Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection

Noah S Butler, Jacqueline Moebius, Lecia L Pewe, Boubacar Traore, Ogobara K Doumbo, Lorraine T Tygrett, Thomas J Waldschmidt, Peter D Crompton, John T Harty, Noah S Butler, Jacqueline Moebius, Lecia L Pewe, Boubacar Traore, Ogobara K Doumbo, Lorraine T Tygrett, Thomas J Waldschmidt, Peter D Crompton, John T Harty

Abstract

Infection of erythrocytes with Plasmodium species induces clinical malaria. Parasite-specific CD4(+) T cells correlate with lower parasite burdens and severity of human malaria and are needed to control blood-stage infection in mice. However, the characteristics of CD4(+) T cells that determine protection or parasite persistence remain unknown. Here we show that infection of humans with Plasmodium falciparum resulted in higher expression of the inhibitory receptor PD-1 associated with T cell dysfunction. In vivo blockade of the PD-1 ligand PD-L1 and the inhibitory receptor LAG-3 restored CD4(+) T cell function, amplified the number of follicular helper T cells and germinal-center B cells and plasmablasts, enhanced protective antibodies and rapidly cleared blood-stage malaria in mice. Thus, chronic malaria drives specific T cell dysfunction, and proper function can be restored by inhibitory therapies to enhance parasite control.

Figures

Figure 1. Human and rodent malaria induce…
Figure 1. Human and rodent malaria induce specific phenotypic and functional characteristics of CD4+ T cell exhaustion
(a) PD-1 expression by CD4+ T cells from Malian children before the malaria season (Before Malaria) and seven days after symptomatic P. falciparum infection (After Malaria). The non-parametric Mann-Whitney test was used to compare continuous variables between groups. (b) Resolution of P. yoelii (Py) blood-stage infection in rodents requires CD4+ T cells and antibody secreting B cells. Survival curves of Py pRBC infected wild-type C57BL/6 mice, wild-type mice depleted of CD4+ or CD8+ T cells on day 10, or C57BL/6 Aicda–/– μs–/– mice. Data represent 2 independent experiments with 5 mice/group. (c) Upregulation of CD49d and CD11a identifies Plasmodium-specific, infection-induced CD4+ T cells. Longitudinal analyses of PBL before (naïve) and 7 days following (Py pRBC) challenge. Data represent 4 independent experiments. (d) Prolonged Py blood-stage infection results in sustained CD4+ T cell proliferation. Histograms show the fraction of CD49dhiCD11ahi Plasmodium-specific (open) or CD49dloCD11alo naïve (filled) CD4+ T cells that have incorporated BrdU following a day 4-8 pulse (top panel) or day 27-30 pulse (bottom panel). Data represent 3 independent experiments with 3 mice/group. (e) Chronic virus (LCMV cl13) and prolonged Plasmodium blood-stage infection (Py pRBC), but not acute virus infection (LCMV Arm) induce T cell inhibitory receptors PD-1 and LAG-3 at day 31 on splenic, pathogen-specific (open) but not naïve (filled), CD4+ T cells. Data represent 3 independent experiments with 3-5 mice/group. (f) Pathogen-specific CD49dhiCD11ahi CD4+ T cells from Py infected mice exhibit dysfunctional IFN-γ, TNF and IL-2 production in response to PMA/ionomycin stimulation. Data (mean±s.d.) are from 4-5 mice/group and are representative of 3 independent experiments. Statistics in (f) were determined by two-tailed, unpaired student's t-test.
Figure 2. Truncation of P. yoelii blood-stage…
Figure 2. Truncation of P. yoelii blood-stage infection with chloroquine reverses CD4+ T cell exhaustion
Groups of C57BL/6 mice were infected with 105 P. yoelii pRBC and subsequently treated with PBS or 80 mg/kg chloroquine/PBS on day 8 and 9 p.i. Spleens were harvested from mice on day 8 (a), or day 24 p.i. (b) and cells were examined for the expression of the indicated cell surface markers or for the functional production of IFN-γ following ex vivo stimulation with PMA-ionomycin (PMA–iono). Numbers in histograms refer to frequency of CD49dhiCD11ahi cells expressing the indicated marker or IFN-γ. Numbers in parentheses show mean fluorescence intensity of T cell inhibitory receptor staining. Data in a,b are representative of 2 independent experiments with 5 mice/group.
Figure 3. Therapeutic in vivo blockade of…
Figure 3. Therapeutic in vivo blockade of PD-1 and LAG-3 in mice improves the anti-Plasmodial CD4+ T cell response and accelerates parasite clearance
(a) Starting on day 14 after infection with 105 Py pRBC, C57BL/6 mice were treated with 200 μg each of anti-PD-L1 and anti-LAG-3, or control rIgG, every 3 days and monitored for parasite burden every two days. Data (mean±s.d.) are from 5 mice/group and are representative of 4 independent experiments. (b) Mice were infected and treated as in (a) and Plasmodium-specific (CD49dhiCD11ahi) splenic CD4+ T cells were enumerated on day 21 p.i. (c) Representative plots showing PMA–iono-induced cytokine expression by CD49dhiCD11ahi CD4+ T cells from rIgG- and inhibitory receptor blockade-treated mice analyzed on day 21p.i. (d) Summary cytokine expression by CD49dhiCD11ahi CD4+ T cells from rIgG- and inhibitory receptor blockade-treated mice. Data (Mean±s.e.m.) in (b) and (d) derive from 2 independent experiments with 3-5 mice/group. Statistics in (a), (d) and (d) were determined by two-tailed, unpaired student's t-test. (*=P<0.05, **P<0.01).
Figure 4. Therapeutic T cell inhibitory blockade…
Figure 4. Therapeutic T cell inhibitory blockade accelerates parasite clearance in genetically diverse backgrounds and prevents chronic Plasmodium infection
(a) Female Swiss Webster mice (n=10/group) were infected with 105 P. yoelii parasitized red blood cells and given 200 μg of anti-PD-L1 and anti-LAG-3, or control rIgG, every three days starting on day 14 p.i. Parasitemia was monitored in individual mice until day 24. Statistics were determined by two-tailed, unpaired student's t-test. Data (Mean±s.d.) are representative of 2 independent experiments. Asterisks indicate P < 0.005. (b-c) Therapeutic in vivo blockade of PD-1 and LAG-3 signaling results in sterilizing clearance of persistent, subpatent P. chabaudi chabaudi (Pcc) infection in the majority of mice. (b) C57BL/6 mice were infected with 104 Pcc blood-stage parasites and given 200 μg of anti-PD-L1 and anti-LAG-3 every three days from days 14 to 32. Parasitemia was monitored on the indicated days. Data (Mean±s.d.) are from 5 mice/group. On day 40 post-challenge, 100 μl of whole blood was collected via cardiac puncture from each donor mouse, diluted 1:2 in saline and injected intravenously into new naïve recipient mice. Recipient mice were monitored for the development of patent Pcc parasitemia from day 2 to day 20 post-transfer. (c) Frequency of patent infection and quantification of parasite burden on day 9 post-transfer in recipient mice are shown. For b-c, the parasitemia limit of detection (L.O.D.) was 0.02%. Data in b-c are representative of 2 independent experiments.
Figure 5. Therapeutic in vivo blockade of…
Figure 5. Therapeutic in vivo blockade of PD-L1 and LAG-3 in mice enhances TFH CD4+ T cell and plasmablast differentiation during clinical malaria
(a) Starting on day 14 after Py pRBC infection, groups of C57BL/6 mice were treated with 200 μg each of anti-PD-L1 and anti-LAG-3, or control rIgG, every 3 days. On day 21 p.i., splenic CD49dhiCD11ahiCXCR5hiCD150lo CD4+ T follicular helper (Tfh) cells were enumerated. (b) Mice were infected and treated as in (a) and CD19hi/intB220hi/int B cells were analyzed for early plasma cell differentiation (evidenced by coordinate downregulation of IgD and upregulation of CD138 prior to loss of CD19 and B220 expression). (c) Summary data (Mean±s.d.) showing total numbers of TFH and pre-plasma cells in rIgG- and inhibitory receptor blockade-treated mice 21 days p.i. Statistics in (c) were determined by two-tailed, unpaired student's t-test. Data in a-c are representative of 3 independent experiments with 5 mice/group.
Figure 6. Enhanced germinal center B cell…
Figure 6. Enhanced germinal center B cell reaction, class switch recombination and functionally protective anti-Plasmodial antibody secretion following therapeutic PDL1 and LAG-3 blockade during clinical malaria
(a) C57BL/6 mice were infected with Py pRBC and subsequently treated with blocking antibodies as described for Figure 2. On day 21 p.i., total CD19+B220+ B cells were enumerated. Data (Mean±s.d.) represent 3 independent experiments with 3 mice/group. (b) Representative dot plots showing enhanced splenic germinal center (B220hi/intPNAhi) B cell responses on day 21 p.i. in mice that received anti-PD-L1 and anti-LAG-3 treatment from days 14 to 20 p.i. Histograms show the frequency of class-switching (IgMlo and IgG2bhi) in germinal center (PNAhi) and non-germinal center (PNAlo) B cells. (c) Summary data (Mean±s.d.) show total numbers of germinal center and class-switched B cells in rIgG- versus anti-PD-L1 and anti-LAG-3 treated mice and are representative of 3 independent experiments with 3 mice/group. Statistics in (a) and (c) were determined by 2-tailed unpaired student's t-test. (d) Sera from Py infected mice that were subsequently treated with rIgG or anti-PDL1 and anti-LAG-3 from days 14 to 32 were collected on day 41 p.i. Total MSP-119-specific IgG antibodies were detected as described in Methods. Data (Mean±s.d.) are expressed as average endpoint titers with absorbance readings below 0.2 (A405) and are representative of 2 experiments with 4 mice/group. (e) Parasite burdens and clearance kinetics following Py pRBC challenge of naïve mice receiving passive transfer of 150 μl of serum from donor mice subjected to the indicated treatments during Py malaria. Donor serum was obtained 41 days after initial Py pRBC challenge (2 weeks after cessation of blockade therapy). Data (Mean±s.d.) in (e) were analyzed by One-way ANOVA with Tukey's post-test of multiple comparisons and are representative of 2 independent experiments with 4 mice/group (*P<0.05; **P<0.01; ***P<0.001 for rIgG versus α-PD-L1+α-LAG-3).

References

    1. Mackinnon MJ, Marsh K. The selection landscape of malaria parasites. Science. 2010;328:866–871.
    1. World Health Organization World Malaria Report 2010. .
    1. Snow RW, Marsh K. The consequences of reducing transmission of Plasmodium falciparum in Africa. Adv Parasitol. 2002;52:235–264.
    1. Wykes M, Good MF. A case for whole-parasite malaria vaccines. Int J Parasitol. 2007;37:705–712.
    1. Good MF, Doolan DL. Malaria vaccine design: immunological considerations. Immunity. 2010;33:555–566.
    1. Pinzon-Charry A, et al. Low doses of killed parasite in CpG elicit vigorous CD4+ T cell responses against blood-stage malaria in mice. J Clin Invest. 2010;120:2967–2978.
    1. Elliott SR, Kuns RD, Good MF. Heterologous immunity in the absence of variant-specific antibodies after exposure to subpatent infection with blood-stage malaria. Infect Immun. 2005;73:2478–2485.
    1. Pombo DJ, et al. Immunity to malaria after administration of ultra-low doses of red cells infected with Plasmodium falciparum. Lancet. 2002;360:610–617.
    1. Riley EM, Wahl S, Perkins DJ, Schofield L. Regulating immunity to malaria. Parasite Immunol. 2006;28:35–49.
    1. Amante FH, Good MF. Prolonged Th1-like response generated by a Plasmodium yoelii-specific T cell clone allows complete clearance of infection in reconstituted mice. Parasite Immunol. 1997;19:111–126.
    1. Amante FH, Good MF. Experimental asexual blood stage malaria immunity. Curr Protoc Immunol. 2001 Chapter 19, Unit 19 14.
    1. Kumar S, Miller LH. Cellular mechanisms in immunity to blood stage infection. Immunol Lett. 1990;25:109–114.
    1. Vinetz JM, et al. Adoptive transfer of CD8+ T cells from immune animals does not transfer immunity to blood stage Plasmodium yoelii malaria. J Immunol. 1990;144:1069–1074.
    1. Miller LH, Good MF, Milon G. Malaria pathogenesis. Science. 1994;264:1878–1883.
    1. Krajden S, Panisko DM, Tobe B, Yang J, Keystone JS. Prolonged infection with Plasmodium falciparum in a semi-immune patient. Trans R Soc Trop Med Hyg. 1991;85:731–732.
    1. Revel MP, et al. Plasmodium falciparum malaria after three years in a non-endemic area. Trans R Soc Trop Med Hyg. 1988;82:832.
    1. Theunissen C, et al. Falciparum malaria in patient 9 years after leaving malaria-endemic area. Emerg Infect Dis. 2009;15:115–116.
    1. Li C, Seixas E, Langhorne J. Rodent malarias: the mouse as a model for understanding immune responses and pathology induced by the erythrocytic stages of the parasite. Med Microbiol Immunol. 2001;189:115–126.
    1. Day CL, et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443:350–354.
    1. Betts MR, et al. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood. 2006;107:4781–4789.
    1. Migueles SA, et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol. 2002;3:1061–1068.
    1. Zajac AJ, et al. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med. 1998;188:2205–2213.
    1. Barber DL, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–687.
    1. Blackburn SD, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10:29–37.
    1. Weiss GE, et al. The Plasmodium falciparum-specific human memory B cell compartment expands gradually with repeated malaria infections. PLoS Pathog. 2010;6:e1000912.
    1. Butler NS, et al. Superior antimalarial immunity after vaccination with late liver stage-arresting genetically attenuated parasites. Cell Host Microbe. 2011;9:451–462.
    1. Rai D, Pham NL, Harty JT, Badovinac VP. Tracking the total CD8 T cell response to infection reveals substantial discordance in magnitude and kinetics between inbred and outbred hosts. J Immunol. 2009;183:7672–7681.
    1. McDermott D, Varga SM. Quantifying antigen-specific CD4 T cells during a viral infection: CD4 T cell responses are larger than we think. J Immunol. 2011;187 (In Press)
    1. Oxenius A, Bachmann MF, Zinkernagel RM, Hengartner H. Virus-specific MHC-class II-restricted TCR-transgenic mice: effects on humoral and cellular immune responses after viral infection. Eur J Immunol. 1998;28:390–400.
    1. Brown KE, Freeman GJ, Wherry EJ, Sharpe AH. Role of PD-1 in regulating acute infections. Curr Opin Immunol. 2010;22:397–401.
    1. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8:239–245.
    1. Goldberg MV, Drake CG. LAG-3 in Cancer Immunotherapy. Curr Top Microbiol Immunol. 2011;344:269–278.
    1. Huang CT, et al. Role of LAG-3 in regulatory T cells. Immunity. 2004;21:503–513.
    1. Achtman AH, Stephens R, Cadman ET, Harrison V, Langhorne J. Malaria-specific antibody responses and parasite persistence after infection of mice with Plasmodium chabaudi chabaudi. Parasite Immunol. 2007;29:435–444.
    1. Langhorne J. The role of CD4+ T-cells in the immune response to Plasmodium chabaudi. Parasitol Today. 1989;5:362–364.
    1. Meding SJ, Langhorne J. CD4+ T cells and B cells are necessary for the transfer of protective immunity to Plasmodium chabaudi chabaudi. Eur J Immunol. 1991;21:1433–1438.
    1. Titanji K, et al. Acute depletion of activated memory B cells involves the PD-1 pathway in rapidly progressing SIV-infected macaques. J Clin Invest. 2010;120:3878–3890.
    1. Velu V, et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature. 2009;458:206–210.
    1. Crotty S. Follicular helper CD4 T cells (TFH). Annu Rev Immunol. 2011;29:621–663.
    1. Fahey LM, et al. Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells. J Exp Med. 2011;208:987–999.
    1. Wipasa J, et al. Effect of Plasmodium yoelii exposure on vaccination with the 19-kilodalton carboxyl terminus of merozoite surface protein 1 and vice versa and implications for the application of a human malaria vaccine. Infect Immun. 2009;77:817–824.
    1. Chandele A, Mukerjee P, Das G, Ahmed R, Chauhan VS. Phenotypic and functional profiling of malaria-induced CD8 and CD4 T cells during blood-stage infection with Plasmodium yoelii. Immunology. 2011;132:273–286.
    1. Kline J, Gajewski TF. Clinical development of mAbs to block the PD1 pathway as an immunotherapy for cancer. Curr Opin Investig Drugs. 2010;11:1354–1359.
    1. Porichis F, et al. Responsiveness of HIV-specific CD4 T cells to PD-1 blockade. Blood. 2011;118:965–974.
    1. Golden-Mason L, et al. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J Virol. 2009;83:9122–9130.
    1. Barber DL, Mayer-Barber KD, Feng CG, Sharpe AH, Sher A. CD4 T cells promote rather than control tuberculosis in the absence of PD-1-mediated inhibition. J Immunol. 2011;186:1598–1607.
    1. Reiley WW, et al. Distinct functions of antigen-specific CD4 T cells during murine Mycobacterium tuberculosis infection. Proc Natl Acad Sci U S A. 2010;107:19408–19413.
    1. Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12:492–499.
    1. White NJ. The treatment of malaria. N Engl J Med. 1996;335:800–806.
    1. Barfod L, et al. Evasion of immunity to Plasmodium falciparum malaria by IgM masking of protective IgG epitopes in infected erythrocyte surface-exposed PfEMP1. Proc Natl Acad Sci U S A. 2011;108:12485–12490.
    1. Wojciechowski W, et al. Cytokine-producing effector B cells regulate type 2 immunity to H. polygyrus. Immunity. 2009;30:421–433.
    1. Nolz JC, Harty JT. Protective capacity of memory CD8(+) T cells is dictated by antigen exposure history and nature of the infection. Immunity. 2011;34:781–793.

Source: PubMed

3
Předplatit