10
REPORTS Cite as: K. E. Pauken et al., Science 10.1126/science.aaf2807 (2016). Persisting antigenic stimulation during chronic infections and cancer can result in T cell exhaustion, a state of impaired effector functions, high inhibitory receptor expression including Programmed Death-1 (PD-1, or CD279), transcriptional reprogramming, and defective immune memory (1). Collectively, these properties prevent optimal control of persisting pathogens and tumors. Blocking the PD-1:PD-L1 pathway can re-invigorate TEX, improving effector functions and enhancing viral and tumor control (1). Recently developed inhibitors of the PD-1 and CTLA-4 pathways represent a new paradigm in cancer treatment (24). Although promising, the majority of patients fail to develop durable responses, and most eventually progress (24). Thus, it is unclear whether blocking PD-1 can promote long-lasting improvements and immunological memory development in TEX. To address this question, we interrogated the cellular, transcriptional and epigenetic changes associated with PD-1 pathway blockade using the mouse model of chronic lym- phocytic choriomeningitis virus (LCMV) infection (fig. S1, A to C) (5, 6). Following anti-PD-L1 treatment, 1080 genes were up-regulated and 1686 genes were down-regulated (p<0.05, LFC≥0.2) (Fig. 1A, fig. S1D, and table S1). Previous studies identified transcriptional (7) or cellular (8, 9) chang- es in metabolic pathways following PD-1 pathway blockade. Indeed, several metabolic genes were altered following PD- L1 blockade (table S1). Gene Set Enrichment Analysis (GSEA), however, identified more prominent changes in cell division pathways (Fig. 1B, table S2) (5, 10). In addition, many effector-related genes were biased toward the anti-PD- L1 group (Fig. 1C-D, table S3). Other genes of interest in- cluded Cxcl9, Il1r2 and Il7r (up) and Klra9, Tnfrsf9, and Cd200r2 (down) (fig. S1D and table S1). Using Leading Edge Metagene (LEM) analysis (11) two metagenes were identified in anti-PD-L1-treated TEX compared to control TEX; one cor- responding to leukocyte activation and one to cell cycle (Fig. 1E; fig. S1, E and F; and table S4). The anti-PD-L1-treated TEX metagenes displayed some overlap with TEFF, largely driven by cell cycle pathways, but minimal overlap with TMEM (Fig. 1E and table S4) suggesting limited acquisition of memory potential upon TEX re-invigoration. PD-1 pathway blockade can re-activate functions in TEX, but whether re-invigoration is sustained is unclear. Here, there was a robust re-invigoration of TEX as expected (Fig. 1, F and G, and figs. S1, A and B, and S2) (5), and expansion peaked ~3 weeks after initiation of blockade. By 8-11 weeks post-treatment, however, this re-invigoration was lost and the quantity, proliferation, effector function and inhibitory receptor expression of LCMV-specific CD8 T cells in the an- ti-PD-L1-treated mice were comparable to control-treated Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade Kristen E. Pauken, 1 Morgan A. Sammons, 2 Pamela M. Odorizzi, 1 Sasikanth Manne, 1 Jernej Godec, 3,4 Omar Khan, 1 Adam M. Drake, 2 Zeyu Chen, 1 Debattama Sen, 3 Makoto Kurachi, 1 R. Anthony Barnitz, 3 Caroline Bartman, 1 Bertram Bengsch, 1 Alexander C. Huang, 5 Jason M. Schenkel, 6 Golnaz Vahedi, 9 W. Nicholas Haining, 3,7,8 Shelley L. Berger, 2 E. John Wherry 1 * 1 Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. 2 Departments of Cell and Developmental Biology, Genetics, and Biology, Penn Epigenetics Program, University of Pennsylvania, Philadelphia, PA, USA. 3 Department of Pediatric Oncology, Dana- Farber Cancer Institute, Boston, MA, USA. 4 Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA. 5 Department of Medicine and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. 6 Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA. 7 Broad Institute of MIT and Harvard, Cambridge, MA, USA. 8 Division of Hematology/Oncology, Children’s Hospital, Harvard Medical School, Boston, MA, USA. 9 Department of Genetics and Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA. *Corresponding author. E-mail: [email protected] Blocking PD-1 can re-invigorate exhausted CD8 T cells (TEX) and improve control of chronic infections and cancer. However, whether blocking PD-1 can reprogram TEX into durable memory T cells (TMEM) is unclear. We found that re-invigoration of TEX in mice by PD-L1 blockade caused minimal memory development. After blockade, re-invigorated TEX became re-exhausted if antigen remained high, and failed to become TMEM upon antigen clearance. TEX acquired an epigenetic profile distinct from effector (TEFF) and TMEM cells that was minimally remodeled following PD-L1 blockade. This suggests TEX are a distinct lineage of CD8 T cells. Nevertheless, PD-1 pathway blockade resulted in transcriptional rewiring and re-engagement of effector circuitry in the TEX epigenetic landscape. These data indicate that epigenetic fate inflexibility may limit current immunotherapies. First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 1 on March 10, 2020 http://science.sciencemag.org/ Downloaded from

Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

REPORTS

Cite as: K. E. Pauken et al., Science 10.1126/science.aaf2807 (2016).

Persisting antigenic stimulation during chronic infections and cancer can result in T cell exhaustion, a state of impaired effector functions, high inhibitory receptor expression including Programmed Death-1 (PD-1, or CD279), transcriptional reprogramming, and defective immune memory (1). Collectively, these properties prevent optimal control of persisting pathogens and tumors. Blocking the PD-1:PD-L1 pathway can re-invigorate TEX, improving effector functions and enhancing viral and tumor control (1). Recently developed inhibitors of the PD-1 and CTLA-4 pathways represent a new paradigm in cancer treatment (2–4). Although promising, the majority of patients fail to develop durable responses, and most eventually progress (2–4). Thus, it is unclear whether blocking PD-1 can promote long-lasting improvements and immunological memory development in TEX.

To address this question, we interrogated the cellular, transcriptional and epigenetic changes associated with PD-1 pathway blockade using the mouse model of chronic lym-phocytic choriomeningitis virus (LCMV) infection (fig. S1, A to C) (5, 6). Following anti-PD-L1 treatment, 1080 genes were up-regulated and 1686 genes were down-regulated (p<0.05, LFC≥0.2) (Fig. 1A, fig. S1D, and table S1). Previous studies identified transcriptional (7) or cellular (8, 9) chang-es in metabolic pathways following PD-1 pathway blockade.

Indeed, several metabolic genes were altered following PD-L1 blockade (table S1). Gene Set Enrichment Analysis (GSEA), however, identified more prominent changes in cell division pathways (Fig. 1B, table S2) (5, 10). In addition, many effector-related genes were biased toward the anti-PD-L1 group (Fig. 1C-D, table S3). Other genes of interest in-cluded Cxcl9, Il1r2 and Il7r (up) and Klra9, Tnfrsf9, and Cd200r2 (down) (fig. S1D and table S1). Using Leading Edge Metagene (LEM) analysis (11) two metagenes were identified in anti-PD-L1-treated TEX compared to control TEX; one cor-responding to leukocyte activation and one to cell cycle (Fig. 1E; fig. S1, E and F; and table S4). The anti-PD-L1-treated TEX metagenes displayed some overlap with TEFF, largely driven by cell cycle pathways, but minimal overlap with TMEM (Fig. 1E and table S4) suggesting limited acquisition of memory potential upon TEX re-invigoration.

PD-1 pathway blockade can re-activate functions in TEX, but whether re-invigoration is sustained is unclear. Here, there was a robust re-invigoration of TEX as expected (Fig. 1, F and G, and figs. S1, A and B, and S2) (5), and expansion peaked ~3 weeks after initiation of blockade. By 8-11 weeks post-treatment, however, this re-invigoration was lost and the quantity, proliferation, effector function and inhibitory receptor expression of LCMV-specific CD8 T cells in the an-ti-PD-L1-treated mice were comparable to control-treated

Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade Kristen E. Pauken,1 Morgan A. Sammons,2 Pamela M. Odorizzi,1 Sasikanth Manne,1 Jernej Godec,3,4 Omar Khan,1 Adam M. Drake,2 Zeyu Chen,1 Debattama Sen,3 Makoto Kurachi,1 R. Anthony Barnitz,3 Caroline Bartman,1 Bertram Bengsch,1 Alexander C. Huang,5 Jason M. Schenkel,6 Golnaz Vahedi,9 W. Nicholas Haining,3,7,8 Shelley L. Berger,2 E. John Wherry1* 1Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. 2Departments of Cell and Developmental Biology, Genetics, and Biology, Penn Epigenetics Program, University of Pennsylvania, Philadelphia, PA, USA. 3Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. 4Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA. 5Department of Medicine and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. 6Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA. 7Broad Institute of MIT and Harvard, Cambridge, MA, USA. 8Division of Hematology/Oncology, Children’s Hospital, Harvard Medical School, Boston, MA, USA. 9Department of Genetics and Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA.

*Corresponding author. E-mail: [email protected]

Blocking PD-1 can re-invigorate exhausted CD8 T cells (TEX) and improve control of chronic infections and cancer. However, whether blocking PD-1 can reprogram TEX into durable memory T cells (TMEM) is unclear. We found that re-invigoration of TEX in mice by PD-L1 blockade caused minimal memory development. After blockade, re-invigorated TEX became re-exhausted if antigen remained high, and failed to become TMEM upon antigen clearance. TEX acquired an epigenetic profile distinct from effector (TEFF) and TMEM cells that was minimally remodeled following PD-L1 blockade. This suggests TEX are a distinct lineage of CD8 T cells. Nevertheless, PD-1 pathway blockade resulted in transcriptional rewiring and re-engagement of effector circuitry in the TEX epigenetic landscape. These data indicate that epigenetic fate inflexibility may limit current immunotherapies.

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 1

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 2: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

mice (Fig. 1, F to H, and figs. S2 to S4). Moreover, although anti-PD-L1 treatment reduced viral load immediately after treatment, 4 months later viral load was similar to control-treated mice (Fig. 1I). Lastly, 18-29 weeks after cessation of blockade, the transcriptional profiles of control- and anti-PD-L1-treated groups were similar (Fig. 1J, figs. S5 and S6, and tables S5 and S6). Collectively, these data indicate that when antigen remains high, TEX re-invigorated by PD-1 pathway blockade become “re-exhausted.”

One possible reason the effects of PD-L1 blockade were not sustained was the infection persisted. We hypothesized that if the infection was cured, anti-PD-L1 might induce dif-ferentiation into TMEM. To test this idea, we transferred equal numbers of control TEX, anti-PD-L1-treated TEX, or TMEM into antigen-free mice and monitored persistence (fig. S7A). Consistent with previous studies (12, 13), TEX survived poorly in antigen-free recipients compared to functional TMEM (Fig. 2A-B). There was a trend toward anti-PD-L1-treated TEX persisting moderately better, though poorly compared to TMEM (Fig. 2A-B). We next interrogated poten-tial mechanisms for this trend. Following PD-1 pathway blockade, interleukin (IL)-7 receptor transcripts (Il7r; CD127) increased significantly (fig. S1D and table S1). There was also a modest increase in CD127 protein on a subset of TEX following anti-PD-L1 (Fig. 2C-2E). Upon stimulation with IL-7, anti-PD-L1-treated TEX also showed more phos-pho-STAT5 compared to control-treated TEX (Fig. 2F and fig. S7B). In contrast, expression of the IL-15 receptor subunit CD122 and responsiveness to IL-15 in vitro were not sub-stantially altered (Fig. 2, C and F, and fig. S7B). These data suggest that anti-PD-L1 treatment may augment activity of the memory-biased IL-7R pathway.

Treating with IL-7 starting in the effector phase can pre-vent development of exhaustion (14, 15). However, later in chronic infection TEX respond poorly to IL-7 (12, 13). Because anti-PD-L1 improved IL-7R signaling, we tested whether combined treatment had additional benefit (fig. S7C). In-deed, while other aspects of the response were less affected, treatment with IL-7 and anti-PD-L1, but not IL-7 alone, re-sulted in more antigen-specific CD8 T cells and improved co-production of IFNγ and TNFα (Fig. 2, G to H, and fig. S7, D to H). Thus, it may be possible to exploit pathways upreg-ulated by PD-L1 blockade including IL-7R to improve check-point blockade.

We next tested whether PD-1 pathway blockade could re-store robust recall potential upon re-infection, a defining property of TMEM. Equal numbers of DbGP33+ CD8 TEX, anti-PD-L1-treated TEX, or TMEM were transferred into antigen-free mice, rested, and then rechallenged with Listeria mono-cytogenes expressing GP33-41. TMEM robustly expanded, and efficiently produced IFNγ (Fig. 3A-D). In contrast, both con-trol- and anti-PD-L1-treated TEX mounted poor responses to

Listeria-GP33 challenge and re-invigorated TEX were as de-fective as control TEX in these key properties (Fig. 3A-D).

After antigen withdrawal, TEX and anti-PD-L1-treated TEX failed to down-regulate PD-1 (Fig. 3E), consistent with Pdcd1 locus DNA methylation and long-term expression of PD-1 (16–18). TEX also have lower global di-acetylated histone H3 (19), but how this relates to differentiation is unclear. We hypothesized that the genome-wide epigenetic landscape of TEX may contribute to the lack of durable improvements following PD-1 pathway blockade. Thus, we performed glob-al chromatin landscape mapping using ATAC-seq (20) (fig. S8). The majority of open chromatin regions (OCRs) identi-fied were in intergenic regions (33.3-43.3%) or introns (43.4-48.5%) (fig. S9A), as expected (21). TEFF, TMEM, and TEX showed substantial chromatin remodeling compared to TN (Fig. 3F and fig. S9, B and C) and genes with transcriptional start sites (TSS) within 20 kb of OCRs tended to be more highly expressed (fig. S10). OCRs at specific genes illustrated distinct patterns for TEFF, TMEM and TEX. For example, TEX lacked several OCRs present the Ifng locus in TEFF and TMEM (Fig. 3G, blue boxes). Similarly, for Pdcd1, TEX-specific OCRs were identified in the “B” and “C” regions (Fig. 3G, black box) (9, 22, 23) and a previously unidentified OCR ~23 kb from the TSS (Fig. 3G, red box). Global hierarchical cluster-ing and co-cluster analysis showed that TEFF and TMEM were more similar to each other than to TEX and that TEX had a distinct global epigenetic landscape (Fig. 3, H to J, and figs. S11 to S13). These data suggest that TEX may represent a dis-tinct lineage of CD8 T cells.

Two subsets of TEX have been defined based on expres-sion of Eomes, T-bet and PD-1 (24, 25) and additional heter-ogeneity has recently been described (26–28). The T-bethiEomeslo PD-1int subset can be re-invigorated by PD-1 pathway blockade while the EomeshiPD-1hi subset is more terminal and responds poorly to blocking PD-1 (24, 25). Here, TEX were ~80% Eomeshi and ~20% T-bethi, and this distribution changed minimally upon anti-PD-L1 treatment (fig. S14, A to C). The transcriptional and epigenetic profiles of TEX and anti-PD-L1-treated TEX were significantly en-riched for genes from the Eomeshi subset (fig. S14, D and E) (29). However, there was also a trend toward enrichment of genes from the PD-1intTbethi TEX subset in the anti-PD-L1-treated group (fig. S14, F and G), perhaps reflecting recent conversion of Tbethi cells into Eomeshi cells or additional heterogeneity.

We next examined the ability of PD-1 pathway blockade to reprogram the epigenetic landscape of TEX. Hierarchical clustering, co-clustering, and principle component analysis showed considerable similarity between control and anti-PD-L1-treated TEX (Fig. 3, H to J, and fig. S11). OCRs prefer-entially found in both TEX and anti-PD-L1-treated TEX were located near Pdcd1, Il10, Ctla4, Cxcr5 and elsewhere suggest-

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 2

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 3: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

ing state-specific regulation that was not substantially al-tered following PD-L1 blockade (fig. S13). While globally the epigenetic changes were modest, co-cluster analysis identi-fied a small subset of OCRs uniquely enriched in TEX (555 peaks) or anti-PD-L1-treated TEX (98 peaks) (Fig. 3, H to I; fig. S13; and table S7). Some of these genes showed the same trend epigenetically and transcriptionally (e.g., CD200r; fig. S10E) and specific biological pathways were enriched in sets of genes near OCRs that changed (fig. S15).

TEX displayed ~6000 unique OCR changes compared to TEFF and TMEM (Fig. 3, F to I). Thus, the ~650 OCR changes induced by PD-L1 blockade was modest by comparison. To determine whether these changes impacted specific tran-scriptional circuits, we identified transcription factor (TF) motifs enriched in peaks gained (e.g., NFκB, Jun:AP1, and CTCF) or lost (e.g., NFATc1, NFAT:AP1, Nur77, Eomes and Egr2) (Fig. 4A). We hypothesized that re-invigoration result-ed from rewired transcriptional control within the existing TEX epigenetic landscape. To test this notion, Wellington bootstrap analysis was performed to predict TF binding ac-tivity (Fig. 4B and table S10). TEX and anti-PD-L1-treated TEX were more similar to each other than to TN, TEFF or TMEM. However, TF motifs biased toward TEX or anti-PD-L1-treated TEX were identified (Fig. 4B and table S10). TF footprinting was then performed to identify TFs with evidence of likely binding (Fig. 4C and figs. S16 and S17). An integrated net-work was then constructed for transcriptional circuitry based on predicted TF activity (Fig. 4D and table S11). This network identified augmented activity of NFκB, IRFs, and bZip factors (AP-1 family) and decreased activity of NFAT, Egr2, and Nur77 upon PD-L1 blockade. Major features of this transcriptional network were recapitulated using a sec-ond network approach where additional TF families were identified (e.g., Runx, Nr2f6, Prdm1, Rarb, Pparg.Rxra and homeobox TFs; fig. S18 and table S12). To further interro-gate how these changes might affect a specific TF, we exam-ined NFAT. NFAT working with AP-1 transactivates many effector-phase genes. In contrast, “partnerless” NFAT that fails to bind AP-1 induces a subset of TEX genes (30). Here, upon anti-PD-L1 treatment, there was significantly reduced expression of targets of partnerless NFAT in re-invigorated TEX (Fig. 4E), suggesting a rewiring of this transcriptional circuit following blockade.

Together these data suggested that, while PD-1 pathway blockade did not fully reprogram TEX into TMEM or TEFF, these cells may (re)acquire some features of TEFF biology. One hypothesis is that upon PD-L1 blockade the rewired tran-scriptional network allows TEX to preferentially re-engage features of their epigenomic program that overlap with TEFF. To test this idea, we separated TF target genes into those containing OCRs that were: a) unique to TEFF; b) unique to TEX; or c) shared between TEFF and TEX (Fig. 4F). We then

examined the change in genes expressed in each category following PD-L1 blockade. For several TFs including T-bet and Eomes there was no redistribution of the pattern of target gene expression (Fig. 4F). However, for many TFs identified above that have a key role in effector biology such as NFκB, IRF1, IRF2, Nur77 and Blimp-1 (encoded by Prdm1), there was an increase in the number of target genes expressed in the TEFF and TEX overlap group compared to the TEX-only group upon PD-L1 blockade (Fig. 4F). Moreover, genes in the shared TEFF and TEX epigenetic module dis-played a substantially greater magnitude of change in ex-pression than genes in the TEX only group (Fig. 4F). These data indicate that PD-1 pathway blockade induces rewired transcriptional activity allowing TEX to more effectively re-engage modules of effector genes contained within the epi-genetic landscape of TEX. Specific TF circuits altered such as NFκB may have implications for co-targeting PD-1 and TNFR family pathways (1, 3, 31) and may be relevant for design of future therapeutics.

Our data suggest that in settings of severe T cell exhaus-tion, re-acquiring durable immune memory may be chal-lenging, especially if tumor or viral antigen persists. However, our data also indicate that PD-1 pathway blockade may reveal opportunities to further augment T cell quality or effector activity (e.g., NFκB, IL-7R). Additional strategies such as priming new T cell responses (3), selectively expand-ing less exhausted subsets (25), or targeting multiple immu-noregulatory or homeostatic pathways (e.g., IL-7, IL-2) simultaneously (1, 3) may also augment acquisition of dura-ble immunity. Finally, these studies provide the impetus for extending epigenetic landscape mapping to human TEX, fu-ture evaluation of checkpoint blockade combined with epi-genetic modifiers, or epigenomic engineering for T cells. Thus, integrated cellular, transcriptional and epigenetic pro-filing of TEX not only reveals mechanistic insights into PD-1 pathway blockade mediated re-invigoration, but also points to key opportunities to improve long-term durability of these effects.

REFERENCES AND NOTES 1. E. J. Wherry, M. Kurachi, Molecular and cellular insights into T cell exhaustion. Nat.

Rev. Immunol. 15, 486–499 (2015). doi:10.1038/nri3862 Medline 2. D. B. Page, M. A. Postow, M. K. Callahan, J. P. Allison, J. D. Wolchok, Immune

modulation in cancer with antibodies. Annu. Rev. Med. 65, 185–202 (2014). doi:10.1146/annurev-med-092012-112807 Medline

3. P. Sharma, J. P. Allison, The future of immune checkpoint therapy. Science 348, 56–61 (2015). doi:10.1126/science.aaa8172 Medline

4. D. S. Shin, A. Ribas, The evolution of checkpoint blockade as a cancer therapy: What’s here, what’s next? Curr. Opin. Immunol. 33, 23–35 (2015). doi:10.1016/j.coi.2015.01.006 Medline

5. D. L. Barber, E. J. Wherry, D. Masopust, B. Zhu, J. P. Allison, A. H. Sharpe, G. J. Freeman, R. Ahmed, Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687 (2006). doi:10.1038/nature04444 Medline

6. See supplementary information on Science Online. 7. M. M. Gubin, X. Zhang, H. Schuster, E. Caron, J. P. Ward, T. Noguchi, Y. Ivanova, J.

Hundal, C. D. Arthur, W.-J. Krebber, G. E. Mulder, M. Toebes, M. D. Vesely, S. S. K.

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 3

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 4: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

Lam, A. J. Korman, J. P. Allison, G. J. Freeman, A. H. Sharpe, E. L. Pearce, T. N. Schumacher, R. Aebersold, H.-G. Rammensee, C. J. M. Melief, E. R. Mardis, W. E. Gillanders, M. N. Artyomov, R. D. Schreiber, Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014). doi:10.1038/nature13988 Medline

8. B. Bengsch, A. L. Johnson, M. Kurachi, P. M. Odorizzi, K. E. Pauken, J. Attanasio, E. Stelekati, L. M. McLane, M. A. Paley, G. M. Delgoffe, E. J. Wherry, Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8+ T cell exhaustion. Immunity 45, 358–373 (2016). doi:10.1016/j.immuni.2016.07.008 Medline

9. M. M. Staron, S. M. Gray, H. D. Marshall, I. A. Parish, J. H. Chen, C. J. Perry, G. Cui, M. O. Li, S. M. Kaech, The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8+ T cells during chronic infection. Immunity 41, 802–814 (2014). doi:10.1016/j.immuni.2014.10.013 Medline

10. N. Patsoukis, J. Brown, V. Petkova, F. Liu, L. Li, V. A. Boussiotis, Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci. Signal. 5, ra46 (2012). doi:10.1126/scisignal.2002796 Medline

11. J. Godec, Y. Tan, A. Liberzon, P. Tamayo, S. Bhattacharya, A. J. Butte, J. P. Mesirov, W. N. Haining, Compendium of immune signatures identifies conserved and species-specific biology in response to inflammation. Immunity 44, 194–206 (2016). doi:10.1016/j.immuni.2015.12.006 Medline

12. H. Shin, S. D. Blackburn, J. N. Blattman, E. J. Wherry, Viral antigen and extensive division maintain virus-specific CD8 T cells during chronic infection. J. Exp. Med. 204, 941–949 (2007). doi:10.1084/jem.20061937 Medline

13. E. J. Wherry, D. L. Barber, S. M. Kaech, J. N. Blattman, R. Ahmed, Antigen-independent memory CD8 T cells do not develop during chronic viral infection. Proc. Natl. Acad. Sci. U.S.A. 101, 16004–16009 (2004). doi:10.1073/pnas.0407192101 Medline

14. M. Pellegrini, T. Calzascia, J. G. Toe, S. P. Preston, A. E. Lin, A. R. Elford, A. Shahinian, P. A. Lang, K. S. Lang, M. Morre, B. Assouline, K. Lahl, T. Sparwasser, T. F. Tedder, J. H. Paik, R. A. DePinho, S. Basta, P. S. Ohashi, T. W. Mak, IL-7 engages multiple mechanisms to overcome chronic viral infection and limit organ pathology. Cell 144, 601–613 (2011). doi:10.1016/j.cell.2011.01.011 Medline

15. S. G. Nanjappa, E. H. Kim, M. Suresh, Immunotherapeutic effects of IL-7 during a chronic viral infection in mice. Blood 117, 5123–5132 (2011). doi:10.1182/blood-2010-12-323154 Medline

16. B. Youngblood, K. J. Oestreich, S.-J. Ha, J. Duraiswamy, R. S. Akondy, E. E. West, Z. Wei, P. Lu, J. W. Austin, J. L. Riley, J. M. Boss, R. Ahmed, Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8+ T cells. Immunity 35, 400–412 (2011). doi:10.1016/j.immuni.2011.06.015 Medline

17. D. T. Utzschneider, A. Legat, S. A. Fuertes Marraco, L. Carrié, I. Luescher, D. E. Speiser, D. Zehn, T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat. Immunol. 14, 603–610 (2013). doi:10.1038/ni.2606 Medline

18. J. M. Angelosanto, S. D. Blackburn, A. Crawford, E. J. Wherry, Progressive loss of memory T cell potential and commitment to exhaustion during chronic viral infection. J. Virol. 86, 8161–8170 (2012). doi:10.1128/JVI.00889-12 Medline

19. F. Zhang, X. Zhou, J. R. DiSpirito, C. Wang, Y. Wang, H. Shen, Epigenetic manipulation restores functions of defective CD8+ T cells from chronic viral infection. Mol. Ther. 22, 1698–1706 (2014). doi:10.1038/mt.2014.91 Medline

20. J. D. Buenrostro, P. G. Giresi, L. C. Zaba, H. Y. Chang, W. J. Greenleaf, Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013). doi:10.1038/nmeth.2688 Medline

21. D. R. Winter, I. Amit, The role of chromatin dynamics in immune cell development. Immunol. Rev. 261, 9–22 (2014). doi:10.1111/imr.12200 Medline

22. K. J. Oestreich, H. Yoon, R. Ahmed, J. M. Boss, NFATc1 regulates PD-1 expression upon T cell activation. J. Immunol. 181, 4832–4839 (2008). doi:10.4049/jimmunol.181.7.4832 Medline

23. C. Kao, K. J. Oestreich, M. A. Paley, A. Crawford, J. M. Angelosanto, M.-A. A. Ali, A. M. Intlekofer, J. M. Boss, S. L. Reiner, A. S. Weinmann, E. J. Wherry, Transcription

factor T-bet represses expression of the inhibitory receptor PD-1 and sustains virus-specific CD8+ T cell responses during chronic infection. Nat. Immunol. 12, 663–671 (2011). doi:10.1038/ni.2046 Medline

24. M. A. Paley, D. C. Kroy, P. M. Odorizzi, J. B. Johnnidis, D. V. Dolfi, B. E. Barnett, E. K. Bikoff, E. J. Robertson, G. M. Lauer, S. L. Reiner, E. J. Wherry, Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225 (2012). doi:10.1126/science.1229620 Medline

25. S. D. Blackburn, H. Shin, G. J. Freeman, E. J. Wherry, Selective expansion of a subset of exhausted CD8 T cells by αPD-L1 blockade. Proc. Natl. Acad. Sci. U.S.A. 105, 15016–15021 (2008). doi:10.1073/pnas.0801497105 Medline

26. R. He, S. Hou, C. Liu, A. Zhang, Q. Bai, M. Han, Y. Yang, G. Wei, T. Shen, X. Yang, L. Xu, X. Chen, Y. Hao, P. Wang, C. Zhu, J. Ou, H. Liang, T. Ni, X. Zhang, X. Zhou, K. Deng, Y. Chen, Y. Luo, J. Xu, H. Qi, Y. Wu, L. Ye, Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 537, 412–428 (2016). doi:10.1038/nature19317 Medline

27. S. J. Im, M. Hashimoto, M. Y. Gerner, J. Lee, H. T. Kissick, M. C. Burger, Q. Shan, J. S. Hale, J. Lee, T. H. Nasti, A. H. Sharpe, G. J. Freeman, R. N. Germain, H. I. Nakaya, H.-H. Xue, R. Ahmed, Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016). doi:10.1038/nature19330 Medline

28. D. T. Utzschneider, M. Charmoy, V. Chennupati, L. Pousse, D. P. Ferreira, S. Calderon-Copete, M. Danilo, F. Alfei, M. Hofmann, D. Wieland, S. Pradervand, R. Thimme, D. Zehn, W. Held, T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016). doi:10.1016/j.immuni.2016.07.021 Medline

29. T. A. Doering, A. Crawford, J. M. Angelosanto, M. A. Paley, C. G. Ziegler, E. J. Wherry, Network analysis reveals centrally connected genes and pathways involved in CD8+ T cell exhaustion versus memory. Immunity 37, 1130–1144 (2012). doi:10.1016/j.immuni.2012.08.021 Medline

30. G. J. Martinez, R. M. Pereira, T. Äijö, E. Y. Kim, F. Marangoni, M. E. Pipkin, S. Togher, V. Heissmeyer, Y. C. Zhang, S. Crotty, E. D. Lamperti, K. M. Ansel, T. R. Mempel, H. Lähdesmäki, P. G. Hogan, A. Rao, The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265–278 (2015). doi:10.1016/j.immuni.2015.01.006 Medline

31. L. K. Ward-Kavanagh, W. W. Lin, J. R. Šedý, C. F. Ware, The TNF receptor superfamily in co-stimulating and co-inhibitory responses. Immunity 44, 1005–1019 (2016). doi:10.1016/j.immuni.2016.04.019 Medline

32. P. M. Odorizzi, K. E. Pauken, M. A. Paley, A. Sharpe, E. J. Wherry, Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J. Exp. Med. 212, 1125–1137 (2015). doi:10.1084/jem.20142237 Medline

33. O. Boyman, C. Ramsey, D. M. Kim, J. Sprent, C. D. Surh, IL-7/anti-IL-7 mAb complexes restore T cell development and induce homeostatic T cell expansion without lymphopenia. J. Immunol. 180, 7265–7275 (2008). doi:10.4049/jimmunol.180.11.7265 Medline

34. J. N. Blattman, E. J. Wherry, S. J. Ha, R. G. van der Most, R. Ahmed, Impact of epitope escape on PD-1 expression and CD8 T-cell exhaustion during chronic infection. J. Virol. 83, 4386–4394 (2009). doi:10.1128/JVI.02524-08 Medline

35. J. Piper, S. A. Assi, P. Cauchy, C. Ladroue, P. N. Cockerill, C. Bonifer, S. Ott, Wellington-bootstrap: Differential DNase-seq footprinting identifies cell-type determining transcription factors. BMC Genomics 16, 1000 (2015). doi:10.1186/s12864-015-2081-4 Medline

36. S. Heinz, C. Benner, N. Spann, E. Bertolino, Y. C. Lin, P. Laslo, J. X. Cheng, C. Murre, H. Singh, C. K. Glass, Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010). doi:10.1016/j.molcel.2010.05.004 Medline

37. M. Reich, T. Liefeld, J. Gould, J. Lerner, P. Tamayo, J. P. Mesirov, GenePattern 2.0. Nat. Genet. 38, 500–501 (2006). doi:10.1038/ng0506-500 Medline

38. J. Piper, M. C. Elze, P. Cauchy, P. N. Cockerill, C. Bonifer, S. Ott, Wellington: A novel method for the accurate identification of digital genomic footprints from DNase-seq data. Nucleic Acids Res. 41, e201 (2013). doi:10.1093/nar/gkt850 Medline

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 4

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 5: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

ACKNOWLEDGMENTS

We thank the Wherry lab for discussions and critically reading the manuscript. We thank C. Surh for providing IL-7 antibody. The IL-7 antibody is available from La Jolla Institute of Allergy and Immunology, and the IL-7 used is available from NCI, both under material transfer agreements with the University of Pennsylvania. The data presented in this manuscript are tabulated in the main paper and in the supplementary materials. Sequencing data are available on GEO (accession numbers GSE86796 (microarray), GSE86881 (RNA seq), and GSE86797 (ATAC seq)). KEP was supported by a Robertson Foundation/Cancer Research Institute Irvington Fellowship, MAS by an American Cancer Society Postdoctoral Fellowship, JMS by F30DK100159, BB by the German Research Foundation Fellowship BE5496/1-1, and ACH by T32 2T32CA009615-26. This work was funded by the National Institutes of Health (CA78831 to SLB and grants AI105343, AI112521, AI082630, AI115712, AI117950, AI108545 to EJW). This research was also supported by the Parker Institute for Cancer Immunotherapy. EJW has a patent licensing agreement on the PD-1 pathway. The authors declare no additional conflicts of interest.

SUPPLEMENTARY MATERIALS www.sciencemag.org/cgi/content/full/science.aaf2807/DC1 Materials and Methods Figs. S1 to S18 Tables S1 to S12 References (32–38) 18 January 2016; accepted 19 September 2016 Published online 27 October 2016 10.1126/science.aaf2807

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 5

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 6: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

Fig. 1. Anti-PD-L1 induces an effector-like transcriptional program in TEX cells that is not sustained after cessation of treatment. (A) Consensus hierarchical clustering by 1-Pearson correlation from the microarray on control- or anti-PD-L1-treated TEX. (B) GSEA of representative Gene Ontology (GO) terms. (C) GSEA of effector genes. (D) Row normalized heat map of effector-associated genes. (E) Circos plots showing overlap in metagenes identified in anti-PD-L1-treated TEX compared to metagenes in TEFF (left) and TMEM cells (right). Ribbons connecting areas of the circos plots indicate shared genes between groups. The microarray includes four independent experiments with 10-12 mice per group per experiment. (F) Frequency of P14 cells among CD8 T cells and (G) Ki67+ P14 cells in the peripheral blood. Grey bar indicates antibody treatment period. (H) Quantification of IFNγ+ TNFα+ P14 cells. (I) Viral load in the kidney. Data in (F-G) are one representative experiment. In (H-I), the +1 day time point is combined from two representative experiments and the +20 week time point is from one representative experiment. Data in (F-I) are representative of at least two independent experiments with at least 4 mice per group per experiment. (J) Principle component analysis of RNA-seq, % of variance (var.) indicated. The RNA-seq was performed on two-four independent experiments with 5-13 mice per group as indicated in the Methods. Each dot represents an independent experiment. Asterisks indicating significance determined by unpaired t tests between groups are * p<0.05, **p<0.01, and ***p<0.001.

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 6

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 7: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

Fig. 2. PD-1 pathway blockade moderately improves antigen-independent persistence and IL-7 signaling in TEX. (A) Number of DbGP33+ donor CD8 T cells per million PBMCs at day 27 (compared to day 1) post-transfer and (B) number recovered from the spleen. (C) Histograms of CD127 and CD122 expression on TEX P14 cells (Day 35 post clone 13) compared to TMEM P14 cells or bulk CD44lo CD62L+ TN cells (Day 167 post LCMV Arm). Values indicate average geometric mean fluorescence intensity (MFI) and standard error of the mean (SEM). (D) Contour plots of PD-1 versus CD127 from mice in (C). (E) Quantification of (D). Data in (A-E) are representative of at least 2 independent experiments with at least 4 mice per group. (F) Quantification of phospho-STAT5 induction by P14 cells at day 39 p.i. following ex vivo stimulation with IL-7 or IL-15 for 30 min. Values indicate fold change over unstimulated controls. (G) Frequency (of CD8+, left) and number (right) of P14 cells in the spleen after two weeks of treatment. (H) Plots (left) and quantification (right) of IFNγ+ TNFα+ P14 cells from (G) following ex vivo peptide stimulation. Data in (F)-(H) are combined from 2 independent experiments with at least 4 mice per group. Asterisks indicating significance are * p<0.05, **p<0.01, and ***p<0.001 determined as described in Supplemental Methods. Blue asterisks indicate ANOVA p values, black asterisks indicate post-test p values.

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 7

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 8: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

Fig. 3. PD-1 pathway blockade fails to restore memory-like recall capacity or reprogram the epigenetic landscape of TEX into TEFF or TMEM cells. (A)-(D) The experimental design outlined in fig. S7A was used except that recipient mice were rechallenged with Listeria-GP33 3.5 weeks post-transfer. (A) Flow cytometry plots of responding TMEM, TEX or anti-PD-L1 treated TEX at 6 days post rechallenge with Listeria-GP33. (B) Concatenated flow cytometry plots gated on P14 cells from mice in (A) following ex vivo peptide stimulation. (C) Quantification of donor (Ly5.2+) DbGP33+ CD8 T cells in the spleens shown in (A). (D) Quantification of IFNγ+ P14 cells shown in (B). (E) Histograms of PD-1 on donor DbGP33+ cells from mice shown in Fig. 2B. Values indicate average geometric MFI and SEM. Data are representative of 2 independent experiments with at least 4 mice per group. Asterisks indicating significance are * p<0.05, **p<0.01, and ***p<0.001 determined as described in Supplemental Methods. Blue asterisks indicate ANOVA p values, black asterisks indicate post-test p values. (F) Venn diagrams of ATAC-seq OCRs compared to TN cells (LFC≥2). Data from the two replicates are combined. (G) Representative ATAC-seq tracks from one independent replicate per group shown at the Ifng and Pcdc1 loci. (H) Co-cluster analysis of variance showing enrichment of OCRs in ATAC-seq data set. Solid lines separate cell types, replicates shown side-by-side. (I) Box and whisker plots showing ATAC-seq enrichment from (H). Whiskers represent the interquartile range. (J) Principle component analysis of all OCRs. For (I) and (J), each replicate is shown. ATAC seq data are from two independent experiments with 2-15 mice per group as described in the Supplemental Methods..

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 8

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 9: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

Fig. 4. Differential transcription factor binding following PD-1 pathway blockade contributes to an altered transcriptional network during TEX re-invigoration. (A) Enrichment of TF binding motifs in OCRs lost or gained following anti-PD-L1 treatment. (B) Wellington bootstrap analysis of TF binding in pairwise comparisons for each cell type, the top 10 TFs (in boxes) enriched in all OCRs is shown. Full list in table S10. (C) TF footprint for NFATc1 in TEX and NFκB-p65 in anti-PD-L1-treated TEX. (D) Integrated network analysis of the transcriptional and epigenetic changes following anti-PD-L1. Lines connect TFs predicted to have altered activity to corresponding genes regulated. Details in table S11. (E) LFC of genes significantly changed by anti-PD-L1 treatment compared to genes significantly induced by the “partnerless” NFAT construct CA-RIT-NFAT1 (30). (F) Venn diagram showing genes near OCRs containing given TF motifs in TEFF, TEX, or both (overlap) (top left). Percent difference in TF target genes changed (p<0.05, LFC≥0.3) with anti-PD-L1 in overlap compared to TEX only (bottom left). Sum of the absolute value of the LFC in expression in TF target genes following anti-PD-L1 (right). ATAC-seq data shown is combined replicates for each condition.

First release: 27 October 2016 www.sciencemag.org (Page numbers not final at time of first release) 9

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 10: Epigenetic stability of exhausted T cells limits ...science.sciencemag.org/content/sci/early/2016/10/26/science.aaf2807.full.pdfanti-PD-L1 treatment reduced viral load immediately

Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade

Schenkel, Golnaz Vahedi, W. Nicholas Haining, Shelley L. Berger and E. John WherryChen, Debattama Sen, Makoto Kurachi, R. Anthony Barnitz, Caroline Bartman, Bertram Bengsch, Alexander C. Huang, Jason M. Kristen E. Pauken, Morgan A. Sammons, Pamela M. Odorizzi, Sasikanth Manne, Jernej Godec, Omar Khan, Adam M. Drake, Zeyu

published online October 27, 2016

ARTICLE TOOLS http://science.sciencemag.org/content/early/2016/10/26/science.aaf2807

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2016/11/04/science.aaf2807.DC1

CONTENTRELATED

http://stke.sciencemag.org/content/sigtrans/9/447/ra94.fullhttp://stm.sciencemag.org/content/scitransmed/7/280/280sr1.fullhttp://stke.sciencemag.org/content/sigtrans/9/419/ec58.abstracthttp://stke.sciencemag.org/content/sigtrans/4/191/ec262.abstracthttp://stke.sciencemag.org/content/sigtrans/8/374/re6.fullhttp://stke.sciencemag.org/content/sigtrans/5/230/ra46.fullhttp://stm.sciencemag.org/content/scitransmed/8/328/328rv4.fullhttp://stm.sciencemag.org/content/scitransmed/6/242/242ra81.fullhttp://science.sciencemag.org/content/sci/354/6316/1165.fullhttp://science.sciencemag.org/content/sci/354/6316/1104.full

REFERENCES

http://science.sciencemag.org/content/early/2016/10/26/science.aaf2807#BIBLThis article cites 37 articles, 12 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Copyright © 2016, American Association for the Advancement of Science

on March 10, 2020

http://science.sciencem

ag.org/D

ownloaded from