Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Sep 9.
Published in final edited form as: Cancer Cell. 2024 Aug 29;42(9):1582–1597.e10. doi: 10.1016/j.ccell.2024.08.007

Combination anti-PD-1 and anti-CTLA-4 therapy generates waves of clonal responses that include progenitor-exhausted CD8+ T cells

Kevin Wang 1, Paulina Coutifaris 1, David Brocks 2, Guanning Wang 1, Tarek Azar 1, Sabrina Solis 3, Ajeya Nandi 1, Shaneaka Anderson 1, Nicholas Han 1, Sasikanth Manne 4, Evgeny Kiner 2, Chirag Sachar 2, Minke Lucas 5, Sangeeth George 6, Patrick K Yan 7, Melanie W Kier 8, Amy I Laughlin 9, Shawn Kothari 10, Josephine Giles 4, Divij Mathew 4, Reem Ghinnagow 11, Cecile Alanio 12,17, Ahron Flowers 1,13, Wei Xu 11, Daniel J Tenney 14, Xiaowei Xu 13,15, Ravi K Amaravadi 1,13, Giorgos C Karakousis 13,16, Lynn M Schuchter 1,13, Marcus Buggert 17, Derek Oldridge 18,19, Andy Minn 11,20, Christian Blank 5,21,22, Jeffrey S Weber 3, Tara C Mitchell 1,13, Michael D Farwell 23,24, Ramin S Herati 3,#, Alexander C Huang 1,20,25,26,27,#
PMCID: PMC11387127  NIHMSID: NIHMS2018187  PMID: 39214097

Summary

Combination checkpoint blockade with anti-PD-1 and anti-CTLA-4 antibodies has shown promising efficacy in melanoma. However, the underlying mechanism in humans remains unclear. Here, we perform paired single-cell RNA and TCR sequencing across time in 36 patients with stage IV melanoma treated with anti-PD-1, anti-CTLA-4, or combination therapy. We develop the algorithm Cyclone to track temporal clonal dynamics and underlying cell states. Checkpoint blockade induces waves of clonal T cell responses that peak at distinct timepoints. Combination therapy results in greater magnitude of clonal responses at 6 and 9 weeks compared to single-agent therapies, including melanoma-specific CD8+ T cells and exhausted CD8+ T cell (TEX) clones. Focused analyses of TEX identify that anti-CTLA-4 induces robust expansion and proliferation of progenitor TEX, which synergizes with anti-PD-1 to reinvigorate TEX during combination therapy. These next generation immune profiling approaches can guide the selection of drugs, schedule, and dosing for novel combination strategies.

Keywords: Checkpoint blockade, T cell exhaustion, PD-1 blockade, CTLA-4 blockade, single-cell sequencing, combination checkpoint blockade, progenitor exhausted CD8+ T cells, clonotypic analysis, melanoma, immunotherapy

eTOC blurb

graphic file with name nihms-2018187-f0001.jpg

Wang et al. analyze longitudinal blood from melanoma patients on checkpoint blockade using paired single-cell RNA and TCR sequencing and uncover waves of clonal responses that peak at different timepoints. By studying anti-CTLA-4, anti-PD-1, and in combination, they show that anti-CTLA-4 reinvigorates progenitor exhausted CD8+ T cells while anti-PD-1 drives their differentiation.

Introduction

Immune checkpoint inhibitors, such as anti-PD-1 therapy (αPD-1) and anti-CTLA-4 (αCTLA-4) therapy, have transformed clinical oncology by inducing long-term remissions in a variety of histologies, even in the metastatic setting. Combination checkpoint blockade with anti-PD-1 and anti-CTLA-4 antibodies (combination therapy) is more effective than single-agent therapy in metastatic melanoma, with a 52% overall survival at 5 years, compared to 44% and 26% in αPD-1 and αCTLA-4, respectively1. A better understanding of the cellular and molecular mechanisms of αPD-1 and αCTLA-4 individually, and in combination, will guide the development of safer and more effective combination immunotherapy strategies.

CTLA-4 and PD-1 blockade each have distinct mechanisms of tumor control that have been defined using pre-clinical models2. CTLA-4 is upregulated early upon T cell activation3,4 and is a competitive inhibitor of the costimulatory molecule CD285-7, attenuating T cell activation in both a T cell intrinsic3,4,8 and extrinsic manner9-12. Thus, CTLA-4 blockade enhances the priming and early activation of effector T cells, resulting in improved tumor control13. Indeed, the human αCTLA-4 antibody ipilimumab results in increased CD8+ tumor infiltrating lymphocytes (TIL)14 as well as broadening of the T cell receptor repertoire15,16 and tumor-specific CD8+ T cell responses17. Ipilimumab also has major pharmacodynamic effects on CD4+ T cells, including the expansion of Th1-like ICOS+Tbet+CD4+ T cells18-21 and regulatory T cells (Tregs)14,22,23, and the generation of CD4+ phenotypes, or archetypes, that are normally absent in physiological settings24.

The major ligand for PD-1 is PD-L1, which is expressed by myeloid cells and cancer cells in the tumor microenvironment. PD-L1 is induced by IFN-γ25, and represents an adaptive resistance mechanism of tumors in response to immune pressure26-29. Exhausted CD8+ T cells (TEX), in particular, are sensitive to PD-1 inhibition as they have the highest expression of PD-1 among CD8+ T cells30. PD-1 blockade reinvigorates TEX, allowing for a temporary burst of proliferation and recovery of effector function31-35, resulting in improved viral and tumor control. In addition, PD-1 blockade has also been shown to modulate effector CD8+ T cells36,37 and Treg populations38,39. Thus CTLA-4 represents an early proximal checkpoint of T cell activation, while PD-1 more subtly modulates T cell differentiation upon T cell activation.

Nevertheless, the cellular and molecular mechanisms of combination checkpoint blockade in humans are still unclear, including the individual contributions of αPD-1, and αCTLA-4. Moreover, the immune responses of patients receiving αPD-1, αCTLA-4, and combination therapy have not been compared directly. Herein, we interrogated the immune responses of cohorts of patients with melanoma receiving αPD-1, αCTLA-4, or combination therapy. We used flow cytometry and single cell transcriptomics paired with clonotypic analyses to study the pharmacodynamic immune properties of checkpoint blockade in several dimensions - in terms of magnitude, breadth, and durability of responses. Using clonotypic trajectory analyses, we determined that these pharmacodynamic responses of CD8+ T cells were composed of waves of clonotypic responses with distinct temporal kinetics and cellular composition. Combination therapy induced large clonal responses at 6 and 9 weeks after treatment, which enriched for exhausted and effector CD8+ T cell clones. Finally, we identified that combination checkpoint blockade resulted in enhanced generation of progenitor TEX, which was largely due to the effect of αCTLA-4.

Results

Combination checkpoint blockade results in exhausted and effector CD8+ T cell responses

By tracking proliferating CD8+ T cells, we and others have identified that PD-1 blockade induces systemic and intra-tumoral immune responses that peak after a single dose of αPD-1 by 3 weeks40-43. We extended these analyses to cohorts of patients treated with αCTLA-4 and αPD-1 plus αCTLA-4 (combination therapy). Similar to αPD-1, αCTLA-4 and combination therapy had maximal pharmacodynamic effects on PD-1 expressing CD8+ T cells (Figures S1A and S1B). Interestingly, treatment-naïve patients who received αPD-1 had a more durable immunologic response than those who had prior αCTLA-4. Combination therapy resulted in a greater magnitude of immunologic response compared to either single-agent therapy alone and had additional effect on PD-1 negative cells, consistent with previous studies44.

To understand the underlying cellular and molecular mechanisms of combination therapy on CD8+ T cells, we performed single-cell analyses of transcriptome and T cell receptor (TCR) clonotypes on 36 patients with melanoma receiving αCTLA-4, αPD-1, or combination therapy across 4 timepoints. We focused on non-naïve CD8+ T cells (Star methods: CD8+ T cell Atlas, Figure 1A), as naïve CD8+ T cells were almost exclusively singlet T cell clones and did not expand in response to therapy. We constructed a batch-corrected non-naïve CD8+ T cell atlas using Harmony45 and Symphony46, with a total of 273,508 non-naïve CD8+ T cells (Figure 1B, Figure S2A, Star methods: CD8+ T cell atlas). Baseline clinical characteristics are summarized in Tables 1 and S1.

Figure 1. Combination therapy induces TEX and effector CD8+ T cell responses.

Figure 1.

A. Study schema. Peripheral blood collected from 36 patients at baseline and every 3 weeks after checkpoint blockade. Clinical metadata. Median age. Response includes complete response and partial response.

B. Uniform Manifold Approximation and Projection (UMAP) representation of non-naive CD8+ T cells (n=273,508). CM indicates central memory, SCM indicates stem cell-like memory.

C. Auto-correlation of top 3000 genes ranked by Hotspot and 4 identified modules by agglomerative clustering.

D. Pathway enrichment analysis of modules, with selected enriched pathways colored by −log 10 of the adjusted p-value (p-adj). Abbreviations: Effector – Eff, Memory – Mem, AP-1/IFN – AP1, Mito/ATP – ATP.

E. UMAPs colored by gene module signature scores.

F. Average module expression by metacluster.

G. Fold change from baseline of frequencies for each metacluster. Significance as indicated by p-value or star is the change in frequency from baseline. One patient in the αPD-1 group with an extremely large exhausted CD8+ T cell population at baseline (15.8%), confirmed by flow cytometry, was excluded from the heatmap. Flow cytometry data is in Figure S1. αCTLA-4 (n=10), αPD-1 (n=15), combination (αPD-1 and αCTLA-4) (n=9). Wilcoxon signed-rank test.

H. Frequency of exhausted and effector CD8+ T cells for combination therapy at indicated times. Transparent lines indicate individual patients (n=9). Opaque lines indicate mean. Wilcoxon signed-rank test.

NS non-significance; *p<0.05.

See also Figures S1 and S2, Tables 1 and S2.

Table 1.

Baseline characteristics of single cell sequencing cohort, related to Figure 1 and Star Methods. See also Table S1.

Pembrolizumab
(n=16)
Ipilimumab
(n=11)
Combination (Ipi+nivo)
(n=9)
Age (Median) 71 (39-83) 59 (50-79) 52 (29-71)
Men 8 (50%) 6 (55%) 7 (78%)
Women 8 (50%) 5 (45%) 2 (22%)
Race
 White 12 (75%) 9 (82%) 9 (100%)
 Other - 2 (18%) -
 Unknown 4 (25%) - -
ECOG performance status
 0-1 15 (94%) 11 (100%) 8 (89%)
 >=2 1 (6%) 1 (11%)
LDH
 Normal 10 (63%) 3 (27%) 5 (56%)
 Elevated 5 (31%) 6 (55%) 2 (22%)
 Unknown 1 (6%) 2 (18%) 2 (22%)
Median number of cycles 10 (3-31) 4 (4-4) 4 (2-4) / 32 (2-55)a
Clinical Response
 Complete response 5 (31%) 3 (27%) 4 (44%)
 Partial response 3 (19%) 3 (27%) 3 (33%)
 Stable disease 2 (12%) - -
 Progressive disease 6 (38%) 3 (27%) 1 (11%)
 Not applicableb - 2 (18%) 1 (11%)
Number of previous lines of therapy
 0 4 (25%) 7 (64%) 5 (56%)
 1 7 (44%) 1 (9%) 2 (22%)
 2 4 (25%) 3 (27%) 1 (11%)
 >3 1 (6%) - 1 (11%)
Previous therapyc
 Immunotherapy 12 (75%) 3 (27%) 3 (33%)
  Ipilimumab 12 (75%) 1 (9%) -
  Pembrolizumab - 1 (9%) 2 (22%)
  Interferon - 1 (9%) 1 (11%)
  IL-2 - - 1 (11%)
 Chemotherapy 5 (31%) 1 (9%) -
 BRAF or MEK inhibitor - 2 (18%) 2 (22%)
 Kinase inhibitor - 1 (9%) -

All patients included in the study had stage IIIC, recurrent, or IV disease. Combination therapy is ipilimumab and nivolumab.

a

Two patients in combination therapy group lost to follow up while on the nivolumab maintenance phase of the therapy

b

Unable to assign clinical response to patients if they received radiation therapy while on treatment

c

Immunotherapy – ipilimumab, pembrolizumab, interferon, IL-2; Chemotherapy – dacarbazine, temozolomide

BRAF or MEK inhibitor – vemurafenib, dabrafenib, encorafenib, binimetinib; Kinase inhibitor – temsirolimus.

We first sought to understand the underlying transcriptional machinery of human non-naïve CD8+ T cells using gene module analysis. We used the algorithm Hotspot47 to identify modules of co-expressed genes through an unsupervised approach. Hotspot identified 4 distinct modules which included co-regulated genes and pathways known to be important in effector, memory, AP-1 and interferon, and mitochondrial/ATPase biology (Figures 1C and 1D, Table S2). The effector module included genes and pathways associated with T cell activation, TCR signaling, migration, and cytotoxicity48,49. The memory module included metabolic pathways known to be used in memory cells such as oxidative phosphorylation, as well as pathways important for translation machinery, cytokine signaling, and survival48,49. We also identified a gene module that enriched for pathways downstream of cellular perturbations, including AP-150, interferon51, and HIF-1α pathways52, which are modulated downstream of T cell activation and cytokine signaling. Finally, a module was identified that included genes related to the mitochondrial electron transport chain and ATP production (ATP5F1E, ATP6V0E1) in conjunction with annexins (ANXA2, ANXA1) and S100 family members (S100A4, S100A10) (Figures 1C and 1D, Table S2).

We then defined non-naïve CD8+ T cell subsets using differential expression of these modules, which allowed us to fine-tune the cluster resolution and generate clusters with distinct molecular identities (Figures 1E and 1F, Star methods: CD8+ T cell atlas). We identified a spectrum of CD8+ T cell differentiation states. Both stem-cell memory (SCM) and memory CD8+ T cells had enrichment of the memory module, but SCM cells were also enriched for a naïve transcriptional signature (Figure S2B); activated CD8+ T cells had high expression of the AP-1/IFN module, and effector and NK-like CD8+ T cells were identified by high expression of the effector module as well as an EMRA gene signature (Figures 1B, 1E, 1F, and S2B).

Exhausted CD8+ T cells in the blood did not cluster independently in unsupervised analysis. This was not surprising due to the low frequency of TEX in the blood and the known heterogeneity of TEX which span a spectrum from progenitor to more differentiated TEX54. Therefore, we defined TEX using a human exhausted gene signature from sorted PD1+CD39+ CD8+ T cells53 (Figure S2C, Table S2, Star methods: Gene lists and exhausted CD8+ T cells). Effector CD8+ T cells had high effector and mitochondrial ATPase activity, while TEX had decreased expression of effector, AP-1, and mitochondrial modules in comparison (Figure 1F). Thus, these analyses capture exhausted CD8+ T cell biology in which partial effector function loss is driven by mitochondrial dysfunction55,56 as well as partnerless NFAT in the absence of AP-157. Altogether, we identified CD8+ T cell states that were consistent with published gene signatures of memory, effector, and exhausted CD8+ T cells (Figures S2B-F) as well as their underlying biology.

Having constructed a non-naïve CD8+ T cell atlas, we sought to identify the cellular changes induced by single-agent and combination checkpoint blockade. αPD-1 induced an expansion of TEX at week 3 as previously reported41, while combination checkpoint blockade significantly increased the frequency of both exhausted and effector CD8+ T cells, which peaked at 6 and 9 weeks, respectively (Figures 1G and 1H). Thus, combination checkpoint blockade therapy had not only a larger magnitude of CD8+ T cell response but also a broad and durable pharmacodynamic effect on CD8+ T cell subsets. These data are consistent with previous studies in patients showing that combination therapy induced a greater magnitude of transcriptional and cytokine changes58 as well as expansion of Tbet+Eomes+CD8+ T cells44 compared to single-agent therapy.

Checkpoint blockade induces waves of clonal CD8+ T cell responses with distinct kinetics and cell state compositions

To understand more precisely how CD8+ T cells respond to checkpoint blockade, we used each cell’s TCR to track clonal dynamics across time. First, to control for normal clonal fluctuations over time, we performed paired single-cell RNA and TCR sequencing on sorted CD8+ T cells from peripheral blood mononuclear cells (PBMC) collected three weeks apart from 2 healthy donors (HD) and assessed the clonal dynamics of non-naïve CD8+ T cells. Overall, healthy donors exhibited minimal clonal variation over time. On the contrary, checkpoint blockade therapy induced significant changes in the frequency of CD8+ T cell clones (Figures S3A-C). Thus, checkpoint blockade therapy induced a greater magnitude of clonal perturbation, above that of normal physiologic variation, consistent with previous reports15,16.

We wanted to focus our analyses on robust clonal responses that were meaningful and therapeutically induced, both in terms of fold change and absolute change in frequency. Only cells with both productive TCR alpha and beta chains were included. We additionally excluded patients with prior immunotherapy to eliminate possible pretreatment effects, except for a large cohort of patients who received αPD-1 and had prior αCTLA-4 therapy with ipilimumab, which we separated into a distinct treatment group (ipi pretreated patients). This resulted in a subset of 83,287 cells constituting a total of 32,080 unique clones across eligible patients and timepoints (Star methods: Cyclone and TCR trajectory). Finally, only significantly changing clones were included in downstream analysis, based on the distribution of clonal changes in the HD reference, which yielded 508 clones, or 1.58% of the total pool (Figures S3D and S3E). Strikingly, each treatment had different frequencies of clones that met the threshold: αCTLA-4 (0.95%), αPD-1 (prior ipi) (1.15%), αPD-1 (2.49%), combination (2.6%), and HD (0.61%) (Figure S3F). αPD-1 and combination therapy had greater dynamic clonal changes than αCTLA-4 and αPD-1 in ipi pretreated patients, suggesting that αCTLA-4 pretreatment affects immune response to subsequent therapies. Thus, immune checkpoint blockade dynamically modulates 1-3% of non-naïve CD8+ T cell clones in the blood, and different treatments induced different magnitudes of clonal responses, consistent with the changes seen in Ki67 (Figure S1).

To understand the pharmacodynamic patterns of T cell clones over time, we developed the algorithm Cyclone59, which treats TCR clonotypic frequencies across time as time-series data and models every clonotype as individual trajectories. Using insights from the field of trajectory clustering, we explored the distinct patterns of TCR expansion and contraction by binning clones through an unsupervised approach based on pairwise frequency correlation (Star methods: Cyclone and TCR trajectory). Using Cyclone, we tracked 508 unique TCR clonotypes in 18 patients across 9 weeks of treatment and identified 6 patterns of clonotypic trajectories (Figures 2A and 2B). Notably, the trajectories generated by immune checkpoint blockade were composed of unique clones that peaked at different timepoints, at weeks 3, 6, and 9, indicating waves of clonal T cell responses. Four out of the six clonotypic patterns consisted of expanding trajectories, including those peaking at week 3 (Traj 3), week 6 (Traj 4 and 5), and week 9 (Traj 6). Traj 1 and 2 were both contracting trajectories, representing clonotypes that decreased in frequency during treatment, albeit with different kinetics.

Figure 2. Checkpoint blockade induces distinct clonal trajectories over time.

Figure 2.

A. Pairwise correlation of clonotypic trajectories using Cyclone.

B. Cumulative frequencies for 6 clonotypic patterns. Each line denotes a single clone, and height between adjacent lines at each timepoint denotes frequency.

C. Cumulative clonotypic frequencies split by treatment. Asterisk indicates significantly greater expansion per clone for the following comparisons: Traj 4: αPD-1 vs αPD-1 (prior ipi) ***, αPD-1 vs αCTLA-4 **, Combination vs αPD-1 (prior ipi) ***; Traj 5: Combination vs αPD-1 *, Combination vs αCTLA-4 **; Traj 6: Combination vs αPD-1 **, Combination vs αPD-1 (prior ipi) **, Combination vs αCTLA-4 ***. *p<0.05, **p<0.01, ***p<0.001, Wilcoxon rank-sum test. See Figure S4 for all comparisons and statistics. Combination therapy indicates αPD-1 and áCTLA-4.

D. Trajectory composition of each treatment, scaled by absolute number of clones.

E. Fold change of cell state composition stratified by pattern, averaged by patient. Percent of cells that have Ki67 gene expression, capped at 10% for visualization. CM indicates central memory, SCM indicates stem cell-like memory.

For all: αCTLA-4 (n=4), αPD-1 (prior ipi) (n=7), αPD-1 (n=4), combination (n=3). Large outlier clones greater than 2.5 standard deviations above mean frequency were excluded from visualization in B-E.

See also Figures S1, S3, S4, and S5.

To understand whether single-agent and combination checkpoint blockade strategies induced distinct clonal dynamics, we separated each clonotypic pattern by treatment (Figures 2C, 2D, S4A, and S4B). While αCTLA-4 induced smaller changes in CD8+ T cell clonal frequency, it has previously been shown to increase clonal diversity15,16, which may be below the sensitivity of detection by single-cell TCR sequencing. αPD-1 in ipi pretreated patients induced Traj 3 that peaked at week 3, consistent with the early kinetics of αPD-1 previously described40-43, although major clonal responses were also seen at week 6 and to a lesser extent at week 9. Treatment-naïve patients, however, had significantly larger expansion of clones at week 6 (Traj 4) (Figures 2C, 2D, and S4C) and generated large novel clones at each timepoint, while ipi pretreated patients had few or no novel clones (Figures S4D-F). Combination therapy, however, induced large clonal responses peaking at weeks 6 and 9 (Traj 4-6) and generated significantly larger novel clones compared to αCTLA-4 or αPD-1 alone. All three treatments resulted in substantial clonal contraction (Traj 1), consistent with the TCR turnover and evolution previously described15,16.

We then analyzed the underlying cell state compositions of these clonal responses using cell state assignments from the non-naïve CD8+ T cell atlas (Figure 1B). Compared to ipi pretreated patients, treatment-naïve patients receiving αPD-1 had clonal expansion across a number of cell states spanning early activated, activated, and early effector at week 3, and early effector and NK-like responses at week 6 (Figures 2E, S5A, and S5B). These phenotypes were not captured amongst the proliferation-based responses that had been described previously40-42, likely due to the fact that these other cell states had a lower Ki67 expression as compared to exhausted CD8+ T cells (Figure 2E: %Ki67 overlay). αCTLA-4 resulted in broad responses with lower magnitude, while combination therapy induced major TEX and effector responses that peaked at 6 and 9 weeks, respectively (Figures 2E, S5A, and S5B).

Combination therapy generates melanoma-specific responses that peak at 3 and 6 weeks.

We next sought to understand the antigen specificity associated with each clonal trajectory to uncover possible clinical relevance. We used a combinatorial tetramer approach to identify antigen-specific responses in serial blood samples from patients expressing A1, A2, A3, or B7 alleles across three treatments: αCTLA-4 (n=2), αPD-1 (n=3), and combination therapy (n=6) (Star methods: Combinatorial Tetramer). In total, we identified 13 melanoma-specific (MART-1, gp100, Tyrosinase, LAGE-1, NY-ESO1, TAG, and MAGE-A1) and 11 viral-specific (CMV, EBV, Flu) CD8+ T cell populations in 9 out of 11 patients (Figure 3A). We detected melanoma-specific CD8+ T cells in 0/2 patients on αCTLA-4, 2/3 patients on αPD-1, and 5/6 patients on combination therapy. Therefore, we primarily focused on combination therapy, which had the majority of detected melanoma-specific responses and had statistically significant expansions at 3 and 6 weeks post-treatment. In contrast, viral-specific CD8+ T cells expanded at 9+ weeks (Figures 3B and 3C).

Figure 3. Combination checkpoint blockade induces melanoma-specific CD8+ T cell responses at 3 and 6 weeks.

Figure 3.

A. Representative flow plots showing combinatorial tetramer gating for HLA-A2 restricted (indicated by A2) melanoma and viral specific CD8+ T cells in one sample.

B. Representative flow plots showing melanoma and viral specific CD8+ T cells at indicated timepoints. Week 9+ indicates weeks 9-15. A2 and A3 indicate HLA-A2 and HLA-A3 restricted epitopes, respectively.

C. Frequencies of HLA-A1, HLA-A2, HLA-A3, and HLA-B7 restricted melanoma and viral specific CD8+ T cells in combination therapy (αPD-1 and αCTLA-4) at indicated timepoints. Each line indicates one patient-specific antigen-specificity. Melanoma (n=6 patients, n=13 patient antigen-specificities); Viral (n=4 patients, n=7 patient antigen-specificities). NS non-significance; *p<0.05, **p<0.01, Wilcoxon signed-rank test.

D. Cumulative frequencies of antigen specific CD8+ T cells. Each line denotes an antigen specificity for individual patients in combination therapy.

E. Phenotypic compositions of antigen specific CD8+ T cells. Each line denotes a phenotype within an antigen specificity for individual patients in combination therapy.

C-E. One large viral clone (CMV pp65 NLV) in patient 16-2189 with frequencies 0.99%, 1.4%, 1.37%, and 1.25% at weeks 0, 3, 6, and 9, respectively, was excluded for clarity.

See also Figure S6 and Table S3.

Trajectory clustering of these antigen-specific responses demonstrated that melanoma-specific CD8+ T cells occupied early trajectories that peaked at 3 or 6 weeks, while viral-specific responses, largely driven by EBV-specific CD8+ T cells, peaked at 9+ weeks (Figure 3D). Finally, CD8+ T cells responding to combination therapy were almost exclusively exhausted or EM1 (CD45RACCR7CD27+) CD8+ T cells, whether melanoma- or viral-specific (Figure 3E).

We also performed bulk TCR sequencing on FFPE tumor specimens from 10 patients in our main sequencing study (4 αCTLA-4, 5 αPD-1, 1 combination) to generate a library of TIL clonotypes, which were mapped back to our main single-cell RNA sequencing dataset. This generated a set of 79 circulating clonotypes that were also present in the tumor. We then ran Cyclone analysis on these 79 clonotypes, yielding 4 trajectories, including clones that peaked at 3, 6, and 9 weeks. For αPD-1 therapy, the majority of tumor-associated clonal responses were in Traj 2, peaking at week 3. In contrast, after combination therapy, tumor-associated clones expanded primarily at week 6 in Traj 3 (Figure S6 and Table S3).

Altogether, combination therapy expanded TEX and effector clones at 6 and 9 weeks after treatment, while antigen-specific and tumor-associated data suggest that the early trajectories, which peak at 3 to 6 weeks, may be relevant for clinical efficacy.

Progenitor exhausted CD8+ T cells identified in peripheral blood

Exhausted CD8+ T cells, as identified by exhaustion signature, had features of memory, effector, and NK-like clusters across the non-naïve CD8+ T cell atlas (Figure 1B), reminiscent of TEX subsets with a parallel differentiation trajectory35,60-67. These TEX are enriched for tumor-specific CD8+ T cells68,69, respond robustly to αPD-131,33,35,41,60,64, and are a major mediator of the clinical efficacy of checkpoint blockade in human cancers70. Moreover, recent studies have revealed the heterogeneity of TEX, including progenitor61,62,65, intermediate/NK-like66,67,71-73, and terminal33,61,62,65 TEX that play complementary roles in tumor control. We therefore subset on TEX and fit a pseudotime trajectory, choosing the more central memory-like TEX as the starting point (Figure 4A). To characterize exhausted CD8+ T cells, we evaluated gene expression across the pseudotime gradient (Figures 4B and 4C, Star methods: Pseudotime), identifying three main subsets with expression patterns consistent with known TEX subsets, including progenitor (CTLA-4, TCF7, SPRY1)61,62,65,74, terminal33,61,62,65 (TOX, LAG3, EOMES), and intermediate/NK-like (ZEB2, BATF, KLRC)66,67,71-73. We additionally analyzed surface protein (antibody-derived tags, ADT) data for a subset of the patients and identified associated protein expression profiles for each subset including progenitor (CD62L, ICOS, CD28), terminal (CD38, CD11a, CD244), and NK-like (CD16, CX3CR1, CD56) exhausted CD8+ T cells (Figure 4D).

Figure 4. Combination therapy reinvigorates progenitor and differentiated exhausted CD8+ T cells.

Figure 4.

A. Monocle 3 tree fitted onto exhausted CD8+ T cells (TEX), colored by pseudotime.

B. Relative expression of top differentially expressed genes for each cluster, across pseudotime.

C. Gene expression for individual cells along pseudotime with fitted spline curve, for selected markers. Cells with 0 expression of a gene are excluded from the respective plot.

D. Relative expression of selected protein markers across pseudotime.

E. Fold change from baseline of frequencies for TEX subsets. Significance indicates change in frequency from baseline. One patient in the αPD-1 group with an extremely large TEX population at baseline (15.8%), confirmed by flow, was excluded from the heatmap. αCTLA-4 (n=10), áPD-1 (n=15), combination (αPD-1 and αCTLA-4) (n=9). Wilcoxon signed-rank test.

F. Frequency of progenitor (Prog) and differentiated (Diff) TEX subsets after combination therapy. Transparent lines indicate individual patients (n=9). Opaque line indicates mean.

*p<0.05, **p<0.01.

See also Figures S7, S8, and S9.

TEX in tumor have a terminally differentiated exhausted signature that clearly separates them from memory and effector CD8+ T cells68,75. However, these circulating TEX did not have the same degree of exhaustion signature. To understand how these blood TEX compared to tumor TEX, we performed single-cell RNA sequencing on paired blood and tumor specimens from 3 patients with melanoma and generated an atlas consisting of CD8+ T cells from both blood and tumor (Figures S7A and S7B, Star methods: Paired blood-tumor atlas). UMAP dimension 1 revealed the path of differentiation as we had seen in the blood, with naïve, memory, effector, and NK-like CD8+ T cells, while UMAP dimension 2 separated blood from tumor. Indeed, the addition of tumor data revealed an additional arm of TEX composed of terminal and progenitor TEX clusters (Figures S7A and S7B). Terminal TEX expressed high levels of exhaustion genes such as TOX, PDCD1, LAG3, and CXCL13. Progenitor TEX had comparatively higher expression of progenitor-associated genes such as TCF7, CCR7, SELL, and TNF (Figure S7C). At the same time, Progenitor TEX also expressed TOX and other exhaustion genes, albeit at a lower amount as compared to terminal TEX but higher than other cell states, consistent with the known biology of Progenitor TEX60,62,67,76. As the distribution of TEX subsets differ based on tissue location60-62,77, we suspected that circulating TEX were enriched for late progenitor and NK/intermediate TEX as described in preclinical models60. We defined circulating and tumor TEX based on the same threshold of human exhaustion signature (Figures S2C, S7D). By this criterion, 2.8% of circulating CD8+ T cells were TEX; in contrast 43% of CD8+ T cells in the tumor were TEX. Tumor TEX also had a greater exhaustion score compared to circulating TEX (Figure S7D). We then mapped these circulating and tumor TEX back onto the blood-tumor atlas, as defined by exhaustion score. Tumor TEX were largely restricted to terminal TEX as previously reported. However, blood TEX largely mapped to progenitor and effector clusters, consistent with the phenotype of progenitor and intermediate/NK TEX (Figure S7E). Given the small numbers of circulating TEX in the paired blood-tumor dataset, we also mapped circulating TEX from the main study, which also fell primarily in progenitor and effector clusters (Figure S7F). Altogether, these data suggest that the TEX identified in circulation are largely composed of progenitor and intermediate TEX while terminal TEX are tissue restricted.

Combination therapy generates progenitor exhausted CD8+ T cell responses that are induced by αCTLA-4

Progenitor TEX (ProgEX) had high expression of CTLA-4 (Figures 4B and 4C) and thus might be modulated by CTLA-4 blockade. To determine the impact of checkpoint blockade on TEX subsets, we defined ProgEX and differentiated TEX (DiffEX), which included terminal and intermediate/NK-like subsets, using a central memory score (Star methods: Defining Progenitor TEX). This revealed expansion of DiffEx by both αCTLA-4 and αPD-1, although changes in ProgEX were underpowered to reach statistical significance (Figure 4E). However, combination therapy was associated with significant expansion of both ProgEX and DiffEX which peaked at weeks 3 or 6 (Figure 4F).

To confirm whether combination checkpoint blockade modulated ProgEX, we analyzed flow cytometry data generated from patients with melanoma treated with combination therapy (Figure S1). We interrogated the proliferating Ki67+ responding cells after combination therapy to understand the cell states that were involved in the pharmacodynamic immune responses. UMAP visualization of these responding cells revealed an enrichment of PD-1, CTLA-4, and TCF-1 expression in responding cells after treatment (Figure S8A), suggesting that combination therapy modulated ProgEX. As CTLA-4 is expressed in both ProgEX and DiffEX62, we defined TEX as PD-1+CTLA-4+CD8+ T cells, consistent with our previously published data (Figure S8B)40,41. Both TCF-1+PD-1+CTLA-4+ (ProgEX) and TCF-1PD1+CTLA-4+ (DiffEX) CD8+ T cells had high expression of classic exhaustion markers, including TIGIT, CD39 and TOX (Figure 5A). Combination therapy generated the largest magnitude of proliferative response in ProgEX and DiffEX compared to other canonical CD8+ T cell differentiation states (Figure 5B), which was associated with expansion of these TEX subsets that peaked at week 6 (Figure S8C). Thus, consistent with single-cell sequencing data, combination therapy robustly reinvigorated both ProgEX and DiffEX subsets of TEX.

Figure 5. CTLA-4 blockade reinvigorates progenitor exhausted CD8+ T cells.

Figure 5.

A. Expression of selected markers for indicated CD8+ T cell subsets 3 weeks after combination therapy (αPD-1 and αCTLA-4). Flow cytometry was performed on PBMC from 8 patients. Central memory (CM), effector memory (EM), terminal effector memory (EMRA).

B. Ki67 expression over time for indicated CD8+ T cell subsets after combination therapy (n=8). Differentiated TEX (Diff), Progenitor TEX (Prog).

C. Study design for sequential treatment cohort. Flow cytometry was performed on blood collected every 3 weeks from a cohort of 8 patients who received ipilimumab followed by pembrolizumab.

D. Ki67 expression in CD8+ T cells over time after ipilimumab (red) and pembrolizumab (blue).

E. Frequency and Ki67 expression for progenitor exhausted CD8+ T cells (n=4).

F. Study design for Checkmate 238 study. Flow cytometry was performed on blood collected before treatment and two weeks post-treatment from patients on ipilimumab (n=24) or nivolumab (n=21).

G. Ki67 expression in progenitor TEX for ipilimumab (red, n=24) and nivolumab (blue, n=21).

H. Frequency of progenitor TEX.

I. Ki67 expression in progenitor TEX at Week 2, comparing ipilimumab and nivolumab. Error bars represent mean and 95th percentile confidence interval.

B, D-I. Transparent lines indicate individual patients. Opaque line indicates mean.

NS non-significance; *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001, Wilcoxon signed-rank test.

See also Figures S8 and S9 and Table S4.

To characterize how each monotherapy contributed to the ProgEX response in combination therapy, we performed flow cytometry on a cohort of 8 patients who received sequential treatment with αCTLA-4 (ipilimumab) followed by αPD-1 (pembrolizumab) to control for patient-specific effects (Figure 5C, 5D). αCTLA-4 induced a greater magnitude of ProgEX expansion and proliferation compared to αPD-1 (Figure 5E), which was also associated with DiffEX responses (Figure S9A). Finally, to further validate these findings and to control for pretreatment effects, flow cytometry was independently performed on peripheral blood at baseline and 2 weeks post treatment from patients with Stage III/IV resectable melanoma treated with adjuvant ipilimumab or nivolumab from a randomized clinical trial (Checkmate 238)78 (Table S4, Figure 5F). αPD-1 resulted in proliferation but no expansion of ProgEX and DiffEX (Figures 5G, 5H and S9B), In fact, there was a trend towards a decrease in the frequency of ProgEX, suggesting differentiation and/or migration events. To understand the relevance of these immune dynamics in the tumor, we performed flow cytometry on baseline and post-treatment tumor samples from a cohort of 17 patients with Stage III melanoma who received neoadjuvant αPD-1 (Figure S9C). ProgEX in the tumor were defined as PD1+TCF-1+Tim3CD8+ T cells, while terminally exhausted CD8+ T cells (TermEX) were defined as PD1+TCF-1Tim3+35,62 (Figure S9C). αPD-1 resulted in proliferation of ProgEX in the tumor microenvironment, which was coupled with a decrease in frequency of ProgEX and increase in TermEX (Figure S9D), consistent with the paradigm that PD-1 blockade drives the differentiation of ProgEX to TermEX62.

In contrast, αCTLA-4 resulted in the expansion of ProgEX in the blood and induced significantly greater proliferation of ProgEX compared to αPD-1 (Figures 5G-I) which was coupled to both expansion and proliferation of DiffEX (Figure S9B). Altogether these data demonstrate that αCTLA-4 results in expansion of ProgEX while αPD-1 drives its differentiation. These immune mechanisms may explain some of the immune and clinical effects of combination therapy.

Discussion

We used single-cell multi-modal analyses to directly compare the pharmacodynamic effects of αPD-1, αCTLA-4, and combination checkpoint blockade therapy in humans being treated for melanoma. By using single-agent therapy cohorts as comparisons, we were able to deconvolute the cellular and molecular effects of combination checkpoint blockade, revealing novel insights.

First, αPD-1 and αCTLA-4 therapy each have their own distinct pharmacodynamic properties, including kinetics of immune responses. In combination, these therapies resulted in immune responses that were both durable and greater in magnitude which may contribute to both enhanced efficacy as well as toxicity.

Second, despite clear pharmacodynamic responses by proliferation (Ki67) and frequency of cell states, these measurements represent snapshots in time and did not inform if cellular responses at different timepoints were clonally related or distinct. Therefore, we developed a novel algorithm Cyclone to characterize patterns of clonotypic trajectories in an unsupervised manner. Cyclone identified distinct waves of clonal CD8+ T cell responses that peaked at different post-treatment timepoints. Thus, in many cases, the sustained immunologic responses seen in flow cytometry reflected the sum of different clonotypes with distinct temporal trajectories. While there was sustained expansion for some clones (e.g. Traj 5), the majority of clones had trajectories characterized by brief expansion at a specific timepoint followed by a return to baseline. Each dose of immune checkpoint blockade induced the activation and expansion of distinct groups of clones which likely recognize distinct groups of epitopes. Moreover, tumor-specific CD8+ T cell clones appear to be primarily modulated within the first 3-6 weeks, consistent with the early pathologic responses seen in neoadjuvant checkpoint blockade trials in melanoma79-83. There also may be a point at which tumor-specific CD8+ T cell clones are depleted by PD-1 blockade, and therapies to promote epitope spreading84,85 or immune rest86-88 may be required to rejuvenate tumor-specific immunity. These observations are consistent with preclinical data demonstrating the importance of sustained and coordinated systemic cellular responses for effective immunotherapy89, as well as the emergence of new exhausted CD8+ T cell clones after PD-1 blockade90. With the recent success of neoadjuvant therapy91, understanding the pharmacodynamics and antigen-specificity of these waves of clonal responses will be critical for defining the optimal time for surgery after neoadjuvant therapy.

Third, immune monitoring through clonal trajectories identified a dimension of immune response distinct from cellular proliferation or Ki67. While clonal responses to αPD-1 peaked at 3 and 6 weeks, consistent with what was seen using Ki6741,42, Cyclone identified αPD-1-responsive cell states (e.g. effector, NK-like)36,37,92 in addition to the TEX responses identified by Ki6740-42. Thus, as proliferation, differentiation, and clonal expansion may have distinct temporal kinetics, clonal monitoring provides the ability to capture pharmacodynamic effects independent of proliferation. Interestingly, prior αCTLA-4 treatment appeared to blunt αPD-1 responses in terms of the magnitude of clonal responses and the diversity of cell states recruited. These data may explain why sequential therapy with nivolumab followed by ipilimumab was superior to ipilimumab followed by nivolumab93, and are consistent with published data demonstrating the presence of distinct predictive features for αPD-1 depending on prior αCTLA-4 treatment history94-96.

Finally, one of the advantages of this large dataset is the ability to study exhausted CD8+ T cell responses, despite their low frequency in the blood. Focused analyses of TEX identified subsets previously defined in preclinical models, including progenitor, terminal, and intermediate/NK-like33,35,60-62,64-67,71-73,77. Combination therapy induced major TEX responses that peaked at 6 weeks, which included progenitor and likely intermediate/NK TEX, which are known to be in circulation60. We demonstrate that αCTLA-4 resulted in the expansion of ProgEX. Thus, in the context of combination therapy, αCTLA-4 may sustain and expand the pool of ProgEX while αPD-1 drives the differentiation of ProgEX into DiffEX.

Altogether, we performed large-scale human immune profiling to study the pharmacodynamic immune responses of checkpoint blockade in several dimensions, by assessing proliferation, changes in cellular frequency, and clonotypic responses. This multidimensional approach to immune monitoring has direct translational implications and can be used to quantify and modulate the magnitude, breadth, and durability of immune responses to novel therapeutic strategies, as well as optimize timings and dosages.

Limitations of the study

There are several limitations to keep in mind for this study. First, because we chose to deeply interrogate three treatment types across several timepoints, the numbers within each treatment type are small, and we were underpowered to find associations with clinical outcomes. This was especially true for the Cyclone analyses because of the requirement for Cyclone for a complete sample set across every timepoint at the required threshold of cell number. These observations need to be validated in larger clinical studies. Second, as many of the clinical progressors were too sick to provide serial samples, some of the cohorts were skewed towards clinical responders. This was especially true for patients treated with αCTLA-4. Finally, the lack of paired tumor specimens before and after treatment limited our ability to study the pharmacodynamics of intra-tumoral T cell clones in relation to the clonal trajectories in the blood. The application of Cyclone to neoadjuvant studies would be valuable.

STAR Methods:

Resource Availability

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Alexander C. Huang (alexander.huang@pennmedicine.upenn.edu)

Materials availability

This study did not generate new unique reagents.

Data and code availability

Raw and processed single-cell sequencing data generated in this study have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus and are publicly available: (Main sequencing study: GSE272993, treatment-naïve aPD-1: GSE272734, healthy donor: GSE272735, paired blood tumor: GSE273718).

All original code for Cyclone has been deposited on GitHub: (https://github.com/jiwen90/cyclone) and Zenodo: (https://doi.org/10.5281/zenodo.12754871).

Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

Experimental model and study participant details

Patients/Specimen Collection

Patients were consented for blood and tissue collection under the University of Pennsylvania Abramson Cancer Center’s melanoma research program tissue collection protocol UPCC 08607 and neoadjuvant clinical trial UPCC02619, in accordance with the Institutional Review Board. Peripheral blood was obtained in sodium heparin tubes at pre-treatment and before each infusion and on-treatment every 3 weeks. Peripheral blood mononuclear cells (PBMC) were isolated using Ficoll gradient and stored in liquid nitrogen. For specimens from Checkmate 238, PBMC were obtained from a randomly selected subset of patients following informed consent under an IRB-approved protocol at NYU.

All relevant clinical characteristics, including age and sex, are listed in Tables 1, S1, and S4.

Method details

Single-cell RNA, TCR, and ADT sequencing

For the 32-patient cohort, following processing of frozen aliquots, cells were stained and sorted via FACS (BD Aria II). For PBMC samples, 300,000 cells from each sample were sorted on a DAPI negative gate for live cells. CD3+ T cell samples were additionally gated on CD3+. Non-naïve T cell sorted samples were additionally gated as cells that were not CD45RA+CCR7+. Non-naïve CD4+ and CD8+ sorted samples were gated on CD4+ and CD8+, respectively, in addition to CD3+ and not CD45RA+CCR7+. Cells were then stained for 30 minutes at room temperature with either a panel of 60 or 101 Total-Seq-C antibodies that also labeled samples individually through sample-specific barcodes, and washed 2x using the HT1000 laminar wash system (Curiox). Cells were then counted using the Cellaca MX High-throughput Automated Cell Counter as described in the manufacturer’s protocol (Nexcelom), pooled and loaded on the 10x Chromium Next GEM Single Cell 5' Kit (v2) using a superloading strategy, mixing cells from different samples in each 10x lane to control for lane-to-lane batch effect. TCR sequences were enriched using the human V(D)J B/T cell enrichment kit. Libraries were prepared according to manufacturer’s protocol (10x Genomics) and sequenced on a NovaSeq 6000 System using the S4 2x150 kit (Illumina). Each sample/lane was sequenced to the average depth of 30,000 to 35,000 reads for gene expression and 10,000 to 15,000 reads for CITE-seq per cell.

For the treatment-naïve patients on αPD-1 and healthy donors, single-cell sequencing was performed on sorted CD3+CD8+ T cells from peripheral blood using the 10x Chromium Next GEM Single Cell 5’ Kit (v2). TCR sequences were enriched using the human V(D)J T cell enrichment kit. Sequencing was performed as described above.

For the samples used to construct the paired blood-tumor atlas, single-cell sequencing was performed on sorted CD4+ and CD8+ T cells from blood and CD45+ cells from tumor biopsies using the 10x Chromium Next GEM Single Cell 5’ Kit (v2). TCR sequences were enriched using the human V(D)J T cell enrichment kit. Sequencing was performed as described above.

Pre-processing of scRNA/TCR-seq libraries

Raw reads were aligned to the human transcriptome (refdata-cellranger-GRCh38-2020-A) to produce cell-by-gene matrices using Cell Ranger count (10x Genomics, version 7.0.0) with default parameters and a Feature Reference CSV file containing the feature barcode library if applicable. For VDJ, raw reads were aligned to the VDJ reference (refdata-cellranger-vdj-GRCh38-alts-ensembl-5.0.0) using Cell Ranger vdj. For the 32-patient cohort, single cells were dehashed into their respective samples by their sample-specific barcodes.

CD8+ T cell atlas construction

Out of the 36 patients in the single-cell sequencing cohort, the 32 patients on αCTLA-4, αPD-1 (prior ipi), and combination therapy were sequenced with one or more of the following 5 sorting strategies: PBMC, CD3+ T cells (CD3+), non-naïve T cells (not CD45RA+CCR7+), non-naïve CD4+ (CD4+ and not CD45RA+CCR7+), and non-naïve CD8+ T cells (CD8+ and not CD45RA+CCR7+). The number of patients for each sorting strategy combination were as follows: PBMC, non-naïve CD4+, and non-naïve CD8+ (2); PBMC and non-naïve T cells (13); CD3+ T cells (17). RNA, TCR, and ADT data were generated for all samples. The CD3+ T cell sort is referred to as PanT in the Seurat object metadata. The non-naïve T cell sort is referred to as Tcells in the Seurat object metadata.

Four additional treatment-naïve patients on αPD-1 and two healthy donors were sorted on CD3+CD8+ T cells. RNA and TCR data were generated for these samples.

Cells with greater than 200 detected genes, less than 2500 detected genes, and less than 10 percent mitochondrial reads were retained.

The 32-patient cohort was used for initial analysis. CD8+ T cell subsets were defined in silico based on protein expression differently for each sorting strategy. PBMC samples were subset on CD3+CD8+CD4, CD3+ T cell sorted samples were subset on CD8+CD4, and non-naïve T cell sorted samples were subset on CD8+CD4. Non-naïve CD8+ T cell sorted samples were included as-is. Naïve cells were removed from PBMC and CD3+ T cell sorts by Azimuth reference mapping.

The standard Seurat workflow was performed separately for the CD3+ T cell sort and all other sorts: NormalizeData, FindVariableFeatures, ScaleData, RunPCA, RunHarmony, FindNeighbors, FindClusters, and RunUMAP. Leftover NK, MAIT, and monocytes were removed by unsupervised clustering and verification with Azimuth. Cells with high TCR-Va7.2 protein expression were excluded. Unsupervised clustering was used to identify clusters with low quality scores, which were removed.

A reference map was created with clean CD3+ T cell sort samples (Figure S2), using Harmony batch correction on patient, timepoint, and sort variables. For dimension reduction, ribosomal genes were excluded. The top 30 PCs were used for Harmony batch correction, and the first 10 Harmony dimensions were used for downstream analysis.

Unsupervised clustering was iteratively performed on the CD3+ T cell sort samples at different resolutions. For each resolution, the object was downsampled to 2000 cells per cluster. The resulting UMI matrix was fed into Hotspot47, excluding genes expressed in less than 3 cells. The Hotspot algorithm was run using the top 10 Harmony embeddings as distance metric and subsequently on the top 3000 informative genes. Gene modules were created using agglomerative clustering with a minimum of 200 genes per module and FDR less than 0.05. Overrepresentation analysis was performed on the top 250 genes of each module, ranked by Z autocorrelation score using a hypergeometric test against the ConsensusPathDB database (http://cpdb.molgen.mpg.de/) with Bonferroni correction. Metaclusters were biologically defined as described in the text. All other samples, including the treatment-naïve αPD-1 patients and healthy donors, were mapped onto the reference map with Symphony46 to create the final non-naïve CD8+ T cell atlas. Cluster labels were transferred to the query using the k-nearest neighbors prediction function in Symphony.

Gene module expression was calculated for each cell using the AddModuleScore function in Seurat, and a final clustering resolution of 0.5 was chosen. Differentially expressed geens were calculated using the FindAllMarkers function in Seurat using default values: a log2 fold change threshold of 0.25 and an adjusted p-value of less than 0.01.

Gene lists and defining exhausted CD8+ T cells

Bulk RNA-seq data from sorted CD8+ T cell subsets were obtained from a previously published paper53. Differential expression analysis one-vs-all for each subset was performed. Patient ID and batch was added as a covariate to the design formula along with its intercept in the linear model in DESeq2. The top 250 genes, ranked by p-value, were chosen for each cell subset.

Seurat’s AddModuleScore was used to calculate an expression score for every cell. The top 3% of cells by exhaustion score were defined as TEX.

Defining Progenitor TEX in scRNA-seq data

Imputation was performed using ALRA97 on the TEX subset to smooth the central memory score mapped onto cells due to dropouts, and progenitor TEX was defined as the top 1/3 of cells by central memory score.

Cyclone and TCR trajectory analyses

Only immunotherapy-naïve patients were included for analysis, in addition to the cohort of αPD-1 patients who received prior αCTLA-4 therapy with ipilimumab. One subject (14-1189_αCTLA-4) on αCTLA-4 was excluded from this analysis because of massive clonal expansion after checkpoint blockade that resulted in a distinctive clonal trajectory.

Only cells with both productive TCR alpha (TRA) and beta (TRB) chains were included (58.7% of CD8+ T cells), with clonotypes defined by the combination of patient ID, TRA, and TRB chain nucleotide sequences. Only TCR clonotypes with an absolute change of magnitude at least 0.25% and a fold change of greater than 2 or less than 0.5 in a 3-week interval were included. Clonotypes with less than 5 cells at every timepoints were filtered out because frequency variance is high with low counts due to Poisson sampling effects.

Increasing clonotypes with fold change greater than 2 were also required to have a peak frequency at a timepoint other than at baseline for inclusion. On the other hand, decreasing clonotypes with fold change of less than 0.5 were required to have a peak frequency at baseline. Pairwise Pearson correlation between each clonotype was calculated based on frequency at each timepoint, and trajectories were created using hierarchical clustering of pairwise Euclidean distances of correlation vectors.

The cell state composition of each trajectory was calculated by breaking down the underlying cell states contained within the cells in a trajectory at each timepoint for each patient, where the frequency is out of all cells in each respective patient sample. In other words, the sum of cell state frequencies for a trajectory at a timepoint equals the total frequency of that trajectory at that timepoint for each patient. Fold change of cell state frequencies were calculated with a 2% regularization term to prevent infinite fold changes. The average fold change across all patients was calculated for plotting.

HLA typing

HLA typing was performed by the University of Pennsylvania Histocompatibility and Immunogenetics Laboratory using one or more of the following molecular methods: SBT, SSP, SSO, RT-PCR, NGS.

Antigen-specific CD8+ T cell trajectory clustering

Unique antigen-specificities for each patient were treated as individual trajectories, which were binned into 4 patterns by the timepoint of maximum frequency (eg Traj 1: week 0, Traj 2: week 3, Traj 3: week 6, Traj 4: week 9+). In order to discover antigen-specific T cell populations that do not change over time, no filtering was performed based on fold change or absolute change as done in Cyclone; however, antigen-specificities with a fold change of less than 1.5 between week 0 and week 3 were binned into Traj 1.

Bulk TCR sequencing

Manual macrodissection was performed on FFPE slides, if necessary, using a scalpel and a slide stained with haematoxylin and eosin (H&E) as a guide. Tissue deparaffinization and DNA extraction were performed using standard methods. DNA amplification, library preparation, sequencing, and preliminary bioinformatics analysis was performed by Adaptive Biotechnologies. Amplification and sequencing of TCRB CDR3 was performed at single-level (ultra-deep) resolution using the immunoSEQ Platform (Adaptive Biotechnologies).

Tumor TCR trajectory clustering

Only productive TCRs were included. TCR beta chain CDR3 amino acid sequences were mapped onto the main single-cell sequencing study by exact match, for each of the 10 patients with bulk TCR data. Clones were binned by the timepoint of maximum frequency.

Pseudotime analysis

Pseudotime analysis was performed using Monocle 3 on the TEX subset, retaining the original UMAP embeddings. Default parameters were used, except for use_partition set to FALSE, close_loop set to FALSE, and a minimal branch length of 8. Central memory-like cells were chosen as the root. A spline curve with 3 degrees of freedom was fit on gene and protein expression across pseudotime. Only the CD3+ sort was used for the protein expression analysis to reduce batch effects because the other sorts used a different ADT panel.

Blood-tumor atlas construction

Cell types were assigned by Azimuth reference mapping, and CD8+ T cells were subset in silico. The standard Seurat workflow was performed: NormalizeData, FindVariableFeatures, ScaleData, RunPCA, RunHarmony, FindNeighbors, FindClusters, and RunUMAP. Samples were batch corrected using Harmony by patient. Circulating exhausted cells from the main study were mapped onto the blood-tumor atlas using Symphony.

Flow cytometry

For samples processed at Penn, cryopreserved PBMC samples were thawed and stained with master mix of antibodies for surface stains including CD4 (Biolegend, SK3), CD8 (ebioscience, RPA-T8), CD45RA (Biolegend, HI100), Tim3 (F38-2E2), IgG4 (Southern Biotech, HP6025), CD39 (Biolegend, A1), CD3 (BD, UCHT1), CD127 (Biolegend, A019D5), CCR7 (Biologend, G043H7), CD28 (BD, CD28.2), CD226 (BD, DX11), CD73 (Biolegend, AD2), TIGIT (Biolegend, VSTM3) and CD27 (BD, L128) and intracellular stains for FoxP3 (Invitrogen, PCH101), CTLA4 (BD, BNI3), Eomes (Invitrogen, WD1928), Tbet (Biolegend, 4B10), Tox (Miltenyi, REA473), TCF-1 (Cell Signaling and Technology, C63D9), and Ki67(BD, B56). Permeabilization was performed using the Foxp3 Fixation/Permeabilization Concentrate and Diluent kit (eBioscience). Cells were resuspended in 1% para-formaldehyde until acquisition on a BD Biosciences LSR II cytometer or Symphony A5.

For samples stained with combinatorial tetramer panels at Penn, cryopreserved PBMC samples were thawed and stained with fluorophore-conjugated tetramers, followed by master mix of antibodies for surface stains, including CD45RA (BD Biosciences, HI100), Blue-Fluorescent Reactive Dye (Invitrogen), CD8 (BD Biosciences, RPA-T8), CD69 (BD Biosciences, FN50), CD226 (BD Biosciences, DX11), CXCR6 (BD Biosciences, 13B 1E5), CD28 (BD Biosciences, CD28.2), PD-1 (BioLegend, EH12.2H7), CD161 (BD Biosciences. DX12), CD14 (BD Biosciences, M5E2), CD19 (BD Biosciences, HIB19), CD41a (BD Biosciences, HIP8), CD4 (BioLegend, SK3), CD127 (BioLegend, A019D5), CCR7 (BioLegend, G043H7), CXCR5 (BD Biosciences, RF8B2), TIGIT (Invitrogen, MBSA43), CX3CR1 (BD Biosciences, 2A9-1), CD103 (BioLegend, Ber-ACT8), TIM-3 (BioLegend, F38-2E2), CD39 (BioLegend, A1), and CD27 (BioLegend, QA17A18) and intracellular stains for LAG-3 (BD Biosciences, T47-530), Ki-67 (BioLegend, Ki-67), CD3 (BioLegend, SK7), CTLA-4 (BioLegend, BNI3), FOXP3 (Invitrogen, PCH101), TBET (BioLegend, 4B10), TOX (Miltenyl Biotec, REA473), and TCF-7/TCF-1 (BD Biosciences, S33-966). Permeabilized was performed using the Foxp3 / Transcription Factor Staining Buffer Set (eBioscience). Cells were resuspension in 1% para-formaldehyde (Electron Microscopy Sciences) until acquisition on a five-laser Aurora cytometer (Cytek Biosciences) and unmixed using non-staining controls and single-color reference libraries.

For samples processed at NYU (Checkmate 238 trial), following thaw, PBMC underwent Fc blockade with Human TruStain FcX (BioLegend) and NovaBlock (Thermo Fisher Scientific) for 10 min at room temperature, followed by surface antibody staining for 20 min at room temperature in the dark. Cells were then permeabilized using the eBioscience Foxp3/Transcription Factor Staining Buffer Set (ThermoFisher) for 20 minutes at room temperature in the dark. Following permeabilization, cells were intracellularly stained for 1 hour at room temperature in the dark, followed by resuspension in 1% para-formaldehyde (Electron Microscopy Sciences). All samples were acquired on a five-laser Aurora cytometer (Cytek Biosciences).

All flow cytometry was analyzed using FlowJo (Becton Dickinson).

Quantification and statistical analysis

All statistical analyses were carried out using base R. Wilcoxon signed-rank test was used for paired comparisons. Wilcoxon rank-sum test was used for unpaired comparisons. Two-sided tests were used for all statistical comparisons. Statistical significance was as follows: p < 0.05 (*), 0.01 (**), 0.001 (***), 0.0001 (***). Error bars represent the 95th percentile confidence interval.

Supplementary Material

1
2

Table S2: Hotspot Module Genes, Pathways, and Human CD8+ T cells Gene Lists. Related to Figure 1.

Hotspot module genes: List of all genes for each of the 4 gene modules generated by Hotspot.

Hotspot module pathways: List of top enriched pathways in the 4 gene modules generated by Hotspot.

Human CD8+ T cell Gene Lists: List of top 250 differentially expressed genes for each CD8+ T cell subset defined by sorted cells from human healthy donors, as described in Star Methods.

3

Table S3: Tumor TCR sequences. Related to Figures 3 and S6.

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
PE/Fire 640 anti-human CD8 BioLegend Cat# 344761; RRID: AB_2860887; clone SK1
PerCP anti-human CD11c BioLegend Cat# 337234; RRID: AB_2566656; clone Bu15
BD Horizon BUV805 Mouse Anti-Human CD14 BD Biosciences Cat# 612902; clone M5E2
BD Horizon BUV496 Mouse Anti-Human CD19 BD Biosciences Cat# 612939; clone 612939
PE/Dazzle 594 anti-human CD20 BioLegend Cat# 302348; RRID: AB_2564387; clone 2H7
BD Pharmingen PE-Cy5 Mouse Anti-Human CD21 BD Biosciences Cat# 551064; RRID: AB_394028; clone B-ly4
BD OptiBuild BUV615 Mouse Anti-Human CD23 BD Biosciences Cat# 751104; RRID: AB_2875134; clone M-L233
BD Horizon BUV563 Mouse Anti-Human CD25 BD Biosciences Cat# 612919; RRID: AB_2870204; clone 2A3
Brilliant Violet 711 anti-human CD27 Antibody BioLegend Cat# 302834; RRID: AB_2563809; clone O323
BD Horizon BV480 Mouse Anti-Human CD28 BD Biosciences Cat# 566110; RRID: AB_2739512; clone CD28.2
APC/Fire 810 anti-human CD38 BioLegend Cat# 303549; RRID: AB_2860783; clone HIT2
Brilliant Violet 605 anti-human CD39 BioLegend Cat# 328236; RRID: AB_2750430; clone A1
Pacific Blue anti-human CD40 BioLegend Cat# 334319; RRID: AB_10612573; clone 5C3
Brilliant Violet 570 anti-human CD45RA BioLegend Cat# 304132; RRID: AB_2563813; clone HI100
Super Bright 780 CD71 (Transferrin Receptor) Invitrogen Cat# 78-0719-42; RRID: AB_2784898; clone OKT9 (OKT-9)
Spark YG 581 anti-human CD127 (IL-7Rα) BioLegend Cat# 351367; RRID: AB_2890778; clone A019D5
Brilliant Violet 510 anti-human CD138 (Syndecan-1) BioLegend Cat# 356517; RRID: AB_2562661; clone MI15
Brilliant Violet 421 anti-human CD152 (CTLA-4) BioLegend Cat# 369606; RRID: AB_2616795; clone BNI3
BD OptiBuild BV750 Mouse Anti-Human CD183 (CXCR3) BD Biosciences Cat# 746895; RRID: AB_2871692; clone 1C6/CXCR3
BD Horizon APC-R700 Rat Anti-Human CXCR5 (CD185) BD Biosciences Cat# 565191; RRID: AB_2739103; clone RF8B2
Brilliant Violet 650 anti-human CD197 (CCR7) BioLegend Cat# 353234; RRID: AB_2563867; clone G043H7
APC/Fire 750 anti-human/mouse/rat CD278 (ICOS) BioLegend Cat# 313536; RRID: AB_2632923; clone C398.4A
BD Horizon BB700 Mouse Anti-Human CD279 (PD-1) BD Biosciences Cat# 566460; RRID: AB_2744348; clone EH12.1
BD Horizon BUV661 Mouse Anti-Human HLA-DR BD Biosciences Cat# 612980; RRID: AB_2870252; clone G46-6
BD Horizon BUV737 Mouse Anti-Human IgD BD Biosciences Cat# 612798; RRID: AB_2870125; clone IA6-2
REAfinity PE anti-human/mouse TOX Miltenyi Biotec Cat# 130-120-716; RRID: AB_2801780; clone REA473
TCF1/TCF7 (C63D9) Rabbit mAb (Alexa Fluor® 647 Conjugate) Cell Signaling Cat# 6709S; RRID: AB_2797631; clone C63D9
BD Horizon BUV395 Mouse Anti-Ki-67 BD Biosciences Cat# 564071; RRID: AB_2738577; clone B56
Anti-human IgG Miltenyi Biotec Cat# 130-119-880; RRID: AB_2784374; clone IS11-3B2.2.3
Mouse Anti-Human IgG4 pFc'-FITC SouthernBiotech Cat# 9190-02; RRID: AB_2796682; clone HP6023
BD Horizon BUV395 Mouse Anti-Human CD45RA BD Biosciences Cat# 568712; clone HI100
LIVE/DEAD Fixable Blue Dead Cell Stain Kit, for UV excitation Invitrogen Cat# L23105; RRID: AB_2796682
BD Horizon BUV496 Mouse Anti-Human CD8 BD Biosciences Cat# 612942; clone RPA-T8
BD OptiBuild BUV563 Mouse Anti-Human CD69 BD Biosciences Cat# 748764; RRID: AB_2873167; clone FN50
BD OptiBuild BUV661 Mouse Anti-Human CD226 BD Biosciences Cat# 749934; RRID: AB_2874171; clone DX11
BD OptiBuild BUV737 Mouse Anti-Human CXCR6 (CD186) BD Biosciences Cat# 748449; RRID: AB_2872865; clone 13B 1E5
BD OptiBuild BUV805 Mouse Anti-Human CD28 BD Biosciences Cat# 742037; RRID: AB_2871331; clone CD28.2
BD Horizon BV421 Mouse Anti-Human CD279 (PD-1) BD Biosciences Cat# 565935; RRID: AB_11153482; clone EH12.1
BD OptiBuild BV480 Mouse Anti-Human CD161 BD Biosciences Cat# 746305; RRID: AB_2743630; clone DX12
BD Horizon V500 Mouse Anti-Human CD14 BD Biosciences Cat# 562693; RRID: AB_2737727; clone MφP9
BD Horizon V500 Mouse anti-Human CD19 BD Biosciences Cat# 561125; RRID: AB_10562391; clone HIB19
D Horizon BV510 Mouse Anti-Human CD41a BD Biosciences Cat# 563250; RRID: AB_2738096; clone HIP8
Spark Violet 538 anti-human CD4 Antibody BioLegend Cat# 344673; RRID: AB_2890774; clone SK3
Brilliant Violet 570 anti-human CD127 (IL-7Rα) Antibody BioLegend Cat# 351308; RRID: AB_2832685 ; clone A019D5
Brilliant Violet 750 anti-human CD197 (CCR7) Antibody BioLegend Cat# 353253; RRID: AB_2800945; clone G043H7
BD Horizon BB700 Rat Anti-Human CXCR5 (CD185) BD Biosciences Cat# 566469; RRID: AB_2869769; clone RF8B2
TIGIT Monoclonal Antibody (MBSA43), PerCP-eFluor 710, eBioscience Invitrogen Cat# 46-9500-42; RRID: AB_10853679; clone MBSA43
BD OptiBuild BB790 Rat Anti-Human CX3CR1 BD Biosciences Custom design; clone 2A9-1
PE/Fire 640 anti-human CD103 (Integrin αE) Antibody BioLegend Cat# 350243; RRID: AB_2924550 ; clone Ber-ACT8
PE/Cyanine5 anti-human CD366 (Tim-3) Antibody BioLegend Cat# 345052; RRID: AB_2819987 ; clone F38-2E2
APC/Fire 750 anti-human CD39 Antibody BioLegend Cat# 328229; RRID: AB_2650838 ; clone A1
APC/Fire 810 anti-human CD27 Recombinant Antibody BioLegend Cat# 393213; RRID: AB_2860961; clone QA17A18
BD OptiBuild BUV615 Mouse Anti-Human LAG-3 (CD223) BD Biosciences Cat# 752362; RRID: AB_2875879; clone T47-530
Brilliant Violet 711 anti-human Ki-67 Antibody BioLegend Cat# 350516; RRID: AB_2563861; clone Ki-67
Spark Blue 550 anti-human CD3 Antibody BioLegend Cat# 344852; RRID: AB_2819985; clone SK7
PE/Dazzle 594 anti-human CD152 (CTLA-4) Antibody BioLegend Cat# 369616; RRID: AB_2632878; clone BNI3
TOX Antibody, anti-human/mouse, APC, REAfinity Miltenyi Biotec Cat# 130-118-335; clone REA473
BD Horizon R718 Mouse Anti-TCF-7/TCF-1 BD Biosciences Cat# 567587; clone S33-966
Brilliant Violet 570 anti-human CD3 Antibody BioLegend Cat# 300435; RRID: AB_10898117; clone UCHT1
PE anti-mouse TIGIT (Vstm3) Antibody BioLegend Cat# 142103; RRID: AB_10895760; clone 1G9
BD Horizon BUV737 Mouse Anti-Human CD27 BD Biosciences Cat# 612829; RRID: AB_2870151; clone L128
EOMES Monoclonal Antibody (WD1928), PE-eFluor 610, eBioscience Invitrogen Cat# 61-4877-42; clone WD1928
BD Pharmingen Alexa Fluor® 700 Mouse anti-Ki-67 BD Biosciences Cat# 561277; RRID: AB_10611571; clone B56
Spark Violet 538 anti-human CD4 BioLegend Cat# 344673; RRID: AB_2890774; clone SK3
PE/Fire 640 anti-human CD8 BioLegend Cat# 344761; RRID: AB_2860887; clone SK1
PerCP anti-human CD11c BioLegend Cat# 337234; RRID: AB_2566656; clone Bu15
BD Horizon BUV805 Mouse Anti-Human CD14 BD Biosciences Cat# 612902; clone M5E2
BD Horizon BUV496 Mouse Anti-Human CD19 BD Biosciences Cat# 612939; clone 612939
PE/Dazzle 594 anti-human CD20 BioLegend Cat# 302348; RRID: AB_2564387; clone 2H7
BD Pharmingen PE-Cy5 Mouse Anti-Human CD21 BD Biosciences Cat# 551064; RRID: AB_394028; clone B-ly4
BD OptiBuild BUV615 Mouse Anti-Human CD23 BD Biosciences Cat# 751104; RRID: AB_2875134; clone M-L233
BD Horizon BUV563 Mouse Anti-Human CD25 BD Biosciences Cat# 612919; RRID: AB_2870204; clone 2A3
Brilliant Violet 711 anti-human CD27 Antibody BioLegend Cat# 302834; RRID: AB_2563809; clone O323
BD Horizon BV480 Mouse Anti-Human CD28 BD Biosciences Cat# 566110; RRID: AB_2739512; clone CD28.2
APC/Fire 810 anti-human CD38 BioLegend Cat# 303549; RRID: AB_2860783; clone HIT2
Brilliant Violet 605 anti-human CD39 BioLegend Cat# 328236; RRID: AB_2750430; clone A1
Pacific Blue anti-human CD40 BioLegend Cat# 334319; RRID: AB_10612573; clone 5C3
Brilliant Violet 570 anti-human CD45RA BioLegend Cat# 304132; RRID: AB_2563813; clone HI100
Super Bright 780 CD71 (Transferrin Receptor) Invitrogen Cat# 78-0719-42; RRID: AB_2784898; clone OKT9 (OKT-9)
Spark YG 581 anti-human CD127 (IL-7Rα) BioLegend Cat# 351367; RRID: AB_2890778; clone A019D5
Brilliant Violet 510 anti-human CD138 (Syndecan-1) BioLegend Cat# 356517; RRID: AB_2562661; clone MI15
Brilliant Violet 421 anti-human CD152 (CTLA-4) BioLegend Cat# 369606; RRID: AB_2616795; clone BNI3
BD OptiBuild BV750 Mouse Anti-Human CD183 (CXCR3) BD Biosciences Cat# 746895; RRID: AB_2871692; clone 1C6/CXCR3
BD Horizon APC-R700 Rat Anti-Human CXCR5 (CD185) BD Biosciences Cat# 565191; RRID: AB_2739103; clone RF8B2
Brilliant Violet 650 anti-human CD197 (CCR7) BioLegend Cat# 353234; RRID: AB_2563867; clone G043H7
APC/Fire 750 anti-human/mouse/rat CD278 (ICOS) BioLegend Cat# 313536; RRID: AB_2632923; clone C398.4A
BD Horizon BB700 Mouse Anti-Human CD279 (PD-1) BD Biosciences Cat# 566460; RRID: AB_2744348; clone EH12.1
BD Horizon BUV661 Mouse Anti-Human HLA-DR BD Biosciences Cat# 612980; RRID: AB_2870252; clone G46-6
BD Horizon BUV737 Mouse Anti-Human IgD BD Biosciences Cat# 612798; RRID: AB_2870125; clone IA6-2
PE-Cyanine5.5 FOXP3 Invitrogen Cat# 35-4776-42; RRID: AB_11218682; clone PCH101
REAfinity PE anti-human/mouse TOX Miltenyi Biotec Cat# 130-120-716; RRID: AB_2801780; clone REA473
TCF1/TCF7 (C63D9) Rabbit mAb (Alexa Fluor® 647 Conjugate) Cell Signaling Cat# 6709S; RRID: AB_2797631; clone C63D9
BD Horizon BUV395 Mouse Anti-Ki-67 BD Biosciences Cat# 564071; RRID: AB_2738577; clone B56
Anti-human IgG Miltenyi Biotec Cat# 130-119-880; RRID: AB_2784374; clone IS11-3B2.2.3
Mouse Anti-Human IgG4 pFc'-FITC SouthernBiotech Cat# 9190-02; RRID: AB_2796682; clone HP6023
BD Horizon BUV395 Mouse Anti-Human CD45RA BD Biosciences Cat# 568712; clone HI100
LIVE/DEAD Fixable Blue Dead Cell Stain Kit, for UV excitation Invitrogen Cat# L23105; RRID: AB_2796682
BD Horizon BUV496 Mouse Anti-Human CD8 BD Biosciences Cat# 612942; clone RPA-T8
BD OptiBuild BUV563 Mouse Anti-Human CD69 BD Biosciences Cat# 748764; RRID: AB_2873167; clone FN50
BD OptiBuild BUV661 Mouse Anti-Human CD226 BD Biosciences Cat# 749934; RRID: AB_2874171; clone DX11
BD OptiBuild BUV737 Mouse Anti-Human CXCR6 (CD186) BD Biosciences Cat# 748449; RRID: AB_2872865; clone 13B 1E5
BD OptiBuild BUV805 Mouse Anti-Human CD28 BD Biosciences Cat# 742037; RRID: AB_2871331; clone CD28.2
BD Horizon BV421 Mouse Anti-Human CD279 (PD-1) BD Biosciences Cat# 565935; RRID: AB_11153482; clone EH12.1
BD OptiBuild BV480 Mouse Anti-Human CD161 BD Biosciences Cat# 746305; RRID: AB_2743630; clone DX12
BD Horizon V500 Mouse Anti-Human CD14 BD Biosciences Cat# 562693; RRID: AB_2737727; clone MφP9
BD Horizon V500 Mouse anti-Human CD19 BD Biosciences Cat# 561125; RRID: AB_10562391; clone HIB19
D Horizon BV510 Mouse Anti-Human CD41a BD Biosciences Cat# 563250; RRID: AB_2738096; clone HIP8
Spark Violet 538 anti-human CD4 Antibody BioLegend Cat# 344673; RRID: AB_2890774; clone SK3
Brilliant Violet 570 anti-human CD127 (IL-7Rα) Antibody BioLegend Cat# 351308; RRID: AB_2832685 ; clone A019D5
Brilliant Violet 750 anti-human CD197 (CCR7) Antibody BioLegend Cat# 353253; RRID: AB_2800945; clone G043H7
BD Horizon BB700 Rat Anti-Human CXCR5 (CD185) BD Biosciences Cat# 566469; RRID: AB_2869769; clone RF8B2
TIGIT Monoclonal Antibody (MBSA43), PerCP-eFluor 710, eBioscience Invitrogen Cat# 46-9500-42; RRID: AB_10853679; clone MBSA43
BD OptiBuild BB790 Rat Anti-Human CX3CR1 BD Biosciences Custom design; clone 2A9-1
PE/Fire 640 anti-human CD103 (Integrin αE) Antibody BioLegend Cat# 350243; RRID: AB_2924550 ; clone Ber-ACT8
PE/Cyanine5 anti-human CD366 (Tim-3) Antibody BioLegend Cat# 345052; RRID: AB_2819987 ; clone F38-2E2
APC/Fire 750 anti-human CD39 Antibody BioLegend Cat# 328229; RRID: AB_2650838 ; clone A1
APC/Fire 810 anti-human CD27 Recombinant Antibody BioLegend Cat# 393213; RRID: AB_2860961; clone QA17A18
BD OptiBuild BUV615 Mouse Anti-Human LAG-3 (CD223) BD Biosciences Cat# 752362; RRID: AB_2875879; clone T47-530
Brilliant Violet 711 anti-human Ki-67 Antibody BioLegend Cat# 350516; RRID: AB_2563861; clone Ki-67
Spark Blue 550 anti-human CD3 Antibody BioLegend Cat# 344852; RRID: AB_2819985; clone SK7
PE/Dazzle 594 anti-human CD152 (CTLA-4) Antibody BioLegend Cat# 369616; RRID: AB_2632878; clone BNI3
FOXP3 Monoclonal Antibody (PCH101), PE-Cyanine5.5, eBioscience Invitrogen Cat# 35-4776-42; RRID: AB_11218682; clone PCH101
PE/Cyanine7 anti-T-bet Antibody BioLegend Cat# 644823; RRID: AB_2561760; clone 4B10
TOX Antibody, anti-human/mouse, APC, REAfinity Miltenyi Biotec Cat# 130-118-335; clone REA473
BD Horizon R718 Mouse Anti-TCF-7/TCF-1 BD Biosciences Cat# 567587; clone S33-966
Brilliant Violet 570 anti-human CD3 Antibody BioLegend Cat# 300435; RRID: AB_10898117; clone UCHT1
PE anti-mouse TIGIT (Vstm3) Antibody BioLegend Cat# 142103; RRID: AB_10895760; clone 1G9
BD Horizon BUV737 Mouse Anti-Human CD27 BD Biosciences Cat# 612829; RRID: AB_2870151; clone L128
EOMES Monoclonal Antibody (WD1928), PE-eFluor 610, eBioscience Invitrogen Cat# 61-4877-42; clone WD1928
TCF1/TCF7 (C63D9) Rabbit mAb (Alexa Fluor® 647 Conjugate) Cell Signaling and Technology Cat# 6709S; clone C63D9
BD Pharmingen Alexa Fluor® 700 Mouse anti-Ki-67 BD Biosciences Cat# 561277; RRID: AB_10611571; clone B56
APC/Cyanine7 anti-human CD3 Antibody BioLegend Cat# 344817; RRID: AB_10644011; clone SK7
PerCP anti-human CD4 Antibody BioLegend Cat# 344623; RRID: AB_2563325; clone SK3
Alexa Fluor® 700 anti-human CD8 Antibody BioLegend Cat# 344723; RRID: AB_2562789; clone SK1
FITC anti-human CD45RA Antibody BioLegend Cat# 304105; RRID: AB_314409; clone HI100
PE anti-human CD197 (CCR7) Antibody BioLegend Cat# 353203; RRID: AB_10916391; clone G043H7
Custom TotalSeq-C Human Cocktail BioLegend Custom
Biological Samples
Human healthy donor PBMC Human Immunology Core, University of Pennsylvania N/A
Patient PBMC and Tumor University of Pennsylvania IRB
Chemicals, Peptides, and Recombinant Proteins
CMV pp65 (YSE) YSEHPTFTSQY GenScript Custom peptide synthesis
FluNP (CTE) CTELKLSDY GenScript Custom peptide synthesis
CMV pp50 (VTE) VTEHDTLLY GenScript Custom peptide synthesis
Tyrosinase (SSD) SSDYVIPIGTY GenScript Custom peptide synthesis
MAGE A1 (EAD) EADPTGHSY GenScript Custom peptide synthesis
MAGE A3 (EVD) EVDPIGHLY GenScript Custom peptide synthesis
AIM2 (RSD) RSDSGQQARY GenScript Custom peptide synthesis
N-RAS Q61R (ILD) ILDTAGREEY GenScript Custom peptide synthesis
CMV pp65 (NLV) NLVPMVATV GenScript Custom peptide synthesis
MART-1 (ELA) ELAGIGILTV GenScript Custom peptide synthesis
gp100 280-9V (YLE) YLEPGPVTV GenScript Custom peptide synthesis
gp100 209-2M (IMD) IMDQVPFSV GenScript Custom peptide synthesis
gp100 (KTW) KTWGQYWQV GenScript Custom peptide synthesis
GnTV (VLP) VLPDVFIRCV GenScript Custom peptide synthesis
LAGE-1 (MLM) MLMAQEALAFL GenScript Custom peptide synthesis
SSX-2 (KAS) KASEKIFYV GenScript Custom peptide synthesis
NY-ESO-1 (SLL) SLLMWITQA GenScript Custom peptide synthesis
PRDX5 (AMA) AMAPIKVRL GenScript Custom peptide synthesis
CMV pp50 (TVR) TVRSHCVSK GenScript Custom peptide synthesis
Flu M1 (SII) SIIPSGPLK GenScript Custom peptide synthesis
EBV BRLF1 (RVR) RVRAYTYSK GenScript Custom peptide synthesis
EBV EBNA 3A (RLR) RLRAEAQVK GenScript Custom peptide synthesis
gp100 (ALL) ALLAVGATK GenScript Custom peptide synthesis
gp100 (LIY) LIYRRRLMK GenScript Custom peptide synthesis
gp100 (ALN) ALNFPGSQK GenScript Custom peptide synthesis
TAG (RLS) RLSNRLLLR GenScript Custom peptide synthesis
MAGE A1 (SLF) SLFRAVIIK GenScript Custom peptide synthesis
Flex-T HLA-A*01:01 Monomer UVX BioLegend Cat# 280001
Flex-T HLA-A*02:01 Monomer UVX BioLegend Cat# 280003
Flex-T HLA-A*03:01 Monomer UVX BioLegend Cat# 280005
Flex-T HLA-B*07:02 Monomer UVX BioLegend Cat# 280009
Critical Commercial Assays
eBioscience Foxp3 / Transcription Factor Staining Buffer Set Invitrogen Cat# 00-5523-00
Next GEM Chip K 10x Genomics Cat# PN-1000286
Next GEM Single Cell 5' Kit v2 10x Genomics Cat# PN-1000263
Chromium Single Cell V(D)J Enrichment Kit, Human T Cell 10x Genomics Cat# PN-1000005
Chromium Single Cell 5' Feature Barcode Library Kit 10x Genomics Cat# PN-1000080
NovaSeq 6000 S4 Reagent Kit v1.5 Illumina Cat# 20028312
Deposited Data
This work (scRNA/TCR/ADT-seq of main 32-patient study) NIH GEO GSE272993
This work (scRNA/TCR-seq of treatment-naive patients on aPD-1) NIH GEO GSE272734
This work (scRNA/TCR-seq of healthy donors) NIH GEO GSE272735
This work (scRNA-seq of paired blood-tumor atlas) NIH GEO GSE273718
Software and Algorithms
R R Version 4.3.1
Python Python Version 3.9.13
FlowJo BD Biosciences Version 10.10.0
CellRanger 10x Genomics Version 7.0.0
Cyclone This paper 10.5281/zenodo.12754871
Seurat https://github.com/jiwen90/seurat Version 4.3.0
Other
CD8 Human Gene Lists, see Table S2 This paper N/A
Bulk TCR-seq of TILs, see Table S3 This paper N/A
Multi-database gene pathways http://cpdb.molgen.mpg.de/ N/A

Highlights.

  • A novel algorithm Cyclone uncovers patterns of clonal trajectories across time

  • Immune checkpoint blockade induces distinct waves of clonal responses after each dose

  • Melanoma-specific CD8+ T cell responses are present at 3 and 6 weeks after treatment

  • CTLA-4 blockade induces robust progenitor-exhausted CD8+ T cell responses

Acknowledgements

This work was supported by National Institutes of Health grants K08 CA230157 (ACH), R01 CA273018 (ACH), RO1CA258113 (XX), P50 CA174523 (ACH), P50CA261608 (ACH, GCK, XX, RKA), P30CA016520 (ACH), R01CA244936 (RSH and JSW). P50CA225450 (JSW and RSH) including a Career Enhancement Program award for RSH, P30CA016087 (RSH).

In addition, ACH was supported by Damon Runyon Clinical Investigator Award, Doris Duke Clinical Scientist Development Award, the W.W. Smith Charitable Trust Award, and the Institute for Translational Medicine and Therapeutics of the Perelman School of Medicine, and the Pew-Stewart Stewart Scholars Program in Cancer Research. C.A. was funded by the Parker Institute for Cancer Immunotherapy. Work in the Huang Lab is funded by the Tara Miller Foundation and the Parker Institute for Cancer Immunotherapy. We thank John Haanen for contribution of tumor specimens. We are grateful to the NIH Tetramer Core for providing biotinylated UV cleavable monomers. We thank E. John Wherry for his continual advice and support. ACH thanks Beatriz Carreno, Gerald Linette, Tara Mitchell, E. John Wherry, Ivan Maillard, and Jesus Christ for making me the investigator I am today.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of interests

ACH performed consulting work for Immunai and received research funding from Bristol Myers Squibb and Merck. RSH has performed consulting work for Bristol Myers Squibb (exclusive of the current work). TCM received honorarium for Scientific Advisory Board participation from: BMS, GigaGen, Merck, Pliant, Pfizer. GCK is on the Merck Advisory Board. JW consulted for and have received less than $10,000 per annum from Merck, Genentech, AstraZeneca, GSK, Novartis, Nektar, Celldex, Incyte, Biond, Moderna, ImCheck, Sellas, Evaxion, Pfizer, Regeneron, and EMD Serono and received $10-$25,000 from BMS for membership on advisory boards. JW also holds equity in Biond, Evaxion, OncoC4, and Instil Bio, and on scientific advisory boards for CytomX, Incyte, ImCheck, Biond, Sellas, Instil Bio, OncoC4, and NexImmune and am remunerated between $10,000-$50,000. In addition, JW is named on a patent filed by Moffitt Cancer Center on an ipilimumab biomarker and on TIL preparation, and also on a PD-1 patent filed by Biodesix; JW receives less than $6000 in royalties. DB, EK, and CS were employed by Immunai when engaged in this project. SG and DT are employees of BMS.

References

  • 1.Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, Cowey CL, Schadendorf D, Wagstaff J, Dummer R, et al. (2019). Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med 381, 1535–1546. 10.1056/NEJMoa1910836. [DOI] [PubMed] [Google Scholar]
  • 2.Wei SC, Duffy CR, and Allison JP (2018). Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov 8, 1069–1086. 10.1158/2159-8290.CD-18-0367. [DOI] [PubMed] [Google Scholar]
  • 3.Brunner MC, Chambers CA, Chan FK, Hanke J, Winoto A, and Allison JP (1999). CTLA-4-Mediated inhibition of early events of T cell proliferation. J Immunol 162, 5813–5820. [PubMed] [Google Scholar]
  • 4.Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, Thompson CB, and Bluestone JA (1994). CTLA-4 can function as a negative regulator of T cell activation. Immunity 1, 405–413. 10.1016/1074-7613(94)90071-x. [DOI] [PubMed] [Google Scholar]
  • 5.Linsley PS, Brady W, Urnes M, Grosmaire LS, Damle NK, and Ledbetter JA (1991). CTLA-4 is a second receptor for the B cell activation antigen B7. J Exp Med 174, 561–569. 10.1084/jem.174.3.561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Linsley PS, Greene JL, Brady W, Bajorath J, Ledbetter JA, and Peach R (1994). Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1, 793–801. 10.1016/s1074-7613(94)80021-9. [DOI] [PubMed] [Google Scholar]
  • 7.van der Merwe PA, Bodian DL, Daenke S, Linsley P, and Davis SJ (1997). CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J Exp Med 185, 393–403. 10.1084/jem.185.3.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Krummel MF, and Allison JP (1995). CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 182, 459–465. 10.1084/jem.182.2.459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bachmann MF, Kohler G, Ecabert B, Mak TW, and Kopf M (1999). Cutting edge: lymphoproliferative disease in the absence of CTLA-4 is not T cell autonomous. J Immunol 163, 1128–1131. [PubMed] [Google Scholar]
  • 10.Friedline RH, Brown DS, Nguyen H, Kornfeld H, Lee J, Zhang Y, Appleby M, Der SD, Kang J, and Chambers CA (2009). CD4+ regulatory T cells require CTLA-4 for the maintenance of systemic tolerance. J Exp Med 206, 421–434. 10.1084/jem.20081811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Peggs KS, Quezada SA, Chambers CA, Korman AJ, and Allison JP (2009). Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med 206, 1717–1725. 10.1084/jem.20082492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Read S, Greenwald R, Izcue A, Robinson N, Mandelbrot D, Francisco L, Sharpe AH, and Powrie F (2006). Blockade of CTLA-4 on CD4+CD25+ regulatory T cells abrogates their function in vivo. J Immunol 177, 4376–4383. 10.4049/jimmunol.177.7.4376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Leach DR, Krummel MF, and Allison JP (1996). Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736. 10.1126/science.271.5256.1734. [DOI] [PubMed] [Google Scholar]
  • 14.Tarhini A, Lin Y, Lin H, Rahman Z, Vallabhaneni P, Mendiratta P, Pingpank JF, Holtzman MP, Yusko EC, Rytlewski JA, et al. (2018). Neoadjuvant ipilimumab (3 mg/kg or 10 mg/kg) and high dose IFN-alpha2b in locally/regionally advanced melanoma: safety, efficacy and impact on T-cell repertoire. J Immunother Cancer 6, 112. 10.1186/s40425-018-0428-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cha E, Klinger M, Hou Y, Cummings C, Ribas A, Faham M, and Fong L (2014). Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med 6, 238ra270. 10.1126/scitranslmed.3008211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Robert L, Tsoi J, Wang X, Emerson R, Homet B, Chodon T, Mok S, Huang RR, Cochran AJ, Comin-Anduix B, et al. (2014). CTLA4 blockade broadens the peripheral T-cell receptor repertoire. Clin Cancer Res 20, 2424–2432. 10.1158/1078-0432.CCR-13-2648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kvistborg P, Philips D, Kelderman S, Hageman L, Ottensmeier C, Joseph-Pietras D, Welters MJ, van der Burg S, Kapiteijn E, Michielin O, et al. (2014). Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med 6, 254ra128. 10.1126/scitranslmed.3008918. [DOI] [PubMed] [Google Scholar]
  • 18.Carthon BC, Wolchok JD, Yuan J, Kamat A, Ng Tang DS, Sun J, Ku G, Troncoso P, Logothetis CJ, Allison JP, and Sharma P (2010). Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin Cancer Res 16, 2861–2871. 10.1158/1078-0432.CCR-10-0569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chen H, Liakou CI, Kamat A, Pettaway C, Ward JF, Tang DN, Sun J, Jungbluth AA, Troncoso P, Logothetis C, and Sharma P (2009). Anti-CTLA-4 therapy results in higher CD4+ICOShi T cell frequency and IFN-gamma levels in both nonmalignant and malignant prostate tissues. Proc Natl Acad Sci U S A 106, 2729–2734. 10.1073/pnas.0813175106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso P, Logothetis C, and Sharma P (2008). CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U S A 105, 14987–14992. 10.1073/pnas.0806075105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ng Tang D, Shen Y, Sun J, Wen S, Wolchok JD, Yuan J, Allison JP, and Sharma P (2013). Increased frequency of ICOS+ CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol Res 1, 229–234. 10.1158/2326-6066.CIR-13-0020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sharma A, Subudhi SK, Blando J, Vence L, Wargo J, Allison JP, Ribas A, and Sharma P (2019). Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3(+) Regulatory T Cells (Tregs) in Human Cancers-Response. Clin Cancer Res 25, 3469–3470. 10.1158/1078-0432.CCR-19-0402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kavanagh B, O'Brien S, Lee D, Hou Y, Weinberg V, Rini B, Allison JP, Small EJ, and Fong L (2008). CTLA4 blockade expands FoxP3+ regulatory and activated effector CD4+ T cells in a dose-dependent fashion. Blood 112, 1175–1183. 10.1182/blood-2007-11-125435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wei SC, Sharma R, Anang NAS, Levine JH, Zhao Y, Mancuso JJ, Setty M, Sharma P, Wang J, Pe'er D, and Allison JP (2019). Negative Co-stimulation Constrains T Cell Differentiation by Imposing Boundaries on Possible Cell States. Immunity 50, 1084–1098 e1010. 10.1016/j.immuni.2019.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, et al. (2002). Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 8, 793–800. 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 26.Zou W, Wolchok JD, and Chen L (2016). PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med 8, 328rv324. 10.1126/scitranslmed.aad7118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson KL, Daniel B, Zimmermann MC, et al. (2003). Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med 9, 562–567. 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
  • 28.Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, and Gajewski TF (2013). Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med 5, 200ra116. 10.1126/scitranslmed.3006504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, Chen S, Klein AP, Pardoll DM, Topalian SL, and Chen L (2012). Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 4, 127ra137. 10.1126/scitranslmed.3003689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pauken KE, and Wherry EJ (2015). Overcoming T cell exhaustion in infection and cancer. Trends Immunol 36, 265–276. 10.1016/j.it.2015.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, and Ahmed R (2006). Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687. 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
  • 32.Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DA, and Wherry EJ (2009). Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 10, 29–37. 10.1038/ni.1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Paley MA, Kroy DC, Odorizzi PM, Johnnidis JB, Dolfi DV, Barnett BE, Bikoff EK, Robertson EJ, Lauer GM, Reiner SL, and Wherry EJ (2012). Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225. 10.1126/science.1229620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, Drake AM, Chen ZY, Sen DR, Kurachi M, et al. (2016). Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165. 10.1126/science.aaf2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MW, Yates KB, Lako A, Felt K, Naik GS, et al. (2019). Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 20, 326–336. 10.1038/s41590-019-0312-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ahn E, Araki K, Hashimoto M, Li W, Riley JL, Cheung J, Sharpe AH, Freeman GJ, Irving BA, and Ahmed R (2018). Role of PD-1 during effector CD8 T cell differentiation. Proc Natl Acad Sci U S A 115, 4749–4754. 10.1073/pnas.1718217115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Yamauchi T, Hoki T, Oba T, Jain V, Chen H, Attwood K, Battaglia S, George S, Chatta G, Puzanov I, et al. (2021). T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors. Nat Commun 12, 1402. 10.1038/s41467-021-21619-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kamada T, Togashi Y, Tay C, Ha D, Sasaki A, Nakamura Y, Sato E, Fukuoka S, Tada Y, Tanaka A, et al. (2019). PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci U S A 116, 9999–10008. 10.1073/pnas.1822001116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kumagai S, Togashi Y, Kamada T, Sugiyama E, Nishinakamura H, Takeuchi Y, Vitaly K, Itahashi K, Maeda Y, Matsui S, et al. (2020). The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat Immunol 21, 1346–1358. 10.1038/s41590-020-0769-3. [DOI] [PubMed] [Google Scholar]
  • 40.Huang AC, Orlowski RJ, Xu X, Mick R, George SM, Yan PK, Manne S, Kraya AA, Wubbenhorst B, Dorfman L, et al. (2019). A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med 25, 454–461. 10.1038/s41591-019-0357-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Huang AC, Postow MA, Orlowski RJ, Mick R, Bengsch B, Manne S, Xu W, Harmon S, Giles JR, Wenz B, et al. (2017). T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 545, 60–65. 10.1038/nature22079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS, Wieland A, Sica GL, Yu K, Koenig L, Patel NT, et al. (2017). Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A 114, 4993–4998. 10.1073/pnas.1705327114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kim KH, Cho J, Ku BM, Koh J, Sun JM, Lee SH, Ahn JS, Cheon J, Min YJ, Park SH, et al. (2019). The First-week Proliferative Response of Peripheral Blood PD-1(+)CD8(+) T Cells Predicts the Response to Anti-PD-1 Therapy in Solid Tumors. Clin Cancer Res 25, 2144–2154. 10.1158/1078-0432.CCR-18-1449. [DOI] [PubMed] [Google Scholar]
  • 44.Wei SC, Anang NAS, Sharma R, Andrews MC, Reuben A, Levine JH, Cogdill AP, Mancuso JJ, Wargo JA, Pe'er D, and Allison JP (2019). Combination anti-CTLA-4 plus anti-PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies. Proc Natl Acad Sci U S A 116, 22699–22709. 10.1073/pnas.1821218116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, Baglaenko Y, Brenner M, Loh PR, and Raychaudhuri S (2019). Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods 16, 1289–1296. 10.1038/s41592-019-0619-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kang JB, Nathan A, Weinand K, Zhang F, Millard N, Rumker L, Moody DB, Korsunsky I, and Raychaudhuri S (2021). Efficient and precise single-cell reference atlas mapping with Symphony. Nat Commun 12, 5890. 10.1038/s41467-021-25957-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.DeTomaso D, and Yosef N (2021). Hotspot identifies informative gene modules across modalities of single-cell genomics. Cell Syst 12, 446–456 e449. 10.1016/j.cels.2021.04.005. [DOI] [PubMed] [Google Scholar]
  • 48.Kaech SM, and Cui W (2012). Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol 12, 749–761. 10.1038/nri3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kaech SM, Wherry EJ, and Ahmed R (2002). Effector and memory T-cell differentiation: implications for vaccine development. Nat Rev Immunol 2, 251–262. 10.1038/nri778. [DOI] [PubMed] [Google Scholar]
  • 50.Foletta VC, Segal DH, and Cohen DR (1998). Transcriptional regulation in the immune system: all roads lead to AP-1. J Leukoc Biol 63, 139–152. 10.1002/jlb.63.2.139. [DOI] [PubMed] [Google Scholar]
  • 51.Crouse J, Kalinke U, and Oxenius A (2015). Regulation of antiviral T cell responses by type I interferons. Nat Rev Immunol 15, 231–242. 10.1038/nri3806. [DOI] [PubMed] [Google Scholar]
  • 52.McGettrick AF, and O'Neill LAJ (2020). The Role of HIF in Immunity and Inflammation. Cell Metab 32, 524–536. 10.1016/j.cmet.2020.08.002. [DOI] [PubMed] [Google Scholar]
  • 53.Giles JR, Manne S, Freilich E, Oldridge DA, Baxter AE, George S, Chen Z, Huang H, Chilukuri L, Carberry M, et al. (2022). Human epigenetic and transcriptional T cell differentiation atlas for identifying functional T cell-specific enhancers. Immunity 55, 557–574 e557. 10.1016/j.immuni.2022.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hashimoto M, Kamphorst AO, Im SJ, Kissick HT, Pillai RN, Ramalingam SS, Araki K, and Ahmed R (2018). CD8 T Cell Exhaustion in Chronic Infection and Cancer: Opportunities for Interventions. Annu Rev Med 69, 301–318. 10.1146/annurev-med-012017-043208. [DOI] [PubMed] [Google Scholar]
  • 55.Bengsch B, Johnson AL, Kurachi M, Odorizzi PM, Pauken KE, Attanasio J, Stelekati E, McLane LM, Paley MA, Delgoffe GM, and Wherry EJ (2016). 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. 10.1016/j.immuni.2016.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Vardhana SA, Hwee MA, Berisa M, Wells DK, Yost KE, King B, Smith M, Herrera PS, Chang HY, Satpathy AT, et al. (2020). Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat Immunol 21, 1022–1033. 10.1038/s41590-020-0725-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Martinez GJ, Pereira RM, Aijo T, Kim EY, Marangoni F, Pipkin ME, Togher S, Heissmeyer V, Zhang YC, Crotty S, et al. (2015). The transcription factor NFAT promotes exhaustion of activated CD8(+) T cells. Immunity 42, 265–278. 10.1016/j.immuni.2015.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H, Callahan M, Wolchok JD, Halaban R, Dhodapkar MV, and Dhodapkar KM (2015). Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol 194, 950–959. 10.4049/jimmunol.1401686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang K. (2024). Cyclone Algorithm for Tracking Clonal Trajectories Across Time. [Google Scholar]
  • 60.Beltra JC, Manne S, Abdel-Hakeem MS, Kurachi M, Giles JR, Chen Z, Casella V, Ngiow SF, Khan O, Huang YJ, et al. (2020). Developmental Relationships of Four Exhausted CD8(+) T Cell Subsets Reveals Underlying Transcriptional and Epigenetic Landscape Control Mechanisms. Immunity 52, 825–841 e828. 10.1016/j.immuni.2020.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.He R, Hou S, Liu C, Zhang A, Bai Q, Han M, Yang Y, Wei G, Shen T, Yang X, et al. (2016). Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 537, 412–428. 10.1038/nature19317. [DOI] [PubMed] [Google Scholar]
  • 62.Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, Hale JS, Lee J, Nasti TH, et al. (2016). Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421. 10.1038/nature19330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Krishna S, Lowery FJ, Copeland AR, Bahadiroglu E, Mukherjee R, Jia L, Anibal JT, Sachs A, Adebola SO, Gurusamy D, et al. (2020). Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 370, 1328–1334. 10.1126/science.abb9847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Siddiqui I, Schaeuble K, Chennupati V, Fuertes Marraco SA, Calderon-Copete S, Pais Ferreira D, Carmona SJ, Scarpellino L, Gfeller D, Pradervand S, et al. (2019). Intratumoral Tcf1(+)PD-1(+)CD8(+) T Cells with Stem-like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy. Immunity 50, 195–211 e110. 10.1016/j.immuni.2018.12.021. [DOI] [PubMed] [Google Scholar]
  • 65.Utzschneider DT, Charmoy M, Chennupati V, Pousse L, Ferreira DP, Calderon-Copete S, Danilo M, Alfei F, Hofmann M, Wieland D, et al. (2016). T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections. Immunity 45, 415–427. 10.1016/j.immuni.2016.07.021. [DOI] [PubMed] [Google Scholar]
  • 66.Daniel B, Yost KE, Hsiung S, Sandor K, Xia Y, Qi Y, Hiam-Galvez KJ, Black M, C JR, Shi Q, et al. (2022). Divergent clonal differentiation trajectories of T cell exhaustion. Nat Immunol 23, 1614–1627. 10.1038/s41590-022-01337-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Giles JR, Ngiow SF, Manne S, Baxter AE, Khan O, Wang P, Staupe R, Abdel-Hakeem MS, Huang H, Mathew D, et al. (2022). Shared and distinct biological circuits in effector, memory and exhausted CD8(+) T cells revealed by temporal single-cell transcriptomics and epigenetics. Nat Immunol 23, 1600–1613. 10.1038/s41590-022-01338-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Li H, van der Leun AM, Yofe I, Lubling Y, Gelbard-Solodkin D, van Akkooi ACJ, van den Braber M, Rozeman EA, Haanen J, Blank CU, et al. (2019). Dysfunctional CD8 T Cells Form a Proliferative, Dynamically Regulated Compartment within Human Melanoma. Cell 176, 775–789 e718. 10.1016/j.cell.2018.11.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Thommen DS, Koelzer VH, Herzig P, Roller A, Trefny M, Dimeloe S, Kiialainen A, Hanhart J, Schill C, Hess C, et al. (2018). A transcriptionally and functionally distinct PD-1(+) CD8(+) T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med 24, 994–1004. 10.1038/s41591-018-0057-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chow A, Perica K, Klebanoff CA, and Wolchok JD (2022). Clinical implications of T cell exhaustion for cancer immunotherapy. Nat Rev Clin Oncol 19, 775–790. 10.1038/s41571-022-00689-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Chen Y, Zander RA, Wu X, Schauder DM, Kasmani MY, Shen J, Zheng S, Burns R, Taparowsky EJ, and Cui W (2021). BATF regulates progenitor to cytolytic effector CD8(+) T cell transition during chronic viral infection. Nat Immunol 22, 996–1007. 10.1038/s41590-021-00965-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hudson WH, Gensheimer J, Hashimoto M, Wieland A, Valanparambil RM, Li P, Lin JX, Konieczny BT, Im SJ, Freeman GJ, et al. (2019). Proliferating Transitory T Cells with an Effector-like Transcriptional Signature Emerge from PD-1(+) Stem-like CD8(+) T Cells during Chronic Infection. Immunity 51, 1043–1058 e1044. 10.1016/j.immuni.2019.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zander R, Schauder D, Xin G, Nguyen C, Wu X, Zajac A, and Cui W (2019). CD4(+) T Cell Help Is Required for the Formation of a Cytolytic CD8(+) T Cell Subset that Protects against Chronic Infection and Cancer. Immunity 51, 1028–1042 e1024. 10.1016/j.immuni.2019.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Liu Z, Zhang Y, Ma N, Yang Y, Ma Y, Wang F, Wang Y, Wei J, Chen H, Tartarone A, et al. (2023). Progenitor-like exhausted SPRY1(+)CD8(+) T cells potentiate responsiveness to neoadjuvant PD-1 blockade in esophageal squamous cell carcinoma. Cancer Cell 41, 1852–1870 e1859. 10.1016/j.ccell.2023.09.011. [DOI] [PubMed] [Google Scholar]
  • 75.Zheng L, Qin S, Si W, Wang A, Xing B, Gao R, Ren X, Wang L, Wu X, Zhang J, et al. (2021). Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474. 10.1126/science.abe6474. [DOI] [PubMed] [Google Scholar]
  • 76.Utzschneider DT, Gabriel SS, Chisanga D, Gloury R, Gubser PM, Vasanthakumar A, Shi W, and Kallies A (2020). Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nat Immunol 21, 1256–1266. 10.1038/s41590-020-0760-z. [DOI] [PubMed] [Google Scholar]
  • 77.Jansen CS, Prokhnevska N, Master VA, Sanda MG, Carlisle JW, Bilen MA, Cardenas M, Wilkinson S, Lake R, Sowalsky AG, et al. (2019). An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470. 10.1038/s41586-019-1836-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance AM, Cowey CL, Dalle S, Schenker M, Chiarion-Sileni V, Marquez-Rodas I, et al. (2017). Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N Engl J Med 377, 1824–1835. 10.1056/NEJMoa1709030. [DOI] [PubMed] [Google Scholar]
  • 79.Menzies AM, Amaria RN, Rozeman EA, Huang AC, Tetzlaff MT, van de Wiel BA, Lo S, Tarhini AA, Burton EM, Pennington TE, et al. (2021). Pathological response and survival with neoadjuvant therapy in melanoma: a pooled analysis from the International Neoadjuvant Melanoma Consortium (INMC). Nat Med 27, 301–309. 10.1038/s41591-020-01188-3. [DOI] [PubMed] [Google Scholar]
  • 80.Rozeman EA, Menzies AM, van Akkooi ACJ, Adhikari C, Bierman C, van de Wiel BA, Scolyer RA, Krijgsman O, Sikorska K, Eriksson H, et al. (2019). Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, randomised, controlled trial. Lancet Oncol 20, 948–960. 10.1016/S1470-2045(19)30151-2. [DOI] [PubMed] [Google Scholar]
  • 81.Amaria RN, Menzies AM, Burton EM, Scolyer RA, Tetzlaff MT, Antdbacka R, Ariyan C, Bassett R, Carter B, Daud A, et al. (2019). Neoadjuvant systemic therapy in melanoma: recommendations of the International Neoadjuvant Melanoma Consortium. Lancet Oncol 20, e378–e389. 10.1016/S1470-2045(19)30332-8. [DOI] [PubMed] [Google Scholar]
  • 82.Blank CU, Rozeman EA, Fanchi LF, Sikorska K, van de Wiel B, Kvistborg P, Krijgsman O, van den Braber M, Philips D, Broeks A, et al. (2018). Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat Med. 10.1038/s41591-018-0198-0. [DOI] [PubMed] [Google Scholar]
  • 83.Amaria RN, Reddy SM, Tawbi HA, Davies MA, Ross MI, Glitza IC, Cormier JN, Lewis C, Hwu WJ, Hanna E, et al. (2018). Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med. 10.1038/s41591-018-0197-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lo JA, Kawakubo M, Juneja VR, Su MY, Erlich TH, LaFleur MW, Kemeny LV, Rashid M, Malehmir M, Rabi SA, et al. (2021). Epitope spreading toward wild-type melanocyte-lineage antigens rescues suboptimal immune checkpoint blockade responses. Sci Transl Med 13. 10.1126/scitranslmed.abd8636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Brossart P. (2020). The Role of Antigen Spreading in the Efficacy of Immunotherapies. Clin Cancer Res 26, 4442–4447. 10.1158/1078-0432.CCR-20-0305. [DOI] [PubMed] [Google Scholar]
  • 86.Zak J, Pratumchai I, Marro BS, Marquardt KL, Zavareh RB, Lairson LL, Oldstone MBA, Varner JA, Hegerova L, Cao Q, et al. (2024). JAK inhibition enhances checkpoint blockade immunotherapy in patients with Hodgkin lymphoma. Science 384, eade8520. 10.1126/science.ade8520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Mathew D, Marmarelis ME, Foley C, Bauml JM, Ye D, Ghinnagow R, Ngiow SF, Klapholz M, Jun S, Zhang Z, et al. (2024). Combined JAK inhibition and PD-1 immunotherapy for non-small cell lung cancer patients. Science 384, eadf1329. 10.1126/science.adf1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Weber EW, Parker KR, Sotillo E, Lynn RC, Anbunathan H, Lattin J, Good Z, Belk JA, Daniel B, Klysz D, et al. (2021). Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science 372. 10.1126/science.aba1786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Spitzer MH, Carmi Y, Reticker-Flynn NE, Kwek SS, Madhireddy D, Martins MM, Gherardini PF, Prestwood TR, Chabon J, Bendall SC, et al. (2017). Systemic Immunity Is Required for Effective Cancer Immunotherapy. Cell 168, 487–502 e415. 10.1016/j.cell.2016.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Yost KE, Satpathy AT, Wells DK, Qi Y, Wang C, Kageyama R, McNamara KL, Granja JM, Sarin KY, Brown RA, et al. (2019). Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med 25, 1251–1259. 10.1038/s41591-019-0522-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Patel SP, Othus M, Chen Y, Wright GP Jr., Yost KJ, Hyngstrom JR, Hu-Lieskovan S, Lao CD, Fecher LA, Truong TG, et al. (2023). Neoadjuvant-Adjuvant or Adjuvant-Only Pembrolizumab in Advanced Melanoma. N Engl J Med 388, 813–823. 10.1056/NEJMoa2211437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Valpione S, Galvani E, Tweedy J, Mundra PA, Banyard A, Middlehurst P, Barry J, Mills S, Salih Z, Weightman J, et al. (2020). Immune-awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat Cancer 1, 210–221. 10.1038/s43018-019-0022-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Weber JS, Gibney G, Sullivan RJ, Sosman JA, Slingluff CL Jr., Lawrence DP, Logan TF, Schuchter LM, Nair S, Fecher L, et al. (2016). Sequential administration of nivolumab and ipilimumab with a planned switch in patients with advanced melanoma (CheckMate 064): an open-label, randomised, phase 2 trial. Lancet Oncol 17, 943–955. 10.1016/S1470-2045(16)30126-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Campbell KM, Amouzgar M, Pfeiffer SM, Howes TR, Medina E, Travers M, Steiner G, Weber JS, Wolchok JD, Larkin J, et al. (2023). Prior anti-CTLA-4 therapy impacts molecular characteristics associated with anti-PD-1 response in advanced melanoma. Cancer Cell 41, 791–806 e794. 10.1016/j.ccell.2023.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Liu D, Schilling B, Liu D, Sucker A, Livingstone E, Jerby-Arnon L, Zimmer L, Gutzmer R, Satzger I, Loquai C, et al. (2020). Author Correction: Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat Med 26, 1147. 10.1038/s41591-020-0975-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, Hodi FS, Martin-Algarra S, Mandal R, Sharfman WH, et al. (2017). Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell 171, 934–949 e916. 10.1016/j.cell.2017.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Linderman GC, Zhao J, Roulis M, Bielecki P, Flavell RA, Nadler B, and Kluger Y (2022). Zero-preserving imputation of single-cell RNA-seq data. Nat Commun 13, 192. 10.1038/s41467-021-27729-z. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

1
2

Table S2: Hotspot Module Genes, Pathways, and Human CD8+ T cells Gene Lists. Related to Figure 1.

Hotspot module genes: List of all genes for each of the 4 gene modules generated by Hotspot.

Hotspot module pathways: List of top enriched pathways in the 4 gene modules generated by Hotspot.

Human CD8+ T cell Gene Lists: List of top 250 differentially expressed genes for each CD8+ T cell subset defined by sorted cells from human healthy donors, as described in Star Methods.

3

Table S3: Tumor TCR sequences. Related to Figures 3 and S6.

Data Availability Statement

Raw and processed single-cell sequencing data generated in this study have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus and are publicly available: (Main sequencing study: GSE272993, treatment-naïve aPD-1: GSE272734, healthy donor: GSE272735, paired blood tumor: GSE273718).

All original code for Cyclone has been deposited on GitHub: (https://github.com/jiwen90/cyclone) and Zenodo: (https://doi.org/10.5281/zenodo.12754871).

Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

RESOURCES