Main

After acute infections or vaccinations, memory T (TMEM) cells form and provide long-term protection against cognate pathogens1. By contrast, the functional exhaustion of T cells has been well characterized in many chronic infections and cancers2. Tumor-infiltrating CD8+ T cells that specifically recognize tumor antigens manifest typical exhaustion-associated phenotypes, including progressive loss of effector functions, proliferative capacities and memory potential, while having elevated and sustained expression of a cluster of inhibitory immune checkpoint receptors (for example, PD-1, CTLA-4 and TIM-3)2. Blocking antibodies targeting these inhibitory immune checkpoint receptors, termed immune checkpoint blockade (ICB), can reinvigorate exhausted CD8+ T (TEX) cells in the TME, which thus lays the foundation for the clinical effects of ICB. Although ICB therapies targeting PD-1–programmed cell death ligand 1 (PD-L1) interaction have documented durable remissions in a subset of patients with cancer, the overall response rates remain modest and even inert in certain cancer types3, which might be partially explained by the fact that ICB therapies do not fundamentally alter the exhaustion-inherited epigenetic program in TEX cells and fail to reprogram these cells to differentiate into TMEM cells4,5.

Accumulating evidence has suggested that the specific recognition of tumor-derived antigens is restricted to a minor proportion of tumor-infiltrating T cells, while the vast majority of tumor-infiltrating T cells recognize tumor-irrelevant antigens, especially common viral antigens, and are thus defined as ‘TBYS cells’ (refs. 6,7,8,9,10,11,12,13,14,15). Tumor-infiltrating TBYS cells were reported to exhibit TMEM-associated phenotypes9,12,14,15; however, their detailed differentiation states are not well characterized. Particularly, it remains unknown whether CD8+ TBYS cells and CD4+ ones share similar memory traits in the TME. Furthermore, owing to the overwhelming number and function of TBYS cells compared to tumor-specific TEX cells in the TME, it would be of great therapeutic value if TBYS cells in situ could be repurposed to eliminate tumor cells.

OV represents a highly versatile platform in delivering various payloads into tumor cells16,17. Many DNA or RNA viruses with potent tumor tropism have been proposed as vectors for OV immunotherapies, such as herpes simplex virus (HSV), adenovirus and Newcastle disease virus (NDV)16,17. To this day, OV immunotherapies have been investigated in ~100 reported clinical trials, in which the modified HSV encoding granulocyte–macrophage colony-stimulating factor (termed T-VEC) has been approved by the US Food and Drug Administration17 and another triple-mutated HSV (termed as G47Δ) has been approved to treat recurrent glioblastoma in Japan18. It remains unknown whether OV can be exploited to drive tumor cells to present TBYS cell-specific antigens, thus causing tumor cells to be eradicated by TBYS cells of functional memory properties in the TME.

Results

Features of tumor-infiltrating virus-specific TBYS cells

Although virus-specific TBYS cells have been reported by accumulating evidence6,7,8,9,10,11,12,13,14,15, a detailed portrait of these specialized T cells, especially CD4+ TBYS cells, is lacking. We thus sought to analyze the kinetics, molecular characteristics and lineage relationship with other memory types of both CD8+ and CD4+ TBYS populations in the TME. To this end, congenic naive (CD45.1+CD44loCD62Lhi) P14 CD8+ T cells recognizing the lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) epitope H-2DbGP33–41 and congenic naive (CD45.1+CD44loCD62Lhi) SMARTA (SM) CD4+ T cells recognizing the LCMV GP epitope I-AbGP66–77 were adoptively transferred into C57BL/6 recipients (CD45.2+), which were then infected with LCMV Armstrong to establish acute viral infection and resultant virus-specific P14 TMEM and SM TMEM cells. On day 60 after infection, recipients were subcutaneously engrafted with syngeneic MC38 colon adenocarcinoma cells and tumor-infiltrating P14 CD8+ and SM CD4+ T cells, and TBYS populations were analyzed on days 10, 15 and 20 after tumor engraftment (Fig. 1a and Extended Data Fig. 1a). As shown, frequencies of tumor-infiltrating P14 TBYS and SM TBYS cells remained stable at the different indicated time points (Fig. 1b,c), suggesting that the infiltrating virus-specific TBYS cells were not altered at the population level during tumor progression.

Fig. 1: Features of tumor-infiltrating virus-specific TBYS cells.
figure 1

a, Schematic of the experimental design. Congenic CD45.1+ P14 CD8+ T cells and CD45.1+ SM CD4+ T cells were adoptively transferred into naive C57BL/6 recipients (CD45.2+), which were then infected with LCMV Armstrong and engrafted with MC38 cells on day (D)60 after infection. On days 10, 15 and 20 after tumor engraftment, tumor-infiltrating P14 and SM cells were analyzed. b, Flow cytometry analysis of MC38 tumor-infiltrating CD8+ (top) and CD4+ (bottom) T cells on days 10, 15 and 20 after tumor engraftment. Numbers adjacent to the outlined areas indicate percentages of CD45.1+Vα2+ P14 cells (blue) of tumor-infiltrating CD8+ T cells or CD45.1+Vα2+ SM cells (red) of tumor-infiltrating CD4+ T cells. c, Frequency of P14 TBYS cells of total MC38 tumor-infiltrating CD8+ T cells (indicated by blue dots, n = 5 mice (day 10) or n = 4 mice (day 15 and day 20)) and frequency of SM TBYS cells of total MC38 tumor-infiltrating CD4+ T cells (indicated by red cubes, n = 5 mice (day 10) or n = 4 mice (day 15 and day 20)) at the indicated time points. d, Frequencies of P14 TMEM, P14 TEX and P14 TBYS cells that express KLRG1 and/or CD127 (top; n = 4 mice (P14 TMEM), n = 3 mice (P14 TEX) or n = 5 mice (P14 TBYS)), coexpress the indicated number of inhibitory receptors (PD-1, TIM-3, LAG-3 and 2B4) (middle; n = 5 mice (P14 TMEM), n = 3 mice (P14 TEX) or n = 4 mice (P14 TBYS)) or coexpress the indicated number of cytokines (IFN-γ and tumor necrosis factor (TNF)-α) and cytotoxic degranulation markers (CD107a and CD107b) (bottom; n = 5 mice (P14 TMEM), n = 3 mice (P14 TEX) or n = 3 mice (P14 TBYS)). e, Comparison of TCF-1, Eomes, T-bet and TOX expression levels among P14 TMEM (n = 3 mice), P14 TEX (n = 3 mice) and P14 TBYS cells (n = 5 mice). MFI, mean fluorescence intensity. f, A Radviz projection of P14 TMEM, P14 TRM (kidney, liver, salivary gland (SG) and small intestine intra-epithelial lymphocyte (siIEL)) and P14 TBYS cells based on nine gene expression programs (Pro_1–Pro_9) inferred by consensus non-negative matrix factorization. A total of 38,880 P14 cells passed quality control. g, Heatmap showing selected DEGs in various P14 cells. h, UMAPs of SM TBYS (left) and SM TMEM cells (right). A total of 3,565 SM TBYS cells are colored in yellow, and a total of 5,648 SM TMEM cells are colored in blue. i, UMAPs showing expression levels of Ly6c2, Gzmb, Foxp3 and Cxcr5 in SM TBYS and SM TMEM cells. j, Frequency of SM TMEM, SM TTST and SM TBYS cells that coexpress the indicated number of inhibitory receptors (PD-1, TIM-3, LAG-3 and 2B4) (top; n = 3 mice (SM TMEM), n = 3 mice (SM TTST) or n = 3 mice (SM TBYS)) or coexpress the indicated number of cytokines (IFN-γ and TNF-α) and cytotoxic degranulation markers (CD107a and CD107b) (bottom; n = 3 mice (SM TMEM), n = 3 mice (SM TTST) or n = 5 mice (SM TBYS)). k, Comparison of TCF-1, Eomes, T-bet and TOX expression levels among SM TMEM (n = 3 mice), SM TTST (n = 3 mice) and SM TBYS (n = 5 mice) cells. TFs, transcription factors. l, Gene set scoring analysis of 3,565 SM TBYS cells and 5,648 SM TMEM cells. Density of cells in the TCM versus TRM score space annotated by cell type is depicted. m, Expression profiles of Cd69, Rgs1, Nr4a1 and Bhlhe40 of SM TBYS and SM TMEM cells in the TCM versus TRM score space. Data (be,j,k) are representative of two independent experiments. One-way ANOVA with Turkey’s test was used in e,k. Center values and error bars (c,e,k) indicate mean and s.e.m.

Source data

Next, we set out to compare the molecular characteristics of virus-specific CD8+ TBYS cells with their counterparts recognizing the same epitope but induced to differentiate into TMEM or TEX cells. Accordingly, congenic naive P14 CD8+ T cells were adoptively transferred into C57BL/6 recipients, which were infected with the LCMV Armstrong strain to induce the differentiation of P14 TMEM cells. Furthermore, a fraction of these infected recipients was subcutaneously engrafted with MC38 cells on day 60 after infection, and tumor-infiltrating P14 TBYS cells were analyzed on day 10 after tumor engraftment. In the scenario of tumor-specific TEX cells, congenic naive P14 CD8+ T cells were adoptively transferred into C57BL/6 recipients, which were then subcutaneously engrafted with syngeneic B16F10 melanoma cells expressing the LCMV GP (hereafter referred to as B16-GP cells)19 and killed on day 15 after tumor engraftment to analyze tumor-specific P14 TEX cells (Extended Data Fig. 1b).

Resembling P14 TMEM cells, we found that P14 TBYS cells are mainly populated by the killer cell lectin-like receptor G1 (KLRG1)loCD127hi subset1 (Fig. 1d). Furthermore, P14 TBYS cells retain an expression pattern of transcription factors similar to that of P14 TMEM cells1, as exemplified by high expression of TCF-1 and Eomes and low expression of T-bet (Fig. 1e). In contrast to TMEM cells, TEX cells are characterized by coexpression of multiple inhibitory receptors, impaired cytokine secretion and the transcription factor TOX associated with T cell exhaustion2,20,21,22,23. Although preserved in the immunosuppressive TME, P14 TBYS cells show limited expression of inhibitory receptors and TOX and exhibit polyfunctional cytokine secretion upon ex vivo re-stimulation (Fig. 1d,e). Thus, these findings confirm that CD8+ TBYS cells phenotypically and functionally resemble CD8+ TMEM cells but not CD8+ TEX cells.

To further define the molecular traits of CD8+ TBYS cells, we performed single-cell RNA sequencing (scRNA-seq) of P14 TBYS cells from the TME of MC38-engrafted mice. Conventional P14 TMEM and tissue-resident memory (TRM) cells from external scRNA-seq datasets24,25 were introduced as controls (Extended Data Fig. 1c). A Radviz projection based on nine gene expression programs inferred by consensus non-negative matrix factorization clearly distinguished P14 TMEM cells from P14 TRM cells of various non-lymphoid tissues (Fig. 1f and Extended Data Fig. 1d,e). Importantly, P14 TBYS cells were drawn toward the groups of P14 TRM cells (Fig. 1f), likely representing the acquisition of certain TRM cell features. Further unbiased hierarchical clustering confirmed that P14 TBYS cells are in close proximity with P14 TRM cells and highlighted by cytolytic activity (Gzmb, Tnf) and tissue residency (Cd69, Nr4a1)26 (Fig. 1g). The potential residency of P14 TBYS cells was also evidenced by high expression of CD69 protein (Extended Data Fig. 2a,b). Therefore, these findings suggest TRM cell features of CD8+ TBYS cells.

Next, we aimed to trace the differentiation of virus-specific CD4+ TBYS cells by performing scRNA-seq of SM TBYS cells from the MC38 TME with conventional SM TMEM cells as controls (Extended Data Fig. 1c). Consistent with previous studies27,28, SM TMEM cells adopt a bifurcation of follicular helper T cells (Cxcr5) and type 1 helper T (TH1) cells (Ly6c2, Gzmb) (Fig. 1h,i). Instead of adopting a follicular helper T/TH1 bifurcation, we found that SM TBYS cells were predominantly fated toward the TH1 lineage (Fig. 1h,i). Indeed, TH1-type SM TBYS cell differentiation was further evidenced by abundant Ly6C protein but rare expression of C–X–C motif chemokine receptor (CXCR)5 (Extended Data Fig. 2c–e). Furthermore, similar to P14 TBYS cells, SM TBYS cells also showed high expression of TCF-1 and Eomes and low expression of T-bet (Fig. 1k), suggesting the retention of memory properties. In addition, SM TBYS cells showed no features related to exhaustion, including multiple inhibitory receptors29 and TOX30, as compared to tumor antigen-specific T (TTST) cells (Fig. 1j,k); instead, SM TBYS cells exhibit a substantial capacity to secrete multiple cytokines upon ex vivo re-stimulation (Fig. 1j). These results define the memory properties of TH1-type CD4+ TBYS cells in the TME.

Previous studies stated distinct molecular signatures between central memory T (TCM) and TRM cells31,32. We therefore examined the enrichment for TCM and TRM cell gene signatures in SM TBYS cells, with conventional SM TMEM cells being introduced as controls, to explore the effector–memory state in the SM TBYS population. We found a largely overlaid TCM signature between SM TBYS and SM TMEM cells, with a noticeable dissection in the TRM signature (Fig. 1l). Indeed, genes related to TRM cells (for example, Cd69, Rgs1, Nr4a1 and Bhlhe40) were highly expressed in SM TBYS cells relative to those in SM TMEM cells (Fig. 1m); meanwhile, genes related to TCM cells (for example, Il7r, Tcf7 and Ccr7) were largely overlapping between SM TBYS and SM TMEM cells (Extended Data Fig. 2f). Distinct from SM TBYS cells, P14 TBYS cells were dominated by the gene signature of TRM cells rather than that of TCM cells (Extended Data Fig. 2g). Thus, these findings highlight a unique hybrid gene signature consisting of both TCM and TRM cell features in CD4+ TBYS cells but not in CD8+ TBYS cells.

We further explored endogenous virus-specific TBYS cells from MC38 tumors engrafted in LCMV-infected mice by quantifying interferon (IFN)-γ-expressing cells upon ex vivo stimulation with cognate viral peptides (Extended Data Fig. 3a). Consistently, we observed the presence of endogenous CD8+ TBYS cells specific to LCMV GP33–41, LCMV GP276–286 and LCMV nucleoprotein (NP)396–404 and endogenous CD4+ TBYS cells specific to LCMV GP66–77 in the MC38 tumors engrafted in LCMV-infected mice (Extended Data Fig. 3b–e). In addition to the MC38 tumor model, endogenous LCMV-specific TBYS cells were also found in the TME of B16F10-engrafted mice (Extended Data Fig. 3f–h). In sum, these findings highlight the functional memory characteristics of virus-specific CD8+ and CD4+ TBYS cells in the TME.

OV-BYTE therapy elicits anti-tumor response

Given the abundant memory pool of virus-specific TBYS cells in the TME, we hypothesized that OV-mediated delivery of TBYS cell epitopes (hereafter referred to as OV-BYTE) to tumor cells might redirect the cytotoxicity of TBYS cells toward tumor cells and thus curtail tumor progression. For this purpose, the gene encoding LCMV GP was inserted into an NDV rSG10 strain (referred to as NDV wild type (WT)) and thus termed NDV-GP (Extended Data Fig. 4a). To assess the ability of NDV-GP to infect and deliver LCMV GP expression in tumor cells, we infected MC38 cells with NDV-GP and then detected LCMV GP expression in the infected MC38 cells. In accordance with the previously reported highly efficient transgene delivery of oncolytic NDV33,34, we found that NDV-GP substantially infected MC38 cells and expressed the LCMV GP transgene (Extended Data Fig. 4b–d). Furthermore, we also found enhanced expression of major histocompatibility complex (MHC)-I and MHC-II molecules in NDV-GP-infected tumor cells and specific killing of these tumor cells by LCMV GP-specific T cells (Extended Data Fig. 4e–g), indicating MHC presentation of LCMV GP epitopes by NDV-GP-infected tumor cells to LCMV GP-specific T cells.

Next, we aimed to assess the in vivo anti-tumor activities of NDV-GP in tumor-bearing mice that have established immune memory for LCMV. To this end, naive C57BL/6 mice were infected with LCMV Armstrong, followed by subcutaneously engraftment with MC38 cells on day 60 after infection and then intratumoral treatment with phosphate-buffered saline (PBS), NDV-WT or NDV-GP (Fig. 2a). Remarkably, NDV-GP treatment delayed tumor growth more efficiently than NDV-WT treatment (Fig. 2b) and led to ~30% complete remission on day 50 after tumor engraftment (Fig. 2c). Moreover, NDV-GP treatment did not show any therapeutic advantages over NDV-WT treatment in tumor-engrafted mice without pre-existing immune memory (Fig. 2d,e); meanwhile, the therapeutic function of NDV-GP to treat MC38 tumors engrafted in LCMV-infected mice was abolished in the presence of neutralizing antibodies specific to LCMV GP proteins (Fig. 2f,g).

Fig. 2: OV-mediated delivery of TBYS cell epitopes to solid tumor cells elicits anti-tumor response.
figure 2

a, Schematic of the experimental design for b,c. C57BL/6 mice were infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally administered PBS, NDV-WT or NDV-GP. b,c, Tumor growth curve (b) and Kaplan–Meier survival curve (c) of MC38 tumor-bearing mice intratumorally treated with PBS (n = 6 mice), NDV-WT (n = 10 mice) or NDV-GP (n = 10 mice) as described in a. d, Schematic of the experimental design for e. Naive C57BL/6 mice were engrafted with MC38 cells and then intratumorally administered PBS, NDV-WT or NDV-GP on days 7, 8, 9, 11 and 12 after tumor engraftment. e, Kaplan–Meier survival curve of MC38 tumor-bearing mice intratumorally treated with PBS (n = 12 mice), NDV-WT (n = 8 mice) or NDV-GP (n = 9 mice) as described in d. f, Schematic of the experimental design for g. LCMV Armstrong-infected C57BL/6 mice were engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally administered PBS, NDV-WT or NDV-GP in the presence or absence of LCMV GP-neutralizing antibodies. g, Tumor growth curve of MC38 tumor-bearing mice intratumorally treated with PBS, NDV-WT or NDV-GP with or without LCMV GP-neutralizing antibodies (NAbs) as described in f. PBS (n = 10 (control) or n = 9 (neutralizing antibodies) mice), NDV-WT (n = 9 (control) or n = 9 (neutralizing antibodies) mice) and NDV-GP (n = 9 (control) or n = 12 (neutralizing antibodies) mice). h, Schematic of the experimental design for i. C57BL/6 mice were infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally administered PBS, NDV-WT or NDV-NP. i, Kaplan–Meier survival curve of MC38 tumor-bearing mice intratumorally treated with PBS (n = 9 mice), NDV-WT (n = 9 mice) or NDV-NP (n = 11 mice) as described in h. j, Schematic of the experimental design for k. C57BL/6 mice were infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally administered PBS, Ad5-WT or Ad5-GP. k, Kaplan–Meier survival curve of MC38 tumor-bearing mice intratumorally treated with PBS (n = 10 mice), Ad5-WT (n = 9 mice) or Ad5-GP (n = 7 mice) as described in j. l, Schematic of the experimental design for m. C57BL/6 mice were infected with LCMV Armstrong and engrafted with B16F10 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally administered PBS, NDV-WT or NDV-GP. m, Kaplan–Meier survival curve of B16F10 tumor-bearing mice intratumorally treated with PBS (n = 6 mice), NDV-WT (n = 8 mice) or NDV-GP (n = 8 mice) as described in l. n, Schematic of the experimental design for o. C57BL/6 mice were infected with LCMV Armstrong, and three plasmids (encoding Myc-tagged NICD1, Myc-tagged AKT and hyperactive sleeping beauty transposase (SB100)) were injected into the hydrodynamic tail vein on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intravenously administered PBS, NDV-WT or NDV-GP. o, Kaplan–Meier survival curve of mice with NICD- and AKT-induced murine intrahepatic cholangiocarcinoma as described in n. PBS (n = 6 mice), NDV-WT (n = 5 mice) and NDV-GP (n = 6 mice). p, Schematic of the experimental design for q,r. NCG mice were engrafted with PBMCs from HLA-A2-positive donors with a history of H1N1 infection. After reconstruction of human T cells, these humanized NCG mice were engrafted with A375 cells and then intratumorally administered PBS, NDV-WT or NDV-H1N1 NP on days 7, 8, 9, 11 and 12 after tumor engraftment. q,r, Tumor growth curve (q) and Kaplan–Meier survival curve (r) of A375 tumor-bearing humanized NCG mice intratumorally treated with PBS (n = 5 mice), NDV-WT (n = 6 mice) or NDV-H1N1 NP (n = 5 mice) as described in p. All data are representative of at least two independent experiments. Two-way ANOVA was used to compare tumor growth curves in b,g,q. The log-rank (Mantel–Cox) test was performed to compare survival curves among groups in c,e,i,k,m,o,r. Center values and error bars (b,g,q) indicate mean and s.e.m.

Source data

In parallel, we also constructed the NDV strain carrying the LCMV NP gene (referred to as NDV-NP) to treat MC38-bearing mice with LCMV immune memory (Fig. 2h and Extended Data Fig. 4a,h). Similarly, NDV-NP treatment led to significant tumor growth delays compared to NDV-WT treatment and generated ~30% complete regression (Fig. 2i). To test whether another non-NDV OV platform could also be used to target TBYS cells to limit tumor progression, LCMV Armstrong-infected C57BL/6 mice were engrafted with MC38 cells and then intratumorally administered the oncolytic adenovirus serotype 5 (Ad5) that carries the LCMV GP gene (referred to here as Ad5-GP) (Fig. 2j and Extended Data Fig. 4i,j). Similarly, Ad5-GP treatment effectively slowed tumor growth and resulted in marked tumor regression (~25% complete regression) compared to control treatments (Fig. 2k).

In addition to MC38 colon adenocarcinoma, the therapeutic functions of NDV-GP were also evident in a B16F10 melanoma model of weak immunogenicity (Fig. 2l,m and Extended Data Fig. 4k). Aside from transplanted tumor models, NDV-GP administration also largely improved the survival rates of mice with LCMV memory and Notch intracellular domain (NICD)- and AKT-induced autochthonous intrahepatic cholangiocarcinoma35 (Fig. 2n,o), suggesting the potential efficacy of OV-BYTE therapy in different cancer types.

To more closely examine the clinical relevance of OV-BYTE therapy, we further recruited a cohort of human leukocyte antigen (HLA)-A2-positive healthy donors with a history of influenza A (H1N1) infection (Extended Data Fig. 4l). The peripheral blood mononuclear cells (PBMCs) of these donors, which contain memory CD8+ and CD4+ T cells specific to the H1N1 NP epitope (Extended Data Fig. 4m,n), were used to develop humanized mice by engraftment of human PBMCs into NCG mice (Fig. 2p). Humanized NCG mice with an appropriate reconstruction of human T cells were engrafted with HLA-A2-matched A375 human melanoma cells to make a cell line-derived xenograft (CDX). Next, these CDX mice were intratumorally administered PBS, NDV-WT or the NDV strain expressing H1N1 NP (referred to as NDV-H1N1 NP) (Fig. 2p and Extended Data Fig. 4o). Remarkably, treatment with NDV-H1N1 NP restricted tumor growth of A375 melanoma and prolonged the overall survival of CDX mice (Fig. 2q,r). Together, these data demonstrate the potential anti-tumor functionality of OV-BYTE therapy.

OV-BYTE therapy is attributed to CD8+ and CD4+ TBYS cells

To determine the cellular mechanism(s) underlying OV-BYTE therapy, we next analyzed the composition of important immune cells within NDV-GP-administered MC38 tumors in mice with LCMV memory (Extended Data Fig. 5a). We found that numbers of CD45+ immune cells were increased in tumors treated with NDV-GP compared with those of tumors treated with PBS or NDV-WT (Extended Data Fig. 5b). Moreover, the NDV-GP-mediated increased immune cell number was mainly attributed to T cells and dendritic cells (DCs) (Extended Data Fig. 5c). Further analysis of T cells revealed increased numbers of both CD8+ and CD4+ T cells in NDV-GP-treated tumors (Extended Data Fig. 5d). In addition, the ratio of CD8+ T cells or conventional CD4+ T cells to regulatory CD4+ T cells was largely enhanced upon NDV-GP treatment (Extended Data Fig. 5e–h). Thus, these findings suggest that NDV-GP treatment favors CD8+ and CD4+ T cell responses in the TME.

To decipher the contributions of CD8+ and CD4+ T cells to the therapeutic effects of NDV-GP administration, we carried out NDV-GP treatment with CD8+ or CD4+ T cell depletion (Fig. 3a). As shown, depletion of either CD8+ or CD4+ T cells similarly abolished the therapeutic effects of NDV-GP and led to a significant reduction in long-term survival of MC38 tumor-bearing mice with LCMV memory (Fig. 3b). To further confirm the important roles of virus-specific CD8+ or CD4+ TBYS cells in the OV-BYTE strategy, LCMV-specific memory CD8+ and/or CD4+ T cells were collected and adoptively transferred into naive C57BL/6 recipients, which were then engrafted with MC38 cells and intratumorally administered PBS, NDV-WT or NDV-GP (Fig. 3c). Compared to the NDV-GP-treated group with no T cell transfer, single adoptive transfer of memory CD8+ or CD4+ T cells resulted in limited tumor retardation in mice administered NDV-GP (Fig. 3d). Remarkably, the combined transfer of memory CD8+ and CD4+ T cells synergized to better control tumor growth in the NDV-GP-treated group than in monotransfer of memory CD8+ or CD4+ T cells (Fig. 3d). Nevertheless, the anti-tumor effects of LCMV-specific memory CD8+ and/or CD4+ T cell transfer were abolished upon PBS or NDV-WT treatment (Fig. 3d). Therefore, these observations indicate that OV-BYTE therapy can target virus-specific CD8+ and CD4+ TBYS cells and efficiently redirect their cytotoxicity toward tumor cells.

Fig. 3: Anti-tumor effects of OV-BYTE therapy are attributed to both virus-specific CD8+ and CD4+ TBYS cells.
figure 3

a, Schematic of the experimental design for b. Naive C57BL/6 mice were infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally administered PBS, NDV-WT or NDV-GP. Meanwhile, recipients were intraperitoneally injected with depleting antibodies for CD8+ or CD4+ T cells at the indicated time points. b, Kaplan–Meier survival curve of MC38 tumor-bearing mice intratumorally treated with PBS or NDV-GP in the presence or absence of depleting antibody for CD8+ (αCD8) or CD4+ (αCD4) T cells as described in a. PBS (n = 11 mice), NDV-GP (n = 11 mice), PBS and anti-CD8 antibody (n = 9 mice), NDV-GP and anti-CD8 antibody (n = 7 mice), PBS and anti-CD4 antibody (n = 12 mice), NDV-GP and anti-CD4 antibody (n = 12 mice). c, Schematic of the experimental design for d. Splenic memory CD8+ and/or CD4+ T cells were collected from LCMV Armstrong-infected mice (day 60) and adoptively transferred into naive C57BL/6 mice. These recipients were then subcutaneously engrafted with MC38 cells 1 day after adoptive cell transfer and intratumorally treated with PBS, NDV-WT or NDV-GP on days 7, 8, 9, 11 and 12 after tumor engraftment. d, Tumor growth curves of PBS (left; n = 9 (no cells), n = 8 (CD8+ T cells), n = 8 (CD4+ T cells) or n = 8 (CD8+ T cells and CD4+ T cells) mice), NDV-WT (middle; n = 9 (no cells), n = 8 (CD8+ T cells), n = 8 (CD4+ T cells) or n = 9 (CD8+ T cells and CD4+ T cells) mice) or NDV-GP (right; n = 9 (no cells), n = 8 (CD8+ T cells), n = 8 (CD4+ T cells) or n = 5 (CD8+ T cells and CD4+ T cells) mice) -treated MC38 tumor-bearing mice receiving adoptive transfer of LCMV Armstrong-activated CD8+ and/or CD4+ T cells or receiving no cell transfer as described in c. All data are representative of at least two independent experiments. The log-rank (Mantel–Cox) test was performed to compare survival curves among groups in b. Two-way ANOVA was used to compare tumor growth curves in d. Center values and error bars (d) indicate mean and s.e.m.

Source data

OV-BYTE therapy provokes cytotoxic CD4+ TBYS cells

To ascertain how TBYS cells respond to OV-BYTE therapy, C57BL/6 mice initially receiving transfer of congenic P14 and SM cells were infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. After five injections of PBS, NDV-WT or NDV-GP, P14 and SM TBYS cells were analyzed (Fig. 4a). Notably, we found that both the frequency and number of total P14 TBYS cells were comparable among PBS, NDV-WT and NDV-GP groups in the TME (Fig. 4b). Consistently, NDV-GP treatment showed no effects in boosting the proliferation of P14 TBYS cells (Fig. 4b). Similarly, the frequencies and numbers of endogenous CD8+ TBYS cells specific to LCMV GP33–41, GP91–101, GP118–125 and GP276–286 were also not influenced by NDV-GP treatment (Extended Data Fig. 5i–m). These results highlighted characteristics similar between CD8+ TBYS cells and TRM cells, both of which immediately differentiate into effector cells but with much less efficient expansion upon antigen rechallenge than TCM cells36.

Fig. 4: OV-BYTE therapy expands and provokes the cytotoxic effector functions of virus-specific CD4+ TBYS cells.
figure 4

a, Schematic of the experimental design. Congenic CD45.1+ P14 CD8+ T cells and CD45.1+ SM CD4+ T cells were adoptively transferred into naive C57BL/6 recipients (CD45.2+), which were then infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally treated with PBS, NDV-WT or NDV-GP. On day 14 after tumor engraftment, tumor-infiltrating P14 and SM cells were analyzed. b, Frequency of P14 TBYS cells of tumor-infiltrating CD8+ T cells (left), number of P14 TBYS cells (middle) and frequency of Ki-67+ cells of P14 TBYS cells (right) from PBS (n = 5 mice), NDV-WT (n = 6 mice) and NDV-GP (n = 5 mice) -treated MC38 tumors. c, Frequency of SM TBYS cells of tumor-infiltrating CD4+ T cells (left), number of SM TBYS cells (middle) and frequency of Ki-67+ cells of SM TBYS cells (right) from PBS (n = 5 mice), NDV-WT (n = 6 mice) and NDV-GP (n = 5 mice) -treated MC38 tumors. d, UMAP analysis of P14 TBYS and SM TBYS cells upon PBS (left, 6,571 cells), NDV-WT (middle, 5,397 cells) or NDV-GP (right, 5,951 cells) treatment. e, Bubble chart showing selected genes in each cluster of P14 TBYS and SM TBYS cells as described in d. f, Volcano plot showing DEGs between SM C2 and SM C1 and SM C3 in d. FC, fold change; NS, not significant. g, Flow cytometry analysis of tumor-infiltrating SM TBYS cells from the PBS-, NDV-WT- or NDV-GP-treated group. Numbers adjacent to the outlined areas indicate percentages of TCF-1hiT-betlo cells (blue) or TCF-1loT-bethi cells (red) of SM TBYS cells. h, Frequencies of TCF-1hiT-betlo cells (left) or TCF-1loT-bethi cells (right) of SM TBYS cells upon PBS (n = 5 mice), NDV-WT (n = 6 mice) or NDV-GP (n = 5 mice) treatment. i, Flow cytometry analysis of total SM TBYS cells of the PBS- or NDV-WT-treated group and TCF-1hiT-betlo or TCF-1loT-bethi SM TBYS cells of the NDV-GP-treated group. Numbers adjacent to the outlined areas indicate percentages of GzmB+ cells of the indicated PBS- or NDV-WT-treated SM TBYS cells or NDV-GP-treated TCF-1hiT-betlo or TCF-1loT-bethi SM TBYS cells. j, Frequency of GzmB+ cells of PBS (n = 6 mice) or NDV-WT (n = 6 mice) -treated SM TBYS cells or NDV-GP-treated (n = 6 mice) TCF-1hiT-betlo or TCF-1loT-bethi SM TBYS cells. k, Ex vitro killing efficiency of SM TBYS cells from the PBS (n = 3 mice) or NDV-WT (n = 3 mice) -treated group and Ly108hiCD39lo or Ly108loCD39hi SM TBYS cells from the NDV-GP-treated group (n = 3 mice). l, Schematic of the experimental design for m. Naive CD45.2+ C57BL/6 mice receiving adoptive transfer of congenic CD45.1+ SM CD4+ T cells were infected with LCMV Armstrong and then engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally treated with NDV-GP. On day 14 after tumor engraftment, tumor-infiltrating Ly108hiCD39lo or Ly108loCD39hi SM TBYS cells were isolated and transferred into a new cohort of C57BL/6 mice, which were not exposed to LCMV Armstrong and engrafted with MC38 cells 6 days before the SM TBYS cell transfer. These new recipients were next intratumorally administered PBS, NDV-WT or NDV-GP on day 7 to day 12 after tumor engraftment. m, Tumor growth curve of MC38 tumor-bearing mice treated with PBS, NDV-WT or NDV-GP upon Ly108hiCD39lo (n = 10 (PBS), n = 10 (NDV-WT) or n = 10 (NDV-GP) mice) or Ly108loCD39hi (n = 7 (PBS), n = 8 (NDV-WT) or n = 8 (NDV-GP) mice) SM TBYS cell transfer or no cell (n = 9 (PBS), n = 9 (NDV-WT) or n = 9 (NDV-GP) mice) transfer as described in l. i.t., intratumoral. n, Schematic of the experimental design for o. Naive WT C57BL/6 mice and naive GzmbKO mice were infected with LCMV Armstrong. On day 60 after infection, splenic CD44+CD127+ memory CD4+ T cells were isolated from WT or GzmbKO mice and then adoptively transferred into a new cohort of C57BL/6 mice. Next, these recipients were engrafted with MC38 cells and intratumorally treated with PBS, NDV-WT or NDV-GP on days 7, 8, 9, 11 and 12 after tumor inoculation. o, Tumor growth curve of MC38 tumor-bearing mice treated with PBS (n = 9 (no cells), n = 8 (WT CD4+ T cells) or n = 8 (GzmbKO CD4+ T cells) mice), NDV-WT (n = 9 (no cells), n = 10 (WT CD4+ T cells) or n = 9 (GzmbKO CD4+ T cells) mice) or NDV-GP (n = 10 (no cells), n = 8 (WT CD4+ T cells) or n = 10 (GzmbKO CD4+ T cells) mice) upon WT or GzmbKO memory CD4+ T cell transfer or no cell transfer as described in n. Data (b,c,gk,m,o) are representative of two independent experiments. One-way ANOVA with Turkey’s test was used in b,c,h,j,k. The Wilcoxon signed-rank test was used in f. Two-way ANOVA was used to compare tumor growth curves in m,o. Center values and error bars (b,c,h,j,k,m,o) indicate mean and s.e.m.

Source data

In the scenario of CD4+ TBYS cells, we found that, in contrast to CD8+ TBYS cells, NDV-GP treatment dramatically boosted both the frequency and number of SM TBYS cells, accompanied by increased cell proliferation (Fig. 4c). Consistently, endogenous LCMV GP66–77-specific CD4+ TBYS cells were also greatly expanded by NDV-GP treatment (Extended Data Fig. 5n). Thus, a biased robust increment of CD4+ TBYS cells over CD8+ TBYS cells is provoked by NDV-GP treatment.

Next, we sought to delineate the molecular patterns of NDV-GP-boosted TBYS cells in the TME. To this end, P14 and SM TBYS cells of MC38 tumors treated with PBS, NDV-WT or NDV-GP were used for scRNA-seq analyses (Fig. 4a). A total of eight clusters were identified by uniform manifold approximation and projection (UMAP) analysis and included four clusters populated by P14 TBYS cells (P14 clusters 1–4, C1–C4) and another four clusters populated by SM TBYS cells (SM C1–C4) (Fig. 4d,e). Within P14 TBYS cells, each cluster was evenly distributed among PBS-, NDV-WT- and NDV-GP-treated groups (Extended Data Fig. 6a,b). Moreover, we found minimally changed gene signatures in NDV-GP-treated P14 TBYS cells as compared to PBS- or NDV-WT-treated ones (Extended Data Fig. 6c), suggesting that NDV-GP administration may not fundamentally alter gene signatures in P14 TBYS cells of inherited TRM properties32. By contrast, cluster composition of SM TBYS cells was dramatically altered upon NDV-GP treatment, with which a pronounced enrichment of SM C2 was observed (Fig. 4d and Extended Data Fig. 6a,b). In agreement with the changed cluster composition, RNA velocity analysis further revealed a SM C2-directed differentiation in NDV-GP-educated SM TBYS cells (Extended Data Fig. 6d), suggesting that SM TBYS cells populating C2 might be unique responders to NDV-GP treatment.

To characterize the molecular signature and functions of NDV-GP-educated SM TBYS cells, we further determined differentially expressed genes (DEGs) between SM C2 and other SM clusters. A total of 1,175 and 264 DEGs were upregulated and downregulated, respectively, in C2 of SM TBYS cells. Specifically, expression of genes involved in cytotoxic functions (for example, Gzmb, Ccl5, Tbx21) was largely enhanced in SM C2, while expression of genes regulating T cell memory such as Tcf7, Il7r and Klf2 was decreased (Fig. 4f). Further gene ontology analysis of SM C2 showed significant enrichment of terms related to regulation of T cell activation and regulation of the immune effector process (Extended Data Fig. 6e), indicative of active effector functions of NDV-GP-educated SM TBYS C2 cells. Furthermore, by virtue of flow cytometry, we confirmed the presence of NDV-GP treatment-induced SM C2 cells as the TCF-1loT-bethi SM TBYS subset (Fig. 4g,h), highly expressing granzyme B (GzmB), which was consistent with transcription profiles of SM C2 cells (Fig. 4i,j). Moreover, NDV-GP-educated SM TBYS cells still preserved TH1 cell identity as indicated by the abundance of Ly6C and the absence of CXCR5 (Extended Data Fig. 7a–c). These findings suggest that a noticeable memory-to-effector transition is involved in the CD4+ TBYS cell response to the OV carrying cognate epitopes.

To further investigate the potential cytotoxic functions of NDV-GP-educated TCF-1loT-bethiCD4+ TBYS cells, we performed ex vivo killing assays with TCF-1loT-bethi and TCF-1hiT-betlo SM TBYS cells from the NDV-GP-treated group as well as SM TBYS cells from PBS- or NDV-WT-treated groups. Here, Ly108 and CD39 were used as surrogate surface markers to distinguish and isolate TCF-1loT-bethi (Ly108loCD39hi) and TCF-1hiT-betlo (Ly108hiCD39lo) SM TBYS cells (Extended Data Fig. 7d,e). We found that NDV-GP-educated TCF-1loT-bethi (Ly108loCD39hi) SM TBYS cells were more efficient at directly killing target cells than TCF-1hiT-betlo (Ly108hiCD39lo) SM TBYS cells from the NDV-GP group and other control SM TBYS cells from the PBS or NDV-WT group (Fig. 4k), indicating superior cytotoxic functions of NDV-GP-educated TCF-1loT-bethi SM TBYS cells. To more directly evaluate the reliance on TCF-1loT-bethiCD4+ TBYS cells for the therapeutic effects of the OV-BYTE strategy, TCF-1loT-bethi (Ly108loCD39hi) or TCF-1hiT-betlo (Ly108hiCD39lo) SM TBYS cells from NDV-GP-administered MC38 tumors were adoptively transferred into MC38 tumor-engrafted recipients with no exposure to LCMV Armstrong infection, and then these recipients were intratumorally administered PBS, NDV-WT or NDV-GP (Fig. 4l). Indeed, only transfer of the TCF-1loT-bethi SM TBYS subset, but not transfer of TCF-1hiT-betlo SM TBYS cells, could mediate the anti-tumor effect of NDV-GP treatment (Fig. 4m).

It is well known that GzmB is important for the cytotoxicity of cytotoxic T cells37. Here, we also found that GzmB was specifically expressed in NDV-GP-induced cytotoxic TCF-1loT-bethi TBYS cells (Fig. 4f,i,j); however, whether GzmB expression is a prerequisite for the cytotoxicity of these TCF-1loT-bethi TBYS cells remains unknown. To this end, LCMV-specific memory CD4+ T cells were isolated from WT C57BL/6 mice or GzmB-deficient (GzmbKO) mice on day 60 after infection and then transferred into naive C57BL/6 recipients as CD4+ TBYS cells. These recipients were then engrafted with MC38 cells and intratumorally treated with PBS, NDV-WT or NDV-GP (Fig. 4n and Extended Data Fig. 7f,g). Consistently, transfer of WT LCMV-specific memory CD4+ T cells potentiated the anti-tumor function of NDV-GP administration in recipients without LCMV Armstrong exposure. Nevertheless, such protection was completely abolished in the absence of GzmB expression in CD4+ T cells (Fig. 4o). In aggregate, these results highlight the GzmB-dependent cytotoxicity of CD4+ TBYS cells as indispensable in achieving therapeutic outcome of OV-BYTE therapy.

OV-BYTE promotes in situ TBYS cells to restrain tumor growth

Reportedly, TTST cells in the tumor-draining lymph node (dLN) serve as a reservoir for their counterparts in the TME that are maintained by continuous migration19,38, which calls into question whether OV-BYTE relies on dLN egressing or TME-resident TBYS cells for its anti-tumor efficacy. To this end, naive C57BL/6 mice with adoptive transfer of P14 and SM cells were infected with LCMV Armstrong and then engrafted with MC38 cells on day 60 after infection. On day 10 after tumor incubation, mice were gavaged with the sphingosine-1-phosphate receptor agonist FTY720 (ref. 39) to block dLN egress and then intratumorally administered PBS or NDV-GP. As a control, MC38-bearing mice gavaged with PBS vehicle were intratumorally administered PBS or NDV-GP (Fig. 5a). As shown, FTY720 treatment blunted the numbers of CD8+ T cells, CD4+ T cells, P14 cells and SM cells in the peripheral blood of mice intratumorally administered PBS or NDV-GP (Fig. 5b–e). In addition, FTY720 treatment yielded reductions of both P14 and SM TBYS cells in mice intratumorally administered PBS (Fig. 5f,g), suggesting that the migration of TMEM cells from dLNs is likely necessary to sustain TBYS cells in the TME. Remarkably, we observed that the FTY720-mediated decrease of P14 and SM TBYS cells in the TME was almost completely rescued by intratumoral administration of NDV-GP (Fig. 5f,g), indicating that NDV-GP treatment can efficiently expand in situ virus-specific TBYS cells even if the replenishment of these cells from dLNs were disrupted. Importantly, we further observed that the FTY720-mediated decrease in PD-1+CD39+ T cells12,40 was also rectified by NDV-GP treatment (Fig. 5h,i), which might be due to enhanced antigen presentation upon NDV-GP treatment (Extended Data Fig. 4e,f) that optimizes T cell responses in the TME.

Fig. 5: TBYS cells bolstered in situ by OV-BYTE therapy contribute to tumor control.
figure 5

a, Schematic of the experimental design. Congenic CD45.1+ P14 CD8+ T cells and CD45.1+ SM CD4+ T cells were adoptively transferred into naive C57BL/6 recipients, which were then infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. On days 12, 13, 14, 16 and 17 after tumor engraftment, recipients were intratumorally treated with PBS or NDV-GP. Meanwhile, the recipients were gavaged with FTY720 drug or vehicle from day 10 to day 18. On day 19 after tumor engraftment, tumor-infiltrating T cells were analyzed. qd, quaque die. be, Numbers of CD8+ T cells (b), CD4+ T cells (c), P14 cells (d) and SM cells (e) in the peripheral blood of mice in each group as described in a. f,g, Numbers of tumor-infiltrating P14 TBYS cells (f) and SM TBYS cells (g). h,i, Numbers of tumor-infiltrating PD-1+CD39+CD8+ T cells (h) and PD-1+CD39+CD4+ T cells (i). PBS and vehicle (n = 4 mice), PBS and FTY720 (n = 4 mice), NDV-GP and vehicle (n = 4 mice) and NDV-GP and FTY720 (n = 4 mice) in bi. j, Schematic of the experimental design for k,l. LCMV Armstrong-infected (day 60) or naive (uninfected) C57BL/6 mice were engrafted with MC38 cells. On day 10 after tumor engraftment, MC38 tumors were dissected from infected or uninfected mice and transplanted to naive C57BL/6 mice, which were then intratumorally treated with PBS, NDV-WT or NDV-GP on days 4, 5, 6, 8 and 9 after tumor transplantation. k,l, Tumor growth curve (k) and Kaplan–Meier survival curve (l) of MC38 tumor mass-recipient mice treated with PBS (n = 5 (naive in k), n = 8 (naive in l), n = 8 (memory in k) or n = 8 (memory in l) mice), NDV-WT (n = 5 (naive in k), n = 8 (naive in l), n = 7 (memory in k) or n = 7 (memory in l) mice) or NDV-GP (n = 7 (naive in k), n = 7 (naive in l), n = 6 (memory in k) or n = 7 (memory in l) mice) as described in j. ‘Naive’ indicates mice transplanted with MC38 tumors originating from uninfected mice; ‘memory’ indicates mice transplanted with MC38 tumors originating from LCMV Armstrong-infected mice. All data are representative of two independent experiments. Two-tailed unpaired Student’s t-test was used in bi. The log-rank (Mantel–Cox) test was performed to compare survival curves among groups in k. Two-way ANOVA was used to compare tumor growth curves in l. Center values and error bars (bi,k) indicate mean and s.e.m.

Source data

To further interrogate whether the in situ amplified TBYS cells were responsible for OV-BYTE therapeutic efficacy, naive and LCMV Armstrong-infected C57BL/6 mice were engrafted with MC38 cells. On day 10 after tumor engraftment, MC38 tumor masses were dissected from naive or infected mice and subcutaneously transplanted to naive C57BL/6 recipients, which were then intratumorally administered PBS, NDV-WT or NDV-GP (Fig. 5j). We found that NDV-GP administration better controlled the growth of transplanted MC38 tumors with pre-existing LCMV GP-specific T cells in the tumor mass and prolonged the survival of mice transplanted with such tumors (Fig. 5k,l), mirroring the long-term protection afforded by NDV-GP in MC38 tumor-engrafted mice with LCMV Armstrong infection (Fig. 2a–c). Hence, in situ amplified virus-specific TBYS cells primarily contribute to the therapeutic effects of OV-BYTE therapy.

OV-BYTE augments TTST cells by inducing epitope spreading

Next, we aimed to evaluate intrinsic TTST cell responses upon OV-BYTE therapy. To this end, we first investigated changes of the T cell receptor (TCR) repertoire in PD-1+CD39+CD8+ T cells originating from PBS-, NDV-WT- or NDV-GP-treated MC38 tumors engrafted on LCMV-infected mice by performing single-cell TCR sequencing (scTCR-seq) analysis (Fig. 6a). The results revealed enhanced oligoclonal expansion of tumor-reactive CD8+ T cells of the NDV-GP group in relation to those of the PBS or NDV-WT groups (Fig. 6b), suggesting potential T cell epitope spreading of tumor-reactive CD8+ T cells in the presence of OV-BYTE therapy. Further examination of endogenous CD8+ T cells specific to the MC38 tumor antigen p15E by both tetramer staining and a relevant peptide stimulation assay highlighted the reinforced p15E-specific CD8+ T cell response, including both quantity and quality, upon NDV-GP treatment (Fig. 6c–f). Moreover, in-depth TCR repertoire analysis indicated that p15E-specific CD8+ T cells from the NDV-GP group exhibited TCR clones of greater breadth and diversity than their counterparts from PBS or NDV-WT groups (Fig. 6g), further corroborating the notion that OV-BYTE therapy targeting virus-specific epitopes virtually led to epitope spreading of tumor-specific antigens and, as a result, enhanced TTST cell responses.

Fig. 6: OV-BYTE therapy triggers TTST cell responses by inducing T cell epitope spreading.
figure 6

a, Schematic of the experimental design. Naive C57BL/6 mice were infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, MC38 tumor-engrafted mice were intratumorally treated with PBS, NDV-WT or NDV-GP. On day 14 after tumor engraftment, tumor-infiltrating T cell responses were analyzed. b, Frequencies of TCR clonotypes in PD-1+CD39+ tumor-reactive CD8+ T cells originating from PBS (n = 3,222 cells), NDV-WT (n = 966 cells) or NDV-GP (n = 1,565 cells) -treated MC38 tumors on day 14 after tumor engraftment as indicated in a. These TCR clonotypes were divided into five groups of different sizes, including hyperexpanded (30 < X ≤ 70), large (10 < X ≤ 30), medium (5 < X ≤ 10), small (1 < X ≤ 5) and single (0 < X ≤ 1). c, Flow cytometry analysis of MC38 tumor-infiltrating CD44+CD8+ T cells from the PBS-, NDV-WT- or NDV-GP-treated group on day 14 after tumor engraftment as indicated in a. Numbers adjacent to the outlined areas indicate percentages of p15E tetramer+CD44hi tumor-specific CD8+ T cells of tumor-infiltrating CD8+ T cells. d, Frequency and number of tumor-infiltrating p15E tetramer+CD44hi tumor-specific CD8+ T cells in c. PBS (n = 6 mice), NDV-WT (n = 5 mice) and NDV-GP (n = 5 mice). e, Flow cytometry analysis of tumor-infiltrating CD44+CD8+ T cells (under the condition of p15E peptide stimulation) from the PBS-, NDV-WT- or NDV-GP-treated group on day 14 after tumor engraftment as indicated in a. Numbers adjacent to the outlined areas indicate frequencies of IFN-γ+CD107+ cells of tumor-infiltrating CD44+CD8+ T cells. f, Frequency and number of tumor-infiltrating p15E peptide-stimulated IFN-γ+CD107+CD8+ T cells in e. PBS (n = 5 mice), NDV-WT (n = 6 mice) and NDV-GP (n = 5 mice). g, Bubble charts showing Shannon entropy analysis of TCR diversity in tumor-infiltrating p15E tetramer+CD39hiCD8+ T cells from the PBS-, NDV-WT- or NDV-GP-treated group on day 14 after tumor engraftment as indicated in a. h, Schematic of the experimental design. MC38 cells were treated in vitro with PBS, Ad5-WT (multiplicity of infection of 1) or Ad5-GP (multiplicity of infection of 1) for 48 h. Next, naive or LCMV Armstrong-infected C57BL/6 mice were inoculated with the same amount of PBS-, Ad5-WT- or Ad5-GP-treated MC38 cells or a 50:50 mix of PBS- and Ad5-GP-treated MC38 cells or a 50:50 mix of PBS- and Ad5-WT-treated MC38 cells. i,j, Tumor growth curves of the MC38 cells described in h engrafted in LCMV Armstrong-infected C57BL/6 mice (i) or naive C57BL/6 mice (j). PBS (n = 6 mice), Ad5-WT (n = 6 mice), Ad5-GP (n = 4 mice), PBS and Ad5-WT (n = 5 mice) and PBS and Ad5-GP (n = 4 mice) in i; PBS (n = 6 mice), Ad5-WT (n = 6 mice), Ad5-GP (n = 5 mice), PBS and Ad5-WT (n = 4 mice) and PBS and Ad5-GP (n = 6 mice) in j. Data (cf,i,j) are representative of two independent experiments. One-way ANOVA with Turkey’s test was used in d,f. Two-way ANOVA was used to compare tumor growth curves in i,j. Center values and error bars (d,f,i,j) indicate mean and s.e.m.

Source data

For further proof of the anti-tumor effects caused by OV-BYTE-driven TCR epitope spreading, we inoculated C57BL/6 naive mice or those with LCMV memory with a 50:50 mix of PBS-treated and Ad5-GP-treated MC38 cells. As controls, C57BL/6 naive mice or those with LCMV memory were inoculated with the same amount of PBS-, Ad5-WT- or Ad5-GP-treated MC38 cells as well as a 50:50 mix of PBS- and Ad5-WT-treated ones (Fig. 6h). It transpired that the tumor growth of Ad5-GP-treated MC38 cells was significantly inhibited in mice with LCMV memory but not in naive mice. Also, such tumor growth inhibition was also preserved in the setting of the mixed PBS- and Ad5-GP-treated MC38 cells engrafted in mice with LCMV memory (Fig. 6i,j), indicating the development of anti-tumor responses against tumor cells expressing no viral antigens upon OV-BYTE therapy.

To further explore the potential mechanism(s) underlying OV-BYTE therapy-driven TCR epitope spreading, mice with LCMV memory were engrafted with MC38 cells expressing ovalbumin (OVA) (hereafter referred to as MC38-OVA) and were then administered PBS, NDV-WT or NDV-GP intratumorally (Extended Data Fig. 8a). Given the amenable role of DCs in fostering TCR epitope spreading of tumor-specific T cells41,42, we next examined the DC response in the aforementioned experimental settings, in which we observed an enhanced DC response in NDV-GP-treated MC38-OVA tumors as compared to that in PBS- or NDV-WT-treated ones (Extended Data Fig. 8b,c). More specifically, we further accessed the presentation of OVA257–264 peptide by MHC-I H-2Kb in DCs and found that DCs presenting OVA257–264 peptide were increased in the NDV-GP group as compared to those in the PBS or NDV-WT group (Extended Data Fig. 8d). In addition, DCs isolated from NDV-GP-treated MC38-OVA tumors rather than PBS- or NDV-WT-treated ones were more efficient in priming the tumor-specific OT-I CD8+ T cell response (Extended Data Fig. 8e,f). In sum, these data provide evidence that OV-BYTE therapy targeting bystander viral epitopes may license DCs to enhance the presentation of spreading tumor-specific epitopes to CD8+ TTST cells.

OV-BYTE and PD-L1 ICB synergize to control tumor progression

Distinct from OV-BYTE therapy that targets TBYS cells, PD-1 and/or PD-L1 ICB therapy is suggested to reinvigorate TEX or TTST cells2. As effective OV treatment has been reported to remodel the TME to favor PD-1 and/or PD-L1 ICB therapy43, we hypothesized that OV-BYTE therapy would show a synergistic anti-tumor response with PD-1 and/or PD-L1 ICB therapy. Indeed, C57BL/6 mice with LCMV memory concurrently treated with NDV-GP and PD-L1 ICB demonstrated ~60% complete protection against MC38 tumor challenge, which was superior to treatment with NDV-GP or PD-L1 ICB therapy alone (Fig. 7a,b). To further understand how TBYS and TTST cells respond to the combined NDV-GP and PD-L1 ICB therapy, C57BL/6 mice with LCMV memory and adoptively transferred P14 and SM cells were engrafted with MC38-OVA cells and then administered NDV-GP and PD-L1 ICB therapy (Fig. 7c). Consistent with the aforementioned data, P14 TBYS cells in MC38-OVA tumors were comparable among PBS, NDV-WT and NDV-GP groups (Fig. 7d). Meanwhile, the SM TBYS cell response of NDV-GP treatment was bolstered compared to that of PBS or NDV-WT treatment (Fig. 7e). However, P14 and SM TBYS cells, both of which barely expressed PD-1 (Fig. 1d,j), were minimally influenced by PD-L1 ICB therapy in all groups (Fig. 7d,e). We then examined TTST cell responses by ex vivo OVA257–264 (CD8+ T cell epitope) or OVA323–339 (CD4+ T cell epitope) peptide stimulation assays. Notably, the OVA257–264-specific CD8+ T cell response was most enhanced by concurrent treatment with PD-L1 ICB and NDV-GP in all experimental settings, evidenced by the highest proportion and number of IFN-γ-producing CD8+ T cells upon OVA257–264 stimulation (Fig. 7f,g). Similarly, the OVA323–339-specific CD4+ T cell response in the NDV-GP and PD-L1 ICB combinatorial treatment group was also superior to that of all other groups (Fig. 7h,i). Collectively, these findings demarcate that OV-BYTE potentiates the anti-tumor efficacy of PD-L1 ICB by augmenting TTST cell responses.

Fig. 7: OV-BYTE therapy and PD-L1 ICB therapy synergize to control tumor progression.
figure 7

a, Schematic of the experimental design for b. Naive C57BL/6 mice were infected with LCMV Armstrong and engrafted with MC38 cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally administered PBS, NDV-WT or NDV-GP. Meanwhile, recipients of each group were administered anti-PD-L1 blocking antibody (αPD-L1) or control immunoglobulin G (IgG) antibody on days 10, 13 and 16 after tumor engraftment. b, Kaplan–Meier survival curve for a. PBS and control (n = 11 mice), PBS and anti-PD-L1 antibody (n = 11 mice), NDV-WT and control (n = 8 mice), NDV-WT and anti-PD-L1 antibody (n = 12 mice), NDV-GP and control (n = 9 mice) and NDV-GP and anti-PD-L1 antibody (n = 8 mice). c, Schematic of the experimental design. Congenic CD45.1+ P14 CD8+ T cells and CD45.1+ SM CD4+ T cells were adoptively transferred into naive C57BL/6 recipients, which were then infected with LCMV Armstrong and engrafted with MC38-OVA cells on day 60 after infection. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally treated with PBS, NDV-WT or NDV-GP. Meanwhile, recipients were treated with anti-PD-L1 antibody or control IgG antibody on days 10, 13 and 16 after tumor engraftment. On day 18 after tumor engraftment, tumor-infiltrating T cells were analyzed. d,e, Frequencies and numbers of P14 TBYS cells of tumor-infiltrating CD8+ T cells (d) and SM TBYS cells of tumor-infiltrating CD4+ T cells (e) from each group described in c. PBS and control (n = 4 mice), PBS and anti-PD-L1 antibody (n = 3 mice), NDV-WT and control (n = 4 mice), NDV-WT and anti-PD-L1 antibody (n = 3 mice), NDV-GP and control (n = 3 mice), NDV-GP and anti-PD-L1 antibody (n = 3 mice) in d,e. f, Flow cytometry analysis of tumor-infiltrating endogenous CD45.2+CD44+CD8+ T cells. Numbers adjacent to outlined areas indicate percentages of IFN-γ+ cells of CD45.2+CD44+CD8+ T cells under the condition of ex vivo OVA257–264 stimulation. g, Frequencies and numbers of OVA257–264-stimulated IFN-γ+CD8+ T cells. PBS and control (n = 4 mice), PBS and anti-PD-L1 antibody (n = 3 mice), NDV-WT and control (n = 4 mice), NDV-WT and anti-PD-L1 antibody (n = 3 mice), NDV-GP and control (n = 3 mice), NDV-GP and anti-PD-L1 antibody (n = 3 mice) in g. h, Flow cytometry analysis of tumor-infiltrating endogenous CD45.2+CD44+CD4+ T cells. Numbers adjacent to outlined areas indicate percentages of IFN-γ+ cells of CD45.2+CD44+CD4+ T cells under the condition of ex vivo OVA323–339 stimulation. i, Frequencies and numbers of OVA323–339-stimulated IFN-γ+CD4+ T cells. PBS and control (n = 4 mice), PBS and anti-PD-L1 antibody (n = 3 mice), NDV-WT and control (n = 4 mice), NDV-WT and anti-PD-L1 antibody (n = 3 mice), NDV-GP and control (n = 3 mice), NDV-GP and anti-PD-L1 antibody (n = 3 mice) in i. All data are representative of two independent experiments. The log-rank (Mantel–Cox) test was performed to compare survival curves among groups in b. Two-tailed unpaired Student’s t-test was used in d,e,g,i. Center value and error bars (d,e,g,i) indicate mean and s.e.m.

Source data

OV-BYTE enables anti-tumoral SARS-CoV-2-specific TMEM cells

Considering that a tremendous population around the globe has been either infected with SARS-CoV-2 or immunized with COVID-19 vaccines, both of which leave behind long-term persistence of SARS-CoV-2-specific T cell memory44, we set out to determine whether such T cell memory could be used by OV-BYTE therapy. To this end, naive BALB/c mice were prime–boost intranasally vaccinated with SARS-CoV-2 receptor-binding domain (RBD) proteins, a core part of the SARS-CoV-2 spike protein and COVID-19 vaccines45, and thus gained CD8+ and CD4+ T cell memory specific to the SARS-CoV-2 RBD (Extended Data Fig. 9a,b). Next, these vaccinated mice received orthotopic injection of murine triple-negative breast cancer 4T1 cells into mammary fat pads on day 60 after vaccination and then were intratumorally treated with an NDV stain expressing SARS-CoV-2 RBD (NDV-RBD). Consistently, intratumoral administration of NDV-RBD protected against the outgrowth of 4T1 tumors in SARS-CoV-2 RBD protein-immunized mice (Fig. 8a–c), with increased SARS-CoV-2 RBD-specific CD4+ TBYS cells in the TME (Extended Data Fig. 9c–i). Furthermore, the protection against 4T1 tumors was reinforced when PD-L1 ICB was given concomitantly with NDV-RBD (Fig. 8a–c).

Fig. 8: OV-BYTE therapy potentiates anti-tumor activities of SARS-CoV-2-specific TMEM cells.
figure 8

a, Schematic of the experimental design for b. Naive BALB/c mice were prime–boost immunized with SARS-CoV-2 RBD protein at an interval of 14 days and engrafted with 4T1 cells on day 60 after prime immunization. On days 7, 8, 9, 11 and 12 after tumor engraftment, recipients were intratumorally administered PBS, NDV-WT or NDV-RBD. Meanwhile, recipients of each group were administered anti-PD-L1 blocking antibody or control IgG antibody on days 10, 13 and 16 after tumor engraftment. b,c, Tumor growth curve (b) and Kaplan–Meier survival curve (c) from a. PBS and control (n = 7 mice), PBS and anti-PD-L1 antibody (n = 6 mice), NDV-WT and control (n = 5 mice), NDV-WT and anti-PD-L1 antibody (n = 6 mice), NDV-RBD and control (n = 7 mice) and NDV-RBD and anti-PD-L1 antibody (n = 7 mice) in b. PBS and control (n = 6 mice), PBS and anti-PD-L1 antibody (n = 4 mice), NDV-WT and control (n = 6 mice), NDV-WT and anti-PD-L1 antibody (n = 6 mice), NDV-RBD and control (n = 8 mice) and NDV-RBD and anti-PD-L1 antibody (n = 9 mice) in c. d, Flow cytometry analysis of CD8+ (top) and CD4+ (bottom) T cells from the PBMCs of SARS-CoV-2-uninfected donors and convalescent donor with COVID-19. Numbers adjacent to the outlined areas indicate percentages of SARS-CoV-2 RBD-stimulated IFN-γ+ cells of CD8+ T cells or IFN-γ+ cells of CD4+ T cells. e, Schematic experimental design for f. NCG mice were engrafted with PBMCs from HLA-A2-positive convalescent patients with COVID-19. After reconstruction of human T cells, these humanized NCG mice were engrafted with A375 cells and then intratumorally administered NDV-WT or NDV-RBD on days 7, 8, 9, 11 and 12 after tumor engraftment. f, Tumor growth curve of A375 tumor-bearing humanized NCG mice intratumorally treated with NDV-WT (n = 5 mice) or NDV-RBD (n = 4 mice) as indicated in e. g, Schematic experimental design for h. NCG mice were engrafted with PBMCs from HLA-A2-positive convalescent patients with COVID-19. Next, these reconstructed humanized NCG mice were engrafted with A375 cells and then intratumorally administered Ad5-WT or Ad5-RBD on days 7, 8, 9, 11 and 12 after tumor engraftment. h, Tumor growth curve of A375 tumor-bearing humanized NCG mice intratumorally treated with Ad5-WT (n = 6 mice) or Ad5-RBD (n = 6 mice) as indicated in g. Data (bd,f,h) are representative of two independent experiments. Two-way ANOVA was used to compare tumor growth curves in b,f,h. The log-rank (Mantel–Cox) test was performed to compare survival curves among groups in c. Center values and error bars (b,f,h) indicate mean and s.e.m.

Source data

To further probe the clinical relevance of OV-BYTE therapy leveraging SARS-CoV-2-specific T cell memory, NCG mice were engrafted with PBMCs from convalescent HLA-A2-typed patients with COVID-19 harboring SARS-CoV-2 RBD-specific TMEM cells46 (Fig. 8d). These NCG mice were inoculated with A375 human melanoma cells and then intratumorally administered NDV-WT or NDV-RBD (Fig. 8e). Noticeably, A375 tumor growth was more restricted in the NDV-RBD-treated group than that in the NDV-WT-treated one (Fig. 8f). Recapitulating NDV treatment-upregulated MHC-I and MHC-II expression in murine tumor cells (Extended Data Fig. 4e,f), both MHC-I (HLA-A2) and MHC-II (HLA-DR) expression levels of A375 tumor cells were reinforced upon NDV treatment (Extended Data Fig. 10a–c). Furthermore, the therapeutic effects of NDV-RBD in A375 tumors engrafted in NCG mice receiving transfer of convalescent COVID-19 patient PBMCs were almost diminished upon CD8+ or CD4+ T cell depletion (Extended Data Fig. 10d,e). These findings indicate that both CD8+ and CD4+ T cells contribute to the anti-tumor functions of OV-BYTE therapy using SARS-CoV-2-specific T cell memory in the humanized NCG model.

We next used replication-deficient Ad5 virus expressing SARS-CoV-2 RBD (Ad5-RBD) to treat A375 tumors inoculated in NCG mice reconstructed by convalescent COVID-19 patient PBMCs (Fig. 8g). Consistently, Ad5-RBD delayed A375 tumor growth as compared to Ad5-WT (Fig. 8h). Therefore, these results together indicate that SARS-CoV-2-specific T cell memory established by infection or vaccination could be exploited by the OV-BYTE strategy to treat cancer.

Discussion

In this study, we provided a proof of concept that redirecting the antigen specificity of tumor cells to tumor-infiltrating CD8+ and CD4+ TBYS cells effectively controls tumor progression. This therapy, called OV-BYTE, was proven to curtail tumor growth and show synergistic effects with PD-L1 ICB therapy in multiple preclinical tumor models. In particular, we identified that TCF-1loT-bethi cytotoxic CD4+ TBYS cells conveyed GzmB-dependent cytotoxicity and were amenable to OV-BYTE-mediated therapeutic effects, thus being aligned with the recently highlighted cytotoxic functions of CD4+ T cells in eliminating cancer cells47 and virus-infected cells48.

We also provided key proof-of-concept data illustrating that OV-BYTE therapy can effectively harness SARS-CoV-2-specific T cell memory to treat cancers in preclinical models. Notably, several SARS-CoV-2-infected patients with cancer have been reported to achieve partial or complete tumor remission49,50. Based on our findings, we speculate that SARS-CoV-2 infection of tumor cells51,52 in these patients may redirect these malignant cells toward the cytotoxicity of SARS-CoV-2-specific TBYS cells. Further clinical trials are urgently needed to validate the therapeutic efficacies of OV-BYTE therapy in patients with cancer and SARS-CoV-2-specific T cell memory.

In conclusion, we revealed that OV-BYTE therapy redirects the cytotoxicity of functional TBYS cells toward tumor cells for improved tumor control and has synergistic effects with PD-L1 ICB. Hence, the combination of OV-BYTE and PD-1 and/or PD-L1 ICB might expand the toolkits for cancer immunotherapy.

Methods

Research involving animal experiments with all relevant ethical regulations is detailed in Mice below. Research involving human samples with all relevant ethical regulations is detailed in Human participants and the humanized mouse model below.

Mice

C57BL/6, BALB/c, OT-I and CD45.1+ (B6.SJL-Ptprca Pepcb/BoyJ) mice were purchased from the Jackson Laboratories. P14 and SM mice were gifts from R. Ahmed (Emory University). CXCR5–GFP knockin mice were generated by inserting an IRES-GFP construct after the open reading frame of Cxcr5 (ref. 53). GzmbKO mice were provided by F. Shao (Chinese Academy of Medical Sciences and National Institute of Biological Sciences). All experiments involving C57BL/6, BALB/c, P14, SM, OT-I, CXCR5–GFP knockin, CD45.1+ and GzmbKO mice were carried out in accordance with procedures approved by the Institutional Animal Care and Use Committees of the Third Military Medical University. Immunodeficient NOD/ShiLtJGpt-Prkdcem26Cd52Il2rgem26Cd22/Gpt (NCG) mice were purchased from GemPharmatech. All experiments involving the generation and characterization of humanized NCG mice were approved by an institutional animal care and use committee at the Model Animal Research Center at Nanjing University (AP LY-01). All mice used for animal experiments were 6–10 weeks of age, and both sexes were included in the study, except only female BALB/c mice were used for the engraftment of 4T1 breast cancer cells. All mice were housed under specific-pathogen free conditions with normal mouse chow (Jiangsu Xietong Shengwu), a 12-h light–12-h dark cycle and controlled temperature (20–22 °C) and humidity (40–60%).

Viral infection and protein immunization

To establish LCMV Armstrong infection, naive C57BL/6 mice were injected intraperitoneally with 2 × 105 plaque-forming units (PFU) of LCMV Armstrong. To establish SARS-CoV-2-specific T cell memory, naive BALB/c mice were intranasally immunized with two doses of 10 μg SARS-CoV-2 RBD protein–mFC tag (Sino Biological, 40592-V05H) conjugated to 10 μg CpG ODN 1826 (Invitrogen, tlrl-1826) at an interval of 14 d.

Construction of modified oncolytic viruses

NDV-LCMV GP, NDV-LCMV NP, NDV-SARS-CoV-2 RBD and NDV-H1N1 NP were generated as previously described54; specifically, the full-length NDV SG10 cDNA clone, pOK-SG10, was used as the backbone to construct a recombinant cDNA clone containing the gene sequences encoding LCMV GP, LCMV NP, SARS-CoV-2 RBD or H1N1 NP between the P and M genes. Ad5-GP and Ad5-RBD were generated by inserting the gene sequences encoding LCMV GP protein and SARS-CoV-2 RBD protein, respectively, into Ad5 lacking 24 base pairs encoding eight amino acids in the pRb-binding domain of E1A and E3.

Tumor models

MC38 (NM-S13) and MC38-OVA (NM-S13-TM56) cells were purchased from the Shanghai Model Organisms Center. B16F10 (CRL-6475), 4T1 (CRL-2539) and A375 (CRL-1619) cells were purchased from ATCC. B16-GP cells were generated by CRISPR–Cas9-mediated insertion of the LCMV GP gene into the genome of B16F10 cells. All tumor cell lines were cultured in complete DMEM-10 medium (Gibco) supplied with 10% FBS (Gibco), 1% penicillin–streptomycin (Gibco) and 1% l-glutamine (Gibco). For subcutaneous tumor models, C57BL/6 mice were implanted with 3–5 × 105 MC38, MC38-OVA, B16F10 or B16-GP cells in the left flank of leg. For the mouse breast cancer model, 2 × 105 4T1 cells were implanted into the mammary fat pad of female BALB/c mice. Tumor volumes were determined in two dimensions with a caliper and calculated according to the formula ((length × width2)/2). For the NICD and AKT plasmid-induced intrahepatic cholangiocarcinoma model, the hydrodynamic tail vein of C57BL/6 mice was injected with a mixture of the three plasmids, including Myc-tagged NICD1 (10 μg per mouse), Myc-tagged AKT (4 μg per mouse) and SB100 (1 μg per mouse), according to a published study35. For the tumor transplantation model, MC38 tumors were surgically dissected from donor mice and cut into pieces of about 5 mm3 in size. Next, tumor specimens were subcutaneously transplanted into the same flank of recipients. Tumor-engrafted mice were killed at the indicated time points on the premise of humane endpoints (tumor volume not exceeding 2,000 mm3).

PCR with reverse transcription

Total RNA from tumor cells was extracted by using TRIzol LS (Life Technologies) and reverse transcribed using the RevertAid Minus First Strand cDNA Synthesis Kit (Thermo Scientific, K1622) according to the manufacturer’s protocol. Relative expression levels of transcripts were determined using AceQ qPCR SYBR Green Master Mix (Vazyme, Q111) with a CFX96 Touch Real-Time System (Bio-Rad). The primers used in the study are as follows: LCMV GP gene (forward, 5′-GCAACTGCTGTGTTCCCGAAAC-3′; reverse, 5′-CATTCACCTGGACTTTGTCAGACTC-3′), LCMV NP gene (forward, 5′-CTCACACGGCATGGATCTTG-3′; reverse, 5′-TCGAAGCTTCCCTGGTCATT-3′) and Hprt1 (forward, 5′-TCAGTCAACGGGGGACATAAA-3′; reverse, 5′-GGGGCTGTACTGCTTAACCAG-3′).

Immunofluorescence

MC38 cells were transferred to 12-mm coverslips in a 12-well plate. After fixation and permeabilization, cells on coverslips were stained with DAPI (Sigma, D9542) and for actin (Cell Signaling Technology, 3134) and the NDV P protein (made in house). Images were acquired with a Zeiss LSM 510 confocal fluorescence microscope and processed with LSM Image Examiner (version 4.0).

Adoptive transfer of T cells

For the LCMV Armstrong infection model, 5 × 104 congenic naive splenic P14 cells and/or SM cells were adoptively transferred into recipients 1 d before infection. For the subcutaneous B16-GP tumor model, 5 × 105 congenic naive splenic P14 cells and/or SM cells were adoptively transferred into recipients 1 d before B16-GP tumor engraftment. For the LCMV Armstrong-specific TMEM cell transfer experiment, 4 × 106 CD8+ and/or CD4+ T cells with LCMV memory were isolated from the spleens of infected mice at day 60 after LCMV Armstrong infection and then adoptively transferred into naive C57BL/6 recipients. For the MC38-OVA tumor model, 2 × 103 congenic OT-I cells were adoptively transferred into recipients on day 10 after MC38-OVA tumor engraftment.

Isolation of lymphocytes

Lymphocytes in spleens were collected by mashing spleens through a cell strainer (BD Falcon). Lymphocytes in the peripheral blood were obtained by using a Ficoll (TBD, LTS107701) density gradient. To obtain liver-resident lymphocytes, mice were euthanized and perfused. Next, livers were dissected and mechanically minced. Liver-resident lymphocytes were acquired using a Percoll (GE Healthcare, 17-0891-09) density gradient. To obtain MC38, MC38-OVA or 4T1 tumor-infiltrating lymphocytes (TILs), tumors were mechanically minced, digested using with 1 mg ml−1 collagenase I (Sigma, c0130) and mashed through filters. Next, we enriched for TILs using a Percoll (GE Healthcare, 17-0891-09) density gradient. To collect B16F10 or B16-GP TILs, tumors were mechanically minced, and we enriched for TILs using a Percoll (GE Healthcare, 17-0891-09) density gradient.

Human participants and the humanized mouse model

We enrolled a cohort containing eight HLA-A2-positive healthy volunteers and a cohort containing four HLA-A2-positive convalescent patients with COVID-19. All volunteers provided written informed consent. The study received institutional review board approvals at Chongqing Public Health Medical Center (2020-023-01-KY). PBMCs from blood donated by these volunteers were isolated using a Ficoll (TBD, LTS107701) density gradient. NCG mice were injected intravenously with 5 × 106 human PBMCs. After the humanization rate reached an appropriate proportion, 5 × 106 A375 cells were subcutaneously injected into each side of recipient mice in a volume of 100 µl PBS. Tumors were measured every day using calipers, and the tumor volume was calculated using the following formula: (length × width2)/2.

Enzyme-linked immunosorbent assay

ELISA plates (Thermo Fisher, 446469) were coated with 50 ng H1N1 NP protein (Sino Biological, 40777-V08B) in 100 μl PBS per well overnight. Next, the ELISA plates were incubated with blocking buffer (5% FBS with 0.1% Tween-20 in PBS) for 1 h. Fifty-fold-diluted sera or blocking buffer (negative control) were next added to each well and incubated for 1 h. The ELISA plates were then washed with PBST (PBS with 0.1% Tween-20), incubated with HRP-conjugated goat anti-human IgG antibody (Bioss Biotech) and washed with PBS, and TMB was added (Beyotime). Samples were allowed to react with TMB for ~5 min, and then the reaction in ELISA plates was stopped by adding 1 M H2SO4 stop buffer. Optical density values were determined at 450 nm.

Administration of reagents

For administration of OVs, mice were treated intratumorally with 2 × 106 PFU of NDV or 1 × 108 PFU of Ad5 in 50 μl PBS. For the LCMV GP-neutralizing antibody experiment, sera containing LCMV GP-neutralizing antibodies were collected from C57BL/6 mice infected with LCMV Cl13 (ref. 55), pathogen inactivated and then incubated in vitro with NDV-GP. For PD-L1 ICB experiments, mice were intraperitoneally injected with 150 μg anti-PD-L1 antibody (BioXCell, clone 10F.9G2) or an isotype-matched control antibody (BioXCell, clone LTF-2) on days 10, 13 and 16 after tumor engraftment. For the depletion of CD8+ or CD4+ T cells in C57BL/6 mice, mice were intraperitoneally injected with 100 μg anti-CD8 antibody (BioXCell, clone 53-6.7) or 100 μg anti-CD4 antibody (BioXCell, clone GK1.5) on days −1, 1, 7 and 12 after tumor engraftment. For the depletion of CD8+ or CD4+ T cells in humanized NCG mice, mice were intraperitoneally injected with 200 μg anti-CD8 antibody (BioXCell, clone OKT8) or 200 μg anti-CD4 antibody (BioXCell, clone OKT4) on days −2 and 2 and then further injected intraperitoneally with 50 μg anti-CD8 antibody or anti-CD4 antibody on days 9 and 16 after tumor engraftment. For FTY720 treatment, mice were intraperitoneally injected with 0.3 mg per kg FTY720 (Merck, SML0700).

Flow cytometry

All information on antibodies for flow cytometry analysis in this study is provided in the Reporting Summary. Cell surface marker staining was performed in PBS containing 2% FBS. For the detection of cytokines, lymphocytes were stimulated for 6 h in the presence of the indicated peptide (0.2 μg ml−1 LCMV GP33–41, LCMV GP66–77, LCMV GP91–101, LCMV GP118–125, LCMV GP276–286, OVA257–264, OVA323–339 or p15E peptide) and anti-CD107a and anti-CD107b antibodies in an incubator at 37 °C. Alternatively, lymphocytes were incubated for 6 h with 1 μg ml−1 of an H1N1 NP peptide pool (Miltenyi Biotec, 130-097-278), for 18 h with 1 μg ml−1 of a SARS-CoV-2 S1 peptide pool (Sino Biological, PP003-A) or for 44 h with 1 μM recombinant SARS-CoV-2 RBD protein in an incubator at 37 °C. Brefeldin A and monensin (BD Biosciences) were added into the culture 4 h before detection. Intracellular cytokine staining for IL-2, IFN-γ and TNF-α was performed with the Cytofix/Cytoperm Fixation/Permeabilization Kit (BD Biosciences, 554714). Staining for T-bet, TCF-1, TOX, Eomes, Ki-67 and GzmB was performed with the Foxp3/Transcription Factor Staining Buffer Set (eBioscience, 00-5523). Dead cells were stained using the LIVE/DEAD Fixable Near-IR Dead Cell Stain Kit (Life Technologies). Flow cytometry data were acquired using FACSDiva software (version 7.0) in the FACSCanto II (BD Biosciences) or the FACSFortessa (BD Biosciences) and analyzed using FlowJo (version 10.4.0). Cell sorting experiments were performed with the BD FACSAria III (BD Biosciences).

Ex vivo killing assay

In the experiment illustrating NDV-GP-mediated GP protein expression in MC38 cells, NDV-WT- and NDV-GP-infected MC38 cells were labeled with CellTrace Violet (Thermo Fisher) at 0.1 μM and 1 μM, respectively, and then mixed at a 1:1 ratio. Next, labeled MC38 cells (target) were mixed with LCMV Armstrong-activated or naive CD8+ T cells (effector) at a 100:1, 50:1 or 25:1 E:T ratio and co-cultured at 37 °C for 8 h. In experiments assessing the killing efficacy of SM TBYS cells, NDV-WT-treated MC38 cells were labeled with CellTrace Violet (Thermo Fisher) at either 0.1 μM or 1 μM. MC38 cells labeled with 1 μM CellTrace Violet were pulsed with 2 μg LCMV GP66–77 peptide and then mixed with MC38 cells labeled with 0.1 μM CellTrace Violet at a 1:1 ratio. These labeled MC38 target cells were further mixed with sorted SM TBYS cells or naive CD4+ T cells (effector) at a 9:1 E:T ratio and co-cultured at 37 °C for 8 h. The killing efficiency was determined as follows: 100 − (((percent MC38-GP with LCMV-specific T cells)/(percent control MC38 with LCMV-specific T cells))/((percent MC38-GP with naive T cells)/(percent control MC38 with naive T cells))) × 100.

scRNA-seq library preparation, sequencing and data processing

scRNA-seq libraries were prepared using Chromium Next GEM Single Cell 3′ Reagent Kits (10x Genomics) according to the manufacturer’s instructions and then sequenced with paired-end sequencing of 150 nucleotides on each end on one lane of a NovaSeq 6000 per sample. Raw gene expression matrices were generated with the Cell Ranger (version 6.1.1) pipeline coupled with the mm10 genome annotation reference, which was then processed using Seurat56, and only cells with >1,000 detected genes or a mitochondrial read percentage <5 were retained. Doublets were automatically detected and removed by scDblFinder57 before all samples were combined into a count matrix using the merge function in Seurat. Data were log normalized and scaled using ‘NormalizeData’ and ‘ScaleData’. The top 2,000 most variable genes were calculated using ‘FindVariableGenes’ and then used in the PCA calculation with RunPCA. The number of top PCs used to calculate a UMAP dimensional reduction was decided by observation of the graphic output of the ‘ElbowPlot’ function.

Identification of differentially expressed genes and functional enrichment

DEG testing was performed using the ‘FindMarkers’ function in Seurat with default parameters. The top 200 DEGs with the highest log2 (fold change) values and a maximum FDR value of 0.01 were filtered and exploited for the enrichment analysis, which was conducted using the Metascape webtool (https://www.metascape.org). Gene sets were derived from the GO Biological Process ontology.

Defining cell state scores

We generated gene sets representing the signatures of TCM cells and TRM cells by conducting DEG identification analysis on external bulk RNA-seq datasets31,32. The resulting gene sets were used as input for the ‘AddModuleScore’ function in Seurat to calculate the corresponding cell scores, which could be further interpreted as phenotypic similarity to TCM and TRM for each cell.

Consensus non-negative matrix factorization analysis

Data from P14 TBYS cells (PBS group) were merged with data from P14 TRM cells (kidney, liver, salivary gland, siIEL)24 and P14 TMEM cells (days 32, 60 and 90)25 from an external dataset to generate a mixed count matrix first, which was the foundation for the inference of consensus gene programs. In detail, we executed the ‘cnmf prepare’ command from the cNMF package58 to normalize the input matrix and prepare the run parameters, the ‘cnmf factorize’ command to factorize the matrix and the ‘cnmf combine’ command to combine the individual spectrum result files for each K (the expected number of gene modules, which should be previously assigned by prior knowledge in the first step) into a merged file. The optimal number for K was decided by observing the output of the ‘cnmf k_selection_plot’ command, considering the tradeoff between stability and error, which distinguished the number 9 from the other candidates for the optimal value of K. At the desired value of K, we obtained consensus estimates for the programs and their usages by executing the ‘cnmf consensus’ command. Calculation and visualization using Radviz was based solely on the results of the cNMF analysis.

RNA velocity analysis

Read annotations for sequenced samples were performed using the ‘velocity run 10X’ command-line tool with BAM (velocyto.py), genome annotation and repeat annotation59. Repeat annotation files were downloaded from the UCSC Genome Browser. The UMAP embedding matrix computed with the Seurat pipeline was used to construct the velocity map with the scVelo Python package60. Briefly, the total procedure involved execution of the pp.filtered_and_normalized, pp.moments, pp.neighbors, tl.recover_dynamics and tl.velocity (with the mode parameter set to ‘dynamical’) functions from the scVelo package.

Bulk TCR amplification, sequencing and data analysis

RNA samples were analyzed by high-throughput sequencing of the TCR β-chain (TRB) using the ImmuHub TCR profiling system61 (ImmuQuad Biotech). The raw sequencing data were then aligned with NCBI and PCR. The resulting nucleotide and amino acid sequences of complementarity-determining region 3 (CDR3) of TRB were determined, and those with out-of-frame and stop codon sequences were removed from the identified TRB repertoire. We further defined the amount of each TRB clonotype by adding numbers of TRB clones sharing the same nucleotide sequence as CDR3. Shannon’s index62 was used to assess the diversity of TCR clones within each sample, which was defined as follows:

$$H^{\prime}=-\sum p_i{\mathrm{ln}}({p_i}),$$

where pi defines the frequency of the i-th specific clonotype.

Single-cell TCR sequencing data analysis

scTCR-seq libraries were prepared using 10x Genomics 5′ Single Cell Immune Profiling technology according to the manufacturer’s instructions. Sequencing was performed by using the NovaSeq 6000. TCR clonotype identification, alignment and annotation were performed using the 10x Genomics Cell Ranger pipeline (version 6.1.1) according to the manufacturer’s instructions. Clonotype alignment was performed against the Cell Ranger human V(D)J reference library 3.1.0 (GRCh38 and Ensembl GTF version 94). TCR clonotypes were defined using the ‘combineTCR’ function in scRepertoire63 (with the ‘cloneCall’ parameter set to ‘CTstrict’). Clonal expansion was determined using a five-group classification. The percentage of cells in single (0–1), small (2–5 cells), medium (6–10 cells), large (11–30) and hyperexpanded (31–70 cells) clones was determined for Pdcd1+Entpd1+Cd8a+ T cells.

Statistics and reproducibility

No statistical method was used to predetermine sample size, but our sample sizes are similar to those reported in previous publications19,27,28,53,64. No data were excluded from the analyses. The experiments were not randomized. The investigators were not blinded to allocation during experiments and outcome assessment. Statistical analysis was performed with Prism 9.0 (GraphPad). Normality and equal variances of data were formally tested. For comparisons between two independent groups, two-tailed unpaired Student’s t-test was used. For comparisons among three or more groups, one-way ANOVA with Turkey’s test was used. Two-way ANOVA was used to compare tumor growth curves. The log-rank (Mantel–Cox) test was performed to compare survival curves among groups. Graphs show individual samples, and center values indicate the mean. P values less than 0.05 were defined as statistically significant.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.