Recombinant human interleukin-7 reverses T cell exhaustion ex vivo in critically ill COVID-19 patients

Frank Bidar, Sarah Hamada, Morgane Gossez, Remy Coudereau, Jonathan Lopez, Marie-Angelique Cazalis, Claire Tardiveau, Karen Brengel-Pesce, Marine Mommert, Marielle Buisson, Filippo Conti, Thomas Rimmelé, Anne-Claire Lukaszewicz, Laurent Argaud, Martin Cour, Guillaume Monneret, Fabienne Venet, RICO Study Group, Remi Pescarmona, Lorna Garnier, Christine Lombard, Magali Perret, Marine Villard, Sébastien Viel, Valérie Cheynet, Elisabeth Cerrato, Estelle Peronnet, Jean-François Llitjos, Laetitia Itah, Inesse Boussaha, Françoise Poitevin-Later, Christophe Malcus, Marine Godignon, Florent Wallet, Marie-Charlotte Delignette, Frederic Dailler, Marie Simon, Auguste Dargent, Pierre-Jean Bertrand, Neven Stevic, Marion Provent, Laurie Bignet, Valérie Cerro, Jean-Christophe Richard, Laurent Bitker, Mehdi Mezidi, Loredana Baboi, Frank Bidar, Sarah Hamada, Morgane Gossez, Remy Coudereau, Jonathan Lopez, Marie-Angelique Cazalis, Claire Tardiveau, Karen Brengel-Pesce, Marine Mommert, Marielle Buisson, Filippo Conti, Thomas Rimmelé, Anne-Claire Lukaszewicz, Laurent Argaud, Martin Cour, Guillaume Monneret, Fabienne Venet, RICO Study Group, Remi Pescarmona, Lorna Garnier, Christine Lombard, Magali Perret, Marine Villard, Sébastien Viel, Valérie Cheynet, Elisabeth Cerrato, Estelle Peronnet, Jean-François Llitjos, Laetitia Itah, Inesse Boussaha, Françoise Poitevin-Later, Christophe Malcus, Marine Godignon, Florent Wallet, Marie-Charlotte Delignette, Frederic Dailler, Marie Simon, Auguste Dargent, Pierre-Jean Bertrand, Neven Stevic, Marion Provent, Laurie Bignet, Valérie Cerro, Jean-Christophe Richard, Laurent Bitker, Mehdi Mezidi, Loredana Baboi

Abstract

Background: Lymphopenia is a hallmark of severe coronavirus disease 19 (COVID-19). Similar alterations have been described in bacterial sepsis and therapeutic strategies targeting T cell function such as recombinant human interleukin 7 (rhIL-7) have been proposed in this clinical context. As COVID-19 is a viral sepsis, the objectives of this study were to characterize T lymphocyte response over time in severe COVID-19 patients and to assess the effect of ex vivo administration of rhIL-7.

Results: Peripheral blood mononuclear cells from COVID-19 patients hospitalized in intensive care unit (ICU) were collected at admission and after 20 days. Transcriptomic profile was evaluated through NanoString technology. Inhibitory immune checkpoints expressions were determined by flow cytometry. T lymphocyte proliferation and IFN-γ production were evaluated after ex vivo stimulation in the presence or not of rhIL-7. COVID-19 ICU patients were markedly lymphopenic at admission. Mononuclear cells presented with inhibited transcriptomic profile prevalently with impaired T cell activation pathways. CD4 + and CD8 + T cells presented with over-expression of co-inhibitory molecules PD-1, PD-L1, CTLA-4 and TIM-3. CD4 + and CD8 + T cell proliferation and IFN-γ production were markedly altered in samples collected at ICU admission. These alterations, characteristic of a T cell exhaustion state, were more pronounced at ICU admission and alleviated over time. Treatment with rhIL-7 ex vivo significantly improved both T cell proliferation and IFN-γ production in cells from COVID-19 patients.

Conclusions: Severe COVID-19 patients present with features of profound T cell exhaustion upon ICU admission which can be reversed ex vivo by rhIL-7. These results reinforce our understanding of severe COVID-19 pathophysiology and opens novel therapeutic avenues to treat such critically ill patients based of immunomodulation approaches. Defining the appropriate timing for initiating such immune-adjuvant therapy in clinical setting and the pertinent markers for a careful selection of patients are now warranted to confirm the ex vivo results described so far. Trial registration ClinicalTrials.gov identifier: NCT04392401 Registered 18 May 2020, http:// clinicaltrials.gov/ct2/show/NCT04392401.

Keywords: Critically ill; Exhaustion; Immunostimulation; Interleukin-7; SARS-CoV-2; T lymphocytes.

Conflict of interest statement

The authors declare no competing financial interests in relation to the work. MAC, KBP and MM are bioMérieux’s employees. This private company had no role in the study design, result analysis and decision to publish this study.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Transcriptomic profile of mononuclear cells in severe COVID-19 patients. RNA extracted from PBMCs of COVID-19 patients at day 0 and day 20 (n = 10) and healthy volunteers (HV, n = 10) was analyzed through NanoString technology. a Volcano plots of differentially expressed genes between patients sampled at D0 or at D20 and HV are showed. Limits of significance are illustrated by red dotted lines (i.e. Log2 Fold Change = −2 or + 2 and −Log 10 P value = 1.3). Selected genes are mentioned. b Venn diagrams of significantly up-regulated (left diagram, n = 21) or down-regulated (right diagram, n = 48) genes between patients and HV are showed. c Ingenuity Pathway Analysis was applied on the list of differentially expressed genes at D0 and D20. Heatmaps of Log 10 P-value (from white indicating the absence of significance to dark red indicating a strong significance) and Z-score (from orange indicating a down-regulation to purple indicating an up-regulation) for pathways related to T cell activation at D0 and D20 are presented
Fig. 2
Fig. 2
Immune-inhibitory receptors expressions on CD4 + and CD8 + T cells in severe COVID-19 patients. PD-1, PD-L1, CTLA-4 and TIM-3 expressions were assessed by flow cytometry in PBMCs from COVID-19 patients (n = 12) at day 0 (D0) and day 20 (D20) and healthy volunteers (HV, n = 10). Results are presented as individual values and boxplots. a Expressions on CD4 + T cells are presented. b Expressions on CD8 + T cells are presented. Nonparametric Mann–Whitney U test was used to compare results between HV and COVID-19 patients. Only statistically significant differences are shown
Fig. 3
Fig. 3
Lymphocyte proliferation and IFN-γ production after TCR stimulation in severe COVID-19 patients. PBMCs were collected from healthy volunteers (HV, n = 7) and COVID-19 patients at D0 and D20 (n = 10). Cells were cultured for 3 days in the presence (TCR stimulation) or not (Control) of anti-CD2/CD3/CD28 Ab-coated beads (ratio of beads/cells = 2:1). Cellular proliferation was assessed by monitoring EdU AF488 incorporation into cells and expressed as percentages of cells incorporating EdU. IFN-γ was measured in cell culture supernatants after stimulation. Results are presented as individual values and boxplots. a CD4 + T cell proliferation is presented. b CD8 + T cell proliferation is presented. c Natural log transformed IFN-γ production in culture supernatants is presented. Nonparametric Mann–Whitney U test was used to compare results between HV and patients. Wilcoxon signed-rank test was used for paired comparisons between D0 and D20. Only statistically significant differences are shown
Fig. 4
Fig. 4
Lymphocyte proliferation and IFN-γ production after rhIL-7 and TCR stimulation in severe COVID-19 patients. PBMCs were collected in COVID-19 patients at D0 (n = 10). Cells were cultured for 3 days in the presence or not of anti-CD2/CD3/CD28 Ab-coated beads (TCR Stimulation, ratio of beads/cells = 2:1) and rhIL-7 (TCR Stimulatio n + rhIL-7,100 ng/ml). Cellular proliferation was assessed by monitoring EdU AF488 incorporation into cells and expressed as percentages of cells incorporating EdU. IFN-γ was measured in cell supernatants after proliferation. Results are presented as individual values and medians. a CD4 + T cell proliferation is presented. b CD8 + T cell proliferation is presented. c Natural log transformed IFN-γ production in culture supernatants is shown. Wilcoxon signed-rank test was used to compare results between groups. Only statistically significant differences are shown

References

    1. Network C-IGobotR, the C-ICUI. Clinical characteristics and day-90 outcomes of 4244 critically ill adults with COVID-19: a prospective cohort study. Intensive Care Med. 2021;47(1):60–73.
    1. Carenzo L, Costantini E, Greco M, Barra FL, Rendiniello V, Mainetti M, et al. Hospital surge capacity in a tertiary emergency referral centre during the COVID-19 outbreak in Italy. Anaesthesia. 2020;75(7):928–934.
    1. Hadjadj J, Yatim N, Barnabei L, Corneau A, Boussier J, Smith N, et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science. 2020;369(6504):718–724.
    1. Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin Infect Dis. 2020.
    1. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8(5):475–481.
    1. Zhou R, To KK, Wong YC, Liu L, Zhou B, Li X, et al. Acute SARS-CoV-2 infection impairs dendritic cell and T cell responses. Immunity. 2020;53(4):864–77.
    1. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13(12):862–874.
    1. Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;13(3):260–268.
    1. Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492–499.
    1. Hotchkiss RS, Moldawer LL. Parallels between cancer and infectious disease. N Engl J Med. 2014;371(4):380–383.
    1. Remy KE, Mazer M, Striker DA, Ellebedy AH, Walton AH, Unsinger J, et al. Severe immunosuppression and not a cytokine storm characterizes COVID-19 infections. JCI Insight. 2020;5:17.
    1. de Roquetaillade C, Monneret G, Gossez M, Venet F. IL-7 and its beneficial role in sepsis-induced T lymphocyte dysfunction. Crit Rev Immunol. 2018;38(6):433–451.
    1. Venet F, Foray AP, Villars-Mechin A, Malcus C, Poitevin-Later F, Lepape A, et al. IL-7 restores lymphocyte functions in septic patients. J Immunol. 2012;189(10):5073–5081.
    1. Unsinger J, Burnham CA, McDonough J, Morre M, Prakash PS, Caldwell CC, et al. Interleukin-7 ameliorates immune dysfunction and improves survival in a 2-hit model of fungal sepsis. J Infect Dis. 2012;206(4):606–616.
    1. Unsinger JMM, Kasten KR, Hoekzema AS, Watanabe E, Muenzer JT, McDonough JS, Tschoep J, Ferguson TA, McDunn JE, Morre M, Hildeman DA, Caldwell CC, Hotchkiss RS. IL-7 Promotes T cell viability, trafficking, and functionality and improves survival in sepsis. J Immunol. 2010;184(7):3768–3779.
    1. Venet F, Demaret J, Blaise BJ, Rouget C, Girardot T, Idealisoa E, et al. IL-7 restores T lymphocyte immunometabolic failure in septic shock patients through mTOR activation. J Immunol. 2017;199(5):1606–1615.
    1. Monneret G, de Marignan D, Coudereau R, Bernet C, Ader F, Frobert E, et al. Immune monitoring of interleukin-7 compassionate use in a critically ill COVID-19 patient. Cell Mol Immunol. 2020;17(9):1001–1003.
    1. Laterre PF, Francois B, Collienne C, Hantson P, Jeannet R, Remy KE, et al. Association of interleukin 7 immunotherapy with lymphocyte counts among patients with severe coronavirus disease 2019 (COVID-19) JAMA Netw Open. 2020;3(7):e2016485.
    1. Venet F, Cour M, Rimmele T, Viel S, Yonis H, Coudereau R, et al. Longitudinal assessment of IFN-I activity and immune profile in critically ill COVID-19 patients with acute respiratory distress syndrome. Crit Care. 2021;25(1):140.
    1. Urrutia A, Duffy D, Rouilly V, Posseme C, Djebali R, Illanes G, et al. Standardized whole-blood transcriptional profiling enables the deconvolution of complex induced immune responses. Cell Rep. 2016;16(10):2777–2791.
    1. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3(7):0034.
    1. Andersen CL, Jensen JL, Orntoft TF. Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res. 2004;64(15):5245–5250.
    1. Poujol F, Monneret G, Friggeri A, Rimmele T, Malcus C, Poitevin-Later F, et al. Flow cytometric evaluation of lymphocyte transformation test based on 5-ethynyl-2'deoxyuridine incorporation as a clinical alternative to tritiated thymidine uptake measurement. J Immunol Methods. 2014;415:71–79.
    1. Thevarajan I, Nguyen THO, Koutsakos M, Druce J, Caly L, van de Sandt CE, et al. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26(4):453–455.
    1. Tan AT, Linster M, Tan CW, Le Bert N, Chia WN, Kunasegaran K, et al. Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell Rep. 2021;34(6):108728.
    1. Rydyznski Moderbacher C, Ramirez SI, Dan JM, Grifoni A, Hastie KM, Weiskopf D, et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell. 2020;183(4):996–1012.
    1. Liu J, Li S, Liu J, Liang B, Wang X, Wang H, et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine. 2020;55:102763.
    1. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130(5):2620–2629.
    1. Stieglitz D, Schmid T, Chhabra NF, Echtenacher B, Mannel DN, Mostbock S. TNF and regulatory T cells are critical for sepsis-induced suppression of T cells. Immun Inflamm Dis. 2015;3(4):374–385.
    1. Venet F, Davin F, Guignant C, Larue A, Cazalis MA, Darbon R, et al. Early assessment of leukocyte alterations at diagnosis of septic shock. Shock. 2010;34(4):358–363.
    1. Avendano-Ortiz J, Lozano-Rodriguez R, Martin-Quiros A, Maroun-Eid C, Terron V, Valentin J, et al. Proteins from SARS-CoV-2 reduce T cell proliferation: a mirror image of sepsis. Heliyon. 2020;6(12):e05635.
    1. Reizine F, Lesouhaitier M, Gregoire M, Pinceaux K, Gacouin A, Maamar A, et al. SARS-CoV-2-induced ARDS associates with MDSC expansion, lymphocyte dysfunction, and arginine shortage. J Clin Immunol. 2021;41(3):515–525.
    1. Coudereau R, Waeckel L, Cour M, Rimmele T, Pescarmona R, Fabri A, et al. Emergence of immunosuppressive LOX-1+ PMN-MDSC in septic shock and severe COVID-19 patients with acute respiratory distress syndrome. J Leukoc Biol. 2021.
    1. Kreutmair S, Unger S, Nunez NG, Ingelfinger F, Alberti C, De Feo D, et al. Distinct immunological signatures discriminate severe COVID-19 from non-SARS-CoV-2-driven critical pneumonia. Immunity. 2021;54(7):1578–93.
    1. Zheng HY, Zhang M, Yang CX, Zhang N, Wang XC, Yang XP, 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):541–543.
    1. Ni L, Cheng ML, Feng Y, Zhao H, Liu J, Ye F, et al. Impaired Cellular Immunity to SARS-CoV-2 in Severe COVID-19 Patients. Front Immunol. 2021;12:603563.
    1. Sattler A, Angermair S, Stockmann H, Heim KM, Khadzhynov D, Treskatsch S, et al. SARS-CoV-2-specific T cell responses and correlations with COVID-19 patient predisposition. J Clin Invest. 2020;130(12):6477–6489.
    1. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, et al. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19) Front Immunol. 2020;11:827.
    1. Chang K, Svabek C, Vazquez-Guillamet C, Sato B, Rasche D, Wilson S, et al. Targeting the programmed cell death 1: programmed cell death ligand 1 pathway reverses T cell exhaustion in patients with sepsis. Crit Care. 2014;18(1):R3.
    1. Snow TAC, Singer M, Arulkumaran N. Immunomodulators in COVID-19: two sides to every coin. Am J Respir Crit Care Med. 2020;202(10):1460–1462.
    1. Andrew D, Aspinall R. Age-associated thymic atrophy is linked to a decline in IL-7 production. Exp Gerontol. 2002;37(2–3):455–463.
    1. Andrew D, Aspinall R. Il-7 and not stem cell factor reverses both the increase in apoptosis and the decline in thymopoiesis seen in aged mice. J Immunol. 2001;166(3):1524–1530.
    1. Nanjappa SG, Walent JH, Morre M, Suresh M. Effects of IL-7 on memory CD8 T cell homeostasis are influenced by the timing of therapy in mice. J Clin Invest. 2008;118(3):1027–1039.
    1. Francois B, Jeannet R, Daix T, Walton AH, Shotwell MS, Unsinger J, et al. Interleukin-7 restores lymphocytes in septic shock: the IRIS-7 randomized clinical trial. JCI Insight. 2018;3(5):e98960.
    1. Mazer MB, Davitt E, Turnbull IR, Caldwell CC, Brakenridge SC, Remy KE, et al. In vitro-administered dexamethasone suppresses t cell function with reversal by interleukin-7 in coronavirus disease 2019. Crit Care Explor. 2021;3(4):e0378.

Source: PubMed

3
Subscribe