Abstract
Acute myeloid leukemia (AML) that is relapsed and/or refractory post-allogeneic hematopoietic cell transplantation (HCT) is usually fatal. In a prior study, we demonstrated that AML relapse in high-risk patients was prevented by post-HCT immunotherapy with Epstein-Barr virus (EBV)-specific donor CD8+ T cells engineered to express a high-affinity Wilms Tumor Antigen 1 (WT1)-specific T-cell receptor (TTCR- C4). However, in the present study, infusion of EBV- or Cytomegalovirus (CMV)-specific TTCR-C4 did not clearly improve outcomes in fifteen patients with active disease post-HCT. TCRC4-transduced EBV-specific T cells persisted longer post-transfer than CMV-specific T cells. Persisting TTCR-C4 skewed towards dysfunctional natural killer-like terminal differentiation, distinct from the dominant exhaustion programs reported for T-cell therapies targeting solid tumors. In one patient with active AML post-HCT, a sustained TTCR-C4 effector-memory profile correlated with long-term TTCR-C4 persistence and disease control. These findings reveal complex mechanisms underlying AML-induced T-cell dysfunction, informing future therapeutic strategies for addressing post-HCT relapse.
Introduction
Relapsed/refractory acute myeloid leukemia (AML) after allogeneic hematopoietic cell transplantation (HCT) poses a major therapeutic challenge,1,2 with 2-year overall survival (OS) rate below 20% and only 4% if relapse occurs within six months post-HCT.1–3 Salvage therapies, including intensive chemotherapy, donor lymphocyte infusions (DLI), and second HCTs have shown limited efficacy, underscoring the urgent need for novel therapies.
The Wilms’ Tumor 1 (WT1) protein is an attractive AML immunotherapy target,4 as WT1 overexpression promotes proliferation and oncogenicity.5–7 In a phase I/II trial, targeting WT1 in the post- HCT setting, we genetically modified matched donor CD8+ T cells to express a high-affinity WT1-specific T cell receptor (TCRC4) specific for the HLA-A*0201-restricted WT1126-134 epitope. To minimize the potential for graft-versus-host disease (GVHD) mediated by infused donor cells,8 Epstein-Barr virus (EBV)-specific or cytomegalovirus (CMV)-specific (in EBV-negative donors) CD8+ T cells were transduced (TTCR-C4). Prophylactic infusion of EBV-specific TTCR-C4 in patients without detectable disease, but at high relapse risk post-transplant (Arm 1), yielded 100% relapse-free survival in 12 patients at a median follow-up of 44 months, and 54% in concurrent controls.9 However, in 15 patients with prior evidence of disease post-HCT discussed here, infused EBV-specific or CMV-specific TTCR-C4 did not yield a superior overall survival compared to historical controls.1,2,10,11
The persistence of functional antigen-specific T cells is required for sustained immunotherapy efficacy.12–14 In solid tumors and lymphomas, reduced persistence generally correlates with T-cell exhaustion,12,15,16 characterized by markers like PD-1, CTLA-4, Tim3, LAG-3, BTLA and/or TIGIT.17 Targeting these immune-inhibitory receptors with checkpoint-blocking antibodies mitigates exhaustion.18,19 In AML, recent studies have questioned the presence of T-cell exhaustion,20 proposing alternative mechanisms of immune dysfunction.21,22 Natural killer-like (NKL) markers expressed on CD8+ T cells have correlated with adverse outcomes, suggesting that NKL skewing may contribute to T-cell dysfunction.22–24 However, whether AML directly induces terminally differentiated, dysfunctional antigen-specific T cells remains unclear.25
Understanding the mechanisms of dysfunction that are operative in AML is critical for designing strategies to overcome the current limitations observed in T-cell therapies. Adoptive transfer of TTCR-C4 in refractory or relapsed AML patients has provided a unique opportunity to track AML-specific T cells, elucidate AML-induced T cell states, identify the mechanisms responsible for T-cell dysfunction, and inform the design of effective anti-AML therapies.
Results
TTCR-C4 is safe, well tolerated, and produces comparable outcomes to conventional treatments for high-risk relapsed/refractory AML patients post-HCT
From December 2012 through March 2020, 15 HLA-A2-expressing patients with relapsed and/or refractory AML post-HCT were enrolled on trial NCT01640301 (Table 1, Extended data Fig. 1). The median age at diagnosis was 40. Pre-HCT, 27% of patients had secondary or treatment-related AML, at diagnosis 47% were adverse risk, and 53% favorable/intermediate risk per European LeukemiaNet (ELN) stratification (Supplementary Table 1).26 Post-HCT, four patients had measurable residual disease (MRD) by ∼28 days (refractory), two relapsed by 3 months, and nine relapsed after day 100. The median time to relapse was 496 days post-HCT. Thirteen patients received salvage therapy before TTCR-C4, including six who underwent a second HCT. Within a median of two weeks before infusion, two patients had overt disease, five were MRD-positive and the rest had no evaluable disease (NED) (Extended data Fig. 2). Ten patients received EBV-specific TTCR-C4 and five received CMV-specific T cells (Table 1). Patients received one to four infusions depending on their place in the TTCR-C4 dose escalation (Extended data Fig. 3).
As in Arm 1,9 TTCR-C4 infusions were safe and well tolerated (Supplementary Table 2). The incidence of acute and chronic GVHD (aGVHD, cGVHD) was lower compared to DLIs or second HCTs as salvage strategies,11,27 with only one patient (7%) developing grade 3 aGVHD and three (20%) developing cGVHD post-TTCR-C4 infusion. Post-infusion cGVHD included one grade 3, one grade 2 and one grade 1 cGVHD. However, the onset of cGVHD occurred when TTCR-C4 constituted <1% of the peripheral blood (PB) CD8+ T cells, making the infusion of TTCR-C4 with an endogenous virus-specific TCR the unlikely cause of cGVHD.
Among the patients enrolled (Table 1), four showed no overt relapse and/or MRD following TTCR- C4 infusion(s) (Extended data Fig. 4) supporting TTCR-C4’s potential biologic activity. However, when all 15 patients were analyzed together, the median OS was 242 days, the 2-year OS was 33%, and the 3-year OS was 20% (Extended data Fig. 5), indicating that these individual outcomes did not translate into a significant survival advantage over historical salvage treatments.1,2,10,11 For example, DLI post-HCT relapse showed a 2-year OS of 21% (±3%) from relapse and 56% (±10%) from DLI in cases of remission or with a favorable karyotype, dropping to 15% (±3%) in aplasia or active disease.27
Virus-specific substrate T cells and AML presence at the time of infusion are major determinants of TTCR-C4 persistence
In our cohort, baseline PB WT1-specific tetramer+ T cells were low (Fig. 1A – red arrows, day 0) indicating limited endogenous WT1-specific T cells. By day 28 post-infusion, four patients showed TTCR- C4 levels exceeding 3% (persistence threshold) of the CD8+ T cells, with three maintaining these levels beyond day 100. Higher frequencies correlated with increased absolute TTCR-C4 counts (Extended data Fig. 6A). Patients with TTCR-C4 frequencies below 3% were eligible for additional infusions (two infusions in 6/15 patients, >2 infusions in 4/15 patients) (Table 1). However, more than two infusions did not significantly increase persistence (Fig. 1B).
We next explored the contribution of substrate cell virus-specificity to post-infusion persistence. Within 28 days, EBV-specific TTCR-C4 were more abundant than CMV-specific TTCR-C4, which remained below the persistence threshold (Fig. 1C). These differences endured when EBV-specific TTCR-C4 recipients from Arm 1 were included (Extended data Fig. 6B).9 As central-memory CD8+ T cells (Tcm) have shown improved persistence compared to more differentiated phenotypes post-transfer,28 we sought to explore whether differences in endogenous virus-specific cells could recapitulate these findings. We analyzed the phenotypes of EBV- and CMV-specific T cells using mass cytometry data29 from 143 healthy individuals and cancer patients. Multidimensional scaling (MDS) showed segregation of CMV- and EBV-specific CD8+ T cells along the first dimension (MDS dim.1) suggesting distinct T-cell differentiation states between these groups (Extended data Fig. 7A). Next, Flow Self-Organizing Maps (FlowSOM)30 metaclustering identified four CD8+ T-cell subsets: naïve (CD95-,CD45RA+, CCR7+, CD27+, CD28+), effector memory (Tem) (CD95+, CD45RA-, CD45RO+, CD27+, CD28+, CD127+), Tcm (CD95+, CD45RA-, CD45RO+, CCR7+, CD27+, CD28+, CD127+), CD45RA+ effector memory (Temra) (CD95+, CD45RA+, CD45RO-, CD57+, KLRG1+) (Extended data Fig. 7B).31,32 Differential abundance analysis revealed a significant (false discovery rate/FDR < 0.05) increase of Temra cells in CMV-specific T cells and of naïve, Tcm and Tem in EBV-specific T cells (Fig. 1D). These results suggest that the Temra state of CMV-specific substrate cells compromises post-infusion persistence, whereas EBV-specific cells, derived from less differentiated precursors, facilitate persistence.
Persistence varied among EBV-specific TTCR-C4 recipients, (Fig. 1C, Extended data Fig. 6A), with only three of ten (4, 8 and 27) exhibiting long-term persistence (>100 days post-infusion) (Fig. 1A, Extended data Fig. 8A). We reasoned that persistence was influenced by AML high-risk factors, including post-transplant remission duration and disease burden at the time of TTCR-C4 infusion.3 Patient 1 was excluded from this analysis due to unavailable WT1-expression data. Six of the remaining patients did not show long-term persistence: four (5,7,14, and 19) had detectable disease (MRD or overt) within two weeks of TTCR-C4 infusion, and two (23, 26) who were disease-free at infusion, had relapsed within 3 months post- HCT, suggesting difficult-to-control disease. Statistical testing revealed a trend (Fisher’s exact test, p = 0.08) linking reduced persistence with “aggressive” disease (defined as having detectable disease pre- infusion or early relapse post-HCT). Patient 8, initially categorized as “aggressive” because MRD-positive before the first infusion, became MRD-negative through salvage therapy before the second infusion, achieving the longest observed TTCR-C4 persistence. Reclassifying this patient as “non-aggressive”, renders the association between disease aggressiveness and persistence statistically significant (p < 0.05).These findings suggest that aggressive disease post-HCT, independently of virus-specificity, was associated with reduced TTCR-C4 persistence in vivo.
Long-term persistent TTCR-C4 acquire NKL/terminal differentiation markers associated with progressive loss of function in vivo
To investigate the fate of TTCR-C4 post-transfer in patients with persistent TTCR-C4, and compare these with endogenous (TCRC4-) T cells, we designed a 24-color spectral flow-cytometry panel (Supplementary Table 3). PB was analyzed at ∼1 (T1), ∼7 (T2), 28 (T3) days, and ∼4 months (T4) post-transfer (Supplementary Table 4). FlowSOM clustering revealed five CD8+ T-cell states categorized into three higher-level clusters (Fig. 2A, top dendrogram): naïve-/cm-like cells (Fig. 2A, dark green) (CD45RA+, CCR7+, CD27+, CD28+); Tem subgroup, including endogenous CD8+ Tem (CD28+, CD27+, Ki67+, CD38+, TIGIT+, PD1+, Tbet+) (Fig. 2A, violet), and phenotypically similar TTCR-C4_Tem (tetramer+) (Fig. 2A, blue); Temra subgroup of endogenous CD8+ T cells (CD45RA+, CD57+, KLRG1+, GZMB+) (Fig. 2A, light green) and closely clustered TTCR-C4_Temra (Fig. 2A, red). Two-dimensional Uniform Manifold Approximation and Projection (UMAP) revealed a progressive increase in TTCR-C4_Temra and decline in TTCR-C4_Tem from T1 to T4 (Fig. 2B). TTCR-C4_Temra expressed minimal Ki67 compared to TTCR-C4_Tem (Fig. 2A), enabling manual gating of these two subsets (Fig. 2C). Over time, TTCR-C4_Temra significantly increased, while TTCR- C4_Tem decreased (p <0.05) (Fig. 2D), suggesting differentiation from proliferative TTCR-C4_Tem to non- proliferative TTCR-C4_Temra. TTCR-C4_Temra predominantly expressed cytotoxic/KLR markers (KLRG1, CD57, GNLY), previously linked to T-cell dysfunction in AML,21–23 rather than classical T-cell exhaustion markers (Tim3, PD1, and TIGIT) (Fig. 2E).17,33 Stratification by timepoint revealed progressive increase of the cytotoxic subset versus the exhausted subset (Fig. 2F). This phenotypic shift concided with a detectable but slow decline in TTCR-C4 CD8+ T cells producing IFNγ and TNFα (Fig. 2G, Extended data Fig. 8B). Thus, long-term persisting EBV-specific TTCR-C4 skew towards a distinct NKL phenotype linked to functional decline.
TTCR-C4 largely represent an intermediate state between effector memory and NK-like/terminally differentiated cells
To explore the relationship between CD8+ endogenous T-cell and TTCR-C4 states, we performed single-cell RNA sequencing (scRNAseq) on available PB and bone marrow (BM) samples from patients with TTCR-C4 exceeding 3% of CD8+ T cells at least until day 28 post-infusion. This analysis included two prophylactic9 and five treatment-arm patients (Supplementary Table 5).
Unsupervised clustering of PB CD8+ T cells (n = 24,472) identified 13 clusters, with the TCRC4 transgene expressed across several clusters, but primarily in cluster 2 (Fig. 3A). Using the marker-based purification algorithm scGate,34 we identified TCRC4+ cells (Fig. 3B) labeled as TTCR-C4, while endogenous (TCRC4−) T cells included clusters 5 and 6,labeled as naïve-/cm-like, expressing CCR7, SELL, TCF7, LEF1, IL7R; clusters 2, 4 and 11, labeled as Tem, expressing genes associated with activation (CD69, TIGIT, GZMK); cluster 12, labeled as interferon signaling genes (ISG), expressing ISG15, ISG20, IRF7, IFI6; clusters 9 and 10, labeled as T memory/proliferative (Tmem/prolif), expressing proliferation (MKI67, MCM5, MCM7) and memory (CD27) genes; clusters 0,1,3,7, and 8, labeled as NKL/Temra, expressing genes associated with cytotoxicity/NKL (KLRF1, KIR3DL1, NKG7, FCGR3A, GZMB, KLRD1, PRF1) (Fig. 3A-C, Extended data Fig. 9A-C, Supplementary Table 6).
To assess transcriptional similarities among cell states, we performed principal component analysis, positioning naïve-/cm-like and NKL/Temra as differentiation spectrum extremes, with TTCR-C4 as the intermediate state between these two extremes (Fig. 3C).
To ensure unbiased labeling of CD8+ T cells, we constructed a scRNAseq reference atlas comprising 109,051 CD8+ T cells, using a published scRNAseq dataset of tumor-infiltrating lymphocytes.35 We aligned and projected our scRNAseq dataset onto the reference atlas (Extended data Fig. 9B), confirming the correspondence between the transcriptional states in our dataset and the reference. Differential gene expression (DGE) analysis using manually curated markers further supported this annotation (Supplementary Table 7, Extended data Fig. 9C).31,32,36,37
Consistent with previous work,32 we observed a progressive reduction of stem-like markers (IL7R, TCF7) from naïve/cm-like cells through Tem and NKL/Temra cells, while activation markers (CD69, GZMK) peaked in Tem. NKL markers, including KLR-exhaustion markers (S1PR5, ZEB2),38,39 were highest in NKL/Temra. TTCR-C4 expressed activation and NKL markers, reflecting an intermediate state between Tem and NKL/Temra (Extended data Fig. 9D).
Monocle40 trajectory inference confirmed TTCR-C4 as an intermediate state between Tem and the terminal NKL/Temra state (Fig. 3E-F), and removing the TCRC4 transgene did not alter this differentiation trajectory (Extended data Fig. 10A-B). Single-gene expression over pseudotime revealed early peaks of naïve/stem-like markers (CCR7, IL7R, TCF7, SELL, LEF1) (Fig. 3G, first row), naïve/memory (CD27, CD28) and activation markers (GZMK, CD69), with the latter remaining high throughout mid-pseudotime before declining.(Fig. 3G, second row). TCRC4 expression peaked at mid-pseudotime, but remained expressed until the end of pseudotime. NKL/Temra-associated genes (PRF1, KLRG1, NKG7, ZEB2, S1PR5) peaked at the end of pseudotime (Fig. 3G, third row). RNA velocity inference41 supported the spectrum of differentiation from naïve-like to Tem and TTCR-C4, culminating in NKL/Temra cells (Fig. 3H).
Overall, our findings suggest that TTCR-C4 largely existed in an intermediate state between the Tem phenotype and the NKL/Temra stage.
AML drives TTCR-C4 towards NKL/terminal differentiation instead of the dominant exhaustion pattern associated with T cells in solid tumors
To investigate AML’s impact on the differentiation of endogenous CD8+ T cells and TTCR-C4, patients samples were grouped as AML- (no AML detected, including two prophylactic arm cases),9 or AML+ (blasts evident in BM and/or PB) (Supplementary Table 8). The increase of ISG and NKL/Temra in AML+ and TTCR-C4 in AML- (Fig. 4A) was significant (Fig. 4B), with a wide confidence interval for ISG, making the magnitude of this difference unclear. TTCR-C4 overexpressed NKL/Temra genes in AML+ compared to AML- (Fig. 4C), suggesting that AML blasts may induce a transcriptional shift towards NKL/Temra in TTCR-C4 and endogenous T cells.
Following previously described methodology,42 we exposed in vitro CD8+ transgenic WT1-specific T cells targeting the HLA A*0201-restricted WT137-45 epitope43 (TTCR37-45) to high-WT1-expressing HLA- A*0201-transduced K562 acutely transformed chronic myelogenous leukemia (Extended data Fig. 11A). As K562 primarily express standard proteasomes and are not readily lysed by TTCR-C4 targeting the immunoproteasome-specific WT1126–134 peptide, we used TTCR37-45 due to its proteasome-agnostic WT1- derived peptide recognition.43 Exposing TTCR37-45 to fresh K562 every 3-4 days at stable effector-to-target (E:T) ratios (1:1, 1:4) caused initial tumor lysis (Extended data Fig. 11B), but control was lost by day 13, marked by a declining TTCR37-45-to-tumor ratio (Extended data Fig. 11C). Irrelevant TCR-expressing T cells exerted no control (Extended data Fig. 11D). Bulk-RNA sequencing on sorted T cells found increased (FDR <0.05) naïve/stem-like genes (IL7R, TCF7, LTB, LEF1, SELL) at day 0, followed at day 13 by increased effector (CCL5, GZMK), activation/exhaustion (CD69, LAG3, CTLA4, PDCD1), and NKL/Temra markers (KLRB1, ENTPD1, KLRC1, KLRC2, NCAM1) (Fig. 4D-E). By day 23, the activation/exhaustion markers decreased, while NKL/Temra persisted or increased (Fig. 4D-E). To integrate these findings with the scRNAseq dataset, we performed a gene-set enrichment analysis with the top 50 differentially expressed genes from each CD8+ subset identified in scRNAseq, including naïve-/cm- like, Tem, NKL/Temra, TTCR-C4, Tmem/prolif and ISG from the scRNAseq dataset. We also included a manually curated exhaustion signature (Supplementary Table 9). Exhaustion and Tem signatures exhibited enrichment in later timepoints, yet there was no further increase at day 23 versus day 13 (Fig. 4F). In contrast, the NKL/Temra increased between days 13 and 23 corresponding to TTCR37-45 loss of tumor control (Extended data Fig. 11C).
We next projected scores from published NKL dysfunction signatures22,38,39 onto our UMAP, revealing overlaps that link the NKL/Temra CD8+ T cells we identified with an end-term dysfunctional differentiation subset (Extended data Fig. 12). To confirm that AML-exposed T cells skewed towards a NKL/Temra rather than exhausted (Tex) phenotype, we projected onto a scRNAseq reference atlas a BM AML CD8+ T-cell dataset, compiled from published sources,20,22,23,44,45 alongside datasets representing pancreatic,46 melanoma,47 and lung48 solid tumors (Fig. 4G-H).35 Unlike solid tumors, which included a Tex cluster, AML datasets aligned with our spectral flow-cytometry and scRNAseq findings confirming the absence of Tex and the presence of NKL/Temra cells (Fig. 4G, Extended data Fig. 13). Although these studies did not specifically analyze tumor-antigen reactive T cells, the findings collectively suggest that the NKL (versus exhaustion) signature is intricately associated with AML-induced T-cell dysfunction.
Prolonged azacitidine exposure enhances self-renewal and Tcm features of TTCR-C4, supporting TTCR- C4-mediated AML control
Given the link between NKL differentiation and AML-induced T-cell dysfunction, we explored the state of TTCR-C4 in patient 8, the only patient of 4 (Extended data Fig. 4) who exhibited prolonged TTCR-C4 and MRD persistence indicative of disease control despite incomplete leukemia clearance, enabling TTCR- C4/AML interplay analysis. This female patient underwent a non-myelo-ablative (Flu/3Gy-TBI) HCT from a male donor, with MRD (0.26% blasts by flow cytometry) detected 28 days post-HCT (Fig. 5A) and received a TTCR-C4 (1010 cells/m2) 77 days post-HCT. AML blasts expressed detectable but limited WT1 protein pre-HCT (Extended data Fig. 14). scRNAseq at day 49 post-infusion revealed circulating CD34+ female-origin blasts (XIST+) expressing WT1 (Fig. 5E-F). Persistent MRD and limited T-cell persistence at day 63 post-infusion (T cells declined to only 0.1 multimer+ CD8+ T cells/µl on day 60), (Fig. 5B, 5C- red arrow, 5D) prompted azacitidine treatment (five days every 28 days). Post-azacitidine, TTCR-C4 counts increased (∼50 multimer+ CD8+ T cells/ul; >3% CD8 multimer+ T cells) without additional TTCR-C4 (Fig. 5D), leading to azacitidine discontinuation after seven cycles when no AML was detected. However, AML recurrence at day 414 post-infusion triggered chemotherapy (mitoxantrone, etoposide and cytarabine), additional azacitidine cycles followed by a second TTCR-C4 (1010 cells/m2) infusion (Fig. 5A). Azacitidine was held post-infusion but restarted upon MRD detection 162 days post-second TTCR-C4 infusion, continuing for over 20 monthly cycles, maintaining marrow MRD below 1% for 21 months (641 days, from day 868 to day 1509) before progression was detected on day 1546 (Fig. 5A-B). During this 21-month period, blasts were detectable via scRNAseq in BM but not in PB (Fig. 5F, Extended data Fig. 15A-B). Throughout the azacitidine cycles, platelet counts decreased (∼100 x 103 platelets/µl) mid-cycle before rising (∼300 x 103 platelets/µl) immediately before the next cycle, consistent with transient azacitidine-mediated myelosuppression. Neutrophil counts, however, paradoxically increased mid-cycle (∼4.5 x 103 neutrophils/µl) and decreased (∼1.6 x 103 neutrophils/µl) just before the next cycle, suggesting that treatment-related AML contraction facilitated enhanced mid-cycle neutrophil production despite azacitidine’s myelosuppressive effect (Extended data Fig. 15C). This 21-month response exceeds the typical 4-month median for post-HCT azacitidine treatment,49 suggesting that persisting TTCR-C4 plus azacitidine contributed to long-term disease control.
TTCR-C4 transcriptomics were analyzed to identify associations with disease control. We scored genes associated with self-renewal and NK-like profiles and found that at earlier timepoints (d49, d256, and d405 post-first infusion), TTCR-C4 were predominantly skewed towards an NKL/Temra transcriptional state. However, at later timepoints (d1322, d1343) although TTCR-C4 remained along the same differentiation trajectory (from less differentiated states to NKL/Temra TTCR-C4), they exhibited fewer NKL features, and showed an enrichment of self-renewal markers (Fig. 5G-I). scTCRseq analysis of PB at days 265, 405, 1322, 1343 post-first TTCR-C4 infusion revealed progressive clonal expansion of TTCR-C4 (Fig. 5K). TTCR-C4 clonal expansion peaked during the 21 months of continued azacitidine treatment, suggesting that during this phase, TTCR-C4 were capable of both self-renewal, which in turn facilitated their long-term persistence, and antigen recognition, which triggered further expansion and disease control. Similarly, BM scRNAseq revealed clonal expansion of TTCR-C4 between d1322 and d1344 post-first infusion (Fig. 5L) despite no additional TTCR-C4. These findings indicate that azacitidine-exposed TTCR-C4, retained a self-renewing (versus NKL) features, which enabled their long-term persistence and enhanced AML-targeting efficacy that may have contributed to long-term disease control (Fig. 5M).
Discussion
We previously demonstrated that EBV-specific TTCR-C4 infusion prevented AML recurrence in patients at high risk of post-HCT relapse.9 In 15 AML patients who received TTCR-C4 post-HCT relapse, we observed indirect anti-leukemic activity, but no survival advantage in those receiving EBV- or CMV- specific TTCR-C4. We analyzed the AML/TTCR-C4 interplay to investigate how AML affects antigen-specific T cells, and identified T-cell characteristics associated with AML control in one case.
The characteristics of substrate cells from which T-cell products are derived can impact post- transfer persistence. In murine and macaque models, antigen-specific Tcm-like CD8+ T cells persist long- term post-adoptive transfer, unlike Temra-like cells.9,28 In our study, TTCR-C4 from EBV-specific persisted longer compared to CMV-specific substrate cells which may reflect the distinct biology of EBV and CMV.50,51 CMV reactivates periodically, requiring rapidly activated responses that promote Temra differentiation,52,53 while EBV tends to remain quiescent, preserving Tcm phenotypes.54–56 While reactivation frequency plays a role, the reactivation microenvironment is also critical. CMV reactivates in non-professional antigen-presenting cells, including fibroblasts, which lack co-stimulatory signals and favor Temra differentiation; EBV reactivates in lymphoid tissues B cells, providing co-stimulation that may preserve the Tcm phenotype.57,58 Our analysis of an independent mass-cytometry dataset29 confirmed that EBV-specific cells better maintain self-renewing features than CMV-specific cells. Taken together these findings establish that self-renewal may predict prolonged T-cell persistence.9,28 However, EBV-specific TTCR-C4 were unable to sustain long-term responses in all patients, suggesting influence by non-intrinsic AML-related factors.
Despite efforts to investigate T cell dysfunction in AML,20,21,23 several critical questions have remained unanswered,25 mainly due to the challenge of identifing AML-specific T cells. In this study, infused TTCR-C4 offered the possibility of examining AML-specific T cell and AML biology. We found that T-cell exhaustion is not the primary cause of AML-specific T-cell dysfunction.20,21,23 Instead, TTCR-C4 with exhaustion markers (PD1, TIGIT, Tim3) also expressed activation markers (CD38, CD69) and proliferation (Ki67), indicating an effector-like T-cell state. Overall, this suggests that AML-exposed T cells follow a differentiation trajectory from effector-memory to NKL cells, compromising TTCR-C4 cell persistence and function.
AML’s distinct properties, including myeloid cells immunosuppressive effects59 via production of reactive oxygen species and other soluble factors, may distinctly redirect T cells toward functional impairment that may preclude transitioning to a classical exhausted phenotype and explain the observed NKL skewing in TTCR-C4 and endogenous CD8+ T cells. The absence of classical exhausted cells in AML has several implications. In solid tumors, chronic antigen stimulation induces a multi-step epigenetic shift of CD8+ T cells into exhaustion,60 starting from PD1+TCF1+ precursor/progenitor exhausted cells,23,24 whose frequency correlates with response to checkpoint inhibitors.9,28 In contrast, our AML findings suggest that exhaustion markers reflect recent T cell activation rather than true exhaustion, which may also explain the limited efficacy of checkpoint inhibitors in AML.20,23,24,61 The presence of a distinct dysfunctional program supports new immunotherapy strategies to enhance the efficacy of anti-AML adoptive T-cell therapy, preventing T-cell dysfunction. For example, knocking out ID3 and SOX4 transcription factors in chimeric antigen receptor-engineered (CAR)-T cells in vitro reduces NKL skewing and enhances effector functions.42 Whether these strategies can improve cell therapy in AML in vivo remains to be determined.
Another strategy to reduce NKL skewing of TTCR-C4, as suggested by the outcome in one of our treated patients, could involve the hypomethylating agent azacitidine. While azacitidine has direct anti- leukemia effects,62 the prolonged disease control observed in our patient was unusual prompting further investigation into the mechanisms at play. Azacitidine administration correlated with clonal expansion and persistence of central memory, as opposed to NKL differentiated TTCR-C4, leading to a prolonged equilibrium between anti-leukemic TTCR-C4 and AML MRD. Before the first T cell infusion in this patient, low AML WT1 expression likely did not activate TTCR-C4, causing their near disappearance. However, after azacitidine introduction, TTCR-C4 frequencies increased, likely driven by azacitidine-induced WT1 expression.63 Intermittent TTCR-C4 activation by WT1 presentation on AML cells transiently induced by cycles of azacitidine with subsequent temporary clearance of AML may have helped sustain TTCR-C4 persistence and response to stimulation. Other studies have shown that azacitidine can have negative effect on T regulatory cells64 and boosts CAR T cell toxicity towards AML.65 Further investigation is needed to determine if azacitidine acts preferentially on leukemia cells by increasing WT1 expression thereby promoting antigen- recognition and/or TTCR-C4 effector functions. However, taken together, these findings support azacitidine as a favorable adjunct to T cell immunotherapy.
Paradoxically, our ability to track dysfunction was limited to patients with persisting TTCR-C4, where T cells were relatively more functional. We hypothesize that patients with more aggressive disease experience accelerated T-cell dysfunction and death rather than establishment of progenitor cells capable of self-renewal; however we were unable to formally test this in our study. Despite a limited sample size, our findings on the nature of T-cell dysfunction in AML were confirmed in larger cohorts of endogenous CD8+ T cells, though further validation in larger trials could provide more detail. Additionally, our interpretation of the effect of azacitidine on TTCR-C4 is based on a single patient. To address this limitation, we are initiating a clinical trial of co-administration of azacitidine and TCR-T cells.
Our study illuminates some of the complex mechanisms underlying AML-induced T cell dysfunction and strengthens support for a distinct pathway outside the traditional paradigm of T cell exhaustion, emphasizing the need to addres this unique dysfunction in future immunotherapy strategies.
Methods
Clinical protocol
The trial was approved by the Fred Hutchinson Cancer Center (FHCC) Institutional Review Board, the US Food and Drug Administration and the National Institutes of Health Recombinant DNA Advisory Committee. It was registered at ClinicalTrials.org under the identifier NCT01640301. Eligible participants included ‘high-risk’ AML patients with relapsed or refractory disease (overt or MRD) post-HCT along with their fully HLA-matched (10 of 10) related or unrelated donors expressing HLA A*0201 (HLA-A2).
Patient selection
HLA-A2 genotype was confirmed by high-resolution typing before enrollment. Exclusion criteria included: refractory central nervous system disease, HIV seropositivity, grade ≥ 3 GVHD and no available CMV/EBV-seropositive matched donor. The sample size for this study was not based on formal power calculations, but on feasibility, the potential to provide descriptive information, determine whether further study was warranted and evaluate toxicity.
Treatment plan
Isolation of TCRC4, lentiviral vector construction, and TTCR-C4 generation were performed as previously described.9 Patients were eligible to receive a first infusion of TTCR-C4 after demonstrating relapse (overt or MRD) at any time post allogeneic HCT. Patients 26,27,28 received lymphodepleting treatment before the first TTCR-C4 infusion with cyclophosphamide (300 mg/m2 IV) and fludarabine phosphate (30 mg/m2 IV) daily on days -4 to -2. A second TTCR-C4 infusion was administered only if the frequency of TTCR-C4 was <3% of total peripheral CD8+ T cells. Patients 5,7,19,23,27 received 1 infusion, patients 4,8,15,26,28 received 2 infusions, patient 1 received 1 infusion, the remainder received 4 infusions (Table 1). Patients were monitored for toxicities, based on Common Toxicity Criteria v.4.0.
Assessment of disease status
Morphology, multiparameter flow cytometry, standard cytogenetics or genomic technologies were routinely performed on bone marrow aspirates and peripheral blood samples that were obtained from all patients. Any level of residual disease was considered to indicate positivity for MRD.
T cell tracking by WT1 peptide/HLA (pHLA) tetramers
WT1 pHLA-specific tetramers (produced by the FHCC Immune Monitoring Core Facility) were used to detect TTCR-C4 in PBMCs collected after infusions, with a staining sensitivity of 0.01% of total CD8+ T cells, as previously described.9 TTCR-C4 percentages were calculated using FlowJo v.10 (Treestar).
Patient outcomes and survival analysis
Kaplan-Meier OS curves were estimated using the survminer (v0.4.9) and survival (v3.3-1) packages in R. OS was calculated from the date of first TTCR-C4 infusion to the date of death or censoring. Outcomes of responding patients were represented using a swimmerplot. Day 0 was defined as the post-HCT relapse date or, for HCT-refractory patients, as day 28 post-HCT, at which timepoint persistent disease was observed. R package swimplot (v1.2.0) was used for visualization.
Analysis of TTCR-C4 persistence
Persistence of TCR-T cells (%TTCR-C4) over time was visualized using ggplot2 (v3.4.4) R package. Wilcoxon rank-sum tests were used for pairwise comparisons between TTCR-C4 with different virus- specificity (EBV vs. CMV) over time. Kruskal-Wallis tests were applied for multi-group comparisons.
Patients were categorized based on disease status prior to infusion: those with detectable MRD or overt disease before TTCR-C4 infusion, or those who relapsed within 3 months post-HCT, were classified as having "aggressive" disease. Patients without these risk factors were considered "non-aggressive." One patient was excluded from the analysis due to the unavailability of WT1-expression data, which could confound interpretation of the persistence results. Fisher’s exact test was used to evaluate the association between disease aggressiveness and TTCR-C4 persistence. The test was chosen due to the small sample size and binary classification of persistence (long-term vs. short-term) and disease aggressiveness (aggressive vs. non- aggressive). Statistical significance was assessed with a p-value threshold of 0.05.
Flow-cytometry
Cryopreserved PBMCs were thawed and allowed to rest overnight in RPMI medium supplemented with 10% fetal bovine serum (R10). The cells underwent stimulation with a cocktail containing the WT1126-134 peptide at a final concentration of 1 μg/ml in R10 and intracellular cytokine staining using a 15-color staining panel, as previously described.9 Flow cytometry was conducted on an LSRII instrument (Becton Dickinson) with data acquisition using FACS-Diva software v.8.0.1. Flow cytometry data were subsequently analyzed using FlowJo v.10 (Treestar). Finally, the percentage of Tetramer+cytokine+ cells gated on the CD8+ T cell population was determined.
Spectral flow cytometry
Post-infusion PB samples were analyzed using a 5-laser Cytek Aurora. Antibodies used are listed in Supplementary Table 1. TTCR-C4 were identified by binding to the APC dye-labeled HLA-A2:WT1126-134 tetramer. Spectral flow-cytometry data were biexponentially transformed, compensated and preprocessed (aggregates and dead cell removal) in FlowJo V10 (TreeStar). Pregated CD8+ T cells were exported from FlowJo and loaded in R (v4.3.2). First we created a flowSet using flowCore (v2.12.2)66and subsequently analyzed the data using CATALYST (v1.24.0).67
Ki67 was then used to manually gate subsets of interest using the FlowJo software. Statistical analysis included Kruskal-Wallis to compare TTCR-C4_Temra and TTCR-C4_Tem percentages over time, and Wilcoxon test for comparing KLRG1+CD57+GNLY+ and Tim3+PD1+TIGIT+ TTCR-C4_Temra.
In-silico mass cytometry validation
Mass cytometry files used to generate an atlas of virus-specific CD8+ T cells29 were downloaded from https://zenodo.org/records/8330231. Data were analyzed using flowCore and CATALYST R packages as specified above.
Single Cell RNA Sequencing
Patients with persisting PB TTCR-C4 (> 3% of the total CD8+ T cells) at least until day 28 after infusion were selected for scRNAseq analysis. Available PBMCs or BMMCs were thawed, washed and loaded on a 10x Chromium Controller based on the 3’ Chromium or 5’ Chromium Single Cell V(D)J Reagent Kit manual (10x Genomics). Library preparation was performed as per manufacturer’s protocol with no modifications. Library quality was confirmed by TapeStation High Sensitivity (Agilent, evaluates library size), Qubit (Thermo Fisher, evaluates dsDNA quantity), and KAPA qPCR analysis (KAPA Biosystems, evaluates quantity of amplifiable transcript). Samples were mixed in equimolar fashion and sequenced on an Illumina HiSeq 2500 rapid run mode according to the standard 10X Genomics protocol. TCR target enrichment, 5’ gene expression library, and TCR library were carried out to the 5’ Chromium Single Cell V(D)J Reagent Kit manual (10x Genomics). The 10X Genomics software Cell Ranger (v2.0.0) was used to process the raw data FASTAQ files with default parameters. The EmptyDrops method68 was used to identify cells with low RNA contents. The “count” function was used to perform alignment, filtering, barcode counting and UMI counting. Reads were aligned to the hg38 human reference genome (Ensembl) and the known transgene codon-optimized sequence using Spliced Transcripts Alignment to a Reference (STAR).69
For V(D)J sequencing assembly and paired clonotype calling, we used CellRanger “vdj” function. This function leverages Chromium cellular barcodes and UMIs to assemble V(D)J transcripts for each cell. CellRanger V(D)J calling produces an output named “filtered_contig_annotations.csv” for each sample, which lists CDR3 amino acid and nucleotide sequences for single cells identified by their barcodes.
scRNAseq quality control and subsetting
The filtered feature matrices generated by the CellRanger pipeline were used for downstream quality control (QC) and analyses. We used the function read10xCounts from the R package DropletUtils (version 1.14.2) to load the CellRanger output in R as a SingleCellExperiment70 object. Doublet cells filtering was performed on each sample using the scds package (v1.10.0).71 QC and filtering were conducted using the scater R package (v1.22.0)72.71 Genes not detected across all the cells were removed and cells were filtered based on feature counts, the percentage of mitochondrial and ribosomal genes, and the number of expressed features. Cells with values beyond a specific threshold, between 1 and 3.5 median absolute deviations (MAD) from the median, were excluded. These MAD thresholds were established according to the quality of each sample. Features with a count greater than 1 in at least 3 cells were retained for downstream analysis. We then split cells by sample in 15 datasets, normalized, found the 2000 most variable genes and scaled for each dataset using Seurat (v4.3.0.9001) SplitObject, NormalizeData, FindVariableFeatures and ScaleData respectively.73,74 For batch correction, we used the FindIntegrationAnchors and IntegrateData functions from Seurat. The integrated dataset was then used for scaling (ScaleData) and dimensionality reduction (PCA and UMAP) using RunPCA and RunUMAP, respectively. UMAP dimension reduction and clustering were computed using the first 20 principal components (PCs). The number of PCs capturing most of the variation in our data was selected using Seurat function ElbowPlot which visualizes the standard deviation of each PC. Clusters were identified via shared-nearest-neighbor-based (SNN) clustering and further analyzed at a resolution of 0.6. CD8+ cell subset was identified using scGate (v1.0.1)34 and subsequently extracted with Seurat subset function.
scRNAseq CD8+ endogenous and TTCR-C4 analysis
The CD8+ subset was used to find the 2000 most variable features (FindVariableFeatures), scale the data (ScaleData), and run dimensionality reduction (runPCA and runUMAP). SNN was used for clustering with a resolution of 0.5 for downstream analysis. Differential gene expression across clusters or condition (e.g. AML(+) vs AML(-)) was computed using Seurat FindAllMarkers with default parameters. TCRC4- transgene+ cells were identified using scGate (v1.0.1). For visualization purposes, we used Seurat built-in functions alongside ComplexHeatmap (v2.15.1)75, scCustomize (v1.1.0.9001) (https://github.com/samuel-marsh/scCustomize) and SCP (https://github.com/zhanghao-njmu/SCP). The pan-cancer CD8+ single-cell reference atlas was built using the CD8.thisStudy_10X.seu.rds file35 downloaded from Zenodo and processed using the make.reference function from the ProjecTILs R package (v3.2.0).76 We then used Run.Projectils function, from the same package, to project the cell states from the query dataset onto the reference.
Cell trajectory inference was computed using the Monocle R package (v1.3.4), with UMAP used as dimensionality reduction. Single-gene expression patterns along the pseudotime were visualized using the plot_genes_in_pseudotime function in Monocle. RNA velocity was conducted by exporting the CD8+ T- cell Seurat object as an h5ad file using the R package seurat-disk (v0.0.0.9020) (https://github.com/mojaveazure/seurat-disk), loading this file in Python (v3.8.14) as AnnData object,77 and estimating velocities with the scvelo Python package (v0.2.4)41 using the deterministic model.77
We classified patients as AML(+) or AML(-) based on the presence of detectable blasts in BM or PB. Three patients lacked BM or PB evaluations at one timepoint each. For two of these patients, we used the donor- recipient (male-female) sex mismatch. Patient 8 at day 49 exhibited cells expressing the female-specific gene XIST alongside the AML-associated genes CD34 and WT1 (Extended data Fig. 15A); similarly, patient 25 expressed XIST in a RPS4Y1 (male-specific gene) negative region at days 7 and 28, thus classified as AML(+) at these timepoints (Extended data Fig. 15B). Patient 4 was categorized as AML(-) at day 100 and AML(+) at day 581, as previously reported.43
To assess the significance of differences in cell proportions per CD8+ cell state between groups (AML(+) vs AML(-)), a permutation test was applied. Specifically, the permutation_test and permutation_plot functions from the R package scProportionTest (version 0.0.0.9000) (https://github.com/rpolicastro/scProportionTest) were used with default parameters.
For scRNAseq in-silico validation, we compiled scRNAseq datasets from independent studies on AML20,22,23,44,45, lung cancer48, pancreatic cancer46 and melanoma.47 CD8+ T cells were identified and extracted using the scGate34 R package. These cells were then projected onto the pan-cancer CD8+ T cell reference atlas35 using ProjecTILs R package.
The TCR repertoire was analyzed using the R package scRepertoire (v1.10.1).78 Initially the degree of single cells clonal expansion was defined based on the number of cells sharing the same clonotype: Hyperexpanded (100 < X <= 500), Large (20 < X <= 100), Medium (5 < X <= 20), Small (1 < X <= 5), Single (0 < X <= 1) The function occupiedscRepertoire was used to visualize the degree of clonal expansion by cell-state over time.
In-vitro chronic antigen stimulation model
The in-vitro dysfunction model was established as previously described.42 The WT1+ K562 tumor cell line was transduced with lentiviral constructs to express HLA-A*02:01 and GFP, then sorted for purity using the Sony MA900 cell sorter. Cells where then cultured in media consisting of IMDM with GlutaMAX (Gibco, Life Technologies, #31980030) with 10% FBS, and 1% of penicillin/streptomycin.
TTCR37-45 cells were obtained following previously published protocols.43 We selected TCR-T cells targeting WT137-45 as K562 cell line primarily express the standard proteasome and is not lysed by TTCR-C4 targeting WT1126-134.43 T cells expressing an irrelevant virus-specific TCR were used as negative control. Co-cultures of T cells with K562 cells were established at a 1:1 and 1:4 E:T ratio (2.5 x 105 T cells, 2.5 x 105 or 1 x 106 tumor cells, respectively). After 3-4 days of coculture, 250 μl of the cell suspension was used for T cell counting and flow-cytometry staining. The remaining cell suspension was spun down, and cells were resuspended in fresh media. A Novocyte 3 lasers flow cytometer was used to quantify GFP+ tumor cells and T cells and to maintain constant E:T ratios by reseeding K562 cells. Notably, during the peak of T-cell expansion (day 9-13), the volumes of the cocultures were reduced to ensure the reseeding of an adequate number of tumor cells, thereby maintaining constant E:T ratios. This protocol was followed for 23 days.
Bulk RNA sequencing
Bulk RNA sequencing was performed using BGISEQ-500 platform at BGI Genomics. Briefly, total RNA was extracted using the Qiagen RNeasy Micro Kit according to the manufacturer’s instructions. For the construction of low input polyA mRNA-seq libraries, the SMARTseq (v4) Package was used. Sequencing was performed on a DNBseq T7 machine (MGI) with paired-end 150 bp reads, generating 30M raw reads per sample. Raw sequencing data were filtered and trimmed using the software Soapnuke developed by BGI Genomics. The filtered reads were then aligned to the reference transcriptome using Bowtie2 (v2.2.5). Gene read counts were subsequently generated from the alignment results using RSEM (v1.2.8).
To assess differences over time across conditions (TTCR37-45_D0, K562_D14, and K562_D23), we used the likelihood ratio test (LRT) with a full model of ∼Condition and a reduced model including only the intercept (reduced = ∼ 1). Normalized counts were obtained using the rlog transformation, and differential expression analysis was performed using the R package DESeq2 (v1.42.0). We manually curated a list of genes of interest (Supplementary Table 10) and filtered the results to retain only those genes that were significant (padj < 0.05) with an absolute log2 fold-change > 1). For visualization purposes, average normalized counts of biological replicates were calculated using the avereps function from the R package limma (v3.56.2) and visualized using the R package pheatmap (v1.0.12). To visualize the expression patterns of manually curated gene signatures, we calculated z-scores for the averaged normalized counts across conditions (TTCR37-45_D0, K562_D14, and K562_D23). Finally, to compare the enrichment of scRNAseq-derived gene signatures (top 50 differentially expressed genes) and of a manually curated exhaustion signature across conditions, we used the hciR (v1.7) function fgsea_all with default parameters and plot_fgsea for plotting.
Funding
We received funding from grant no. P01CA18029-41 (P.D.G.), grant no. NIH-5K08CA169485 (A.G.C.), the Immunotherapy Integrated Research Center at the Fred Hutchinson Cancer Research Center (A.G.C.), Damon Runyon (A.G.C.), the Guillot Family ZachAttacksLeukemia Foundation, Parker Institute for Cancer Immunotherapy (P.D.G.), Gabrielle’s Angel Foundation, the V Foundation, and Juno Therapeutics.
Conflicts of interest
A.G.C. has received support from Juno Therapeutics, Lonza, and Affini-T. P.D.G. is a consultant, has received support from and had ownership interest in Juno Therapeutics and Affini-T Therapeutics. He has also received support from Lonza, and consults and has ownership interest in Rapt Therapeutics, Elpisciences, Immunoscape, Earli, Metagenomi. Catalia, and Nextech. P.D.G., T.M.S., H.N.N. and the Fred Hutchinson Cancer Research Center have intellectual property related to TCR-C4. A.G.C. and K.G.P. have received reagents from 10X Genomics. R.G. has received consulting income from Takeda, Arcellx and Sanofi, and declares ownership in Ozette Technologies.. The authors declare no other competing interests.
Data sharing
For original data, please contact fmazziot{at}fredhutch.org and achapuis{at}fredhutch.org. Raw data will be deposited and made publicly available as per journal requirements.
Data Availability
Raw data will be deposited and made publicly available after peer-reviewed publication
Contributions
Conception and design were performed by P.D.G., A.G.C., F.M., M.B., and T.M.S. Collection and assembly of data were carried out by F.M., L.M., A.G.C., D.N.E., M.B., V.V., D.H. Data analysis and interpretation were performed by F.M., L.M., T.T., V.V., R.G., F.M., A.G.C., and P.D.G. All of the authors contributed to the writing of the manuscript. Final approval of the manuscript was given by all of the authors.
Acknowledgements
We thank all members of the Chapuis and Greenberg lab for their contribution to the manuscript; the Fred Hutchinson Cancer Center Good Manufacturing Practice Cell Processing Facility for generating TTCR-C4; the Immune Monitoring Laboratory for generating tetramers; the Flow Cytometry Facility for providing instruments and assistance in flow cytometry assays. We received funding from grant no. P01CA18029-41 (P.D.G.), grant no. NIH-5K08CA169485 (A.G.C.), NIH-T32CA080416 (M.C.L.), the Immunotherapy Integrated Research Center at the Fred Hutchinson Cancer Center (A.G.C., F.M.), Damon Runyon (A.G.C.), the Guillot Family ZachAttacksLeukemia Foundation, Parker Institute for Cancer Immunotherapy (P.D.G.), Gabrielle’s Angel Foundation, the V Foundation, and Juno Therapeutics. We thank all the patients who participated in this study.
References
- 1.↵
- 2.↵
- 3.↵
- 4.↵
- 5.↵
- 6.↵
- 7.↵
- 8.↵
- 9.↵
- 10.↵
- 11.↵
- 12.↵
- 13.
- 14.↵
- 15.↵
- 16.↵
- 17.↵
- 18.↵
- 19.↵
- 20.↵
- 21.↵
- 22.↵
- 23.↵
- 24.↵
- 25.↵
- 26.↵
- 27.↵
- 28.↵
- 29.↵
- 30.↵
- 31.↵
- 32.↵
- 33.↵
- 34.↵
- 35.↵
- 36.↵
- 37.↵
- 38.↵
- 39.↵
- 40.↵
- 41.↵
- 42.↵
- 43.↵
- 44.↵
- 45.↵
- 46.↵
- 47.↵
- 48.↵
- 49.↵
- 50.↵
- 51.↵
- 52.↵
- 53.↵
- 54.↵
- 55.
- 56.↵
- 57.↵
- 58.↵
- 59.↵
- 60.↵
- 61.↵
- 62.↵
- 63.↵
- 64.↵
- 65.↵
- 66.
- 67.↵
- 68.↵
- 69.↵
- 70.↵
- 71.↵
- 72.↵
- 73.↵
- 74.↵
- 75.↵
- 76.↵
- 77.↵
- 78.↵
- 79.↵
- 80.↵