Skip to main content
Immunotherapy Advances logoLink to Immunotherapy Advances
. 2021 Jul 2;1(1):ltab015. doi: 10.1093/immadv/ltab015

Peripheral T cell lymphopenia in COVID-19: potential mechanisms and impact

Sifan Zhang 1, Becca Asquith 2, Richard Szydlo 3, John S Tregoning 4,#, Katrina M Pollock 5,#,
PMCID: PMC9364037  PMID: 35965490

Summary

Immunopathogenesis involving T lymphocytes, which play a key role in defence against viral infection, could contribute to the spectrum of COVID-19 disease and provide an avenue for treatment. To address this question, a review of clinical observational studies and autopsy data in English and Chinese languages was conducted with a search of registered clinical trials. Peripheral lymphopenia affecting CD4 and CD8 T cells was a striking feature of severe COVID-19 compared with non-severe disease. Autopsy data demonstrated infiltration of T cells into organs, particularly the lung. Seventy-four clinical trials are on-going that could target T cell-related pathogenesis, particularly IL-6 pathways. SARS-CoV-2 infection interrupts T cell circulation in patients with severe COVID-19. This could be due to redistribution of T cells into infected organs, activation induced exhaustion, apoptosis, or pyroptosis. Measuring T cell dynamics during COVID-19 will inform clinical risk-stratification of hospitalised patients and could identify those who would benefit most from treatments that target T cells.

Keywords: CD4 cell, CD8 cell, SARS-CoV-2 virus, COVID-19, T cell biology

Graphical Abstract

graphic file with name ltab015_iffig1.jpg

In this review, we demonstrate that CD4+ and CD8+ T cell lymphopenia is a feature of severe COVID-19 compared with moderate disease in hospitalised patients. Pathology data indicate T cell infiltration into infected organs, particularly the lung. Potential mechanisms affecting T cells in severe disease due to SARS-CoV-2 infection include T cell redistribution and sequestration, pyroptosis, and apoptosis.

Introduction

SARS-CoV-2 is the causative agent of the COVID-19 pandemic, responsible for a global health crisis unprecedented in recent times. A number of immunological, pathological, and histological studies indicate a role for T cells in the pathogenesis underlying COVID-19 [1–3]. However, the speed and spread of SARS-CoV-2, coupled with challenges in collecting experimental clinical evidence means that characterisation of immunopathogenesis has thus far been limited.

CD4+ and CD8+ T cells play key roles in containing and resolving viral infections. Suppression of T cell responses is associated with a failure to achieve sterilising immunity against viral infection, classically demonstrated by HIV infection [4, 5]. Viral pathogenicity can suppress T cell function through a number of mechanisms including direct and indirect cytotoxicity, organ-based sequestration, and suppression of both antigen recognition and the downstream effector mechanisms that contain infection [6–10]. Damage to the T cell compartment can also have longer term clinical and immunological sequelae, limiting responses to other pathogens even in recovered individuals [11–14].

Evidence from observational studies indicates that immunopathology is an important driver of the clinical features of severe COVID-19 [15, 16]. The mechanisms of this immunopathology in COVID-19 are unclear. An excess inflammatory response has been widely observed and is associated with the later stages of disease and multi-organ failure. Dysregulation of both CD8+ and CD4+ T cell circulation and function within specific tissues could contribute to this immune injury.

The quality and quantity of T cell function is important in all stages of SARS-CoV-2 infection, once viral replication is established, including viral containment, the resolution of infection and recovery. Severe COVID-19 therefore mainly represents a failure of normal T cell function to contain and resolve SARS-CoV-2 infection. We systematically reviewed clinical observational data from histopathology and immunological studies of autopsies and in vivo clinical observational cohorts to identify whether the data gathered so far supports this idea.

Search strategy and selection criteria

Observational studies for this review were identified through a search of PubMed for articles published from 1 December 2019 to 31 December 2020, by use of the terms ‘COVID-19’ and ‘T cells’. Articles published in English and Chinese were included. Observational studies were selected if they reported original T cell counts in patients with severe and non-severe COVID-19. The selection process is shown as a flow chart (Fig. 1).

Figure 1.

Figure 1.

Flow chart of the study selection process.

Autopsy studies for this review were identified through a search of PubMed for articles published from 1 December 2019 to 31 December 2020, by use of the terms ‘COVID-19’ and ‘Autopsy’. Articles published in English and Chinese were included. The inclusion criteria for post-mortem studies were measurement of T lymphocytes in patients who died of COVID-19 by haematoxylin and eosin (H&E) or immunohistochemistry (IHC) staining. The selection process is shown as a flow chart (Fig. 1).

Clinical trials for this review were identified through a search of ClinicalTrials.gov database for trials started from 1 December 2019 to 31 December 2020, by use of the terms ‘COVID-19’ and ‘CD147/IL-6/CCR5/PD-1/mTOR’. Phase 1 clinical trials were excluded; phase 2, 3, and 4 clinical trials were included.

Clinical classification of COVID-19 and data extraction

Clinical descriptions in the eligible publications, which were from China, the Republic of Korea, Italy, France, Poland, Turkey, and Spain, followed broadly similar guidelines to classify patients with COVID-19 by disease severity. Four categories of COVID-19 disease were described, all of which had confirmed COVID-19 by polymerase chain reaction (PCR) testing or suspected COVID-19 based on clinical diagnostic criteria such as the Chinese Clinical Guidance of COVID-19 Pneumonia Diagnosis and Treatment [17] (Supplementary Table 1). Cases were asymptomatic, mild-moderate, severe, or critical. Mild-moderate cases had fever and respiratory symptoms such as dry cough, nasal obstruction, sore throat; severe cases were defined as those with pneumonia causing respiratory compromise and an oxygen saturation ≤93% when breathing room air at rest, and critical illness was defined as cases of COVID-19 where invasive ventilation was required. A summary of clinical features measured in the included publications is shown in the Supplementary Table 1. The reported CD3+ cell count (×106 cells/L), CD4+ cell count (×106 cells/L), CD8+ cell count (×106 cells/L), and CD4:CD8 ratio were extracted from the selected publications. Cases that were admitted to intensive care unit (ICU), did not survive or were designated as critical or severe in the original publication were allocated to the severe group, cases that did not meet these criteria were allocated as non-severe. Infection with SARS-CoV-2 was confirmed in the autopsy cases by PCR, spike protein under transmission electron microscopy, IHC, or RNA hybrid assay.

Peripheral T cell lymphopenia in severe COVID-19

Data on peripheral blood white cell counts and immune cell subsets have been widely collected during hospital admission for COVID-19. Forty studies have overwhelmingly reported that peripheral T cell lymphopenia was worse in patients with severe COVID-19 compared with those with mild disease (Table 1). Although a wide range of T cell counts were reported with differences evident in standardised reported values for each study, the direction of change between severe and non-severe COVID-19 in these studies was consistent (Table 1). For all studies reporting CD3, CD4, and CD8 counts, the standardised mean difference in severe versus non-severe COVID-19 was significantly lower in severe disease (P < 0.00001 for all three; Fig. 2). For studies which only presented the median and interquartile range (27/40), mean and standard deviations (SD) were estimated using mathematical methods [18].

Table 1.

Raw data of T cell subsets in patients with severe and non-severe COVID-19

Paper Location N
(Non-severe: severe)
CD3+ T cell count (×106/L) CD4+ T cell count
(×106/L)
CD8+ T cell count
(×106/L)
CD4:CD8 Ratio Reference number (DOI)
Non severe Severe Non severe Severe Non severe Severe Non severe Severe
Calvet, J. 2020 [19] Spain 17:13 725 (497–1119) 647 (375–1113) 545 (445–767) 278 (178–663) 253 (145–319) 237 (87–586) 3.12 (1.58–3.99) 1.72 (0.78–2.52) 10.3390/v12111277
Cantenys-Molina, S. 2020 [20] Spain 590:112 662 (464–890) 363 (251–581) 412 (288–568) 242 (154–353) 213 (137–331) 116 (67–210) 1.90 (1.33–2.71) 1.98 (1.18–3.14) 10.1111/cei.13547
Chen, G. 2020 [21] China 10:11 640.5 (588.3–789.5) 294.0 (169.3–415.3) 381.5 (255–451) 177.5 (104–249.8) 254 (183.3–312.8) 89 (61.5–130.3) N/A N/A 10.1172/JCI137244
Chen, R. 2020 [22] China 345:155 662.00 (452.00–950.00) 367.5 (210.00–556.00) 386 (287.50–691.00) 226.5 (144.25–362.25) 252.5 (168.75–371.75) 126.5 (61.25–164.50) 1.52 (1.17–2.36) 1.94 (1.55–2.58) 10.1016/j.jaci.2020.05.003
Cui, N. 2020 [23] China 118:13 1047 (760–1330) 122 (57–322) 610 (446–808) 93 (30–225) 336 (245–449) 51 (17–122) 1.88 (1.36–2.52) 2.01 (1.32–4.04) 10.3389/fcimb.2020.595333
Demaret, J. 2020 [24] France 10:24 1000 (700–1400) 1300 (1000–1800) 700 (400–900) 700 (600–1100) 300 (200–500) 500 (400–600) N/A N/A 10.1002/cti2.1217
Diao, B. 2020 [25] China 479:20 652 (351–977) 261 (157–457) 342 (192–559) 198 (100–279) 208 (118–356) 64.3 (40.7–160) 1.60 (1.17–2.28) 2.43 (1.50–4.25) 10.3389/fimmu.2020.00827
Du, R. H. 2020 [26] China 158:21 N/A N/A 128.3 (73.5–201.7) 68 (55.1–148.8) 104.5 (58.5–142.7) 47.9 (25.4–73.8) NA NA 10.1183/13993003.00524-2020
Fu, Y. Q. 2020 [27] China 71:14 609.00 (410.00–905.00) 339.50 (217.50–524.25) 368.00 (246.00–549.00) 203.00 (126.50–284.25) 205.00 (111.00–303.00) 145.00 (70.00–213.00) 1.93 (1.26–2.68) 1.59 (1.13–2.47) 10.1371/journal.pone.0240751
Gutierrez-Bautista, J. F. 2020 [28] Spain 100:17 859 (549–1278) 630 (500–876) 387 (258–573) 313 (262–467) 172 (101–303) 167 (123–334) 2.41 (1.50–3.53) 1.96 (1.04–3.03) 10.3389/fimmu.2020.596553
Han, M. 2020 [29] China 122:32 864 (598–1125) 477 (337.675) 462 (314–621) 221 (185–407) 326 (224–478) 172 (119–269) 1.37 (1.09–1.83) 1.36 (0.95–2.09) 10.1007/s00430-020-00693-z
He, B. 2020 [30] China 32:21 794 (586–1112) 221 (168–414) 433 (318–651) 146 (107–277) 297 (230–388) 59 (33–109) 1.45 (1.24–1.80) 2.38 (1.62–4.63) 10.3389/fimmu.2020.02075
He, S. 2020 [31] China 48:25 821.86 (526.19) 465.28 (505.67) 475.68 (263.57) 277.07 (306.09) 302.35 (256.25) 185.75 (166.3) 1.68 (0.9) 1.46 (0.96) 10.1016/j.ijid.2020.06.059
Kalicinska, E. 2020 [32] Poland 11:16 530 (360–980) 540 (175–1080) 320 (200–530) 185 (95–525) 200 (98–380) 320 (60–630) 1.65 (1.18–2.75) 1.01 (0.59–1.2) 10.1016/j.tranon.2020.100943
Kalpakci, Y. 2020 [33] Turkey 20:20 N/A N/A 958.83 ± 416.24 395.45 ± 237.59 504.15 (313.61–786.22) 192.00 (135.52–261.80) 1.57 (1.38–2.85) 1.81 (1.16–3.13) 10.1590/1806-9282.66.12.1666
Kang, C. K. 2020 [34] Republic of Korea 8:3 N/A N/A 435.5 ±57.5 306.2 ±79.0 296.1 ±60.9 143.2 ±13.1 N/A N/A 10.1016/j.ijid.2020.05.106
Ke, C. 2020 [35] China 148:46 965.06 ±421.16 322.88 ±223.97 612.47 ±277.33 218.31 ±179.59 316.26 ±181.77 90.31 ±76.92 2.28 ±1.39 3.33 ±2.11 10.1016/j.medcli.2020.06.055
Kwiecien, I. 2020 [36] Poland 9:14 951 (683–2253) 691 (524–1416) 619 (533–1210) 319 (239–584) 313 (225–862) 330 (160–549) 2.1 (1.4–3.6) 1.3 (0.6–2.4 10.3390/cells9122615
Li, S. 2020 [37] China 43:26 991 (740–1154) 378 (258–576) 544 (364–667) 199 (128–325) 417 (309–539) 134 (91–237) 1.18 (0.96–1.58) 1.40 (0.79–2.08) 10.1172/jci.insight.138070
Liu, F. 2020 [38] China 32:8 857.74 ± 283.59 582.25 ± 305.80 454.16 ± 193.24 317.63 ± 162.30 385.74 ± 142.82 273.63 ± 168.54 N/A N/A 10.1038/s41598-020-70387-2
Liu, Q. 2020 [39] China 310:30 773 ± 549
678 (317.4–1105.1)
228 ± 168
170 (94.0–339.1)
457 ± 342
370 (182.8–651.0)
139 ± 98
115 (62.8–195.1)
297 ± 220
249 (118.8–425.1)
80.9 ± 97.7
56.8 (26.8–80.8)
1.71 ± 0.894
1.59 (1.06–2.04)
2.41 ± 1.28
2.25 (1.70–2.93)
10.1371/journal.pone.0239695
Liu, R. 2020 [40] China 49:61 509 (336.5–873.5) 517 (376–651.5) 290 (172.5–506) 292 (237.5–399.5) 161 (101–302.5) 200 (93–261) 1.89 (1.25–2.64) 1.75 (1.09–2.75) 10.1016/j.cca.2020.05.019
Luo, M. 2020 [41] China 817:201 611.01 (420.12–858.10) 391.20 (262.95–505.46) 367.99 (242.39–543.00) 245.00 (161.64–317.64) 203.98 (142.54–313.05) 96.89 (60.65–140.08) 1.70 (1.30–2.21) 2.37 (1.77–3.36) 10.1172/jci.insight.139024
Pallotto, C. 2020 [42] Italy 25:13 N/A N/A 461 (275–654) 348 (206–616) 184 (132–334) 100 (83–198) 2.4 (1.5–2.7) 2.8 (2.3–4) 10.1080/23744235.2020.1778178
Shao, L. 2020 [43] China 104:25 824.54 ± 469.11 447.25 ± 177.74 504.39 ± 367.90 255.00 ± 88.37 327.29 ± 212.36 179.33 ± 119.96 N/A N/A 10.3389/fmed.2020.00246
Sun, D. W. 2020 [44] China 25:11 1089.680 ± 290.154 698.455 ± 393.675 686.960 ± 225.383 427.091 ± 251.712 359.84 ± 111.279 247.818 ± 153.683 1.993 ± 0.606 1.957 ± 0.905 10.1016/j.cca.2020.05.027
Sun, H. B. 2020 [45] China 13:12 N/A N/A 576 ± 234 293 ± 76 492 ± 214 323 ± 56 1.23 ± 0.37 0.91 ± 0.15 10.1371/journal.pone.0239532
Sun, Y. 2020 [46] China 36:10 808.97 ± 371.22 522.57 ± 318.73 436.8 ± 225.08 257.8 6± 129.48 355.33 ± 166.86 205.14 ± 153.09 1.62 ± 1.86 1.28 ± 0.76 10.1016/j.jaut.2020.102473
Urra, J. M. 2020 [47] Spain 145:27 701.0 ± 408.5 528.3 ± 350.9 395.9 ± 241.0 340.3 ± 251.9 287.6 ± 223.8 172.4 ± 123.9 1.9 ± 1.6 2.4 ± 1.4 10.1016/j.clim.2020.108486
Varchetta, S. 2020 [48] Italy 17:15 N/A N/A 280 (144–562) 291 (132–734) 189 (32–505) 125 (35–411) N/A N/A 10.1038/s41423-020-00557-9
Wang, F. 2020a [49] China 33:32 N/A N/A 363.7± 225.5 179.5± 110.1 206.3± 137.0 53± 35.14 N/A N/A 10.1038/s41423-020-0483-y
Wang, F. 2020b [50] China 253:70 1071 (772.5–1399) 529 (387.0–712.5) 596.5 (452.5–757.0) 302.0 (204.5–383.0) 402.5 (273.0–546.5) 201.0 (134.5–294.0) 1.48 (1.12–1.96) 1.61 (1.02–1.94) 10.1186/s12967-020-02423-8
Wang, H. 2020 [51] China 48:47 774 (572–1095) 324 (195–455) 513 (304–625) 180 (109–274) 312 (197–423) 123 (71–179) 1.50 (1.11–2.04) 1.54 (1.04–2.51) 10.1016/j.intimp.2020.106683
Wu, Y. 2020 [52] China 31:29 399 (324–626) 306 (167–422) 234 (156–401) 153 (102–289) 191 (125–288) 88 (45–147) 1.46 (0.78–2.11) 1.99 (1.28–3.75) 10.1128/mSphere.00362-20
Xie, L. 2020 [53] China 322:51 1342.67 ± 493.06 938.79 ± 429.62 810.72 ± 321.02 530.61 ± 269.47 455.71 ± 198.07 357.58 ± 198.21 2.02 ± 0.96 1.87 ± 1.27 10.1177/1753466620942129
Xu, B. 2020 [54] China 117:28 894.50 (662.75–1192.00) 593.00 (412.00–725.00) 573.50 (426.75–771.00) 299.00 (249.00–460.00) 323.50 (232.75–448.75) 188.00 (134.00–274.00) 1.68 (0.96–2.18) 1.96 (1.02–2.70) 10.1016/j.jinf.2020.04.012
Yang, A. P. 2020 [55] China 69:24 763.8 ± 483.3 222.2 ± 195.2 448.7 ± 254.9 132.6 ± 98.5 264.6 ± 217.4 83.9 ± 97.2 2.01 ± 0.98 2.00 ± 0.97 10.18632/aging.103255
Yang, P. H. 2020 [56] China 22:48 975.00 (685.00–1365.00) 731.00 (377.25–1019.50) 540.00 (364.00–724.00) 377.00 (196.00–513.00) 417.00 (295.00–545.00) 326.50 (137.00–490.50) 1.41 (1.07–1.59) 1.13 (0.85–1.53) 10.1186/s40249-020-00780-6
Zhang, X. 2020 [57] China 293:12 778 (553–1041) 500 (379–705) 455 (314–650) 332 (226–541) 265 (171–393) 133 (102–199) N/A N/A 10.1038/s41586-020-2355-0
Zhao, Y. 2020 [58] China 414:125 814 (516–1088) 277 (163.5–430) 473 (291–657.75) 172 (99.5–267.5) 262.5 (163–405.25) 84 (39.5–155.5) N/A N/A 10.1186/s40249-020-00723-1

Data are presented as median (IQR) and/or mean ± SD.

Figure 2.

Figure 2.

Forest plot of T cell subsets in patients with severe and non-severe COVID-19. CD3+, CD4+ and CD8+ T cell counts are significantly lower in patients with severe COVID-19 compared with those in patients with non-severe COVID-19 (P < 0.00001). The effect of severity of disease on the CD4:CD8 ratio was inconclusive (P = 0.08). Black diamond represents test for overall effect of 40 studies.

Where reported (in 30/40 studies), data on the CD4:CD8 ratio were more variable compared with findings for the lymphocyte subsets (Table 1). Overall, the CD4:CD8 ratio was not significantly different in those patients with non-severe disease compared with patients with severe disease (P = 0.08), and the direction of change varied across the studies (Table 1, Fig. 2). Normal CD4:CD8 ratio is variously described as ≥ 1 or ≥ 1.2 and decreases with age [59]. Only one study reported a CD4:CD8 ratio <1.2, in either severe or non-severe COVID-19, the CD4:CD8 ratio therefore remained within normal range in both non-severe and severe COVID-19. Taken together, and compared with non-severe COVID-19, there is mounting evidence that severe COVID-19 disease is associated with a reduced frequency of CD3+ T cells affecting both CD4+ and CD8+ T cells in the peripheral circulation.

Given that lymphopaenia is known to be a feature of viral infection, rather than unique to severe COVID-19 we searched for observational studies that had measured CD4 and CD8 counts in other respiratory viral infections. Only two studies that reported comparative lymphocyte levels comparing between severe and non-severe infections were found, one in influenza [60] and one for SARS-CoV-1 [61]. Both indicated acute lymphopenia in severe disease (Supplementary Table 2). One study has compared lymphocyte levels between patients with SARS-CoV-2 and influenza, and the counts were similar [62]. This suggests that lymphopenia may be a common feature of severe respiratory viral infections and not unique to SARS-CoV-2 and therefore strategies that target it may be more broadly beneficial.

Clinical, virological, and immunological significance of T cell lymphopenia in COVID-19

Lymphopenia could be a risk factor for severity, mortality, and poor prognosis in COVID-19, with T cells the most affected compared with B cells and natural killer (NK) cells [63, 64]. T cell counts negatively correlated with survival and CD4+ and CD8+ T lymphocytes decreased significantly in patients with severe disease [40]. In the early stage of the COVID-19 outbreak, a hospital in Wuhan studied lymphocyte subsets in 27 patients; lymphopenia was common (70.4%, 19/27) in severe cases, and T lymphocytes decreased more than B lymphocytes. Interestingly, CD4+ and CD8+ T cells but not B lymphocytes or NK cells, were significantly elevated after treatment of symptomatic disease suggesting that clinical and immunological recovery may be correlated and can be measured by an improving peripheral CD4 and CD8 count [65]. In another study, although CD4:CD8 ratio remained in the normal range, CD8+ T lymphocytes were the most improved subsets after treatment [66]. While only one study reported that CD4+ T cell count was independently associated with ICU admission [67], several results highlighted the role of CD8+ T cells in COVID-19. CD8+ T cell lymphopenia was analysed as an independent predictor for the prognosis of COVID-19 [26, 46]. Notably, an association of decreased lymphocytes with disease severity, has been observed suggesting that lymphopenia is an important predictive factor for the severity of COVID-19 [57,68,69].

The nature of the association between lymphopenia and failure of viral containment has not been established, however a negative correlation between T cell count and virus detection could indicate that T cells contribute to the limitation of viral load. Peripheral blood lymphocyte counts on admission were significantly and negatively correlated with SARS-CoV-2 nucleic acid-positive duration in 18 patients. Amongst lymphocytes, the T cell count but not B cell or NK cell count that negatively correlated with nucleic acid-positive duration [70]. Another study which analysed T cells in 66 recovered COVID-19 patients, found that CD4+ T cell count was predictive of the duration of viral RNA detection in the stool sample [71]. These data suggest that depletion of T lymphocytes is associated with a delay in viral clearance; however, the direction of causality is yet to be established.

While clinical data indicate T cell lymphopenia is associated with persistent SARS-CoV-2 viraemia, some studies also indicate that infection may alter the quality and phenotype of the CD4+ T cell response. Surprisingly, a higher than normal naive-to-memory CD4+ T cell ratio has been observed suggesting an impact on the differentiation of naive to memory T cells in COVID-19 patients [64]. There was a lower frequency of regulatory T cells (T regs) in patients with severe disease compared with those with mild disease [34]. Patients had reduced levels of CD4+ Th1 cells [72], with a reported Th2 skewed response in peripheral blood smears from ICU patients [73]. An upregulated Th17 response was found in 39 COVID-19 patients [74]. Bioinformatics analysis shows that genes involved in Th17 cell differentiation were enriched in patients with both mild and severe COVID-19 [75]. Altogether these observations of skewed T cell helper phenotypes suggest that the inhibition of Treg-induced anti-inflammatory responses and increased Th2 or Th17 responses, which are inflammatory without necessarily leading to viral control, could be involved in the pathogenesis of COVID-19 [76].

Possible mechanisms for T cell lymphopenia

Dissection of the mechanism for the universal finding of peripheral T cell lymphopenia in SARS-CoV-2 infection particularly in severe COVID-19, will be important for the development of treatments and identifying who is at risk of severe disease.

SARS-CoV-2 infection of T cells

It is not yet clear whether SARS-CoV-2 can cause cytopathology through directly infecting T cells. Viral gene and angiotensin-converting enzyme 2(ACE2) expression were not detectable in the peripheral blood mononuclear cells (PBMCs) of COVID-19 patients [77]. Findings that the spike protein can bind to CD147 also known as basigin, and mediate viral invasion have not been corroborated [78, 79]. More recently a study has suggested that SARS-CoV-2 could interact with the CD4 molecule and that CD4-positive cells are permissive to viral infection [80]. However in vivo infection of CD4+ but not CD8+ T cells is not supported by published studies. Given the very early and conflicting nature of the data, caution is needed when interpreting these reports.

T cell redistribution

Although SARS-CoV-2 replication occurs principally in the lung, in severe cases, patients present with multi-organ failure in the late stage. This begs the question as to whether this is due to direct viral infection of these organs or due to an indirect effect of viral replication in the lung. To investigate this, we evaluated 123 autopsy case series of SARS-CoV-2 patients. In the respiratory tract, lung congestion, patchy lesions, diffuse alveolar damage, widespread vascular thrombosis, and new vessel growth were distinct characteristics of COVID-19 [81, 82]. Of significance for the interaction of the virus with T cells, 74 out of 123 studies reported mild-to-prominent infiltration of lymphocytes into organs by H&E staining, 36 out of 74 studies additionally confirmed T cell infiltration by IHC staining (Fig. 1). Both T cell infiltration and viral infection, as confirmed by presence of spike protein and/or viral nucleic acid, was observed in the heart [83], spleen [84], pharynx [82, 85], liver [83], and kidney [86]. Activated T cells in lymph nodes have also been reported [87]. In the spleen, the commonly reported lymphoid hypoplasia and lymphocytic depletion, especially CD8+ T cells indicates a decrease in the source of circulating lymphocytes [88]. The absence of germinal centres in hilar and posterior mediastinal lymph nodes suggests that the differentiation and maturation of B cells is also impacted [87].

The movement of T cells into tissues could be driving some of the observed damage. Cardiovascular changes reported were cardiomyocyte hypertrophy, degeneration, necrosis, congestion, and oedema of interstitial tissue [89]. H&E staining and caspase 3 staining support a model of inflammatory infiltration consistent with a pattern of endothelial apoptosis [90]. CD8+ T cell activation could contribute to cardiac injury in patients with severe disease [91]. The myocarditis found in histopathological studies could result from hyper inflammation of pathological T cells and cytokine storm [92]. Finally, the intense inflammation observed in severe COVID-19 could be associated with aberrant T-cell endothelial cell interactions leading to altered tethering and adhesion of cells [93]. Taken together, these autopsy findings indicate that normal T cell circulation is interrupted during severe disease due to SARS-CoV-2 infection (Fig. 3).

Figure 3.

Figure 3.

Hypothesis for peripheral T cell lymphopenia during SARS-CoV-2 infection and severe disease. According to experimental data from peripheral blood of patients with severe COVID-19, we propose two drivers for peripheral lymphopenia. Firstly, T lymphocytes in the periphery are attracted by chemokines released by infected cells and immune cells at the site of disease and migrate out of the periphery to infected organs, mainly the lungs. Secondly, functionally exhausted T lymphocytes and activation of Th1/Th2/Th17 responses at the site of disease, fail to achieve viral containment, and undergo cell death through a variety of mechanisms including apoptosis and pyroptosis. It is likely that interruption of the normal circulation of T cells is the key component in this cycle.

One potential driver for T cell redistribution contributing to the observed lymphopenia is the cytokine environment. Type I interferons are associated with SARS-CoV-2 infection, and in mouse models, lymphopenia was dependent on IFN signalling [94]. This may be a greater problem in the prolonged viral infection associated with severe COVID-19.

T cell activation, exhaustion, and apoptosis

Given that lymphopenia was associated with higher viral loads, dysregulation of T cell circulation could be associated with antigen-driven over-activation and exhaustion, which may in turn lead to their absence in peripheral circulation. Data to support this hypothesis are scarce. The abnormal cytokine profile in COVID-19 has been well described and also in association with expression of apoptosis pathway transcripts in PBMC but more data are needed to demonstrate whether these are linked [77, 95]. Several studies have indicated expression of exhaustion and activation markers in severe COVID-19. Kinetic studies found that CD4+ and CD8+ T cells expressed higher levels of the exhaustion markers PD-1, Tim-3, and NKG2A in the symptomatic stage, compared with the prodromal and recovery stages [25, 96]. Analysis of a peripheral blood sample taken from a deceased patient showed that the remaining CD4+ and CD8+ T cells were hyperactivated, indicated by the expression of both CD38 and HLA-DR. The increased frequency of CCR6+ Th17 CD4+ T cells and perforin and granulysin positive CD8+ T cells suggests that T cell activation accounted for severe immune injury in this patient [97]. Downregulation of the costimulatory molecule CD28 in patients with severe disease also suggests activation of T cells might contribute to aberrant signalling [98]. Different levels of activation markers were observed in different classes of T cells, with a very high level of activation in CD8+ T cells observed compared to CD4+ T cells [99]. An incompetent or aberrant CD8+ T-cell response could limit antigen-specific immunity, and there is some evidence this might be occurring in older people, although our review has not demonstrated lymphopaenia to be uniquely CD8+ T cell biased [100, 101]. It was notable that in clonal analysis of CD8+ T cells, they were highly activated but not exhausted in COVID-19 patients compared with healthy controls [102]. While data are accumulating indicating T cell hyperactivation in COVID-19, whether this is mechanistically linked to lymphopaenia remains to be seen.

Cell death

Finally, T cell lymphopenia could also be a function of SARS-CoV-2 infection-induced cell death. The cytokine storm produced by T cells and other inflammatory cells can promote apoptosis, pyroptosis, and necrosis of T cells in turn [25, 103]. A study of SARS-CoV found higher plasma Fas-ligand level in patients, which is associated with a higher level of caspase-3, which plays a key role in cell apoptosis, in CD4+ and CD8+ lymphocytes [104]. Since there are similarities between SARS-CoV-1 and SARS-CoV-2, it was proposed that lymphocyte apoptosis is one of the causes of lymphopenia in COVID-19. Genes involved in apoptosis and P53 signalling pathways were enriched in PBMCs and cells of bronchoalveolar lavage fluid sample in three COVID-19 patients, indicating cell apoptosis could contribute to lymphopenia [77].

Another form of inflammation induced cell death is pyroptosis, which is caspase-1 triggered cell death via cleavage of gasdermin family members. Pyroptosis is triggered by inflammatory cytokines. While no studies have directly reported T cell pyroptosis during COVID-19 infection, one study proposed a mechanism for cytokine induced cell death [105]. This cell death was linked to IFNγ and TNF, both of which are elevated in patients with severe COVID-19, so it could potentially be a mechanism. Type I interferons have also been shown to prime cells for Fas-mediated apoptosis [106] which may drive the cell death seen.

Treatment strategies and ongoing clinical trials targeting T cells

The link between lymphopenia and severe disease opens up a number of therapeutic strategies, to target the different mechanisms driving the lymphopenia. Treatment strategies that have been considered include blocking SARS-CoV-2 from infecting T cells [78], inhibition of cytokine secretion [107–109], mitigation of T cell exhaustion [110, 111], blockade of the chemokine receptor CCR5 [112, 113], and normalisation of Th1/Th2/Th17 differentiation [114, 115]. Despite limited understanding of the pathogenesis and processes involving T cells in SARS-CoV-2 infection, some of these have already entered clinical trials (Table 2). Anti-IL-6 treatment has been tested in a number of different settings, with mixed results. A recently published study indicates that in critically ill patients anti-IL-6 receptor drugs can improve outcomes [116] but in other settings it has been less effective.

Table 2.

Clinical trials using therapeutics targeting T cells

Rationale Target Drug ClinicalTrials.gov Identifier
Proposed viral entry (mechanism to be confirmed) CD147 Meplazumab
(Anti-CD147 antibody)
NCT04275245 Phase 1, 2
NCT04586153 Phase 2, 3
Target a downstream component of aberrant immune cell communication Reduce cytokine-storm, inflammation and exhaustion IL-6 Tocilizumab
(Anti-IL-6R antibody)
ChiCTR2000029765
NCT04320615 Phase 3
NCT04330638 Phase 3
NCT04345445 Phase 3
NCT04347031 Phase 2, 3
NCT04349410 Phase 2, 3
NCT04356937 Phase 3
NCT04359095 Phase 2, 3
NCT04361032 Phase 3
NCT04372186 Phase 3
NCT04377750 Phase 4
NCT04380519 Phase 2, 3
NCT04381936 Phase 2, 3
NCT04403685 Phase 3
NCT04409262 Phase 3
NCT04412772 Phase 3
NCT04423042 Phase 3
NCT04424056 Phase 3
NCT04577534 Phase 3
NCT04600141 Phase 3
NCT04678739 Phase 3
NCT04730323 Phase 4
Siltuximab
(Anti-IL-6 antibody)
NCT04322188 Not shown
NCT04329650 Phase 2
NCT04330638 Phase 3
NCT04486521 Not shown
Clazakizumab
(Anti-IL-6 antibody)
NCT04381052 Phase 2
NCT04343989 Phase 2
NCT04363502 Phase 2
NCT04348500 Phase 2
NCT04494724 Phase 2
NCT04659772 Phase 2
Sarilumab
(Anti-IL-6 Receptor antibody)
NCT04315298 Phase 2, 3
NCT04324073 Phase 2, 3
NCT04322773 Phase 2
NCT04327388 Phase 3
NCT04341870 Phase 2, 3
NCT04357808 Phase 2
NCT04357860 Phase 2
NCT04359901 Phase 2
NCT04661527 Phase 2
Fluoxetine
(SSRI inhibitor)
NCT04377308 Phase 4
Ruxolitinib
(JAK inhibitor)
NCT04331665 Not shown
NCT04334044 Phase 1, 2
NCT04338958 Phase 2
NCT04348071 Phase 2, 3
NCT04348695 Phase 2
NCT04355793 Not shown
NCT04359290 Phase 2
NCT04361903 Not shown
NCT04362137 Phase 3
NCT04374149 Phase 2
NCT04377620 Phase 3
NCT04403243 Phase 2
NCT04414098 Phase 2
NCT04424056 Phase 3
NCT04477993 Phase 2, 3
NCT04581954 Phase 1, 2
Reduce aberrant T cell migration CCR5 Maraviroc
(CCR5 antagonist)
NCT04441385 Phase 2
NCT04475991 Phase 2
NCT04710199 Phase 2
Leronlimab
(Anti-CCR5 antibody)
NCT04343651 Phase 2
NCT04347239 Phase 2
NCT04678830 Phase 2
Limit T cell exhaustion PD-1 PD-1 blocking antibody NCT04268537 Phase 2
Nivolumab
(Anti-PD1 antibody)
NCT04356508 Phase 2
NCT04413838 Phase 2
Limit T cell exhaustion mTOR Rapamycin/ Sirolimus
(mTOR inhibitor)
NCT04341675 Phase 2
NCT04482712 Phase 1, 2
NCT04461340 Phase 2
RTB101
(PI3K/ mTOR) inhibitor
NCT04584710 Phase 2
NCT04409327 Phase 2

Dexamethasone, which suppresses the immune response via the glucocorticoid receptor, is the only drug so far to have shown to have consistent efficacy against COVID-19 [117]. Randomised clinical trials have shown that intravenous dexamethasone treatment significantly prolonged ventilator-free days [118] and reduced mortality [119]. A meta-analysis demonstrated lower 28-day all-cause mortality in the corticosteroid treatment group [120]. Despite clinical improvements, the side effect of dexamethasone including dampening viral clearance, suppressing bone marrow, and interrupting metabolism need attention [121]. The impact of dexamethasone on T cell circulation and function in COVID-19 will be important to study. Why dexamethasone, which acts pleiotropically has been more effective than targeted cytokine blocking drugs is not clear, there may be a goldilocks effect, where both too little and too great a cytokine response is detrimental, so dexamethasone dampens but does not completely block inflammation while specific cytokine drugs remove the beneficial role of the cytokine as well as the excess inflammation.

Vaccines that induce a T cell response

Priming the T cell response prior to infection with vaccination is one strategy to improve protection from disease. To date, the published human clinical vaccine studies including T cell data have been largely phase I/II trials and are not designed to evaluate the protective role of T cells. Several of the studies have reported T cell responses after vaccination [122]. The mRNA vaccine BNT162b1-induced receptor binding domain-specific CD4+ and CD8+ Th1 responses with IFN γ production [123]. A combination of rAd26 and rAd5 vector-based vaccines induced proliferation of antigen-specific CD4+ and CD8+ T cells [124]. Adenovirus type-5-vectored COVID-19 vaccine successfully induced IFN γ-producing T cells in phase 1 and phase 2 clinical trial [125, 126]. ChAdOx1 nCoV-19 vaccine also elicited IFN γ-producing T cells to a similar degree [127]. Induction of a robust SARS-CoV-2-specific T cell memory response may be different in natural infection compared with immunisation, and the frequency of SARS-CoV-2-specific T cells may be contingent upon both vaccine design and whether the initial response to the prime dose has been boosted [128]. Larger, follow-up studies will be required to identify the role of vaccine-induced T cells in protection from infection and disease.

Long COVID-19

The sequalae of infection with SARS-CoV-2 can be chronic and leave symptoms even in recovered nucleic-negative individuals such as fatigue, chest heaviness, and breathlessness [129]. These symptoms have been grouped together as long COVID. Although data are at present limited, persistence of symptoms is common, particularly in patients who are hospitalised, with only 13% of 143 patients symptom-free after discharge in one Italian study [130]. The role of T cells in this condition is yet to be identified, but aberrant T cell circulation, tissue infiltration, and immune damage during acute infection may all be important in determining longer-term outcomes.

Conclusion

Peripheral circulation T cell lymphopenia affecting both CD4+ and CD8+ T cells is a universal finding in case series of COVID-19 and is associated with severe disease. Our findings are in line with other publications [131–134]. Current evidence from autopsy and in vivo studies indicates the most likely mechanism is the interruption of normal lymphocyte circulation and organ-based, and/or endothelial sequestration of T cells. This may be due to the intense inflammation caused by SARS-CoV-2 replication. Coupled with aberrant T cell function, T cell exhaustion, and persistent failure of viral containment, these findings indicate a self-amplifying inflammatory cycle in severe COVID-19, which should be prevented or interrupted with rationally designed vaccines and therapeutics.

Supplementary material

Supplementary data are available at Immunotherapy Advances online.

Supplementary Table 1. Summary of studies included

Supplementary Table 2. Lymphocyte count of patients with SARS-CoV, MERS-CoV and Influenza. Cell count was shown as mean ± SD and/or median (IQR).

ltab015_suppl_Supplementary_Material

Acknowledgements

The Editor-in-Chief, Tim Elliott, would like to thank Tao Dong and Laura Denney for their contribution to the peer review of this article.

Glossary

Abbreviations

ACE2

Angiotensin-converting enzyme 2

CCR5

C-C chemokine receptor type 5

CD

Cluster of differentiation

COVID-19

Coronavirus disease 2019

H&E

Haematoxylin and eosin

HIV

Human immunodeficiency virus

HLA-DR

Human leukocyte antigen – DR isotype

ICU

Intensive care units

IFN

Interferon

IHC

Immunohistochemistry

IL

Interleukin

mTOR

mechanistic target of rapamycin

NK

Natural killer

P53

Tumour protein P53

PBMC

Peripheral blood mononuclear cell

PCR

Polymerase chain reaction

PD-1

Programmed cell death protein 1

RNA

Ribonucleic acid

SARS-CoV

Severe acute respiratory syndrome coronavirus

Th

T helper

Tim-3

T-cell immunoglobulin mucin 3

Treg

regulatory T cell

Contributor Information

Sifan Zhang, Department of Infectious Disease, Imperial College London, London, UK.

Becca Asquith, Department of Infectious Disease, Imperial College London, London, UK.

Richard Szydlo, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK.

John S Tregoning, Department of Infectious Disease, Imperial College London, London, UK.

Katrina M Pollock, Department of Infectious Disease, Imperial College London, London, UK.

Author contributions

Conceptualisation: K.P.; Writing-Original Draft: S.Z.; Figures and tables: S.Z., J.S.T., and K.P.; Writing-Review and Editing: S.Z., J.S.T., B.A., R.S., and K.P.

Funding

This research was supported by the National Institute for Health Research (NIHR) Imperial Biomedical Research Centre and the NIHR Imperial Clinical Research Facility. The views expressed are those of the author(s) and not necessarily those of the National Health Service, the NIHR or the Department of Health and Social Care.

Conflict of interest

K.P. reports grants from the National Institute for Health Research and the Medical Research Council UK Research and Innovation. K.P. is chief investigator for the Imperial College London COVID-19 vaccine development programme and principal investigator for the University of Oxford COVID-19 vaccine development trials, personal fees from Sanofi, outside the submitted work; and honoraria received for Vaccines for COVID-19 e-Learning course; British Society for Antimicrobial Therapy and ITV PLC.

Data availability

No new datasets were generated for this manuscript.

References

  • 1. Zhou  R, To  KK, Wong  YC  et al.  Acute SARS-CoV-2 infection impairs dendritic cell and T cell responses. Immunity  2020;53(4):864–77.e5. 10.1016/j.immuni.2020.07.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Grifoni  A, Weiskopf  D, Ramirez  SI  et al.  Targets of T Cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell  2020;181(7):1489–501.e15. 10.1016/j.cell.2020.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Ackermann  M, Verleden  SE, Kuehnel  M  et al.  Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19. N Engl J Med  2020;383(2):120–8. 10.1056/NEJMoa2015432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Fenwick  C, Joo  V, Jacquier  P  et al.  T-cell exhaustion in HIV infection. Immunol Rev  2019;292(1):149–63. 10.1111/imr.12823 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Douek  DC, Brenchley  JM, Betts  MR  et al.  HIV preferentially infects HIV-specific CD4+ T cells. Nature  2002;417(6884):95–8. 10.1038/417095a [DOI] [PubMed] [Google Scholar]
  • 6. Gougeon  ML. To kill or be killed: how HIV exhausts the immune system. Cell Death Differ  2005;12 Suppl 1:845–54. 10.1038/sj.cdd.4401616 [DOI] [PubMed] [Google Scholar]
  • 7. Younan  P, Santos  RI, Ramanathan  P  et al.  Ebola virus-mediated T-lymphocyte depletion is the result of an abortive infection. PLoS Pathog  2019;15(10):e1008068. 10.1371/journal.ppat.1008068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Malacarne  F, Webster  GJ, Reignat  S  et al.  Tracking the source of the hepatitis B virus-specific CD8 T cells during lamivudine treatment. J Infect Dis  2003;187(4):679–82. 10.1086/368369 [DOI] [PubMed] [Google Scholar]
  • 9. McSharry  BP, Samer  C, McWilliam  HEG  et al.  Virus-mediated suppression of the antigen presentation molecule MR1. Cell Rep  2020;30(9):2948–62-e2944. 10.1016/j.celrep.2020.02.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Ho  MR, Tsai  TT, Chen  CL  et al.  Blockade of dengue virus infection and viral cytotoxicity in neuronal cells in vitro and in vivo by targeting endocytic pathways. Sci Rep  2017;7(1):6910. 10.1038/s41598-017-07023-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Li  J, Wang  J, Kang  AS  et al.  Mapping the T cell response to COVID-19. Signal Transduct Target Ther  2020;5(1):112. 10.1038/s41392-020-00228-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Gomez-Mora  E, Garcia  E, Urrea  V  et al.  Preserved immune functionality and high CMV-specific T-cell responses in HIV-infected individuals with poor CD4+ T-cell immune recovery. Sci Rep  2017;7(1):11711. 10.1038/s41598-017-12013-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Bajwa  M, Vita  S, Vescovini  R  et al.  CMV-specific T-cell responses at older ages: broad responses with a large central memory component may be key to long-term survival. J Infect Dis  2017;215(8):1212–20. 10.1093/infdis/jix080 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Papagno  L, Appay  V, Sutton  J  et al.  Comparison between HIV- and CMV-specific T cell responses in long-term HIV infected donors. Clin Exp Immunol  2002;130(3):509–17. 10.1046/j.1365-2249.2002.02005.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Nicolai  L, Leunig  A, Brambs  S  et al.  Immunothrombotic dysregulation in COVID-19 pneumonia is associated with respiratory failure and coagulopathy. Circulation  2020;142(12): 1176–89. 10.1161/CIRCULATIONAHA.120.048488 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Yang  L, Liu  S, Liu  J  et al.  COVID-19: immunopathogenesis and immunotherapeutics. Signal Transduct Target Ther  2020;5(1):128. 10.1038/s41392-020-00243-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Commission CNH.  Chinese Clinical Guidance for COVID-19 Pneumonia Diagnosis and Treatment (7th edition). 2020. http://kjfy.meetingchina.org/msite/news/show/cn/3337.html [Google Scholar]
  • 18. Wan  X, Wang  W, Liu  J  et al.  Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Methodol  2014;14:135. 10.1186/1471-2288-14-135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Calvet  J, Gratacos  J, Amengual  MJ  et al.  CD4 and CD8 lymphocyte counts as surrogate early markers for progression in SARS-CoV-2 pneumonia: a prospective study. Viruses  2020;12(11):1277. 10.3390/v12111277 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Cantenys-Molina  S, Fernandez-Cruz  E, Francos  P  et al.  Lymphocyte subsets early predict mortality in a large series of hospitalized COVID-19 patients in Spain. Clin Exp Immunol  2021;203(3):424–32. 10.1111/cei.13547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Chen  G, Wu  D, Guo  W  et al.  Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest  2020;130(5):2620–9. 10.1172/JCI137244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Chen  R, Sang  L, Jiang  M  et al.  Longitudinal hematologic and immunologic variations associated with the progression of COVID-19 patients in China. J Allergy Clin Immunol  2020;146(1):89–100. 10.1016/j.jaci.2020.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Cui  N, Yan  R, Qin  C  et al.  Clinical characteristics and immune responses of 137 deceased patients with COVID-19: a retrospective study. Front Cell Infect Microbiol  2020;10:595333. 10.3389/fcimb.2020.595333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Demaret  J, Lefevre  G, Vuotto  F  et al. ; Lille Covid Research Network (LICORNE). Severe SARS-CoV-2 patients develop a higher specific T-cell response. Clin Transl Immunology  2020;9(12):e1217. 10.1002/cti2.1217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Diao  B, Wang  C, Tan  Y  et al.  Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19). Front Immunol  2020;11:827. 10.3389/fimmu.2020.00827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Du  RH, Liang  LR, Yang  CQ  et al.  Predictors of mortality for patients with COVID-19 pneumonia caused by SARSCoV-2: a prospective cohort study. Eur Respir J  2020;55(5):2000524. 10.1183/13993003.00524-2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Fu  YQ, Sun  YL, Lu  SW  et al.  Effect of blood analysis and immune function on the prognosis of patients with COVID-19. PLoS One  2020;15(10):e0240751. 10.1371/journal.pone.0240751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Gutiérrez-Bautista  JF, Rodriguez-Nicolas  A, Rosales-Castillo  A  et al.  Negative clinical evolution in COVID-19 patients is frequently accompanied with an increased proportion of undifferentiated Th cells and a strong underrepresentation of the Th1 subset. Front Immunol  2020;11:596553. 10.3389/fimmu.2020.596553 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Han  M, Xu  M, Zhang  Y  et al.  Assessing SARS-CoV-2 RNA levels and lymphocyte/T cell counts in COVID-19 patients revealed initial immune status as a major determinant of disease severity. Med Microbiol Immunol  2020;209(6):657–68. 10.1007/s00430-020-00693-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. He  B, Wang  J, Wang  Y  et al.  The metabolic changes and immune profiles in patients with COVID-19. Front Immunol  2020;11:2075. 10.3389/fimmu.2020.02075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. He  S, Zhou  C, Lu  D  et al.  Relationship between chest CT manifestations and immune response in COVID-19 patients. Int J Infect Dis  2020;98:125–9. 10.1016/j.ijid.2020.06.059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Kalicińska  E, Szymczak  D, Andrasiak  I  et al.  Lymphocyte subsets in haematological patients with COVID-19: multicentre prospective study. Transl Oncol  2021;14(1):100943. 10.1016/j.tranon.2020.100943 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Kalpakci  Y, Hacibekiroglu  T, Trak  G  et al.  Comparative evaluation of memory T cells in COVID-19 patients and the predictive role of CD4+CD8+ double positive T lymphocytes as a new marker. Rev Assoc Med Bras (1992)  2020;66(12):1666–72. 10.1590/1806-9282.66.12.1666 [DOI] [PubMed] [Google Scholar]
  • 34. Kang  CK, Han  GC, Kim  M  et al.  Aberrant hyperactivation of cytotoxic T-cell as a potential determinant of COVID-19 severity. Int J Infect Dis  2020;97:313–21. 10.1016/j.ijid.2020.05.106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Ke  C, Yu  C, Yue  D, Zeng  X, Hu  Z, Yang  C. Clinical characteristics of confirmed and clinically diagnosed patients with 2019 novel coronavirus pneumonia: a single-center, retrospective, case-control study. Med Clin (Barc)  2020;155(8):327–34. 10.1016/j.medcli.2020.06.055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kwiecien  I, Rutkowska  E, Klos  K  et al.  Maturation of T and B lymphocytes in the assessment of the immune status in COVID-19 patients. Cells  2020;9(12):2615. 10.3390/cells9122615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Li  S, Jiang  L, Li  X, et al.  Clinical and pathological investigation of patients with severe COVID-19. JCI Insight. 2020;5(12):e138070. 10.1172/jci.insight.138070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Liu  F, Ji  C, Luo  J  et al.  Clinical characteristics and corticosteroids application of different clinical types in patients with corona virus disease 2019. Sci Rep  2020;10(1):13689. 10.1038/s41598-020-70387-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Liu  Q, Fang  X, Tokuno  S  et al.  A web visualization tool using T cell subsets as the predictor to evaluate COVID-19 patient’s severity. PLoS One  2020;15(9):e0239695. 10.1371/journal.pone.0239695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Liu  R, Wang  Y, Li  J  et al.  Decreased T cell populations contribute to the increased severity of COVID-19. Clin Chim Acta  2020;508:110–14. 10.1016/j.cca.2020.05.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Luo  M, Liu  J, Jiang  W, Yue  S, Liu  H, Wei  S. IL-6 and CD8+ T cell counts combined are an early predictor of in-hospital mortality of patients with COVID-19. JCI Insight  2020;5(13):e139024. 10.1172/jci.insight.139024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Pallotto  C, Suardi  LR, Esperti  S  et al.  Increased CD4/CD8 ratio as a risk factor for critical illness in coronavirus disease 2019 (COVID-19): a retrospective multicentre study. Infect Dis (Lond)  2020; 52(9): 675–7. 10.1080/23744235.2020.1778178 [DOI] [PubMed] [Google Scholar]
  • 43. Shao  L, Li  X, Zhou  Y  et al.  Novel insights into illness progression and risk profiles for mortality in non-survivors of COVID-19. Front Med (Lausanne)  2020;7:246. 10.3389/fmed.2020.00246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Sun  DW, Zhang  D, Tian  RH  et al.  The underlying changes and predicting role of peripheral blood inflammatory cells in severe COVID-19 patients: A sentinel?  Clin Chim Acta  2020;508:122–9. 10.1016/j.cca.2020.05.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Sun  HB, Zhang  YM, Huang  LG  et al.  The changes of the peripheral CD4+ lymphocytes and inflammatory cytokines in Patients with COVID-19. PLoS One  2020;15(9):e0239532. 10.1371/journal.pone.0239532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Sun  Y, Dong  Y, Wang  L  et al.  Characteristics and prognostic factors of disease severity in patients with COVID-19: the Beijing experience. J Autoimmun  2020;112:102473. 10.1016/j.jaut.2020.102473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Urra  JM, Cabrera  CM, Porras  L  et al.  Selective CD8 cell reduction by SARS-CoV-2 is associated with a worse prognosis and systemic inflammation in COVID-19 patients. Clin Immunol  2020;217:108486. 10.1016/j.clim.2020.108486 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Varchetta  S, Mele  D, Oliviero  B  et al. Unique immunological profile in patients with COVID-19. Cell Mol Immunol  2021;18(3):604–12. 10.1038/s41423-020-00557-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Wang  F, Hou  H, Yao  Y  et al.  Systemically comparing host immunity between survived and deceased COVID-19 patients. Cell Mol Immunol  2020; 17(8):875–7. 10.1038/s41423-020-0483-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Wang  F, Qu  M, Zhou  X  et al.  The timeline and risk factors of clinical progression of COVID-19 in Shenzhen, China. J Transl Med  2020;18(1):270. 10.1186/s12967-020-02423-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wang  H, Zhang  Y, Mo  P  et al.  Neutrophil to CD4+ lymphocyte ratio as a potential biomarker in predicting virus negative conversion time in COVID-19. Int Immunopharmacol  2020;85:106683. 10.1016/j.intimp.2020.106683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Wu  Y, Huang  X, Sun  J  et al.  Clinical characteristics and immune injury mechanisms in 71 patients with COVID-19. mSphere  2020;5(4):e00362–20. 10.1128/mSphere.00362-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Xie  L, Wu  Q, Lin  Q  et al.  Dysfunction of adaptive immunity is related to severity of COVID-19: a retrospective study. Ther Adv Respir Dis  2020;14:1753466620942129. 10.1177/1753466620942129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Xu  B, Fan  CY, Wang  AL  et al.  Suppressed T cell-mediated immunity in patients with COVID 19: A clinical retrospective study in Wuhan, China. J Infect  2020;81(1):e51–e60. 10.1016/j.jinf.2020.04.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Yang  AP, Li  HM, Tao  WQ  et al.  Infection with SARS-CoV-2 causes abnormal laboratory results of multiple organs in patients. Aging (Albany NY)  2020;12(11):10059–69. 10.18632/aging.103255 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Yang  PH, Ding  YB, Xu  Z  et al.  Increased circulating level of interleukin-6 and CD8+ T cell exhaustion are associated with progression of COVID-19. Infect Dis Poverty  2020;9(1):161. 10.1186/s40249-020-00780-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Zhang  X, Tan  Y, Ling  Y  et al.  Viral and host factors related to the clinical outcome of COVID-19. Nature  2020;583(7816):437–40. 10.1038/s41586-020-2355-0 [DOI] [PubMed] [Google Scholar]
  • 58. Zhao  Y, Nie  HX, Hu  K  et al.  Abnormal immunity of nonsurvivors with COVID-19: predictors for mortality. Infect Dis Poverty  2020;9(1):108. 10.1186/s40249-020-00723-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Francis-Morris  A, Mackie  NE, Eliahoo  J  et al.  Compromised CD4:CD8 ratio recovery in people living with HIV aged over 50 years: an observational study. HIV Med  2020;21(2):109–18. 10.1111/hiv.12800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Wang  N, Liu  X, Zhang  Y, Xie  Y, Zhao  W. Hematologic markers of influenza A H1N1 for early laboratory diagnosis and treatment assessment. Laboratory Medicine  2011;42(10):607–11. 10.1309/lmrvljy3brxlzge3 [DOI] [Google Scholar]
  • 61. He  Z, Zhao  C, Dong  Q  et al. Effects of severe acute respiratory syndrome (SARS) coronavirus infection on peripheral blood lymphocytes and their subsets. Int J Infect Dis  2005;9(6):323–30. 10.1016/j.ijid.2004.07.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Qian  F, Gao  G, Song  Y  et al.  Specific dynamic variations in the peripheral blood lymphocyte subsets in COVID-19 and severe influenza A patients: a retrospective observational study. BMC Infect Dis  2020;720(1):910. 10.1186/s12879-020-05637-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Bao  J, Li  C, Zhang  K, Kang  H, Chen  W, Gu  B. Comparative analysis of laboratory indexes of severe and non-severe patients infected with COVID-19. Clin Chim Acta  2020;509:180–94. 10.1016/j.cca.2020.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Qin  C, Zhou  L, Hu  Z  et al.  Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin Infect Dis  2020;71(15):762–8. 10.1093/cid/ciaa248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Ni  M, Tian  FB, Xiang  DD  et al.  Characteristics of inflammatory factors and lymphocyte subsets in patients with severe COVID-19. J Med Virol  2020;92(11):2600–06. 10.1002/jmv.26070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Ganji  A, Farahani  I, Khansarinejad  B  et al.  Increased expression of CD8 marker on T-cells in COVID-19 patients. Blood Cells Mol Dis  2020;83:102437. 10.1016/j.bcmd.2020.102437 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Chen  J, Qi  T, Liu  L  et al.  Clinical progression of patients with COVID-19 in Shanghai, China. J Infect  2020;80(5):e1–e6. 10.1016/j.jinf.2020.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Tan  L, Wang  Q, Zhang  D  et al.  Lymphopenia predicts disease severity of COVID-19: a descriptive and predictive study. Signal Transduct Target Ther  2020;5(1):33. 10.1038/s41392-020-0148-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Tan  L, Kang  X, Ji  X  et al.  Validation of predictors of disease severity and outcomes in COVID-19 patients: a descriptive and retrospective study. Med (NY)  2020;1(1):128–38-e123. 10.1016/j.medj.2020.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Dong  X, Wang  M, Liu  S  et al.  Immune characteristics of patients with coronavirus disease 2019 (COVID-19). Aging Dis  2020;11(3):642–8. 10.14336/AD.2020.0317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Ling  Y, Xu  SB, Lin  YX  et al.  Persistence and clearance of viral RNA in 2019 novel coronavirus disease rehabilitation patients. Chin Med J (Engl)  2020;133(9):1039–43. 10.1097/CM9.0000000000000774 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Cao  M, Zhang  D, Wang  Y  et al.  Clinical features of patients infected with the 2019 novel coronavirus (COVID-19) in Shanghai, China. medRxiv  2020;2020.03.04.20030395. 10.1101/2020.03.04.20030395 [DOI] [Google Scholar]
  • 73. Roncati  L, Nasillo  V, Lusenti  B  et al.  Signals of Th2 immune response from COVID-19 patients requiring intensive care. Ann Hematol  2020;99(6):1419–20. 10.1007/s00277-020-04066-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. De Biasi  S, Meschiari  M, Gibellini  L  et al.  Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat Commun  2020;11(1):3434. 10.1038/s41467-020-17292-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Ouyang  Y, Yin  J, Wang  W  et al.  Down-regulated gene expression spectrum and immune responses changed during the disease progression in COVID-19 patients. Clin Infect Dis  2020;71(16):2052–60. 10.1093/cid/ciaa462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Muyayalo  KP, Huang  DH, Zhao  SJ  et al.  COVID-19 and Treg/Th17 imbalance: Potential relationship to pregnancy outcomes. Am J Reprod Immunol  2020;84(5):e13304. 10.1111/aji.13304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Xiong  Y, Liu  Y, Cao  L  et al.  Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients. Emerg Microbes Infect  2020;9(1):761–70. 10.1080/22221751.2020.1747363 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Wang  K, Chen  W, Zhang  Z, et al.  CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells. Signal Transduct Target Ther  2020;5(1):283. 10.1038/s41392-020-00426-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Shilts  J, Crozier  TWM, Greenwood  EJD, Lehner  PJ, Wright  GJ. No evidence for basigin/CD147 as a direct SARS-CoV-2 spike binding receptor. Sci Rep  2021;11(1):413. 10.1038/s41598-020-80464-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Davanzo  GG, Codo  AC, Brunetti  NS  et al.  SARS-CoV-2 Uses CD4 to infect T helper lymphocytes. medRxiv  2020;.2009.2025.20200329. 10.1101/2020.09.25.20200329 [DOI] [Google Scholar]
  • 81. Liu  Q, Wang  RS, Qu  GQ  et al.  Gross examination report of a COVID-19 death autopsy. Fa Yi Xue Za Zhi  2020;36(1):21–3. 10.12116/j.issn.1004-5619.2020.01.005 [DOI] [PubMed] [Google Scholar]
  • 82. Wichmann  D, Sperhake  JP, Lutgehetmann  M  et al.  Autopsy findings and venous thromboembolism in patients withCOVID-19: a prospective cohort study. Ann Intern Med  2020;173(4):268–77. 10.7326/M20-2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Schaller  T, Hirschbuhl  K, Burkhardt  K  et al.  Postmortem examination of patients with COVID-19. Jama  2020;323(24):2518–20. 10.1001/jama.2020.8907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Buja  LM, Wolf  DA, Zhao  B  et al.  The emerging spectrum of cardiopulmonary pathology of the coronavirus disease 2019 (COVID-19): report of 3 autopsies from Houston, Texas, and review of autopsy findings from other United States cities. Cardiovasc Pathol  2020;48:107233. 10.1016/j.carpath.2020.107233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Edler  C, Schroder  AS, Aepfelbacher  M  et al.  Correction to: dying with SARS-CoV-2 infection-an autopsy study of the first consecutive 80 cases in Hamburg, Germany. Int J Legal Med  2020; 134(5):1977. 10.1007/s00414-020-02336-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Su  H, Yang  M, Wan  C  et al.  Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int  2020;98(1):219–27. 10.1016/j.kint.2020.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Lax  SF, Skok  K, Zechner  P  et al.  Pulmonary arterial thrombosis in COVID-19 with fatal outcome: results from a prospective, single-center, clinicopathologic case series. Ann Intern Med  2020;173(5):350–61. 10.7326/M20-2566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Gu  J, Korteweg  C. Pathology and pathogenesis of severe acute respiratory syndrome. Am J Pathol  2007;170(4):1136–47. 10.2353/ajpath.2007.061088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Yao  XH, Li  TY, He  ZC  et al.  A pathological report of three COVID-19 cases by minimally invasive autopsies. Zhonghua Bing Li Xue Za Zhi  2020;49(5):411–7. 10.3760/cma.j.cn112151-20200312-00193 [DOI] [PubMed] [Google Scholar]
  • 90. Varga  Z, Flammer  AJ, Steiger  P  et al.  Endothelial cell infection and endotheliitis in COVID-19. Lancet  2020;395(10234):1417–8. 10.1016/S0140-6736(20)30937-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Zhang  C, Wang  FS, Silvestre  JS  et al.  Is aberrant CD8+ T cell activation by hypertension associated with cardiac injury in severe cases of COVID-19?  Cell Mol Immunol  2020;17(6):675–6. 10.1038/s41423-020-0454-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Bavishi  C, Bonow  RO, Trivedi  V, Abbott  JD, Messerli  FH, Bhatt  DL. Acute myocardial injury in patients hospitalized with COVID-19 infection: a review. Prog Cardiovasc Dis  2020;63(5):682–9. 10.1016/j.pcad.2020.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Muller  WA. Leukocyte-endothelial-cell interactions in leukocyte transmigration and the inflammatory response. Trends Immunol  2003;24(6):327–34. 10.1016/s1471-4906(03)00117-0 [DOI] [PubMed] [Google Scholar]
  • 94. Kamphuis  E, Junt  T, Waibler  Z  et al.  Type I interferons directly regulate lymphocyte recirculation and cause transient blood lymphopenia. Blood  2006;108(10):3253–61. 10.1182/blood-2006-06-027599 [DOI] [PubMed] [Google Scholar]
  • 95. Lucas  C, Wong  P, Klein  J  et al.  Yale IMPACT Team. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature  2020;584(7821):463–9. 10.1038/s41586-020-2588-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Zheng  M, Gao  Y, Wang  G  et al.  Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol Immunol  2020;17(5):533–5. 10.1038/s41423-020-0402-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Xu  Z, Shi  L, Wang  Y  et al.  Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med  2020;8(4):420–2. 10.1016/S2213-2600(20)30076-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Wang  F, Hou  H, Luo  Y  et al.  The laboratory tests and host immunity of COVID-19 patients with different severity of illness. JCI Insight  2020;5(10):e137799. 10.1172/jci.insight.137799 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Zheng  HY, Zhang  M, Yang  CX  et al.  Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients. Cell Mol Immunol  2020;17(5):541–3. 10.1038/s41423-020-0401-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Westmeier  J, Paniskaki  K, Karakose  Z  et al.  Impaired cytotoxic CD8(+) T cell response in elderly COVID-19 patients. mBio  2020;11(5):e02243–20. 10.1128/mBio.02243-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Chen  Z, John Wherry  E. T cell responses in patients with COVID-19. Nat Rev Immunol  2020;20(9): 529–36. 10.1038/s41577-020-0402-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Liu  C, Martins  AJ, Lau  WW  et al.  NIAID COVID Consortium; COVID Clinicians. Time-resolved systems immunology reveals a late juncture linked to fatal COVID-19. Cell  2021;184(7): 1836–57:e1822. 10.1016/j.cell.2021.02.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Li  S, Zhang  Y, Guan  Z  et al.  SARS-CoV-2 triggers inflammatory responses and cell death through caspase-8 activation. Signal Transduct Target Ther  2020;5(1):235. 10.1038/s41392-020-00334-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Chen  RF, Chang  JC, Yeh  WT  et al.  Role of vascular cell adhesion molecules and leukocyte apoptosis in the lymphopenia and thrombocytopenia of patients with severe acute respiratory syndrome (SARS). Microbes Infect  2006;8(1):122–7. 10.1016/j.micinf.2005.06.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Karki  R, Sharma  BR, Tuladhar  S  et al.  Synergism of TNF-α and IFN-γ triggers inflammatory cell death, tissue damage, and mortality in SARS-CoV-2 infection and cytokine shock syndromes. Cell  2021;184(1), 149–168 e117. 10.1016/j.cell.2020.11.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Dondi  E, Roué  G, Yuste  VJ  et al.  A dual role of IFN-alpha in the balance between proliferation and death of human CD4+ T lymphocytes during primary response. J Immunol  2004;173(6):3740–47. 10.4049/jimmunol.173.6.3740 [DOI] [PubMed] [Google Scholar]
  • 107. Gonzalez-Gay  MA, Mayo  J, Castaneda  S, Cifrian  JM, Hernandez-Rodriguez  J. Tocilizumab: from the rheumatology practice to the fight against COVID-19, a virus infection with multiple faces. Expert Opin Biol Ther  2020;20(7):717–23. 10.1080/14712598.2020.1770222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Benucci  M, Giannasi  G, Cecchini  P  et al.  COVID-19 pneumonia treated with Sarilumab: A clinical series of eight patients. J Med Virol  2020;92(11):2368–70. 10.1002/jmv.26062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Addeo  A, Obeid  M, Friedlaender  A. COVID-19 and lung cancer: risks, mechanisms and treatment interactions. J Immunother Cancer  2020;8(1):e000892. 10.1136/jitc-2020-000892 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Liu  Y, Pang  Y, Hu  Z  et al.  Thymosin alpha 1 (Talpha1) reduces the mortality of severe COVID-19 by restoration of lymphocytopenia and reversion of exhausted T cells. Clin Infect Dis  2020;71(16):2150–7. 10.1093/cid/ciaa630 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Riva  G, Nasillo  V, Tagliafico  E  et al.  COVID-19: room for treating T cell exhaustion?  Crit Care  2020;24(1):229. 10.1186/s13054-020-02960-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Patterson  BK, Seethamraju  H, Dhody  K  et al.  Disruption of the CCL5/RANTES-CCR5 pathway restores immune homeostasis and reduces plasma viral load in critical COVID-19. medRxiv  2020;2020.05.02.20084673. 10.1101/2020.05.02.20084673 [DOI] [Google Scholar]
  • 113. Chua  RL, Lukassen  S, Trump  S  et al.  COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat Biotechnol  2020;38(8):970–9. 10.1038/s41587-020-0602-4 [DOI] [PubMed] [Google Scholar]
  • 114. Omarjee  L, Janin 1, Perrot  F  et al.  Targeting T-cell senescence and cytokine storm with rapamycin to prevent severe progression in COVID-19. Clin Immunol  2020;216:108464. 10.1016/j.clim.2020.108464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Zhou  Y, Hou  Y, Shen  J  et al.  Network-based drug repurposing for novel coronavirus 2019-nCoV/SARS-CoV-2. Cell Discov  2020;6:14. 10.1038/s41421-020-0153-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Gordon  AC, Mouncey  PR, Al-Beidh  F  et al. ; REMAPCAP Investigators.  Interleukin-6 receptor antagonists in critically ill patients with covid-19. N Engl J Med  2021;384(16):1491–502. 10.1056/NEJMoa2100433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Andreakos  E, Papadaki  M, Serhan  CN. Dexamethasone, pro-resolving lipid mediators and resolution of inflammation in COVID-19. Allergy  2020;76(3):626–8. 10.1111/all.14595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Tomazini  BM, Maia  IS, Cavalcanti  AB  et al.  COALITION COVID-19 Brazil III Investigators. Effect of dexamethasone on days alive and ventilator-free in patients with moderate or severe acute respiratory distress syndrome and COVID-19: the CoDEX randomized clinical trial. JAMA  2020;324(13):1307–16. 10.1001/jama.2020.17021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Group  RC, Horby  P, Lim  WS  et al.  Dexamethasone in hospitalized patients with covid-19 - preliminary report. N Engl J Med  2020;384(8):693–704. 10.1056/NEJMoa2021436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group, Cain  DW, Cidlowski  JA. After 62 years of regulating immunity, dexamethasone meets COVID-19. Nat Rev Immunol  2020;324(13):1330–41. 10.1001/jama.2020.17023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Cain  DW, Cidlowski  JA. After 62 years of regulating immunity, dexamethasone meets COVID-19. Nat Rev Immunol  2020;20(10):587–8. 10.1038/s41577-020-00421-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Tregoning  JS, Brown  ES, Cheeseman  HM  et al.  Vaccines for COVID-19. Clin Exp Immunol  2020;202(2):162–92. 10.1111/cei.13517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Sahin  U, Muik  A, Derhovanessian  E  et al.  COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses. Nature  2020;586(7830):594–9. 10.1038/s41586-020-2814-7 [DOI] [PubMed] [Google Scholar]
  • 124. Logunov  DY, Dolzhikova  IV, Zubkova  OV  et al.  Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in formulations: two open, non randomised phase 1/2 studies from Russia. Lancet  2020; 396(10255):887–97. 10.1016/S0140-6736(20)31866-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Zhu  FC, Li  YH, Guan  XH  et al.  Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. Lancet  2020;395(10240):1845–54. 10.1016/S0140-6736(20)31208-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Zhu  FC, Guan  XH, Li  YH  et al.  Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet  2020;396(10249):479–88. 10.1016/S0140-6736(20)31605-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Folegatti  PM, Ewer  KJ, Aley  PK  et al. ; Oxford COVID Vaccine Trial Group. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet  2020;396(10249):467–78. 10.1016/S0140-6736(20)31604-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Prendecki  M, Clarke  C, Brown  J  et al.  Effect of previous SARS-CoV-2 infection on humoral and T-cell responses to single-dose BNT162b2 vaccine. Lancet  2021;397(10280):1178–81. 10.1016/S0140-6736(21)00502-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Mahase  E. Covid-19: what do we know about “long covid”?  BMJ  2020;370:m2815. 10.1136/bmj.m2815 [DOI] [PubMed] [Google Scholar]
  • 130. Carfi  A, Bernabei  R, Landi  F; Gemelli Against COVID-19 Post-Acute Care Study Group. Persistent symptoms in patients after acute COVID-19. JAMA  2020;324(6):603–5. 10.1001/jama.2020.12603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Zhang  H, Wu  T. CD4+T, CD8+T counts and severe COVID-19: a meta-analysis. J Infect  2020; 81(3):e82–e84. 10.1016/j.jinf.2020.06.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Hu  D, Li  L, Shi  W  et al.  Less expression of CD4+ and CD8+ T cells might reflect the severity of infection and predict worse prognosis in patients with COVID-19: Evidence from a pooled analysis. Clin Chim Acta  2020;510:1–4. 10.1016/j.cca.2020.06.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Huang  W, Berube  J, McNamara  M  et al.  Lymphocyte subset counts in COVID-19 patients: a meta-analysis. Cytometry A  2020;97(8):772–6. 10.1002/cyto.a.24172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Akbari  H, Tabrizi  R, Lankarani  KB  et al.  The role of cytokine profile and lymphocyte subsets in the severity of coronavirus disease 2019 (COVID-19): a systematic review and meta-analysis. Life Sci  2020;258:118167. 10.1016/j.lfs.2020.118167 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ltab015_suppl_Supplementary_Material

Data Availability Statement

No new datasets were generated for this manuscript.


Articles from Immunotherapy Advances are provided here courtesy of Oxford University Press

RESOURCES