tcellexhaustion the epigenetic landscape of t cell exhaustion · 1166 2 december 2016• vol 354...

6
but also points to key opportunities for improving the long-term durability of these effects. REFERENCES AND NOTES 1. E. J. Wherry, M. Kurachi, Nat. Rev. Immunol. 15, 486499 (2015). 2. D. B. Page, M. A. Postow, M. K. Callahan, J. P. Allison, J. D. Wolchok, Annu. Rev. Med. 65, 185202 (2014). 3. P. Sharma, J. P. Allison, Science 348, 5661 (2015). 4. D. S. Shin, A. Ribas, Curr. Opin. Immunol. 33, 2335 (2015). 5. D. L. Barber et al., Nature 439, 682687 (2006). 6. See supplementary information on Science Online. 7. M. M. Gubin et al., Nature 515, 577581 (2014). 8. B. Bengsch et al., Immunity 45, 358373 (2016). 9. M. M. Staron et al., Immunity 41, 802814 (2014). 10. N. Patsoukis et al., Sci. Signal. 5, ra46 (2012). 11. J. Godec et al., Immunity 44, 194206 (2016). 12. H. Shin, S. D. Blackburn, J. N. Blattman, E. J. Wherry, J. Exp. Med. 204, 941949 (2007). 13. E. J. Wherry, D. L. Barber, S. M. Kaech, J. N. Blattman, R. Ahmed, Proc. Natl. Acad. Sci. U.S.A. 101, 1600416009 (2004). 14. M. Pellegrini et al., Cell 144, 601613 (2011). 15. S. G. Nanjappa, E. H. Kim, M. Suresh, Blood 117, 51235132 (2011). 16. B. Youngblood et al., Immunity 35, 400412 (2011). 17. D. T. Utzschneider et al., Nat. Immunol. 14, 603610 (2013). 18. J. M. Angelosanto, S. D. Blackburn, A. Crawford, E. J. Wherry, J. Virol. 86, 81618170 (2012). 19. F. Zhang et al., Mol. Ther. 22, 16981706 (2014). 20. J. D. Buenrostro, P. G. Giresi, L. C. Zaba, H. Y. Chang, W. J. Greenleaf, Nat. Methods 10, 12131218 (2013). 21. D. R. Winter, I. Amit, Immunol. Rev. 261,922 (2014). 22. K. J. Oestreich, H. Yoon, R. Ahmed, J. M. Boss, J. Immunol. 181, 48324839 (2008). 23. C. Kao et al., Nat. Immunol. 12, 663671 (2011). 24. M. A. Paley et al., Science 338, 12201225 (2012). 25. S. D. Blackburn, H. Shin, G. J. Freeman, E. J. Wherry, Proc. Natl. Acad. Sci. U.S.A. 105, 1501615021 (2008). 26. R. He et al., Nature 537, 412428 (2016). 27. S. J. Im et al., Nature 537, 417421 (2016). 28. D. T. Utzschneider et al., Immunity 45, 415427 (2016). 29. T. A. Doering et al., Immunity 37, 11301144 (2012). 30. G. J. Martinez et al., Immunity 42, 265278 (2015). 31. L. K. Ward-Kavanagh, W. W. Lin, J. R. Šedý, C. F. Ware, Immunity 44, 10051019 (2016). ACKNOWLEDGMENTS We thank the Wherry lab for discussions and critically reading the manuscript. We thank C. Surh for providing the antibody against IL-7 (anti-IL-7). The anti-IL-7 antibody is available from La Jolla Institute of Allergy and Immunology, and the IL-7 used is available from the National Cancer Institute, 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 at Gene Expression Omnibus [accession numbers GSE86796 (microarray), GSE86881 (RNA seq), and GSE86797 (ATAC seq)]. This study was supported by a Robertson FoundationCancer Research Institute Irvington Fellowship (K.E.P.), an American Cancer Society Postdoctoral Fellowship (M.A.S.), National Institutes of Health grant F30DK100159 (J.M.S.), German Research Foundation Fellowship BE5496/1-1 (B.B.), and National Institutes of Health grant T32 2T32CA009615-26 (A.C.H.). This work was funded by the National Institutes of Health (grant CA78831 to S.L.B. and grants AI105343, AI112521, AI082630, AI115712, AI117950, and AI108545 to E.J.W.). This research was also supported by the Parker Institute for Cancer Immunotherapy. E.J.W. has a patent licensing agreement on the PD-1 pathway. The authors declare no additional conflicts of interest. SUPPLEMENTARY MATERIALS www.sciencemag.org/content/354/6316/1160/suppl/DC1 Materials and Methods Figs. S1 to S18 Tables S1 to S12 References (3238) 18 January 2016; accepted 19 September 2016 Published online 27 October 2016 10.1126/science.aaf2807 T CELL EXHAUSTION The epigenetic landscape of T cell exhaustion Debattama R. Sen, 1,2 * James Kaminski, 3 * R. Anthony Barnitz, 1 Makoto Kurachi, 4,5 Ulrike Gerdemann, 1 Kathleen B. Yates, 1 Hsiao-Wei Tsao, 1 Jernej Godec, 1,2 Martin W. LaFleur, 1,2 Flavian D. Brown, 1,2 Pierre Tonnerre, 6 Raymond T. Chung, 6 Damien C. Tully, 7 Todd M. Allen, 7 Nicole Frahm, 8 Georg M. Lauer, 6 E. John Wherry, 4,5 Nir Yosef, 3,7,9 †‡ W. Nicholas Haining 1,10,11 †‡ Exhausted T cells in cancer and chronic viral infection express distinctive patterns of genes, including sustained expression of programmed cell death protein 1 (PD-1). However, the regulation of gene expression in exhausted Tcells is poorly understood. Here, we define the accessible chromatin landscape in exhausted CD8 + T cells and show that it is distinct from functional memory CD8 + T cells. Exhausted CD8 + T cells in humans and a mouse model of chronic viral infection acquire a state-specific epigenetic landscape organized into functional modules of enhancers. Genome editing shows that PD-1 expression is regulated in part by an exhaustion-specific enhancer that contains essential RAR, T-bet, and Sox3 motifs. Functional enhancer maps may offer targets for genome editing that alter gene expression preferentially in exhausted CD8 + T cells. T cell exhaustionan acquired state of T cell dysfunctionis a hallmark of cancer and chronic viral infection (1, 2), and clinical trials of checkpoint blockade, which aim to reverse T cell exhaustion in cancer, have proven strikingly effective (3, 4). Chimeric antigen receptor (CAR)T cell therapy has also proven highly effective for hematologic malig- nancies (5), but the development of exhaustion in T cells engineered to treat solid tumors re- mains a substantial barrier to its broader use (6). The identification of mechanisms that reg- ulate exhausted T cells is therefore a major goal in cancer immunotherapy. To identify regulatory regions in the genome of exhausted CD8 + T cells, we used an assay for transposase-accessible chromatin with high- throughput sequencing (ATAC-seq) (7) to demar- cate areas of accessible chromatin in mouse antigen-specific CD8 + T cells differentiating in response to lymphocytic choriomeningitis virus (LCMV) infection (fig. S1A and table S1). Acute LCMV infection elicits highly functional effec- tor CD8 + T cells, whereas chronic LCMV infection gives rise to exhausted CD8 + T cells (13, 8, 9). Analysis of high-quality ATAC-seq profiles (fig. S1, B to H) from naïve CD8 + T cells and those at day 8 and day 27 postinfection (p.i.) (d8 and d27, respectively) revealed that naïve CD8 + T cells underwent large-scale remodeling (Fig. 1A and fig. S2A) during differentiation [as de- tected by DESeq2, with a false discovery rate (FDR) < 0.05]. The majority (71%) (fig. S2A) of chromatin-accessible regions (ChARs) either emerged (e.g., those at the Ifng locus) or disap- peared (e.g., Ccr7) (Fig. 1A) as naïve CD8 + T cells underwent differentiation. The gain and loss of ChARs were not balanced; a much larger frac- tion of regions emerged at d8 p.i. and persisted or emerged only at d27 than were either tran- siently detected at d8 p.i. or lost from naïve cells (Fig. 1B). Thus, differentiation from a naïve CD8 + T cell state is associated with a net increase, rather than decrease, in chromatin accessibility (fig. S2B). Comparison of ChARs from exhausted CD8 + T cells with those found in functional effector or memory CD8 + T cells revealed marked differ- ences in the pattern of regulatory regions. Dif- ferential regulatory regions between acute and chronic infection (Fig. 1C and fig. S2C) showed features of enhancers: They tended to be depleted of transcription start sites (TSSs) and enriched for intergenic and intronic areas (Fig. 1D), and found distal to gene promoters (fig. S2D). The magnitude of difference in the profile of regula- tory regions between exhausted and functional CD8 + T cells was greater than that seen in gene expression. We found that 44.48% of all ChARs SCIENCE sciencemag.org 2 DECEMBER 2016 VOL 354 ISSUE 6316 1165 1 Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA. 2 Division of Medical Sciences, Harvard Medical School, Boston, MA 02115, USA. 3 Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA. 4 Institute of Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA. 5 Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA. 6 Gastrointestinal Unit and Liver Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA. 7 Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Boston, MA 02139, USA. 8 Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. 9 Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA. 10 Division of Pediatric Hematology and Oncology, Childrens Hospital, Boston, MA 02115, USA. 11 Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA. *These authors contributed equally to this work. These authors contributed equally to this work. Corresponding author. Email: [email protected] (N.Y.); [email protected]. edu (W.N.H.) RESEARCH | REPORTS on February 14, 2021 http://science.sciencemag.org/ Downloaded from

Upload: others

Post on 04-Oct-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: TCELLEXHAUSTION The epigenetic landscape of T cell exhaustion · 1166 2 DECEMBER 2016• VOL 354 ISSUE 6316 sciencemag.org SCIENCE Fig. 1. CD8+ Tcell exhaustion is associated with

but also points to key opportunities for improvingthe long-term durability of these effects.

REFERENCES AND NOTES

1. E. J. Wherry, M. Kurachi, Nat. Rev. Immunol. 15, 486–499(2015).

2. D. B. Page, M. A. Postow, M. K. Callahan, J. P. Allison,J. D. Wolchok, Annu. Rev. Med. 65, 185–202 (2014).

3. P. Sharma, J. P. Allison, Science 348, 56–61 (2015).4. D. S. Shin, A. Ribas, Curr. Opin. Immunol. 33, 23–35

(2015).5. D. L. Barber et al., Nature 439, 682–687 (2006).6. See supplementary information on Science Online.7. M. M. Gubin et al., Nature 515, 577–581 (2014).8. B. Bengsch et al., Immunity 45, 358–373 (2016).9. M. M. Staron et al., Immunity 41, 802–814 (2014).10. N. Patsoukis et al., Sci. Signal. 5, ra46 (2012).11. J. Godec et al., Immunity 44, 194–206 (2016).12. H. Shin, S. D. Blackburn, J. N. Blattman, E. J. Wherry, J. Exp.

Med. 204, 941–949 (2007).13. E. J. Wherry, D. L. Barber, S. M. Kaech, J. N. Blattman,

R. Ahmed, Proc. Natl. Acad. Sci. U.S.A. 101, 16004–16009(2004).

14. M. Pellegrini et al., Cell 144, 601–613 (2011).15. S. G. Nanjappa, E. H. Kim, M. Suresh, Blood 117, 5123–5132

(2011).16. B. Youngblood et al., Immunity 35, 400–412 (2011).17. D. T. Utzschneider et al., Nat. Immunol. 14, 603–610

(2013).18. J. M. Angelosanto, S. D. Blackburn, A. Crawford, E. J. Wherry,

J. Virol. 86, 8161–8170 (2012).19. F. Zhang et al., Mol. Ther. 22, 1698–1706 (2014).20. J. D. Buenrostro, P. G. Giresi, L. C. Zaba, H. Y. Chang,

W. J. Greenleaf, Nat. Methods 10, 1213–1218 (2013).21. D. R. Winter, I. Amit, Immunol. Rev. 261, 9–22 (2014).22. K. J. Oestreich, H. Yoon, R. Ahmed, J. M. Boss, J. Immunol. 181,

4832–4839 (2008).23. C. Kao et al., Nat. Immunol. 12, 663–671 (2011).24. M. A. Paley et al., Science 338, 1220–1225 (2012).25. S. D. Blackburn, H. Shin, G. J. Freeman, E. J. Wherry, Proc.

Natl. Acad. Sci. U.S.A. 105, 15016–15021 (2008).26. R. He et al., Nature 537, 412–428 (2016).27. S. J. Im et al., Nature 537, 417–421 (2016).28. D. T. Utzschneider et al., Immunity 45, 415–427 (2016).29. T. A. Doering et al., Immunity 37, 1130–1144 (2012).30. G. J. Martinez et al., Immunity 42, 265–278 (2015).31. L. K. Ward-Kavanagh, W. W. Lin, J. R. Šedý, C. F. Ware,

Immunity 44, 1005–1019 (2016).

ACKNOWLEDGMENTS

We thank the Wherry lab for discussions and critically reading themanuscript. We thank C. Surh for providing the antibody against IL-7(anti-IL-7). The anti-IL-7 antibody is available from La Jolla Institute ofAllergy and Immunology, and the IL-7 used is available from theNational Cancer Institute, both under material transfer agreementswith the University of Pennsylvania. The data presented in thismanuscript are tabulated in the main paper and in the supplementarymaterials. Sequencing data are available at Gene ExpressionOmnibus [accession numbers GSE86796 (microarray), GSE86881(RNA seq), and GSE86797 (ATAC seq)]. This study was supported bya Robertson Foundation–Cancer Research Institute IrvingtonFellowship (K.E.P.), an American Cancer Society PostdoctoralFellowship (M.A.S.), National Institutes of Health grant F30DK100159(J.M.S.), German Research Foundation Fellowship BE5496/1-1 (B.B.),and National Institutes of Health grant T32 2T32CA009615-26 (A.C.H.).This work was funded by the National Institutes of Health (grantCA78831 to S.L.B. and grants AI105343, AI112521, AI082630, AI115712,AI117950, and AI108545 to E.J.W.). This research was also supportedby the Parker Institute for Cancer Immunotherapy. E.J.W. has a patentlicensing agreement on the PD-1 pathway. The authors declare noadditional conflicts of interest.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/354/6316/1160/suppl/DC1Materials and MethodsFigs. S1 to S18Tables S1 to S12References (32–38)

18 January 2016; accepted 19 September 2016Published online 27 October 201610.1126/science.aaf2807

T CELL EXHAUSTION

The epigenetic landscape ofT cell exhaustionDebattama R. Sen,1,2* James Kaminski,3* R. Anthony Barnitz,1 Makoto Kurachi,4,5

Ulrike Gerdemann,1 Kathleen B. Yates,1 Hsiao-Wei Tsao,1 Jernej Godec,1,2

Martin W. LaFleur,1,2 Flavian D. Brown,1,2 Pierre Tonnerre,6 Raymond T. Chung,6

Damien C. Tully,7 Todd M. Allen,7 Nicole Frahm,8 Georg M. Lauer,6 E. John Wherry,4,5

Nir Yosef,3,7,9†‡ W. Nicholas Haining1,10,11†‡

Exhausted T cells in cancer and chronic viral infection express distinctive patternsof genes, including sustained expression of programmed cell death protein 1 (PD-1).However, the regulation of gene expression in exhausted T cells is poorly understood.Here, we define the accessible chromatin landscape in exhausted CD8+ T cells andshow that it is distinct from functional memory CD8+ T cells. Exhausted CD8+ T cellsin humans and a mouse model of chronic viral infection acquire a state-specificepigenetic landscape organized into functional modules of enhancers. Genome editingshows that PD-1 expression is regulated in part by an exhaustion-specific enhancer thatcontains essential RAR, T-bet, and Sox3 motifs. Functional enhancer maps may offertargets for genome editing that alter gene expression preferentially in exhaustedCD8+ T cells.

Tcell exhaustion—an acquired state of T celldysfunction—is a hallmark of cancer andchronic viral infection (1, 2), and clinicaltrials of checkpoint blockade, which aimto reverse T cell exhaustion in cancer,

have proven strikingly effective (3, 4). Chimericantigen receptor (CAR)–T cell therapy has alsoproven highly effective for hematologic malig-nancies (5), but the development of exhaustionin T cells engineered to treat solid tumors re-mains a substantial barrier to its broader use(6). The identification of mechanisms that reg-ulate exhausted T cells is therefore a major goalin cancer immunotherapy.To identify regulatory regions in the genome

of exhausted CD8+ T cells, we used an assayfor transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) (7) to demar-cate areas of accessible chromatin in mouse

antigen-specific CD8+ T cells differentiating inresponse to lymphocytic choriomeningitis virus(LCMV) infection (fig. S1A and table S1). AcuteLCMV infection elicits highly functional effec-tor CD8+ T cells, whereas chronic LCMV infectiongives rise to exhausted CD8+ T cells (1–3, 8, 9).Analysis of high-quality ATAC-seq profiles (fig.S1, B to H) from naïve CD8+ T cells and thoseat day 8 and day 27 postinfection (p.i.) (d8 andd27, respectively) revealed that naïve CD8+

T cells underwent large-scale remodeling (Fig.1A and fig. S2A) during differentiation [as de-tected by DESeq2, with a false discovery rate(FDR) < 0.05]. The majority (71%) (fig. S2A) ofchromatin-accessible regions (ChARs) eitheremerged (e.g., those at the Ifng locus) or disap-peared (e.g., Ccr7) (Fig. 1A) as naïve CD8+ T cellsunderwent differentiation. The gain and loss ofChARs were not balanced; a much larger frac-tion of regions emerged at d8 p.i. and persistedor emerged only at d27 than were either tran-siently detected at d8 p.i. or lost from naïve cells(Fig. 1B). Thus, differentiation from a naïve CD8+

T cell state is associated with a net increase,rather than decrease, in chromatin accessibility(fig. S2B).Comparison of ChARs from exhausted CD8+

T cells with those found in functional effectoror memory CD8+ T cells revealed marked differ-ences in the pattern of regulatory regions. Dif-ferential regulatory regions between acute andchronic infection (Fig. 1C and fig. S2C) showedfeatures of enhancers: They tended to be depletedof transcription start sites (TSSs) and enrichedfor intergenic and intronic areas (Fig. 1D), andfound distal to gene promoters (fig. S2D). Themagnitude of difference in the profile of regula-tory regions between exhausted and functionalCD8+ T cells was greater than that seen in geneexpression. We found that 44.48% of all ChARs

SCIENCE sciencemag.org 2 DECEMBER 2016 • VOL 354 ISSUE 6316 1165

1Department of Pediatric Oncology, Dana-Farber CancerInstitute, Boston, MA 02115, USA. 2Division of MedicalSciences, Harvard Medical School, Boston, MA 02115, USA.3Center for Computational Biology, University of California,Berkeley, Berkeley, CA 94720, USA. 4Institute ofImmunology, University of Pennsylvania, Philadelphia, PA19104, USA. 5Department of Microbiology, University ofPennsylvania, Philadelphia, PA 19104, USA. 6GastrointestinalUnit and Liver Center, Massachusetts General Hospital,Harvard Medical School, Boston, MA 02115, USA. 7RagonInstitute of Massachusetts General Hospital, MassachusettsInstitute of Technology, and Harvard University, Boston, MA02139, USA. 8Vaccine and Infectious Disease Division, FredHutchinson Cancer Research Center, Seattle, WA 98109,USA. 9Department of Electrical Engineering and ComputerScience, University of California, Berkeley, Berkeley, CA94720, USA. 10Division of Pediatric Hematology andOncology, Children’s Hospital, Boston, MA 02115, USA.11Broad Institute of Harvard and Massachusetts Institute ofTechnology, Cambridge, MA 02142, USA.*These authors contributed equally to this work. †These authorscontributed equally to this work. ‡Corresponding author. Email:[email protected] (N.Y.); [email protected] (W.N.H.)

RESEARCH | REPORTSon F

ebruary 14, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 2: TCELLEXHAUSTION The epigenetic landscape of T cell exhaustion · 1166 2 DECEMBER 2016• VOL 354 ISSUE 6316 sciencemag.org SCIENCE Fig. 1. CD8+ Tcell exhaustion is associated with

1166 2 DECEMBER 2016 • VOL 354 ISSUE 6316 sciencemag.org SCIENCE

Fig. 1. CD8+ Tcell exhaustion is associated with extensive changes in accessible chromatin. (A) Representative ATAC-seq tracks at the Ccr7 and Ifnggene loci. (B) Developmental trajectory of new regions at each time point. (C) Overlap in ChARs between cell states. (D) Distribution of nondifferential (left) anddifferential (right) regions between acute and chronic CD8+ T cell states. TSS, transcription start site. (E) Correlation network of similarity between statesmeasured by gene expression (left) and chromatin accessibility (right). Edge length corresponds to similarity (Spearman correlation).

Fig. 2. State-specific enhancers in CD8+ T cells form modules that map to functionally distinct classes of genes. (A) Heat map of peak intensityfor all differentially accessible regions (rows) clustered by similarity across cell states (columns). Shown are normalized numbers of cut sites(supplementary methods), scaled linearly from row minimum (white) to maximum (purple). (B) Heat map showing row-normalized average mRNAexpression of neighboring genes within each module in (A) in each cell state. Informative genes from each module are shown on right. (C) Heat mapshowing enrichment of Gene Ontology (GO) terms (rows) in each module (columns). P-values (hypergeometric test) presented as –log10.

RESEARCH | REPORTSon F

ebruary 14, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 3: TCELLEXHAUSTION The epigenetic landscape of T cell exhaustion · 1166 2 DECEMBER 2016• VOL 354 ISSUE 6316 sciencemag.org SCIENCE Fig. 1. CD8+ Tcell exhaustion is associated with

were differentially present between functionaland exhausted cells at each time point, com-pared with only 9.75% of differentially expressedgenes (both values estimated at FDR < 0.05).Consistent with this, the rank correlation betweeneach T cell state by gene expression was muchhigher than at the level of regulatory regions(Fig. 1E). Thus, state change during CD8+ T celldifferentiation is accompanied by a larger reor-ganization of accessible chromatin than is ap-parent by examination of gene expression.Unsupervised clustering identified “modules”

of differential ChARs with similar patterns ofactivity across T cell states (Fig. 2A and fig. S3A).We found a highly significant positive correlationbetween the average peak intensity of ChARs

within each module and the average gene ex-pression of the adjacent genes (F test, P < 0.001)(Fig. 2B and fig. S3B). This suggests that, onaverage, the ChARs contained in each moduletended to be associated with the activation,rather than repression, of corresponding genes.Genes adjacent to ChARs in each state-specific

module included many with known functions inthe corresponding T cell state. For example, mod-ule d, active in mouse T cells experiencing chronicLCMV infection on d8 and d27 p.i., containedChARs adjacent to the inhibitory receptors Pdcd1and Havcr2 (which encodes Tim3) and the trans-cription factor Batf, all genes that are up-regulated in exhausted CD8+ T cells (Fig. 2B)(1, 8). Moreover, the functional classes of genes

in each module were distinct on the basis ofpathway enrichment (Fig. 2C and table S2).Thus, ChARs that distinguish naïve, effector,memory, and exhausted CD8+ T cells are or-ganized into state-specific modules that posi-tively regulate functionally distinct programs ofgenes.We next sought to test whether regulatory re-

gions specific to exhausted cells could regulategenes differentially expressed in exhausted CD8+

T cells. Persistent expression of PD-1 is a cardi-nal feature of exhausted CD8+ T cells, but PD-1 isalso transiently expressed by effector CD8+ T cellsduring acute LCMV infection (3, 8). We identifiednine ChARs within 45 kb of the Pdcd1 gene locus(Fig. 3A) and found several that correspond to

SCIENCE sciencemag.org 2 DECEMBER 2016 • VOL 354 ISSUE 6316 1167

Fig. 3. High-resolution functional mapping of an exhaustion-specificenhancer identifies minimal sequences that regulate PD-1. (A) ATAC-seq tracks from CD8+ Tcells, EL4 cell line, and regulatory CD4+ Tcells (12).Arrowheads indicate individual ChARs. (B) Cell sorting gates (top) and cor-responding genomic polymerase chain reaction amplification for thePD-1 enhancer region (bottom) showing proportion of wild-type (WT)or deleted (Del) alleles in EL4 cells transfected with control (left) ordouble-cut sgRNAs (right). Representative data shown from two replicates.(C) PD-1 expression of EL4 WT (light gray) or representative enhancer-deleted (red) single-cell clone out of 46 clones. (D) Normalized enrichmentof sgRNAs (gray symbols) within PD-1–high and PD-1–low populations atlocations shown (supplementary methods). Control nontargeting sgRNAs are

pseudo mapped with 5-bp spacing. Red symbols correspond to the 21sgRNAs with the largest effect within the –23.8 kb enhancer, for which isogeniccell lines were later produced. (E) Overlap of TF footprints and sgRNA activitywithin the –23.8 kb enhancer. TF footprints with binding probability >0.9 inchronic d27 are shown on top. Lines represent cut sites of top-scoring sgRNAs.Change in PD-1 mean fluorescence intensity (MFI) relative to control guidetransfected populations for each sgRNA (red symbol, left axis); 10-bp runningaverage of PD-1 MFI changes caused by sgRNA activity shown in black (rightaxis). (F) sgRNA cut sites within the SOX3, TBX21, and RAR motifs. (G) Logfold enrichment of predicted TF footprints in acute d27 versus chronic d27CD8+ Tcells (x axis) (see supplementary methods) are plotted against thecorresponding P-value (hypergeometric).

RESEARCH | REPORTSon F

ebruary 14, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 4: TCELLEXHAUSTION The epigenetic landscape of T cell exhaustion · 1166 2 DECEMBER 2016• VOL 354 ISSUE 6316 sciencemag.org SCIENCE Fig. 1. CD8+ Tcell exhaustion is associated with

previously described regions with enhancer ac-tivity (–1.5 kb and –3.7 kb) (Fig. 3A) (10); thesewere present in both acute and chronic infection.We also identified an additional region (–23.8 kb)that only showed appreciable chromatin acces-sibility in exhausted CD8+ T cells at d8 and d27p.i. from chronic infection (Fig. 3A).We hypothesized that this ChAR might func-

tion as an enhancer of PD-1 that is required forpersistent, high levels of expression in exhaustedCD8+ T cells. Analysis of chromatin accessibilityat this region in previously published deoxyribo-nuclease I–hypersensitive site–mapping (11) orATAC-seq data (12) showed that it was not activein other types of hematopoietic cells, exceptthe murine T cell line EL4 and regulatory CD4+

T cells, both of which can constitutively expresshigh levels of PD-1 (10, 13) (Fig. 3A and fig. S4A).We cloned a 781–base pair (bp) fragment corre-sponding to this region into a reporter constructand found that it induced a 10- to 12-fold in-crease in reporter gene expression, confirmingthat it could function as an enhancer (fig. S4B).

We then tested whether the –23.8 kb enhancerwas necessary for high-level PD-1 expression. Weused the CRISPR-Cas9 nuclease to delete a 1.2-kbfragment at that position in EL4 cells, whichhave both sustained high-level PD-1 expressionand open chromatin at that enhancer site (14, 15)(fig. S4, C to G). In Cas9-expressing EL4 cellstransduced with a pair of single-guide RNAs(sgRNAs) flanking the enhancer, cells with thelowest PD-1 expression had the highest amountof the enhancer deletion (Fig. 3B). We confirmedthis finding in single-cell clones and found thatthe expression of PD-1 in clones with a biallelicdeletion of the target ChAR was significantlylower (P > 0.0002, Mann-Whitney U test) thanexpression in nondeleted clones (fig. S4, H toJ). Deletion of this region resulted in decreasedbut not abrogated PD-1, suggesting that addi-tional regulatory regions in EL4 cells are alsoinvolved in regulating PD-1 expression (Fig. 3C).Among all genes within 1.5 Mb of the Pdcd1 locus,only PD-1 mRNA expression was significantly de-creased by deletion of the –23.8 kb ChAR (fig.

S3K). This suggests that the –23.8 kb ChARpresent in exhausted, but not functional, CD8+ Tcells serves as an enhancer that is required tomaintain high levels of PD-1 expression.We next sought to identify the functional con-

tribution of specific sequences within enhancerregions to the regulation of PD-1 expression. Weused Cas9-mediated in situ saturation mutagen-esis and designed all possible sgRNAs within the–23.8 kb enhancer and eight other regulatorysequences near the Pdcd1 locus (15, 16) (Fig. 3A).We transduced Cas9-expressing EL4 cells witha pool of 1754 enhancer-targeting sgRNAs, 117sgRNAs targeting the Pdcd1 exons as positivecontrols, and 200 nontargeting sgRNAs as neg-ative controls (fig. S5, A and B). We sorted trans-duced EL4s into populations on the basis of highor low PD-1 expression and quantified the abun-dance of individual sgRNAs (fig. S5C).In comparison with nontargeting sgRNAs,

which were equivalently distributed betweenPD-1–high and PD-1–low fractions, sgRNAs tar-geting Pdcd1 exons were highly enriched in the

1168 2 DECEMBER 2016 • VOL 354 ISSUE 6316 sciencemag.org SCIENCE

Fig. 4. Exhaustion-specific epigenetic profiles in the mouse are con-served in antigen-specific exhausted human T cells in HIV-1 infec-tion. (A) Representative ATAC-seq tracks from naïve, HIV-1 tetramer+,and CMV tetramer+ samples at the IFNG gene locus (top). Orthologousregions from five mouse cell states at the IFNG locus, based on mapping ofmouse ChARs to the human genome (colored blocks, bottom). (B) Sche-matic diagram of mouse and human comparative analysis. (C) Heat mapof average chromatin accessibility at regions orthologous to mouse naïve,

memory, and exhaustion enhancers in human samples indicated. Color scale as in Fig. 2A. (D) PD-1 and CD39 expression measured by flow cytometry inHCV C63B tetramer+, HCV 174D tetramer+, and influenza (flu) matrix peptide (MP) tetramer+ populations from a single HCV-infected donor. (E) Viral sequencesencoding C63B and 174D epitopes. (F) Heat map of average chromatin accessibility at regions orthologous to mouse naïve, memory, and exhaustion enhancersin human samples indicated from a single HCV-infected donor.

RESEARCH | REPORTSon F

ebruary 14, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 5: TCELLEXHAUSTION The epigenetic landscape of T cell exhaustion · 1166 2 DECEMBER 2016• VOL 354 ISSUE 6316 sciencemag.org SCIENCE Fig. 1. CD8+ Tcell exhaustion is associated with

PD-1–low fraction as expected (Fig. 3D and fig.S5, D and E). sgRNAs targeting eight of the nineregulatory regions were also significantly en-riched in the PD-1–low fraction to varying de-grees (P < 0.00001 to P < 0.01, see supplementarymethods), suggesting that critical sequencesaffecting PD-1 expression are densely repre-sented within each of the eight regulatoryregions. However, sgRNAs in the –35.6 kb ChARhad no significant effect on PD-1 expression,consistent with prior observations that thisregion falls outside the CCCTC-binding factor(CTCF)–mediated boundaries of the Pdcd1locus (10).We focused on sgRNAs inducing cleavage in

the –23.8 kb enhancer (fig. S5F) and found astrong correlation between the predicted activ-ity in a pooled setting (PD-1 high:low ratio >1 SDbelow mean) and their effect on PD-1 mean fluo-rescence intensity in individual cell lines (P =0.0041) (fig. S5, G and H). Inspection of the pre-dicted cleavage-site locations revealed three crit-ical regions of the enhancer in which cleavagemarkedly affected PD-1 expression (Fig. 3E, grayshading).We next asked whether these critical regions

in the –23.8 kb enhancer were associated withdistinct patterns of transcription factor (TF) bind-ing in exhausted CD8+ T cells in vivo. We iden-tified TF footprints (17) using ATAC-seq cut sitesfrom CD8+ T cells experiencing chronic infec-tion, which allowed us to infer TF binding with-in the –23.8 kb enhancer (Fig. 3E; fig. S6, A toD; fig. S7A; and tables S3 to S6). We found thatcleavage sites of sgRNAs that reduced PD-1expression in EL4 cells were significantly en-riched in TF footprints found in exhausted CD8+

cells in vivo (P = 8.63 × 104, hypergeometrictest). The three TF footprints with greatest sen-sitivity to disruption corresponded to motifsfor Sox3, T-bet (encoded by Tbx21), and retinoicacid receptor (RAR) in exhausted CD8+ T cellsin vivo (Fig. 3F and fig. S7B). Indeed, compar-ison of genome-wide TF footprinting betweenchronic and acute infection at d27 to identifyTF motifs that showed significantly differentialinferred binding (Fig. 3G, fig. S7C, and tablesS3 and S5) confirmed that Rara binding wassignificantly enriched in exhausted CD8+ T cells(FDR = 3.14 × 10−13) compared with their func-tional counterparts.To test whether T cell exhaustion is also asso-

ciated with a distinct epigenetic state in humanexhausted CD8+ T cells, we analyzed global pat-terns of chromatin accessibility in tetramer+

CD8+ T cells from four subjects with chronicprogressive HIV-1 who were not on therapy(Fig. 4, A and B; fig. S8, A and B; and table S7).We successfully mapped 80 to 85% of ChARsidentified in the mouse model to their humanorthologous regions (Fig. 4A, colored blocks,and fig. S8C) (18, 19) and found them to beenriched for disease-associated single-nucleotidepolymorphisms (SNPs) (probabilistic identifica-tion of causal SNPs, P < 2.77 × 10−8; hyper-geometric test) (fig. S8D) (20) and, in particular,immune-related National Human Genome Re-

search Institute genome-wide association studySNPs (P < 3.70 × 10−3) (fig. S8, E to G). Thisenrichment strongly suggested that mapped re-gions corresponded to functional regulatory re-gions within the immune system. Regions atthe Pdcd1 locus were not among those mappedfrom the mouse model, as previously observed(10), which limited our ability to detect an or-tholog to the –23.8 kb enhancer observed inthe mouse model.Human naïve CD8+ T cells from the majority

of donors showed greater chromatin accessi-bility in naïve-specific regions defined in themouse than in memory- or exhaustion-specificregions. In the healthy donor, CMV-specifictetramer+ CD8+ T cells, and effector memorycells were enriched for memory-specific regions(Mann-Whitney U test, P = 0.01 to P < 0.0001)(Fig. 4C and fig. S8H). In contrast, HIV-specifictetramer+ cells from three out of the four subjectsshowed significantly greater chromatin acces-sibility in exhaustion-specific regions (Mann-Whitney U test, P = 0.05 to P < 0.001) than inmemory-specific regions.Finally, we confirmed these findings in a sub-

ject with chronic hepatitis C virus (HCV) infectionin whom CD8+ T cell responses to two epitopesof HCV could be detected (Fig. 4D). Sequencingof the HCV genome in this subject revealedthat, unlike the C63B epitope, the 174D epitopehad undergone extensive viral escape, and nowild-type viral sequence could be detected (Fig.4E). We found that the C63B tetramer+ cellshad a phenotype consistent with exhaustionand showed significantly greater chromatin ac-cessibility at exhaustion-specific regions (Mann-Whitney U test, P = 0.01) than memory regions(Fig. 4F). In contrast, 174D tetramer+ cells, whichwere specific for the escape mutant epitope,lacked exhaustion-specific surface markers andshowed greater chromatin accessibility in memory-specific regions, as did influenza-specific CD8+

T cells (Mann-Whitney U test, P = 0.04) (Fig. 4F).Thus, the state-specific pattern of chromatinaccessibility found in mouse exhausted CD8+

T cells is conserved in human exhausted CD8+

T cells.We find that CD8+ T cell exhaustion occurs

with a broad remodeling of the enhancer land-scape and TF binding. This suggests that ex-hausted CD8+ T cells occupy a differentiationstate distinct from functional memory CD8+ T cells.Identifying the plasticity of this state and whetheror how it could be reverted becomes a criticalquestion for immunotherapy applications. Ourdata also suggest that mapping state-specific en-hancers in exhausted T cells could enable moreprecise genome editing for adoptive T cell therapy.Genome editing of CAR-T cells to make them re-sistant to exhaustion is an appealing conceptand has led to recent studies investigating thedeletion of the PD-1 gene locus (21, 22). Editingexhaustion-specific enhancers (15) may providea more “tunable” and state-specific approach tomodulate T cell function than deleting codingregions of genes. Functional maps of enhancersspecific to exhausted CD8+ T cells may therefore

provide a crucial step toward the rational engi-neering of T cells for therapeutic use.

REFERENCES AND NOTES

1. E. J. Wherry et al., Immunity 27, 670–684(2007).

2. A. J. Zajac et al., J. Exp. Med. 188, 2205–2213(1998).

3. D. L. Barber et al., Nature 439, 682–687(2006).

4. S. L. Topalian et al., N. Engl. J. Med. 366, 2443–2454(2012).

5. D. L. Porter, B. L. Levine, M. Kalos, A. Bagg, C. H. June, N. Engl.J. Med. 365, 725–733 (2011).

6. A. H. Long et al., Nat. Med. 21, 581–590(2015).

7. J. D. Buenrostro, P. G. Giresi, L. C. Zaba, H. Y. Chang,W. J. Greenleaf, Nat. Methods 10, 1213–1218 (2013).

8. T. A. Doering et al., Immunity 37, 1130–1144(2012).

9. M. A. Paley et al., Science 338, 1220–1225(2012).

10. J. W. Austin, P. Lu, P. Majumder, R. Ahmed, J. M. Boss,J. Immunol. 192, 4876–4886 (2014).

11. J. Vierstra et al., Science 346, 1007–1012(2014).

12. D. Lara-Astiaso et al., Science 345, 943–949(2014).

13. G. Raimondi, W. J. Shufesky, D. Tokita, A. E. Morelli,A. W. Thomson, J. Immunol. 176, 2808–2816(2006).

14. O. Shalem et al., Science 343, 84–87(2014).

15. M. C. Canver et al., Nature 527, 192–197(2015).

16. J. Vierstra et al., Nat. Methods 12, 927–930(2015).

17. R. Pique-Regi et al., Genome Res. 21, 447–455(2011).

18. E. Gjoneska et al., Nature 518, 365–369(2015).

19. D. Villar et al., Cell 160, 554–566(2015).

20. K. K. Farh et al., Nature 518, 337–343(2015).

21. A. Hendel et al., Nat. Biotechnol. 33, 985–989(2015).

22. K. Schumann et al., Proc. Natl. Acad. Sci. U.S.A. 112,10437–10442 (2015).

ACKNOWLEDGMENTS

The authors thank members of the Yosef and Haininglaboratories for their input, the research subjects fortheir participation, and J. Doench and the entire GeneticPerturbation Platform at the Broad Institute for theiradvice on Cas9-mediated screening technology. Theauthors are grateful for input from the Cancer Center forGenome Discovery. The data reported in this manuscriptare tabulated in the main paper and in the supplementarymaterials. Genome-wide data generated in this study canbe accessed via GEO accession no. GSE87646. This researchwas supported by AI115712, AI091493, and AI082630 to W.N.H.from the NIH; by the BRAIN Initiative grants MH105979 andHG007910 from the NIH to N.Y.; and by 1R21AI078809-01 andUM1 AI068618 from the NIH to N.F. The authors declare nopotential conflicts of interest. D.R.S., J.K., N.Y., E.J.W., andW.N.H. are inventors on a patent application (U.S. PatentApplication no. 62/310,903) held and submitted by Dana-FarberCancer Institute.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/354/6316/1165/suppl/DC1Materials and MethodsFigs. S1 to S8Tables S1 to S7References (23–43)

18 December 2015; accepted 7 October 2016Published online 27 October 201610.1126/science.aae0491

SCIENCE sciencemag.org 2 DECEMBER 2016 • VOL 354 ISSUE 6316 1169

RESEARCH | REPORTSon F

ebruary 14, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 6: TCELLEXHAUSTION The epigenetic landscape of T cell exhaustion · 1166 2 DECEMBER 2016• VOL 354 ISSUE 6316 sciencemag.org SCIENCE Fig. 1. CD8+ Tcell exhaustion is associated with

The epigenetic landscape of T cell exhaustion

Allen, Nicole Frahm, Georg M. Lauer, E. John Wherry, Nir Yosef and W. Nicholas HainingTsao, Jernej Godec, Martin W. LaFleur, Flavian D. Brown, Pierre Tonnerre, Raymond T. Chung, Damien C. Tully, Todd M. Debattama R. Sen, James Kaminski, R. Anthony Barnitz, Makoto Kurachi, Ulrike Gerdemann, Kathleen B. Yates, Hsiao-Wei

originally published online October 27, 2016DOI: 10.1126/science.aae0491 (6316), 1165-1169.354Science 

, this issue p. 1104, p. 1165; see also p. 1160ScienceT cell phenotype. Thus, epigenetic regulation may limit the success of immunotherapies.

memoryof exhausted T cells after immunotherapy. Although there was transcriptional rewiring, the cells never acquired a examined the epigenetic profileet al.modules of enhancers that are also conserved in exhausted human T cells. Pauken

defined specific functionalet al.exhausted T cells are a distinct lineage (see the Perspective by Turner and Russ). Sen epigenetic profile of exhausted T cells differs substantially from those of effector and memory T cells, suggesting thatImmunotherapies aim to reverse this state. Using a mouse model of chronic infection, two studies now show that the

During cancer or chronic infection, T cells become dysfunctional, eventually acquiring an ''exhausted'' phenotype.The epigenetics of exhaustion

ARTICLE TOOLS http://science.sciencemag.org/content/354/6316/1165

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2016/11/04/science.aae0491.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/354/6316/1160.fullhttp://science.sciencemag.org/content/sci/354/6316/1104.full

REFERENCES

http://science.sciencemag.org/content/354/6316/1165#BIBLThis article cites 43 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 February 14, 2021

http://science.sciencem

ag.org/D

ownloaded from