       as preprints is provided in Table 1 to aid the     the number of T cells, in particular, in the
of T cells6,8,10, have a prominent role in       reader in establishing the weight of evidence      periphery is a prominent feature of many
SARS-​CoV-2 infection. However, it remains       for particular features.                           individuals with severe disease. It remains
unclear whether T cell responses are helpful                                                        unclear why the lymphopenia is T cell
or harmful in COVID-19, and whether T cell       Lymphopenia in COVID-19                            biased and perhaps specifically CD8+
responses are suboptimal and dysfunctional       One prominent feature of SARS-​CoV-2               T cell biased. In animal models, lymphopenia
or excessive, with evidence having been          infection is lymphopenia1,6,12,13. Lymphopenia     can augment T cell activation and
provided for both ends of the spectrum.          is associated with severe disease10,12,13 but is   proliferation22. Therefore, determining how
     Here, we summarize some of the recent       reversed when patients recover4,12. In some        lymphopenia in patients with COVID-19
data on conventional T cell responses            patients, lymphopenia has been reported            might impact T cell hyperactivation
in COVID-19, noting, however, that               to affect CD4+ T cells, CD8+ T cells, B cells      and, potentially, immunopathology is an
emerging data also highlight impacts on          and natural killer cells4,6, whereas other data    important future goal, as therapeutics such
other lymphocyte populations, including          suggest that SARS-​CoV-2 infection has a           as IL-7 could be beneficial in this regard.
B cells4,8,11, innate lymphoid cells4, natural   preferential impact on CD8+ T cells8,9,14.
killer cells4,11, mucosa-​associated invariant   Transient lymphopenia is a common feature          CD8+ T cell responses in COVID-19
T cells4 and γδT cells11. We highlight some of   of many respiratory viral infections, such         Early studies of small numbers of patients,
the key observations made for conventional       as with influenza A H3N2 virus, human              or sometimes even a single patient, reported
αβ CD8+ and CD4+ T cells in COVID-19,            rhinovirus or respiratory syncytial virus,         alterations in the activation and/or
including the prominent lymphopenia              but lymphopenia in these other infections          differentiation status of CD8+ T cells in

Nature ReviewS | Immunology                                                                                    volume 20 | September 2020 | 529
Progress

 Table 1 | Studies reporting T cell analysis in patients with COVID-19
 Donor cohort          Sample origin   Profiling technology Major conclusions for αβT cells                                                        Refs
                                       used
 3 healthy, 3 mild/    Bronchoalveolar 10x Genomics           Greater clonal expansion of T cells in moderate disease than severe                       5

 moderate disease,     lavage fluid    scRNA-​seq, 10×        disease; T cells in moderate disease have stronger signatures of tissue
 6 severe disease                      Genomics scTCR-​seq    residency
 8 moderate disease, Nasopharyngeal 10x Genomics              Fewer CTLs in severe disease than moderate disease; hyperactivation of                    9

 11 severe disease   and bronchial  scRNA-​seq                CTLs in the respiratory tract, with a signature of interacting with epithelial
                     samples                                  cells and other immune cell types
 5 healthy,            PBMCs           10x Genomics           Greater clonal expansion of T cells in late-​recovered than in early-​                   28

 5 early-​recovered,                   scRNA-​seq, 10x        recovered patients; fewer CD8+ T cells but greater cytotoxic signatures
 5 late-​recovered                     Genomics scTCR-​seq    in early-​recovered than in late-​recovered patients
 6 healthy,            PBMCs           Seq-​Well scRNA-​seq   Heterogeneity of immune responses, including of interferon-​stimulated                    7

 3 non-​ventilated,                                           genes; no transcriptional signature of exhaustion; features of T cell
 4 with ARDS                                                  hyperactivation in some of the patients with ARDS
 3 healthy, 6 mild/    PBMCs           10x Genomics           Strong T cell lymphopenia in severe disease with potential systemic                      25

 moderate disease,                     scRNA-​seq, flow       adaptive immune dysregulation; altered T cell differentiation and a              (Preprint)
 4 severe disease                      cytometry              hyperactivation stage in severe disease; thymosin α1 can expand the
                                                              memory-​like T cell population and prevent T cell hyperactivation
 15 healthy,           PBMCs           10x Genomics           Similar total and activated T cell counts for influenza and COVID-19                     40

 79 COVID-19                           scRNA-​seq (3          groups; higher IFNα-​responding and IFNγ-​responding signatures in the           (Preprint)
 (15 with ARDS),                       influenza and 4        severe influenza groups than in the COVID-19 groups
 26 influenza                          COVID-19),
 (7 with ARDS)                         flow cytometry
 28 with ARDS,       PBMCs             Flow cytometry         Stronger T cell lymphopenia in more severe COVID-19; lower CD4+ T cell                    6

 26 non-​ARDS, other                                          counts in COVID-19 (n = 17) than in influenza (H1N1; n = 4)
 infection controls
 12 healthy,         PBMCs             High-​dimensional      Stronger T cell lymphopenia in more severe disease; heterogeneity of                      4

 7 recovered,                          flow cytometry         T cell responses related to activation and cytotoxicity signatures; T cells
 7 moderate disease,                                          express more markers of terminal differentiation or exhaustion in severe
 27 severe disease                                            disease
 60 healthy,           PBMCs           High-​dimensional      Stronger T cell lymphopenia in severe disease, with a bias towards CD8+                   8

 36 recovered,                         flow cytometry         T cells; heterogeneity of T cell responses based on high-​dimensional
 125 hospitalized                                             immune profiling, with three potential immune subtypes; T cells more
 patients (NIH                                                activated but also express more markers of terminal differentiation
 ordinal score 2–5)                                           and exhaustion in patients with COVID-19 than in individuals who are
                                                              healthy or who recovered
 40 healthy, 522 with PBMCs            Flow cytometry         Stronger T cell lymphopenia in ICU patients, elderly patients and severe                 10

 varying disease                                              disease, for both CD4+ and CD8+ T cells; IL-6, IL-10 and TNF levels
 severity                                                     negatively correlate with lymphocyte count; T cells express higher levels
                                                              of PD1 and TIM3 in ICU patients than in non-​ICU individuals
 55 healthy,           PBMCs           High-​dimensional      Stronger T cell lymphopenia in severe disease; increased number of                       11

 6 mild disease,                       flow cytometry         hyperactivated proliferating CD4+ and CD8+ T cells in severe disease;            (Preprint)
 26 moderate                                                  increased markers of terminal differentiation or exhaustion in severe
 disease, 31 severe                                           disease compared with milder disease
 disease
 10 moderate           PBMCs           Flow cytometry         Higher lymphocyte counts in moderate disease than in severe disease,                     13

 disease, 11 severe                                           for both CD4+ and CD8+ T cells; more IFNγ-​producing T cells in moderate
 disease                                                      disease than severe disease
 20 healthy, 30 with   PBMCs           Flow cytometry         T cell lymphopenia in patients compared with healthy controls, with                      14

 varying disease                                              an increased ratio of CD4+ T cells to CD8+ T cells; increased proportion
 severity                                                     of terminally differentiated or senescent CD8+ T cells in patients, with
                                                              a reduced proportion of IFNγ-​producing cells; tocilizumab improves
                                                              lymphocyte counts (n = 5)
 30 healthy, 55 mild   PBMCs           Flow cytometry         Stronger T cell lymphopenia in severe disease than in mild disease                       23

 disease, 13 severe                                           or healthy controls, with recovery of T cell numbers in convalescent
 disease                                                      individuals; reduced levels of multiple cytokines in CD8+ T cells in
                                                              disease groups, with higher levels of NKG2A expression than healthy
                                                              controls
 6 healthy, 10 mild    PBMCs           Flow cytometry         Increased cytotoxicity but decreased cytokine secretion of T cells,                      24

 disease, 6 severe                                            particularly CD8+ T cells, in severe disease compared with mild disease;
 disease                                                      CD8+ T cells in severe disease express more inhibitory receptors
 Case report,         PBMCs            Flow cytometry         ICOS+PD1+ circulating TFH cells increase during recovery; activated                      26

 multiple time points                                         CD4+ and CD8+ T cells peak at day 9 post disease onset and decline after
                                                              recovery

530 | September 2020 | volume 20                                                                                                   www.nature.com/nri
Progress

 Table 1 (cont.) | Studies reporting T cell analysis in patients with COVID-19
 Donor cohort             Sample origin         Profiling technology Major conclusions for αβT cells                                                                    Refs
                                                used
 20 healthy,       PBMCs                        Flow cytometry              CD4+ and CD8+ T cells from convalescent patients respond to                                     29

 20 convalescent,                                                           SARS-​CoV-2 epitopes, including S, M and N proteins and other ORFs;
 other common cold                                                          T cell reactivity to SARS-​CoV-2 also detected in non-​exposed donors,
 coronaviruses                                                              with potential cross-​reactivity to other common cold coronaviruses
 14 convalescent          PBMCs                 Flow cytometry              CD4+ and CD8+ T cells from convalescent patients respond to                                     30

                                                                            SARS-​CoV-2 epitopes
 16 healthy,              PBMCs                 Flow cytometry              T cells from convalescent patients with mild or severe disease respond                          31

 28 recovered                                                               to SARS-​CoV-2 epitopes; convalescent patients with mild disease have                  (Preprint)
 from mild disease,                                                         a better memory CD8+ T cell response than convalescent patients with
 14 recovered from                                                          severe disease
 severe disease
 8 healthy, 8 with        PBMCs                 Flow cytometry              T cell lymphopenia in COVID-19 compared with healthy controls, with                             35

 varying disease                                                            an increase of T cell activation phenotypes; SARS-​CoV-2-​specific T cells
 severity                                                                   mainly produce TH1-​type cytokines
 10 healthy,              PBMCs                 Flow cytometry              CD4+ T cells in ICU patients produce more GM-​CSF and IL-6 than                                 37

 21 non-​ICU,                                                               non-​ICU individuals and healthy controls
 12 in ICU
 245 healthy,             PBMCs                 Flow cytometry              Stronger T cell lymphopenia in severe disease compared with mild                                38

 19 mild disease,                                                           disease and healthy controls; IL-6 levels negatively correlate with
 41 severe disease                                                          lymphocyte count; patients who respond to treatment recover
                                                                            lymphocyte numbers
 15 male and              PBMCs                 High-​dimensional           Male and female patients have T cell lymphopenia; female patients                               60

 29 female healthy,                             flow cytometry              have more T cell activation than male patients; male patients with                     (Preprint)
 17 male and                                                                severe disease have greater reduction of T cell activation and loss
 21 female with                                                             of IFNγ-​producers than those with stable disease
 COVID-19
 ARDS, acute respiratory distress syndrome; CTL, cytotoxic T lymphocyte; GM-​CSF, granulocyte–macrophage colony-​stimulating factor; ICOS, inducible co-​stimulator;
 ICU, intensive care unit; IFNγ, interferon-​γ; ORF, open reading frame; PBMC, peripheral blood mononuclear cell; PD1, programmed cell death protein 1; scRNA-​seq,
 single-​cell RNA sequencing; scTCR-​seq, single-​cell T cell receptor sequencing; TFH cell, T follicular helper cell; TH1 cell, T helper 1 cell; TIM3, T cell immunoglobulin
 and mucin domain-​containing protein 3; TNF, tumour necrosis factor.

severe COVID-19 (Fig. 1a). For example,                    observed in many, but not all, patients4,8,11,26.           responses5,16, and recent scRNA-​seq data
there is evidence of terminally differentiated             In other settings, CD38+HLA-​DR+ or Ki67+                   from the upper respiratory tract imply that
T cells or possibly exhausted T cells in                   CD8+ T cells that are present in the blood                  interactions occur there between epithelial
severe disease, with reported increases in                 during the acute phase of a viral infection                 cells and cytotoxic T lymphocytes, in
expression levels of the inhibitory receptors              or after live-​attenuated vaccination contain               particular through an interferon-​γ (IFNγ)
PD1, TIM3, LAG3, CTLA4, NKG2A and                          virus-​specific T cells27, which indicates that             axis9. Indeed, more robust clonal expansion
CD39 (refs4,8,10,11,23,24). However, expression            there might be virus-​specific CD8+ T cell                  of CD8+ T cells in peripheral blood28 or
of these receptors could also reflect recent               responses in those patients with COVID-19                   bronchoalveolar lavage fluid5 may be
activation, and it is not clear whether the                who have robust CD38+HLA-​DR+ or Ki67+                      associated with milder disease or recovery,
T cells in patients with COVID-19 are                      CD8+ T cell responses. However, not all                     although it is not clear whether this CD8+
exhausted or just highly activated. By                     patients have this T cell activation phenotype              T cell clonal expansion is the cause or
contrast, at least one blood scRNA-​seq study              and current data point to potentially diverse               consequence of the disease recovery. Last,
has reported limited expression of inhibitory              patterns of CD8+ T cell responses in patients               there is evidence of SARS-​CoV-2-​specific
receptors by CD8+ T cells in patients                      with COVID-19 (ref.8).                                      CD8+ T cells in patients who have recovered,
with COVID-19 compared with healthy                            Several important considerations arise                  which provides evidence not only of
controls7. In one report, CD8+ T cells from                from these studies: how does heterogeneity                  virus-​specific CD8+ T cell responses but
patients with severe COVID-19 had reduced                  of the CD8+ T cell response relate to disease;              also of CD8+ T cell memory in many
cytokine production upon stimulation23.                    how do T cell responses in peripheral blood                 convalescent patients29–32. The precise role of
Alternatively, other data suggest that                     reflect events in the respiratory tract; and are            these virus-​specific CD8+ T cells in control
CD8+ T cells might have a hyperactivation                  the CD8+ T cell responses antigen specific?                 of initial acute SARS-​CoV-2 infection
signature, including high levels of                        First, multiple studies have indicated                      and their capacity to protect from future
expression of natural killer cell-​related                 heterogeneity in immune responses to                        infection remain to be determined.
markers and increased cytotoxicity4,23,25.                 SARS-​CoV-2, including in CD8+ T cells,
Some of these studies imply the presence                   and emerging data identify potentially                      CD4+ T cell responses in COVID-19
of an overaggressive CD8+ T cell response                  distinct patient immunotypes6,8 that might,                 Similar to CD8+ T cells, there is evidence
or a hyperactive state in patients with                    in some cases, be related to disease features.              of functional impairment and increased
COVID-19 (ref.25). Increased numbers of                    Second, autopsy studies and scRNA-​seq data                 expression of activation and/or exhaustion
CD38+HLA-​DR+ activated CD8+ T cells or                    from bronchoalveolar lavage fluid suggest                   markers by CD4+ T cells in patients with
Ki67+ proliferating CD8+ T cells were also                 the importance of respiratory CD8+ T cell                   COVID-19 (refs10,14) (Fig. 1b). Case reports

Nature ReviewS | Immunology                                                                                                          volume 20 | September 2020 | 531
Progress

a Proposed CD8+ T cell response during COVID-19

                                                                                                                                 Cytokines,
                                  Activated                                                                                      chemokines
                                  T cells                                                  Cell          TCR          Cell cycle and          Co-inhibitory
                 Mild                                                                      number        clonality    state      cytotoxicity signals
                 disease                                           Effector T cells
               IL-2, type I                                        and terminally                                                  IFNγ++
                                                                   differentiated T cells                                                           PD1+/–
               and type III                                                                          Dominant                      IL-2+
                                                                                           Normal or                                               TIM3+/–
               interferon                                                                            clones   Ki67+/–              CD107a++
                                                                                           increased                                               CD38+
                                 Clonal                                                              detected                      TNF+            CD39+/–
                                 expansion                                                                                         GZMB+
                                                                    Memory T cells

 Naive                                                                                                                                             PD1++
              IL-6, IL-10,        Less activated                                                                                   IFNγ +          TIM3++
 T cell       IL-1β, CXCL8        T cells?                                                                                         IL-2+/–         CD38++
              and other                                                                              Lack of                       CD107a+         CD39+
                                                              Terminally    Exhausted-     Strongly
              CXCLs, TNF                                                                             dominant Ki67+/+              GZMB++          NKG2A+
                                                              differentiated like T cell    decreased
                                                                                                     clones                        Perforin++      KLRs+
                 Severe                                       T cell
                                                                                                                                   CCL3+           LAG3+
                 disease                                                                                                           CCL4++          CTLA4+
                                 Reduced                                    Memory                                                 IL-1β+          TIGIT+
                                 T cell pool?                  ?            T cell

b Proposed CD4+ T cell response during COVID-19
                                  Proliferating                                                                                                  Co-stimulatory
                                  TH cells                                                                               Cell      Humoral       and
                 Mild                                                                      Cell           Lineage        cycle     immune        co-inhibitory
                 disease                                            Systemic               number         specificity     state     memory        signals
               IL-2,                                                antiviral response                                                           CD38+/–
               type I and                                                                                 TH1 cell,                              ICOS+/–
               type III                                             Germinal-centre                       TH2 cell,                              CD95–
                                                                                           Normal or                                             CX3CR1+/–
               interferon                                           B cell activation      increased      germinal-      Ki67+/–   Efficient
                                                                                                          centre                                 PD1+/–
                                 Germinal-                                                                TFH cell                               TIM3+/–
                                 centre TFH cell                                                                                                 CD39+/–

 Naive                                                              Plasmablast
 T cell                                                             hyperactivation
                                                               ?
               IL-6, IL-10,     ICOS CD38
                                     +        +
                                                                                                                                                 CD38++
               IL-1β, CXCL8     circulating TFH cells                                                   Circulating
                                                                                                        TFH cell,                                ICOS++
               and other                                                                   Immune                                                CD95+
               CXCLs, TNF                                           Systemic                            TH17 cell,
                                                                    antiviral response     cell subtype hyper-T    1 Ki67
                                                                                                                         +/+
                                                                                                                             Unknown             CX3CR1+
                                                                                           dependent cell (T H                                   PD1+
                 Severe                                                                                        reg
                                                                                                                                                 TIM3+
                 disease         Hyperactivated                                                         cell?)
                                                                                                                                                 CD39+
                                 TH1 cells                                                                                                       TIGIT+
                                                               ?
                                                                    Immunopathology

                               CCR6+ TH17 cell

Fig. 1 | Potential model of T cell responses during COVID-19 progres-            CX3CR1, CX3C-​chemokine receptor 1; CXCL, CXC-​chemokine ligand;
sion. A proposed model of CD8+ T cell responses (a) and CD4+ T cell              GZMB, granzyme B; ICOS, inducible co-​stimulator; IFNγ, interferon-​γ; KLR,
responses (b) during COVID-19 progression in mild versus severe disease.         killer cell lectin-​like receptor; LAG3, lymphocyte activation gene 3; TCR,
Tables show the immune parameters that have been reported to differ              T cell receptor; TFH cell, T follicular helper cell; TH1 cell, T helper 1 cell; TH2
between mild and severe COVID-19. Phenotype data are collated from the           cell, T helper 2 cell; TH17 cell, T helper 17 cell; TIGIT, T cell immunoreceptor
references cited in this Progress article. Results that have been confirmed      with immunoglobulin and ITIM domains; TIM3, T cell immunoglobulin and
by multiple studies are indicated in bold type. CCL, CC-​chemokine ligand;       mucin domain-​containing protein 3; TNF, tumour necrosis factor; Treg cell,
CCR6, CC-​chemokine receptor 6; CTLA4, cytotoxic T lymphocyte antigen 4;         regulatory T cell.

have suggested that CD8+ T cell activation              SARS-​CoV-2 spike protein have been                  indicates a potential role for T helper 17
might be greater than CD4+ T cell activation,           identified in acute infection and have a             (TH17) cell-​mediated immuno­pathology,
as defined by activation markers such as                TH1 cell cytokine profile35. A role for T            whereas another study identified possible
CD38 and HLA-​DR26,33,34. However, another              helper 2 (TH2) cell-​type responses in severe        increases in the number of CD4+ T cells
study identified a subset of patients with              COVID-19 is unclear, although patients               producing transforming growth factor-​β
higher levels of CD4+ T cell activation                 with mild disease may have a normal TH2              (TGFβ)36. A population of CD4+ T cells
who possibly do worse clinically8. One                  cell response11. Given the prominent role of         producing granulocyte–macrophage
report has described a higher proportion                TH2 cell responses in other lung diseases,           colony-​stimulating factor (GM-​CSF) or
of IFNγ-​producing T helper 1 (TH1)-​like               this is an important area for further study.         IL-6 has also been reported in patients with
cells in patients with moderate disease                 Two individual reports have also described           COVID-19 (ref.37), although T cells do not
than in patients with severe disease13.                 a strong CCR6+CD4+ T cell signature                  typically produce IL-6. Regulatory T cells36
Moreover, CD4+ T cells specific for the                 in severe COVID-19 (refs2,34), which                 and ICOS+CD38+ circulating T follicular

532 | September 2020 | volume 20                                                                                                            www.nature.com/nri
Progress

helper (activated cTFH) cells8 may also be        consistent with a recent observation of            may also paradoxically be pro-​inflammatory
altered in patients with COVID-19, with           reduced numbers of plasmacytoid dendritic          and therefore perhaps contribute to
increased numbers of activated cTFH cells         cells11. This observation, however, contrasts      immunopathology. Older patients
in some settings perhaps connected to             with the robust and consistent upregulation        experience more severe lymphopenia during
a reported increase in the number of              of the interferon-​inducible chemokine             COVID-19 (ref.10), although it is not clear
circulating plasmablasts4.                        CXCL10 observed in one patient cohort11,           whether ageing-​associated lymphopenia is
    Lymphopenia also affects CD4+ T cells,        perhaps owing to non-​interferon inducers          causal to disease severity or vice versa.
although some studies suggest that the            of expression of this chemokine.                       Men with COVID-19 have higher
impact is less than that for CD8+ T cells8,38.        Another key gap in our understanding is        rates of hospitalization and mortality than
It remains to be determined how                   the relationship between viral load and the        women58, and among severe cases of disease,
lym­phopenia might relate to CD4+ T cell          immune response. Viral load could have a           men have more severe lymphopenia59.
activation and/or dysfunction. Moreover, in       major impact on the magnitude and quality          There may also be a bias to stronger CD4+
addition to memory CD8+ T cells, patients         of the T cell response, and future studies that    and CD8+ T cell activation in women with
who have recovered from SARS-​CoV-2               quantify virus replication should provide          COVID-19 (ref.60). It is unclear whether these
infection have virus-​specific memory             context for understanding evolving T cell          sex biases relate to X chromosome-​encoded
CD4+ T cells29–32, which bodes well for the       responses during SARS-​CoV-2 infection.            immune genes58 and/or the role of sex
possibility of T cell memory and, perhaps,        A key feature of patients with COVID-19            hormones in regulating immune responses61.
protective immunity. In individuals               is the enrichment for co-​morbidities such         Nevertheless, these data highlight the
who recovered from mild COVID-19,                 as hypertension and diabetes42 (see next           importance of accounting for sex in ongoing
CD4+ T cells gained a typical memory              section), but it remains to be defined             clinical trials.
phenotype with high levels of expression of       how an individual patient’s background                 A very high proportion of patients
IL-7Rα (ref.32). Interestingly, cross-​reactive   ‘immune health’ landscape shapes responses         with COVID-19 who are hospitalized have
memory CD4+ T cells are also found in             to SARS-​CoV-2 infection. Although                 one or more underlying cardiovascular
subjects who have never been exposed to           lymphopenia is not a unique feature of             co-​morbidities44,45,62–64. Moreover, many
SARS-​CoV-2 (ref.29). It is unclear how these     SARS-​CoV-2 infection43, it may be more            of the manifestations of severe disease
pre-​existing memory CD4+ T cells, which          prolonged in patients with COVID-19,               relate to coagulopathies and vascular
are presumably generated in response              and lymphopenia-​induced proliferation             complications62,64. Even the newly recognized
to human endemic coronaviruses, affect            can clearly affect T cell differentiation and      SARS-​CoV-2-​associated multisystem
immunity or pathology upon SARS-​CoV-2            activation; however, the contribution of           inflammatory syndrome in children has
infection. There are other settings in which      lymphopenia to T cell differentiation in           similarities to Kawasaki disease, which
the presence of memory CD4+ T cells               patients with COVID-19 has not yet been            often involves inflammation of the coronary
in the absence of effective CD8+ T cells or       investigated.                                      artery65,66. Other viral infections, including
antibodies can cause pathology39. Thus,                                                              Ebola virus and virulent lymphocytic
CD4+ T cell responses are present in patients     Co-​morbidities and COVID-19                       choriomeningitis virus in mice, can cause
with COVID-19 and perhaps form memory             Co-​morbidities such as cardiovascular             damage to the vascular endothelium. These
following resolution of the disease. Whether      disease44,45, diabetes42 and obesity46 are         viruses infect the vascular endothelium,
CD4+ T cells responding in acute infection        some of the most common underlying                 and T cells can then cause vascular damage
with SARS-​CoV-2 are functionally impaired        conditions associated with worse clinical          by attacking the virus-​infected cells67–69.
or hyperactivated, and how these responses        outcome and severe COVID-19. Moreover,             However, disease presentation in these
relate to disease outcome, remain important       older patients experience greater clinical         settings is distinct from that for COVID-19,
unanswered questions.                             severity of COVID-19 (refs1,47), males             and extensive SARS-​CoV-2 infection of
                                                  may experience more severe disease than            the vascular endothelium is unlikely in
T cell differentiation in COVID-19                females1,47 and genetic variations, including      most patients.
Most acute viral infections in humans             the ABO blood type8,48,49, have been reported          Obesity also contributes to more
induce the activation and proliferation           to affect the clinical outcomes for patients       severe COVID-19 and higher rates of
of both CD4+ T cells and CD8+ T cells,            with COVID-19. However, how these                  hospitalization46,70, which are perhaps
so SARS-​CoV-2 infection may not be unique        co-​morbidities are associated with T cell         associated with the role of body mass index
in this regard. However, hyperactivation          responses during COVID-19 remains                  in cardiovascular health. However, obesity
or hypoactivation of T cells, or skewing          largely unknown.                                   can also directly affect T cell responses
towards an ineffective differentiation state          Advanced age is a common co-​morbidity         to influenza vaccination71 or infection72,
(for example, TH17 cells, exhausted T cells or    for severity of disease during respiratory         and a role for obesity in altering T cell
terminally differentiated T cells), might not     viral infections, and disease severity             differentiation has been observed in asthma73
be optimal in some patients with COVID-19.        may be associated with altered T cell              and chronic inflammation74. Nevertheless,
Several features that have been described         responses50,51. Ageing affects T cell repertoire   the precise role of T cells in driving the
in patients with severe COVID-19 —                diversity52, both for CD4+ T cells53 and for       coagulopathies or inflammation that are
including high levels of systemic cytokines       CD8+ T cells54. This age-​related reduction        characteristic of severe COVID-19 in
or chemokines, most notably IL-6, CXCL8,          in T cell clonal diversity is associated with      patients with cardiovascular co-​morbidities
CXCL9 and CXCL10 (refs8,11,40), or delayed        impaired responses to viral infections such        is unclear.
or defective type I interferon responses41 —      as influenza55. Advanced age can also be               Finally, a recent genetic association study
could potentially skew T cell responses.          associated with T cell senescence, which           compared patients with severe COVID-19
The potential type I interferon deficiency        perhaps contributes to ineffective responses       and ARDS with healthy controls49. Several
described in patients with COVID-19 is            to infections56,57. However, senescent T cells     chemokine receptor genes, including CCR9,

Nature ReviewS | Immunology                                                                                     volume 20 | September 2020 | 533
Progress

CXCR6 and XCR1, and the locus controlling         different stages of disease or with different     be important. Although evidence of
the ABO blood type were identified as being       immune response characteristics and then          T cell memory to other coronaviruses is
associated with severe disease. Whether           to match them with treatment strategies.          encouraging, such immune natural history
these genes are directly or indirectly related    Such a precision medicine strategy                studies for SARS-​CoV-2 will likely be
to T cell responses in COVID-19, however,         based on analysis of ‘immune health’              valuable for examining vaccine-​induced
remains unknown and needs further                 could be used to better tailor the use of         T cell responses.
investigation.                                    immunostimulatory strategies such as
    Thus, although severe COVID-19 is             thymosin α1 (ref.25) or type I interferon75       Conclusions
partially characterized by patients with          versus immunosuppressive drugs such               The accumulating evidence supports a role
co-​morbidities, how these co-​morbidities        as tocilizumab76–78, ruxolitinib79 or             for T cells in COVID-19 and probably in
relate to T cell responses remains poorly         dexamethasone80 to patients who will receive      the immunological memory that forms
understood. Causal associations may               most benefit. These efforts should be aided       following recovery from SARS-​CoV-2
exist, perhaps connected to some of the           by the development of better preclinical          infection. Most, although not all, patients
underlying genetics. However, it is also          models, including human ACE2-​expressing          who are hospitalized seem to mount both
possible that certain co-​morbidities             mice81–85. It will be interesting in the future   CD8+ and CD4+ T cell responses, and
or pre-​existing conditions make                  to use such models to address key questions       evidence points to possible suboptimal,
patients with severe disease less able to         including: what is an ‘appropriate’ profile       excessive or otherwise inappropriate T cell
tolerate the severe virus-​mediated and           of T cell activation and differentiation for a    responses associated with severe disease.
immune-​mediated pathology associated             successful antiviral response to SARS-​CoV-2      In fact, multiple distinct patterns of T cell
with SARS-​CoV-2 infection.                       without triggering severe pathology;              response may exist in different patients,
                                                  how does viral load or initial inoculum of        which suggests the possibility of distinct
Strength of COVID-19 T cell response              SARS-​CoV-2 impact T cell responses and           clinical approaches tailored to the particular
The published data discussed here indicate        pathology; how do T cell responses in the         immunotype of a specific patient. Much
that patients with severe COVID-19 can            blood relate to lung-​infiltrating T cells,       of the available data on T cell responses
have either insufficient or excessive T cell      considering that many respiratory viruses         in patients with COVID-19 who are
responses. It is possible, therefore, that        are controlled by tissue-​resident immune         hospitalized is still available only in preprint
disease might occur in different patients         cells that may not be present in the blood;       form. This format has been essential for
at either end of this immune response             and what are the molecular mechanisms             rapid dissemination of information in the
spectrum, in one case from virus-​mediated        that support a successful T cell response         setting of pandemic urgency. However,
pathology and in the other case from              during SARS-​CoV-2 infection?                     as these studies move through peer review,
T cell-​driven immunopathology4. However,                                                           we should gain increasing confidence and
it is unclear why some patients respond           T cell memory in COVID-19                         clarity about the nature of T cell responses
too little and some patients too much, and        A key question is whether protective              to SARS-​CoV-2 infection. More carefully
whether the strength of the T cell response       T cell memory can form following either           defining distinct classes of T cell response
in the peripheral blood reflects the T cell       SARS-​CoV-2 infection or vaccination.             types in COVID-19 and delineating how
response intensity in the respiratory tract and   Although data on vaccination will await           pre-​existing conditions, co-​morbidities,
other SARS-​CoV-2-​infected organs. Some          trial results, early data from patients           race, ‘immune health’ status and other
information exists on SARS-​CoV-2-​specific       with COVID-19 who have recovered are              variables influence T cell responses should
T cells in patients who are hospitalized, and     promising. Memory CD4+ T cells and                reveal novel opportunities for treatment
approaches using activation markers (such         CD8+ T cells were detected in 100%                and prevention.
as Ki67) likely capture virus-​specific T cell    and 70% of patients who recovered,                Zeyu Chen and E. John Wherry          ✉
responses; recent studies are beginning to fill   respectively29. Moreover, memory T cell
                                                                                                    Department of Systems Pharmacology and
in this picture by identifying virus-​specific    responses were detected for multiple              Translational Therapeutics, Institute for Immunology,
CD4+ T cells and CD8+ T cells in acute            SARS-​CoV-2 proteins, including not only          Perelman School of Medicine, University of
disease5,29,30. However, one missing set of       spike protein but also nucleoprotein and          Pennsylvania, Philadelphia, PA, USA.
information is a careful immune analysis          membrane protein31. Whether these T cells         ✉e-​mail: wherry@pennmedicine.upenn.edu

of outpatients who, during acute infection        provide protective immunity is unclear,           https://doi.org/10.1038/s41577-020-0402-6
with SARS-​CoV-2, are undergoing a                and addressing this question for T cells          Published online 29 July 2020
‘successful’ immune response. Moreover,           alone will be confounded by the presence of       1.   Huang, C. et al. Clinical features of patients infected
it will be crucial to incorporate into any        SARS-​CoV-2-​specific antibodies in patients           with 2019 novel coronavirus in Wuhan, China.
                                                                                                         Lancet 395, 497–506 (2020).
model of COVID-19 how the magnitude and           who recover. Nevertheless, ongoing analysis       2.   Xu, Z. et al. Pathological findings of COVID-19
quality of T cell responses relate to signals     of patients who have recovered should                  associated with acute respiratory distress syndrome.
                                                                                                         Lancet Resp. Med. 8, 420–422 (2020).
from other parts of the immune system, such       provide insights into the protective capacity     3.   Matthay, M. A., Aldrich, J. M. & Gotts, J. E.
as innate cytokines, antigen presentation         of immune memory, including humoral                    Treatment for severe acute respiratory distress
                                                                                                         syndrome from COVID-19. Lancet Resp. Med. 8,
and myeloid cells. Lastly, the strength of the    and cellular memory, as these individuals              433–434 (2020).
T cell response may also relate to the stage      are potentially re-​exposed to SARS-​CoV-2        4.   Kuri-​Cervantes, L. et al. Comprehensive mapping
                                                                                                         of immune perturbations associated with severe
of disease or infection. Accurately assessing     in the community. Further studies should               COVID-19. Sci. Immunol. 5, eabd7114 (2020)
the different ‘phases’ of acute SARS-​CoV-2       also define the differentiation state and         5.   Liao, M. et al. Single-​cell landscape of bronchoalveolar
                                                                                                         immune cells in patients with COVID-19. Nat. Med. 8,
infection may also be valuable in interpreting    durability of T cell memory. Moreover,                 842–844 (2020).
T cell responses.                                 defining how T cell memory forms in               6.   Giamarellos-​Bourboulis, E. J. et al. Complex immune
                                                                                                         dysregulation in COVID-19 patients with severe
     Thus, future studies should provide          patients who experience mild symptoms                  respiratory failure. Cell Host Microbe 27, 992–1000
insights into how to identify patients at         of COVID-19 versus severe disease will                 (2020).

534 | September 2020 | volume 20                                                                                                       www.nature.com/nri
Progress

7.    Wilk, A. J. et al. A single-​cell atlas of the peripheral          lack of terminal differentiation, and high proliferative    58. Scully, E. P., Haverfield, J., Ursin, R. L., Tannenbaum, C.
      immune response in patients with severe COVID-19.                  potential. Preprint at bioRxiv https://doi.org/10.1101/         & Klein, S. L. Considering how biological sex impacts
      Nat. Med. 26, 1070–1076 (2020).                                    2020.06.08.138826 (2020).                                       immune responses and COVID-19 outcomes. Nat. Rev.
8.    Mathew, D. et al. Deep immune profiling of COVID-19          33.   Vabret, N. et al. Immunology of COVID-19:                       Immunol. 20, 442–447 (2020).
      patients reveals distinct immunotypes with therapeutic             current state of the science. Immunity 52, 910-941          59. Meng, Y. et al. Sex-​specific clinical characteristics
      implications. Science https://doi.org/10.1126/                     (2020).                                                         and prognosis of coronavirus disease-19 infection in
      science.abc8511 (2020).                                      34.   Qin, C. et al. Dysregulation of immune response in              Wuhan, China: a retrospective study of 168 severe
9.    Chua, R. L. et al. COVID-19 severity correlates with               patients with coronavirus 2019 (COVID-19) in Wuhan,             patients. PLoS Pathog. 16, e1008520 (2020).
      airway epithelium–immune cell interactions identified              China. Clin. Infect. Dis. https://doi.org/10.1093/cid/      60. Takahashi, T. et al. Sex differences in immune
      by single-​cell analysis. Nat. Biotech. https://doi.org/           ciaa248 (2020).                                                 responses to SARS-​CoV-2 that underlie disease
      10.1038/s41587-020-0602-4 (2020).                            35.   Weiskopf, D. et al. Phenotype and kinetics of                   outcomes. Preprint at medRxiv https://doi.org/
10.   Diao, B. et al. Reduction and functional exhaustion                SARS-​CoV-2-specific T cells in COVID-19 patients with          10.1101/2020.06.06.20123414 (2020).
      of T cells in patients with coronavirus disease 2019               acute respiratory distress syndrome. Sci. Immunol. 5,       61. Bouman, A., Heineman, M. J. & Faas, M. M.
      (COVID-19). Front. Immunol. 11, 827 (2020).                        eabd2071 (2020).                                                Sex hormones and the immune response in humans.
11.   Laing, A. G. et al. A consensus Covid-19 immune              36.   Wang, W. et al. High-​dimensional immune profiling              Hum. Reprod. Update 11, 411–423 (2005).
      signature combines immuno-​protection with discrete                by mass cytometry revealed immunosuppression                62. Zheng, Y.-Y., Ma, Y.-T., Zhang, J.-Y. & Xie, X. COVID-19
      sepsis-​like traits associated with poor prognosis.                and dysfunction of immunity in COVID-19 patients.               and the cardiovascular system. Nat. Rev. Cardiol. 17,
      Preprint at medRxiv https://doi.org/10.1101/                       Cell. Mol. Immunol. 17, 650–652 (2020).                         259–260 (2020).
      2020.06.08.20125112 (2020).                                  37.   Zhou, Y. et al. Pathogenic T-​cells and inflammatory        63. Magro, C. et al. Complement associated microvascular
12.   Tan, L. et al. Lymphopenia predicts disease severity               monocytes incite inflammatory storms in severe                  injury and thrombosis in the pathogenesis of severe
      of COVID-19: a descriptive and predictive study.                   COVID-19 patients. Natl Sci. Rev. 7, 998–1002                   COVID-19 infection: a report of five cases. Transl Res.
      Signal. Transduct. Target. Ther. 5, 33 (2020).                     (2020).                                                         220, 1–13 (2020).
13.   Chen, G. et al. Clinical and immunological features          38.   Wang, F. et al. Characteristics of peripheral lymphocyte    64. Klok, F. A. et al. Incidence of thrombotic complications
      of severe and moderate coronavirus disease 2019.                   subset alteration in COVID-19 pneumonia. J. Infect. Dis.        in critically ill ICU patients with COVID-19. Thromb. Res.
      J. Clin. Invest. 130, 2620–2629 (2020).                            221, 1762–1769 (2020).                                          191, 145–147 (2020).
14.   Mazzoni, A. et al. Impaired immune cell cytotoxicity         39.   Penaloza-​MacMaster, P. et al. Vaccine-​elicited CD4        65. Viner, R. M. & Whittaker, E. Kawasaki-​like disease:
      in severe COVID-19 is IL-6 dependent. J. Clin. Invest.             T cells induce immunopathology after chronic LCMV               emerging complication during the COVID-19 pandemic.
      https://doi.org/10.1172/JCI138554 (2020).                          infection. Science 347, 278–282 (2015).                         Lancet 395, 1741–1743 (2020).
15.   McClain, M. T. et al. Longitudinal analysis of leukocyte     40.   Mudd, P. A. et al. Targeted immunosuppression               66. Belhadjer, Z. et al. Acute heart failure in multisystem
      differentials in peripheral blood of patients with acute           distinguishes COVID-19 from influenza in moderate               inflammatory syndrome in children (MIS-​C) in the
      respiratory viral infections. J. Clin. Virol. 58, 689–695          and severe disease. Preprint at medRxiv https://                context of global SARS-​CoV-2 pandemic. Circulation
      (2013).                                                            doi.org/10.1101/2020.05.28.20115667 (2020).                     https://doi.org/10.1161/CIRCULATIONAHA.120.
16.   Wichmann, D. et al. Autopsy findings and venous              41.   Blanco-​Melo, D. et al. Imbalanced host response                048360 (2020).
      thromboembolism in patients with COVID-19:                         to SARS-​CoV-2 drives development of COVID-19.              67. Frebel, H. et al. Programmed death 1 protects
      a prospective cohort study. Ann. Intern. Med.                      Cell 181, 1036–1045 (2020).                                     from fatal circulatory failure during systemic virus
      https://doi.org/10.7326/M20-2003 (2020).                     42.   Fang, L., Karakiulakis, G. & Roth, M. Are patients              infection of mice. J. Exp. Med. 209, 2485–2499
17.   Taga, K. & Tosato, G. IL-10 inhibits human T cell                  with hypertension and diabetes mellitus at increased            (2012).
      proliferation and IL-2 production. J. Immunol. 148,                risk for COVID-19 infection? Lancet Resp. Med. 8,           68. Yang, Z. Y. et al. Identification of the Ebola virus
      1143–1148 (1992).                                                  e21 (2020).                                                     glycoprotein as the main viral determinant of vascular
18.   Böttcher, J. P. et al. IL-6 trans-​signaling-dependent       43.   Panesar, N. S. Lymphopenia in SARS. Lancet 361,                 cell cytotoxicity and injury. Nat. Med. 6, 886–889
      rapid development of cytotoxic CD8+ T cell function.               1985 (2003).                                                    (2000).
      Cell Rep. 8, 1318–1327 (2014).                               44.   Li, B. et al. Prevalence and impact of cardiovascular       69. Wolf, T. et al. Severe Ebola virus disease with vascular
19.   Malefyt, R. et al. Interleukin 10 (IL-10) and viral                metabolic diseases on COVID-19 in China. Clin. Res.             leakage and multiorgan failure: treatment of a
      IL-10 strongly reduce antigen-​specific human T cell               Cardiol. 109, 531–538 (2020).                                   patient in intensive care. Lancet 385, 1428–1435
      proliferation by diminishing the antigen-​presenting         45.   Guo, T. et al. Cardiovascular implications of fatal             (2015).
      capacity of monocytes via downregulation of class II               outcomes of patients with coronavirus disease 2019          70. Stefan, N., Birkenfeld, A. L., Schulze, M. B. &
      major histocompatibility complex expression.                       (COVID-19). JAMA Cardiol. 5, 811-818 (2020).                    Ludwig, D. S. Obesity and impaired metabolic health
      J. Exp. Med. 174, 915–924 (1991).                            46.   Lighter, J. et al. Obesity in patients younger than 60          in patients with COVID-19. Nat. Rev. Endocrinol. 16,
20.   Liu, Y. et al. Neutrophil-​to-lymphocyte ratio as an               years is a risk factor for COVID-19 hospital admission.         341–342 (2020).
      independent risk factor for mortality in hospitalized              Clin. Infect. Dis. https://doi.org/10.1093/cid/ciaa415      71. Sheridan, P. A. et al. Obesity is associated with
      patients with COVID-19. J. Infect. 81, e6–e12 (2020).              (2020).                                                         impaired immune response to influenza vaccination
21.   Ma, Y. et al. Predictive value of the neutrophil-​to-        47.   Richardson, S. et al. Presenting characteristics,               in humans. Int. J. Obes. 36, 1072–1077 (2012).
      lymphocyte ratio (NLR) for diagnosis and worse clinical            comorbidities, and outcomes among 5700 patients             72. Paich, H. A. et al. Overweight and obese adult
      course of the COVID-19: findings from ten provinces                hospitalized with COVID-19 in the New York City area.           humans have a defective cellular immune response
      in China. Preprint at SSRN https://doi.org/10.2139/                JAMA 323, 2052–2059 (2020).                                     to pandemic H1N1 influenza A virus. Obesity 21,
      ssrn.3569838 (2020).                                         48.   Zhao, J. et al. Relationship between the ABO blood              2377–2386 (2013).
22.   Surh, C. D. & Sprent, J. Homeostasis of naive and                  group and the COVID-19 susceptibility. Preprint             73. Leiria, L. O. S., Martins, M. A. & Saad, M. J. A.
      memory T cells. Immunity 29, 848–862 (2008).                       at medRxiv https://doi.org/10.1101/2020.03.11.                  Obesity and asthma: beyond TH2 inflammation.
23.   Zheng, M. et al. Functional exhaustion of antiviral                20031096 (2020).                                                Metabolism 64, 172–181 (2015).
      lymphocytes in COVID-19 patients. Cell. Mol. Immunol.        49.   Ellinghaus, D. et al. Genomewide association study          74. DeFuria, J. et al. B cells promote inflammation in
      17, 533–535 (2020).                                                of severe Covid-19 with respiratory failure. N. Engl.           obesity and type 2 diabetes through regulation of
24.   Zheng, H.-Y. et al. Elevated exhaustion levels and                 J. Med. https://doi.org/10.1056/NEJMoa2020283                   T-​cell function and an inflammatory cytokine profile.
      reduced functional diversity of T cells in peripheral              (2020).                                                         Proc. Natl Acad. Sci. USA 110, 5133–5138 (2013).
      blood may predict severe progression in COVID-19             50.   Lee, N., Shin, M. S. & Kang, I. T-​cell biology in aging,   75. Sallard, E., Lescure, F.-X., Yazdanpanah, Y., Mentre, F.
      patients. Cell. Mol. Immunol. 17, 541–543 (2020).                  with a focus on lung disease. J. Gerontol. A Biol. Sci.         & Peiffer-​Smadja, N. Type 1 interferons as a potential
25.   Yu, K. et al. Thymosin α-1 protected T cells from                  Med. Sci. 67, 254–263 (2012).                                   treatment against COVID-19. Antivir. Res. 178,
      excessive activation in severe COVID-19. Preprint at         51.   Goronzy, J. J. & Weyand, C. M. Successful and                   104791 (2020).
      Research Square https://doi.org/10.21203/                          maladaptive T cell aging. Immunity 46, 364–378              76. Xu, X. et al. Effective treatment of severe COVID-19
      rs.3.rs-25869/v1 (2020).                                           (2017).                                                         patients with tocilizumab. Proc. Natl Acad. Sci. USA
26.   Thevarajan, I. et al. Breadth of concomitant immune          52.   Naylor, K. et al. The influence of age on T cell                117, 10970–10975 (2020).
      responses prior to patient recovery: a case report                 generation and TCR diversity. J. Immunol. 174,              77. Luo, P. et al. Tocilizumab treatment in COVID-19:
      of non-​severe COVID-19. Nat. Med. 26, 453–455                     7446–7452 (2005).                                               a single center experience. J. Med. Virol. 92, 814–818
      (2020).                                                      53.   Pourgheysari, B. et al. The cytomegalovirus-​specific           (2020).
27.   Miller, J. D. et al. Human effector and memory                     CD4+ T-​cell response expands with age and markedly         78. Guo, C. et al. Tocilizumab treatment in severe COVID-19
      CD8+ T cell responses to smallpox and yellow fever                 alters the CD4+ T-​cell repertoire. J. Virol. 81,               patients attenuates the inflammatory storm incited
      vaccines. Immunity 28, 710–722 (2008).                             7759–7765 (2007).                                               by monocyte centric immune interactions revealed by
28.   Wen, W. et al. Immune cell profiling of COVID-19             54.   Messaoudi, I., Lemaoult, J., Guevara-​Patino, J. A.,            single-​cell analysis. Preprint at bioRxiv https://doi.org/
      patients in the recovery stage by single-​cell sequencing.         Metzner, B. M. & Nikolich-​Zugich, J. Age-​related CD8          10.1101/2020.04.08.029769 (2020).
      Cell Discov. 6, 31 (2020).                                         T cell clonal expansions constrict CD8 T cell repertoire    79. Cao, Y. et al. Ruxolitinib in treatment of severe
29.   Grifoni, A. et al. Targets of T cell responses to                  and have the potential to impair immune defense.                coronavirus disease 2019 (COVID-19): a multicenter,
      SARS-​CoV-2 coronavirus in humans with COVID-19                    J. Exp. Med. 200, 1347–1358 (2004).                             single-​blind, randomized controlled trial. J. Allergy
      disease and unexposed individuals. Cell 181,                 55.   Yager, E. J. et al. Age-​associated decline in T cell           Clin. Immunol. 146, 137–146 (2020).
      1489–1501.e15 (2020).                                              repertoire diversity leads to holes in the repertoire       80. Abdolahi, N. et al. Letter to the editor: efficacy
30.   Ni, L. et al. Detection of SARS-​CoV-2-specific humoral            and impaired immunity to influenza virus. J. Exp. Med.          of different methods of combination regimen
      and cellular immunity in COVID-19 convalescent                     205, 711–723 (2008).                                            administrations including dexamethasone, intravenous
      individuals. Immunity 52, 971–977 (2020).                    56.   Goronzy, J. J., Li, G., Yu, M. & Weyand, C. M.                  immunoglobulin, and interferon-​β to treat critically ill
31.   Dong, T. et al. Broad and strong memory CD4                        Signaling pathways in aged T cells—a reflection                 COVID-19 patients: a structured summary of a study
      and CD8 T cells induced by SARS-​CoV-2 in UK                       of T cell differentiation, cell senescence and host             protocol for a randomized controlled trial. Trials 21,
      convalescent COVID-19 patients. Preprint at bioRxiv                environment. Semin. Immunol. 24, 365–372 (2012).                549 (2020).
      https://doi.org/10.1101/2020.06.05.134551 (2020).            57.   Yu, H. T., Park, S., Shin, E.-C. & Lee, W.-W. T cell        81. Goldman-​Israelow, B., Song, E., Mao, T., Lu, P.
32.   Neidleman, J. et al. SARS-​CoV-2-specific T cells exhibit          senescence and cardiovascular diseases. Clin. Exp. Med.         & Meir, A. Mouse model of SARS-​CoV-2 reveals
      unique features characterized by robust helper function,           16, 257–263 (2016).                                             inflammatory role of type I interferon signaling.

Nature ReviewS | Immunology                                                                                                                         volume 20 | September 2020 | 535
Progress

      Preprint at bioRxiv https://doi.org/10.1101/2020.         Acknowledgements                                                 Peer review information
      05.27.118893 (2020).                                      Z.C. is supported by grant CA234842 from the National            Nature Reviews Immunology thanks Susan L. Swain and the
82.   Sun, J. et al. Generation of a broadly useful model for   Institutes of Health (NIH). Work in the Wherry laboratory        other, anonymous, reviewer(s) for their contribution to
      COVID-19 pathogenesis, vaccination, and treatment.        is supported by NIH grant AI082630, the Allen Institute          the peer review of this work.
      Cell https://doi.org/10.1016/j.cell.2020.06.010           for Immunology and the Parker Institute for Cancer
      (2020).                                                   Immunotherapy.                                                   Publisher’s note
83.   Hassan, A. O. et al. A SARS-​CoV-2 infection model                                                                         Springer Nature remains neutral with regard to jurisdictional
      in mice demonstrates protection by neutralizing           Author contributions                                             claims in published maps and institutional affiliations.
      antibodies. Cell https://doi.org/10.1016/                 The authors contributed equally to all aspects of the article.
      j.cell.2020.06.011 (2020).
84.   Bao, L. et al. The pathogenicity of SARS-​CoV-2           Competing interests                                               Related links
      in hACE2 transgenic mice. Nature https://doi.org/         E.J.W. has consulting agreements with and/or is a scientific      CoVID-19 Map of the Johns Hopkins Coronavirus resource
      10.1038/s41586-020-2312-y (2020).                         advisor for Merck, Roche, Pieris, Elstar and Surface Oncology.    Center: https://coronavirus.jhu.edu/map.html
85.   Jiang, R.-D. et al. Pathogenesis of SARS-​CoV-2 in        E.J.W. has a patent licensing agreement on the PD1 pathway
      transgenic mice expressing human angiotensin-​            with Roche/Genentech. E.J.W. is a founder of Arsenal
      converting enzyme 2. Cell 182, 50–58 (2020).              Biosciences. Z.C. declares no competing interests.               © Springer Nature Limited 2020

536 | September 2020 | volume 20                                                                                                                                   www.nature.com/nri
You can also read