The overall response rate for anti–PD-1 therapy remains modest in hepatocellular carcinoma (HCC). We found that a combination of IFNα and anti–PD-1–based immunotherapy resulted in enhanced antitumor activity in patients with unresectable HCC. In both immunocompetent orthotopic and spontaneous HCC models, IFNα therapy synergized with anti–PD-1 and the combination treatment led to significant enrichment of cytotoxic CD27+CD8+ T cells. Mechanistically, IFNα suppressed HIF1α signaling by inhibiting FosB transcription in HCC cells, resulting in reduced glucose consumption capacity and consequentially establishing a high-glucose microenvironment that fostered transcription of the T-cell costimulatory molecule Cd27 via mTOR–FOXM1 signaling in infiltrating CD8+ T cells. Together, these data reveal that IFNα reprograms glucose metabolism within the HCC tumor microenvironment, thereby liberating T-cell cytotoxic capacities and potentiating the PD-1 blockade–induced immune response. Our findings suggest that IFNα and anti–PD-1 cotreatment is an effective novel combination strategy for patients with HCC.

Significance:

Our study supports a role of tumor glucose metabolism in IFNα-mediated antitumor immunity in HCC, and tumor-infiltrating CD27+CD8+ T cells may be a promising biomarker for stratifying patients for anti–PD-1 therapy.

See related commentary by Kao et al., p. 1615.

This article is highlighted in the In This Issue feature, p. 1599

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide (1). In the past few years, immune-checkpoint blockade (ICB) has revolutionized the approach to cancer therapy, and ICB has become a second-line option for patients with advanced HCC who fail sorafenib. Current ICB agents for HCC treatment mainly consist of anti–CTLA-4 and anti–PD-1/PD-L1 antibodies (2–4). However, objective response rates (ORR) were found to be approximately 15% in an Asian cohort of HCC patients treated with a single agent, such as nivolumab, pembrolizumab, or camrelizumab (2, 3, 5), suggesting that the complex immunosuppressive microenvironment of HCC leads to evasion of ICB via unknown mechanisms. Elucidating the mechanisms underlying the immunosuppressive microenvironment and subsequent remodeling to guide rational combination therapy remains a major challenge for therapeutic intervention in patients with HCC. Thus, the development of novel strategies to overcome intrinsic or adaptive resistance to immunotherapy in HCC has become an urgent need.

Responses to cancer immunotherapy are largely dampened by the immunosuppressive tumor microenvironment (TME; ref. 6). Due to increased glucose consumption in tumor cells, the TME becomes glucose-deprived in some tumor types (7). Glucose deprivation and hypoxia, induced by metabolically dysregulated cancer cells, restrict antitumor immunity by impeding the metabolic fitness of tumor-infiltrating cytotoxic CD8+ T cells (8). Hypoxia-inducible factor 1α (HIF1α) is the key regulator of the response to hypoxia in normal and cancer tissues; moreover, it is well-known that HIF1α regulates glycolysis and is a target for cancer therapy (9). HIF1α was shown to increase PI3K/AKT/mTOR activity in a mouse model, and it mediates glycolytic metabolic alterations that drive cancer progression and resistance to therapy (10). PD-L1 has recently been identified as a direct target of HIF1α (11), and elevated expression levels of HIF1α and PD-L1 have been associated with a poor prognosis in HCC (12).

Furthermore, metabolic pathways specifically regulate T-cell development, differentiation, and function in the TME (13). Glucose competition, amino acid competition, oxygen competition, and lactic acid secretion promote the formation of an immunosuppressive phenotype in the TME (13). The hypoxic TME also affects T-cell function, in which HIF1α drives CD8+ T-cell migration and effector function (14). Metabolically fit T cells contribute to relatively strong antitumor immunity by destroying tumor cells (15). Infiltrating immunomodulatory cells with different metabolic preferences also facilitate the establishment of an immunosuppressive TME (16). Blockade of PD-L1 under hypoxic conditions enhances myeloid-derived suppressor cell (MDSC)–mediated suppression of T cells present in tumors to construct an immunosuppressive TME (11). Therefore, simultaneous blockade of PD-L1 and inhibition of HIF1α may represent a novel approach for cancer immunotherapy. T-cell metabolism can be altered to perform different cellular functions in the TME (17). Although the metabolic interplay between tumor and immune cells plays an important role in immune response regulation (18), how metabolic changes in the tumor niche affect the development and maintenance of a favorable immune response and how the metabolic function of tumor or immune cells contributes to the outcome of ICB remain unclear.

IFNα is a cytokine belonging to the type I IFN family that exerts multiple biological effects, including antiviral and antitumor activities, in patients with defined types of cancer or viral diseases (19). IFNα has been widely applied to treat patients with chronic hepatitis B infection (20), but it is also a potential drug for combination with novel strategies for cancer therapy. Indeed, IFNα is considered a potential agent for tumor treatment, considering that it has been shown to directly block cell-cycle progression and induce apoptosis, which in turn inhibits tumor expansion by inducing the expression of tumor suppressor proteins and antitumor IFN-stimulated gene products (21–23). IFNα increases the surface expression of class I and II MHC molecules on tumor cells to enhance immune recognition (24). IFNα administration in combination with PD-1 blockade effectively augments antitumor immunity in tumor-bearing mice (25). The combination of an anti–PD-L1 antibody and IFNα has been reported to improve tumor targeting and antigen presentation as well as antitumor responses in a mouse model (26). Thus, a recent clinical trial identified IFNα plus an anti–PD-1 antibody as a promising treatment strategy in melanoma (27), indicating the potential value of IFNα-based combination ICB. Despite such results, the mechanism by which IFNα synergizes with ICB and whether the combination of IFNα with ICB can benefit HCC are still poorly understood.

Therefore, we aimed to investigate the mechanism by which IFNα improves the anti–PD-1 antibody–induced immune response in HCC. Here, we show for the first time that combination therapy comprising IFNα and an anti–PD-1 antibody could strongly inhibit HCC progression in both HCC mouse models and HCC patients. IFNα and anti–PD-1 combination treatment was found to impair glycolysis and glucose uptake in HCC cells and enhance glycolysis in tumor-infiltrating CTLs, which was attenuated after anti–PD-1 antibody administration. Our data reveal that combination therapy with IFNα and an anti–PD-1 antibody is a promising ICB strategy for HCC.

Patients with Unresectable HCC Have Remarkable Clinical Responses to Anti–PD-1–Based Immunotherapy Combined with IFNα

Although both PD-1 inhibitors and IFNα have been evaluated separately in HCC patients in clinical trials and showed promising activity, the combined effect of these two agents remains elusive in HCC. To this end, we observed the therapeutic efficacy of 15 patients with unresectable HCC who were treated with anti–PD-1–based immunotherapy combined with IFNα. Unexpectedly, we found that the ORR was 40.0% (6 of 15) and the disease control rate (DCR) was 80.0% (12 of 15), and only 20.0% of the patients (3 of 15) still progressed on therapy (Fig. 1A and B). Above all, no treatment-related deaths occurred. The clinical characteristics of all patients are listed in Supplementary Table S1. Here, we report on two patients with unresectable HCC who had previously failed PD-1 blockade but later showed a remarkable clinical response after treatment with an anti–PD-1 antibody in combination with IFNα.

Figure 1.

IFNα enhances the efficacy of anti–PD-1–based immunotherapy in HCC. A and B, Clinical outcome assessments of the cohort of 15 patients with unresectable HCC treated with anti–PD-1–based immunotherapy combined with IFNα. The ORR was 40.0% (6 of 15 patients), and the DCR was 80.0% (12 of 15 patients). Six patients (40.0%) had partial responses (PR), six patients (40.0%) had stable disease (SD), and three patients (20.0%) had progressive disease (PD; A). Best change from baseline in the tumor burden per patient referring to the modified RECIST (mRECIST) guidelines (B). C and D, Two representative patients with unresectable HCC who were initially treated with PD-1 blockade and then treated with PD-1 blockade combined with IFNα. Red arrows/dotted circles indicate the target lesions/metastasis. C, Representative MRI scans of target lesions in patient #1. D, Representative CT scans of lung metastasis in patient #2 during the indicated treatment. E and F, Flow cytometry analysis was performed using longitudinal peripheral blood samples collected (at pretreatment and late during treatment) from patients undergoing treatment with anti–PD-1–based immunotherapy combined with IFNα. Percentages of CD8+ and CD4+ cells in the peripheral blood collected before treatment versus those collected after treatment with PD-1 blockade and IFNα (n = 8). SSC, side scatter. All data presented are shown as the mean ± SD. *, P < 0.05; ns, not significant.

Figure 1.

IFNα enhances the efficacy of anti–PD-1–based immunotherapy in HCC. A and B, Clinical outcome assessments of the cohort of 15 patients with unresectable HCC treated with anti–PD-1–based immunotherapy combined with IFNα. The ORR was 40.0% (6 of 15 patients), and the DCR was 80.0% (12 of 15 patients). Six patients (40.0%) had partial responses (PR), six patients (40.0%) had stable disease (SD), and three patients (20.0%) had progressive disease (PD; A). Best change from baseline in the tumor burden per patient referring to the modified RECIST (mRECIST) guidelines (B). C and D, Two representative patients with unresectable HCC who were initially treated with PD-1 blockade and then treated with PD-1 blockade combined with IFNα. Red arrows/dotted circles indicate the target lesions/metastasis. C, Representative MRI scans of target lesions in patient #1. D, Representative CT scans of lung metastasis in patient #2 during the indicated treatment. E and F, Flow cytometry analysis was performed using longitudinal peripheral blood samples collected (at pretreatment and late during treatment) from patients undergoing treatment with anti–PD-1–based immunotherapy combined with IFNα. Percentages of CD8+ and CD4+ cells in the peripheral blood collected before treatment versus those collected after treatment with PD-1 blockade and IFNα (n = 8). SSC, side scatter. All data presented are shown as the mean ± SD. *, P < 0.05; ns, not significant.

Close modal

Patient #1 was a 67-year-old female with unresectable HCC who underwent transarterial chemoembolization (TACE). Thereafter, the patient received PD-1 blockade therapy followed by TACE. However, she did not have a significant clinical response to the treatment and eventually progressed on PD-1 blockade. Abdominal MRI was performed, and her liver tumors had typical enhancement. The largest tumor (2.3 × 4.1 cm) was in segment VIII adjacent to branches of the hepatic portal veins. Unexpectedly, MRI scans taken about 7 weeks after the IFNα and anti–PD-1 combination treatment showed notable tumor shrinkage and necrosis in this patient, and a subsequent scan after 3 months of treatments confirmed the patient's remarkable response with minimal tumor activity (Fig. 1C). In addition, no further progression was noticeable on serial imaging. Notably, patient #1 had no other toxic effects or immune-related adverse events.

Patient #2 was a 34-year-old man with giant HCC. Previously, he had undergone multiple cycles of TACE as well as two hepatic resections, followed by administration of PD-1 blockade therapy. He appeared well on follow-up with no evidence of recurrence for 1 year. Two years later, he presented with dyspnea, and a chest CT scan showed >10 lung metastatic nodules, which were confirmed as metastatic HCC by thoracoscopic biopsy. Surprisingly, after 2-month IFNα and anti–PD-1 combination treatment, the restaging chest CT was performed and showed the complete eradication of numerous lung metastases in cross-sections taken at several levels and shrinkage of the largest nodule (1.4 cm) without evidence of newly developed metastatic disease (Fig. 1D).

Additionally, we analyzed the immune cells in the peripheral blood collected from patients who received an anti–PD-1–based immunotherapy combined with IFNα compared with those from pretreatment samples. In the analysis of the main immune cell types, we detected enrichment of CD8+ T cells but not CD4+ T cells in the peripheral blood using flow cytometry (Fig. 1E and F). We also noticed that there was no significant change of naïve CD8+ T cells, central memory CD8+ T cells, effector CD8+ T cells, and effector memory CD8+ T cells between pretreatment and posttreatment (Supplementary Fig. S1A–S1D). Collectively, our results demonstrate that the combination of IFNα and an anti–PD-1–based immunotherapy exhibits an acceptable toxicity profile with promising evidence of enhanced clinical benefit in patients with unresectable HCC.

IFNα Treatment Potentiates the Efficacy of an Anti–PD-1 Antibody in Murine HCC Models

To explore the mechanism underlying the improved efficacy of the combination therapy, we established orthotopic syngeneic mouse models of HCC with Hepa1-6 or H22 cells. We first assessed the effects of the combination therapy on immunocompetent orthotopic HCC mouse models (Fig. 2A; see Methods for the dosing schedule). There were no changes in body weight among the tested mice, suggesting the gross safety of the combination therapy (Supplementary Fig. S2A and S2B). In the Hepa1-6 model, compared with monotherapy with IFNα, an anti–PD-1 antibody, or an IgG control, the combination therapy resulted in significant Hepa1-6 tumor regression and tumor size reduction detected by an ultrasound imaging platform (Fig. 2B). Similar phenotypes were also observed in the spontaneous HCC model driven by c-Myc and immunocompetent H22 orthotopic tumor–bearing model mice (Fig. 2C; Supplementary Fig. S2C): The combination therapy led to greater tumor regression than either IFNα or anti–PD-1 treatment alone. Importantly, the overall survival (OS) of several HCC models including the c-Myc-driven spontaneous HCC model, the Hepa1-6 tumor–bearing mouse model, and the H22 tumor–bearing mouse model that received the combination therapy was significantly improved compared with that of corresponding mice that received either treatment alone (Fig. 2D and E; Supplementary Fig. S2D). IHC analysis further confirmed the spontaneous induction of HCC, as evidenced by universal nuclear staining of hepatocytic-specific transcription factor HNF4α but not biliary-specific transcription factor CK-19 (Supplementary Fig. S2E). We also observed that the combination therapy resulted in severe tumor necrosis and complete elimination of metastatic lesions in the lungs (Fig. 2F and G; Supplementary Fig. S2F–S2H).

Figure 2.

IFNα enhances the efficacy of anti–PD-1 immunotherapy and transforms the immune landscape of the TME. A, Schematic representation of the therapy schedule for IFNα, anti–PD-1, or combination therapy. B, Growth curves of Hepa1-6 tumors in C57BL/6J mice (vehicle + IgG, 10 mg/kg, n = 16; IFNα, 1 × 104 IU, n = 12; anti–PD-1, 10 mg/kg, n = 15; combination therapy, n = 15). C, Representative images and the statistical results from spontaneous HCC models that received the indicated treatments (6 mice per group; vehicle + IgG, 10 mg/kg; IFNα, 1 × 104 IU; anti–PD-1, 10 mg/kg; combination therapy). Scale bars: 1 cm. D, Survival curves of C57BL/6J mice bearing Hepa1-6 tumors (n = 15 per group) that received 10 mg/kg IgG, 1 × 104 IU IFNα, 10 mg/kg anti–PD-1, or the combination therapy. E, Survival curves of the spontaneous HCC model (n = 15 per group) that received 10 mg/kg IgG, 1 × 104 IU IFNα, 10 mg/kg anti–PD-1, or the combination therapy. F and G, Representative hematoxylin and eosin staining of lungs from mice bearing Hepa1-6 tumors (vehicle + IgG, 10 mg/kg, n = 16; IFNα, 1 × 104 IU, n = 12; anti–PD-1, 10 mg/kg, n = 15; combination therapy, n = 15) and the incidence of lung metastasis in the indicated groups. Arrows in F indicate metastasis. H, t-Distributed stochastic neighbor embedding (t-SNE) plots of tumor-infiltrating T cells, B cells, natural killer (NK) cells, myeloid cells, CD4+ cells, CD8+ cells, and double-negative (DN) T cells in the Hepa1-6 tumors by CyTOF analysis from Hepa1-6 tumors given the indicated treatment. I, Quantification of tumor-infiltrating T cells, B cells, NK cells, myeloid cells, CD4+ cells, CD8+ cells, and DN T cells in Hepa1-6 tumors given the indicated treatment, assessed by CyTOF, and analyzed by FlowJo. The results are shown as the mean ± SD, and the statistical significance of differences between groups was determined by an unpaired Student t test. **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 2.

IFNα enhances the efficacy of anti–PD-1 immunotherapy and transforms the immune landscape of the TME. A, Schematic representation of the therapy schedule for IFNα, anti–PD-1, or combination therapy. B, Growth curves of Hepa1-6 tumors in C57BL/6J mice (vehicle + IgG, 10 mg/kg, n = 16; IFNα, 1 × 104 IU, n = 12; anti–PD-1, 10 mg/kg, n = 15; combination therapy, n = 15). C, Representative images and the statistical results from spontaneous HCC models that received the indicated treatments (6 mice per group; vehicle + IgG, 10 mg/kg; IFNα, 1 × 104 IU; anti–PD-1, 10 mg/kg; combination therapy). Scale bars: 1 cm. D, Survival curves of C57BL/6J mice bearing Hepa1-6 tumors (n = 15 per group) that received 10 mg/kg IgG, 1 × 104 IU IFNα, 10 mg/kg anti–PD-1, or the combination therapy. E, Survival curves of the spontaneous HCC model (n = 15 per group) that received 10 mg/kg IgG, 1 × 104 IU IFNα, 10 mg/kg anti–PD-1, or the combination therapy. F and G, Representative hematoxylin and eosin staining of lungs from mice bearing Hepa1-6 tumors (vehicle + IgG, 10 mg/kg, n = 16; IFNα, 1 × 104 IU, n = 12; anti–PD-1, 10 mg/kg, n = 15; combination therapy, n = 15) and the incidence of lung metastasis in the indicated groups. Arrows in F indicate metastasis. H, t-Distributed stochastic neighbor embedding (t-SNE) plots of tumor-infiltrating T cells, B cells, natural killer (NK) cells, myeloid cells, CD4+ cells, CD8+ cells, and double-negative (DN) T cells in the Hepa1-6 tumors by CyTOF analysis from Hepa1-6 tumors given the indicated treatment. I, Quantification of tumor-infiltrating T cells, B cells, NK cells, myeloid cells, CD4+ cells, CD8+ cells, and DN T cells in Hepa1-6 tumors given the indicated treatment, assessed by CyTOF, and analyzed by FlowJo. The results are shown as the mean ± SD, and the statistical significance of differences between groups was determined by an unpaired Student t test. **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

To test the extent to which the TME contributed to the antitumor effect of IFNα, immunodeficient NOD CRISPR Prkdc II2rg (NCG) mice with established Hepa1-6 tumors were treated with IFNα (Supplementary Fig. S2I). We found that IFNα induced a minimal delay in tumor growth in the NCG mice, as indicated by the tumor size compared with that of vehicle control–treated mice, suggesting that the observed antitumor effect of IFNα largely depended on immune cells. To clarify whether IFNα directly acts on immune cells, we compared the expression pattern of IFNAR1, the specific receptor of IFNα, between HCC cells and CD8+ T cells derived from orthotopic models. Public single-cell sequencing data based on Ma and colleagues (28) indicated that tumor parenchymal cells show higher expression of IFNAR1 than CD8+ T cells (Supplementary Fig. S2J and S2K). Similarly, we found that HCC cells exhibited drastically higher expression of IFNAR1 in these orthotopic models compared with tumor-infiltrating CD8+ T cells (Supplementary Fig. S2L). Importantly, we also constructed Ifnar1-knockdown (KD) HCC cells by short hairpin RNA (shRNA)–mediated downregulation of Ifnar1. Orthotopic models were subsequently established with Ifnar1-KD Hepa1-6 cells and received the indicated treatment (Supplementary Fig. S2M). We found that knocking down Ifnar1 in HCC cells abolished the therapeutic potential of IFNα. Notably, IFNα treatment failed to synergize with anti–PD-1 antibody after silencing Ifnar1 expression (Supplementary Fig. S2M), highlighting that IFNα targeted tumor cells and tumor-intrinsic IFNAR1 was essential for IFNα-based therapy. Taken together, our data demonstrate that IFNα enhances the efficacy of anti–PD-1 therapy through the intact immune system.

Given the apparent dependency of the combination treatment on immune cell infiltration, we wanted to investigate the infiltrating immune cells in the TME. To conduct a high-dimensional characterization of the TME immune landscape of Hepa1-6 syngeneic mouse tumors from the four treatment groups, we first assessed the expression of 42 surface and intracellular immune markers on CD45+ cells from disaggregated Hepa1-6 tumors using time-of-flight mass cytometry (CyTOF). Our metal-labeled antibody panel was devised to identify the major tumor-infiltrating immune cell populations (Supplementary Table S2) and comprised cell lineage markers, function-related markers (homing and activation receptors), and cytokine-related markers. With this approach, we were able to divide CD45+ cells within TME into lymphoid and myeloid compartments (Fig. 2H; Supplementary Fig. S2N) and analyzed the expression of the corresponding lineage markers in each group (Supplementary Fig. S2O). However, after manually gating the percentages of each major immune lineage in each individual sample from the different treatment groups, we found that the tumor-infiltrating lymphocyte (TIL) density as well as the other major lineages showed no significant differences among the four groups, suggesting that the potential subgroup of immune cells might contribute to the antitumor effect of combination therapy (Fig. 2I). To explore the differentiation states of TILs within tumors derived from mouse models receiving different treatments, we further applied flow cytometry analysis to evaluate the exhausted states. We found anti–PD-1 treatment decreased the proportion of progenitor-exhausted CD8+ T cells (defined as TCF+TIM3) compared with the vehicle group, while IFNα treatment exerted no significant effect on progenitor exhausted CD8+ T cells. Importantly, combination anti–PD-1 antibody and IFNα resulted in a decrease of progenitor-exhausted CD8+ T cells to the greatest extent (Supplementary Fig. S2P). Interestingly, although terminally exhausted CD8+ T cells (defined as TCFTIM3+) were decreased after anti–PD-1 treatment, IFNα exerted more powerful induction effect on decreasing terminally exhausted CD8+ T cells than anti–PD-1 did. However, combination therapy showed no obvious synergic effects on reducing terminally exhausted CD8+ T cells (Supplementary Fig. S2Q). Thus, we found that IFNα treatment showed moderate effect on ameliorating the exhausted state of CD8+ T cells, and more importantly, IFNα potentiated the effect of anti–PD-1 antibody on reversing exhausted effector T cells in HCC. In summary, our results further validated the clinical efficacy of PD-1 blockade/IFNα combination therapy in immunocompetent orthotopic and spontaneous HCC model mice.

High-Dimensional Analysis Shows Enrichment of CD27+CD8+ T Cells upon PD-1 Blockade in Combination with IFNa Treatment

Given that cancer immunotherapy depends mainly on T-cell infiltration in the tumor immune microenvironment (TIME; ref. 29), we depleted CD4+ T cells, CD8+ T cells, or both to further assess which T-cell populations contribute to the improved anticancer effect of IFNα combined with an anti–PD-1 antibody (Fig. 3A). To deplete CD4+ T cells and CD8+ T cells in the Hepa1-6 tumor mouse model, we treated mice with anti-CD4– and anti-CD8–neutralizing antibodies or isotype control IgG. We found that depletion of CD8+ T cells or both CD4+ T cells and CD8+ T cells completely blocked the tumor regression induced by the combination therapy, whereas depletion of CD4+ T cells alone, similar to control treatment, had no such effect (Fig. 3B; Supplementary Fig. S3A and S3B). These results demonstrate that the combination of IFNα and an anti–PD-1 antibody eliminates tumor cells in a CD8+ T cell–dependent manner; thus, we focused on characterizing the role of CD8+ T cells in further analyses.

Figure 3.

In-depth characterization of the T-cell compartment by CyTOF. A, Schedule of tumor implantation and injection of antibodies for cell depletion in mice. B, Depletion of CD8+ or CD4+ cells in the setting of combination therapy with IFNα and anti–PD-1. Cumulative graph of the mean tumor size per group (n = 5) after CD8+ and CD4+ T-cell depletion. For CD8+ and CD4+ T-cell depletion, mice were injected i.p. with 10 mg/kg anti-CD8α and anti-CD4α antibodies twice weekly beginning 1 day before therapy as indicated. C, Heat map shows normalized expression of selected T-cell panel markers for the 21 T-cell clusters displayed on the t-SNE plot shown in E. CM, central memory; DN, double-negative; EM, effector memory. D, t-Distributed stochastic neighbor embedding (t-SNE) plots of infiltrating T-cell populations in each indicated group. E, t-SNE plots of intratumoral T cells from all groups merged with populations and dot plots showing the frequency of CD27+CD8+ T cells for each treatment group. F, Representative images of multiplex immunofluorescence results for Hepa1-6 tumor tissue stained for nuclei using DAPI (blue), CD8 (green), and CD27 (red) and quantification of CD27+CD8+ T cells in mice bearing Hepa1-6 tumors from the indicated groups. Scale bars: 50 μm. G, Evaluation of the influence of CD27 on CD8+ T-cell function after transfection of NT-siRNA or Cd27-siRNA by flow cytometry. H, Uniform manifold approximation and projection (UMAP) plot visualization of CD27 expression in CD8+ T-cell clusters detected in patients with HCC. I and J, Single-cell sequencing analysis of T-cell populations from patients with HCC showing the correlations between CD27 expression in CD8+ T cells and effector T-cell activation markers. I, Violin plots showing differentially expressed genes between CD27hiCD8 T cells and CD27loCD8 T cells from patients with HCC. J, Volcano plot showing the indicated genes with differential expression in CD27hiCD8 T cells compared with CD27loCD8 T cells from patients with HCC. Each dot represents a gene, with color annotation inside. FC, fold change. K and L, Flow cytometry analysis of PD-1 expression in CD27hiCD8+ T cells compared with CD27loCD8+ T cells derived from human HCC sample. MFI, mean fluorescence intensity. M, Representative HCC tumor samples showing the expression of CD8, CD27, Foxp3, and CD206. Scale bars: 100 μm. Scatter plot analysis showing the correlations between the number of intratumoral CD27+CD8+ cells and macrophages or Tregs. The results are shown as the mean ± SD for experiments performed in triplicate. Each experiment was repeated three times, and the statistical significance of differences between groups was determined by an unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 3.

In-depth characterization of the T-cell compartment by CyTOF. A, Schedule of tumor implantation and injection of antibodies for cell depletion in mice. B, Depletion of CD8+ or CD4+ cells in the setting of combination therapy with IFNα and anti–PD-1. Cumulative graph of the mean tumor size per group (n = 5) after CD8+ and CD4+ T-cell depletion. For CD8+ and CD4+ T-cell depletion, mice were injected i.p. with 10 mg/kg anti-CD8α and anti-CD4α antibodies twice weekly beginning 1 day before therapy as indicated. C, Heat map shows normalized expression of selected T-cell panel markers for the 21 T-cell clusters displayed on the t-SNE plot shown in E. CM, central memory; DN, double-negative; EM, effector memory. D, t-Distributed stochastic neighbor embedding (t-SNE) plots of infiltrating T-cell populations in each indicated group. E, t-SNE plots of intratumoral T cells from all groups merged with populations and dot plots showing the frequency of CD27+CD8+ T cells for each treatment group. F, Representative images of multiplex immunofluorescence results for Hepa1-6 tumor tissue stained for nuclei using DAPI (blue), CD8 (green), and CD27 (red) and quantification of CD27+CD8+ T cells in mice bearing Hepa1-6 tumors from the indicated groups. Scale bars: 50 μm. G, Evaluation of the influence of CD27 on CD8+ T-cell function after transfection of NT-siRNA or Cd27-siRNA by flow cytometry. H, Uniform manifold approximation and projection (UMAP) plot visualization of CD27 expression in CD8+ T-cell clusters detected in patients with HCC. I and J, Single-cell sequencing analysis of T-cell populations from patients with HCC showing the correlations between CD27 expression in CD8+ T cells and effector T-cell activation markers. I, Violin plots showing differentially expressed genes between CD27hiCD8 T cells and CD27loCD8 T cells from patients with HCC. J, Volcano plot showing the indicated genes with differential expression in CD27hiCD8 T cells compared with CD27loCD8 T cells from patients with HCC. Each dot represents a gene, with color annotation inside. FC, fold change. K and L, Flow cytometry analysis of PD-1 expression in CD27hiCD8+ T cells compared with CD27loCD8+ T cells derived from human HCC sample. MFI, mean fluorescence intensity. M, Representative HCC tumor samples showing the expression of CD8, CD27, Foxp3, and CD206. Scale bars: 100 μm. Scatter plot analysis showing the correlations between the number of intratumoral CD27+CD8+ cells and macrophages or Tregs. The results are shown as the mean ± SD for experiments performed in triplicate. Each experiment was repeated three times, and the statistical significance of differences between groups was determined by an unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

Several studies have discovered dynamic changes in CD8+ T-cell subpopulations during the antitumor immune response (30, 31). To further identify the specific subpopulations of CD8+ T cells that mediate the potent antitumor effect of the combination therapy, we computationally identified lymphoid cells in the CD45+ tumor-infiltrating cell population and reutilized the CyTOF-based systems approach to analyze 27 markers specific to CTL functions. This method yielded 21 distinct lymphoid subpopulations (including four distinct CD8+ T-cell clusters) broadly defined by the markers (Fig. 3C and D). Moreover, this method identified a significant subpopulation shift in CD8+ T cells within Hepa1-6 tumors among the four treatment groups. Notably, we found markedly elevated infiltration of the CD27+CD8+ T-cell subset in tumor tissues from mice that received the combination IFNα and anti–PD-1 therapy compared with those from mice on other treatments (Fig. 3E; Supplementary Fig. S3C), which was further confirmed by multicolor immunofluorescence (Fig. 3F). Furthermore, we adopted the HCC patient-derived organoid (PDO) and autologous intratumoral immune cell coculture model based on previously published methods (32, 33), which served as an in vitro platform to mimic the development of checkpoint inhibition blockade (Supplementary Fig. S3D). Similar to our in vivo results, IFNα was able to increase CD27 on CD8+ T cells, while the anti–PD-1 antibody failed to increase CD27. Combination IFNα and anti–PD-1 led to the increase of CD27 to the highest extent among all groups (Supplementary Fig. S3E and S3F).

We hypothesized that CD27 expression on CD8+ T cells might correlate with T-cell antitumor activities. We knocked down Cd27 in CD8+ T cells via Cd27-specific siRNA. After activation by anti-CD3 and anti-CD28 antibodies, CD8+ T cells derived from Hepa1-6 tumors with Cd27 KD exhibited attenuated effector function, as indicated by reduced secretion of proinflammatory cytokines such as TNFα, IFNγ, and GZMB compared with control CD8+ T cells transduced with nontargeting siRNA (Fig. 3G). Our findings were further supported by analysis of single-cell sequencing data. We generated integrated single-cell RNA sequencing (scRNA-seq) data of HCC TILs merged from two public HCC data sets (GSE140228 and GSE125449; refs. 28, 34) using Seurat's canonical correlation analysis (CCA; ref. 35). In accordance with our previous findings, the integrated single-cell data showed a differential CD27 expression profile among CD8+ T-cell subclusters (Fig. 3H), and the expression of genes important for T-cell cytotoxicity and proliferation (IFNG, ICOS, GZMA, GZMK, and STMN1) was increased in CD27hiCD8+ T cells (Fig. 3I and J). Surprisingly, we noted that the expression of PDCD1 (encoding PD-1) was also elevated in CD27hiCD8+ T cells (Fig. 3I and J). To confirm this finding, we separated tumor-infiltrating CD8+ T cells from patients with HCC and found that PD-1 expression in CD27hiCD8+ T cells was significantly higher than in CD27loCD8+ T cells (Fig. 3K and L). These findings provided a rationale for anti–PD-1 antibody–based immunotherapy. Additionally, the expression of genes involved in the effector memory CD8+ T-cell signature was elevated in CD27hi T cells (Supplementary Fig. S3G). Furthermore, using tissue microarrays from our in-house HCC cohort, we found that CD27 was negatively correlated with FOXP3 and CD206, markers of regulatory T cells (Treg) and immunosuppressive macrophages, respectively (Fig. 3M).

Overall, we speculated that the CD27+CD8+ population is the subpopulation of CD8+ T cells that mediates the observed antitumor effects of the combination of IFNα and anti–PD-1 and that CD27 is the key factor in the effector function of CD8+ T cells.

IFNa Enhances Anti–PD-1 Efficacy by Suppressing HIF1α-Induced Glycolysis in HCC

To further elucidate the mechanism by which IFNα enhances the efficacy of anti–PD-1, RNA sequencing (RNA-seq) of tumor samples from the Hepa1-6 model was carried out to identify the transcriptional alterations after anti–PD-1 treatment with or without IFNα administration. Considering tumor purity, we selected samples composed of at least 80% tumor nuclei, which was validated by pathologists to reflect the alteration in malignant cells according to The Cancer Genome Atlas (TCGA) tissue sample requirements (available at https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga/studied-cancers; ref. 36). Gene set enrichment analysis (GSEA) showed that HIF1α signaling was positively enriched in the anti–PD-1 group compared with vehicle + IgG as the control group (P = 0.039; Fig. 4A). More importantly, HIF1α signaling was significantly inhibited following combination therapy administration (P < 0.001; Fig. 4B). To verify these findings, we separated malignant as well as CD8+ T cells from tumor tissues of all four groups via flow cytometry sorting and conducted immunoblotting analysis. We observed higher expression of HIF1α and its downstream targets, such as PKM2, LDHA, and HK1, in the surviving Hepa1-6 and H22 cells derived from C57BL/6J or BALB/c mice, respectively, that received anti–PD-1 compared with control-treated mice. IFNα treatment dramatically repressed the expression levels of these markers (Fig. 4C; Supplementary Fig. S4A). In contrast, the expression of HIF1α and its downstream targets in CD8+ T cells derived from both C57BL/6J and BALB/c mouse models was diminished by PD-1 blockade but could be successfully rescued by IFNα, indicating that the combination treatment maintained infiltrating CD8+ T cells in a highly glycolytic state (Fig. 4C; Supplementary Fig. S4A). Moreover, Hepa1-6 cells that survived anti–PD-1 treatment showed the highest proliferating cell nuclear antigen (PCNA) expression, accompanied by the lowest cleaved caspase-3 expression. In contrast, CD8+ T cells derived from residual foci in anti–PD-1 antibody–treated C57BL/6J mice showed the lowest expression of PCNA but the highest expression of cleaved caspase-3 (Supplementary Fig. S4B). As expected, the combination treatment reversed the expression patterns of PCNA and cleaved caspase-3. Glycolysis is a feature of both cancer cell progression and T-cell activation (17). Previous reports have suggested that microenvironmental glucose competition may be a driver of immunosuppression by depleting necessities required to maintain effector T-cell function, whereas blockade of glycolysis in malignant cells can boost the efficiency of immunotherapy and metabolic remodeling between tumor cells and T cells (37). By analyzing the fluorescence signal of CFSE and the percentage of IFNγ+ cells, we confirmed that increased glucose uptake drives CD8+ T-cell proliferation and activation (Supplementary Fig. S4C and S4D).

Figure 4.

Aberrant HIF1α signaling activation in HCC cells results in glucose restriction to alter the T-cell glycolytic state, and IFNα treatment restores T-cell cytotoxic capacities. A, GSEA results indicated that hyperactivation of HIF1α signaling was observed in surviving HCC cells after anti–PD-1 treatment compared with surviving cells following control treatment. NES, normalized enrichment score. B, GSEA results showed that HIF1α signaling was significantly inhibited in HCC cells treated with IFNα and anti–PD-1 compared with HCC cells treated with anti–PD-1 alone. C, Immunoblot assays for PKM2, LDHA, and HK1 in Hepa1-6 cells (left) and CD8+ T cells (right) derived from indicated Hepa1-6 tumors that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. D, ECAR results for tumor cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. E, ECAR results for CD8+ T cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. F, Glucose concentration in the extracellular milieu of HCC tissues derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. G, IFNγ+CD8+ T cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. H, CD27+CD8+ T cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. I, Diagram illustrations for the coculture system established to evaluate the impacts of HIF1α signaling on the function of CD8+ T cells. HIF1αi, HIF1α inhibitor. J, ECAR results for Hepa1-6 cells that received IgG or 100 μg/mL anti–PD-1 treatment with or without 10 μmol/L KC7F2. K, ECAR results for CD8+ T cells after coculture with Hepa1-6 cells that received IgG or 100 μg/mL anti–PD-1 treatment with or without 10 μmol/L KC7F2. L, CD27+ proportions of CD8+ T cells after coculture with tumor cells that received IgG or 100 μg/mL anti–PD-1 treatment with or without 10 μmol/L KC7F2. M, IFNγ+ proportions of CD8+ T cells after coculture with tumor cells that received IgG or 100 μg/mL anti–PD-1 treatment with or without 10 μmol/L KC7F2. N, Representative B-ultrasound (B-US) and general images of tumors derived from Hif1α-KD C57BL/6J mouse models that received IgG or 10 mg/kg anti–PD-1. O, Growth curves for Hif1α-KD C57BL/6J mouse models (n = 6 per group) that received IgG or 10 mg/kg anti–PD-1. P, Glucose concentration in the extracellular milieu of HCC tissues derived from indicated Hepa1-6 tumors that received Hif1α silencing and/or an anti–PD-1 concentration of 10 mg/kg. Q, CD27+CD8+ T cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received IgG or 10 mg/kg anti–PD-1. The results are shown as the mean ± SD for experiments performed in triplicate. Each experiment was repeated three times, and the statistical significance of differences between groups was determined by an unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 4.

Aberrant HIF1α signaling activation in HCC cells results in glucose restriction to alter the T-cell glycolytic state, and IFNα treatment restores T-cell cytotoxic capacities. A, GSEA results indicated that hyperactivation of HIF1α signaling was observed in surviving HCC cells after anti–PD-1 treatment compared with surviving cells following control treatment. NES, normalized enrichment score. B, GSEA results showed that HIF1α signaling was significantly inhibited in HCC cells treated with IFNα and anti–PD-1 compared with HCC cells treated with anti–PD-1 alone. C, Immunoblot assays for PKM2, LDHA, and HK1 in Hepa1-6 cells (left) and CD8+ T cells (right) derived from indicated Hepa1-6 tumors that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. D, ECAR results for tumor cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. E, ECAR results for CD8+ T cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. F, Glucose concentration in the extracellular milieu of HCC tissues derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. G, IFNγ+CD8+ T cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. H, CD27+CD8+ T cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments. IFNα concentration: 1 × 104 IU; anti–PD-1 concentration: 10 mg/kg. I, Diagram illustrations for the coculture system established to evaluate the impacts of HIF1α signaling on the function of CD8+ T cells. HIF1αi, HIF1α inhibitor. J, ECAR results for Hepa1-6 cells that received IgG or 100 μg/mL anti–PD-1 treatment with or without 10 μmol/L KC7F2. K, ECAR results for CD8+ T cells after coculture with Hepa1-6 cells that received IgG or 100 μg/mL anti–PD-1 treatment with or without 10 μmol/L KC7F2. L, CD27+ proportions of CD8+ T cells after coculture with tumor cells that received IgG or 100 μg/mL anti–PD-1 treatment with or without 10 μmol/L KC7F2. M, IFNγ+ proportions of CD8+ T cells after coculture with tumor cells that received IgG or 100 μg/mL anti–PD-1 treatment with or without 10 μmol/L KC7F2. N, Representative B-ultrasound (B-US) and general images of tumors derived from Hif1α-KD C57BL/6J mouse models that received IgG or 10 mg/kg anti–PD-1. O, Growth curves for Hif1α-KD C57BL/6J mouse models (n = 6 per group) that received IgG or 10 mg/kg anti–PD-1. P, Glucose concentration in the extracellular milieu of HCC tissues derived from indicated Hepa1-6 tumors that received Hif1α silencing and/or an anti–PD-1 concentration of 10 mg/kg. Q, CD27+CD8+ T cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received IgG or 10 mg/kg anti–PD-1. The results are shown as the mean ± SD for experiments performed in triplicate. Each experiment was repeated three times, and the statistical significance of differences between groups was determined by an unpaired Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

Therefore, in light of the necessity of HIF1α signaling in regulating tumoral glycolysis, we hypothesized that IFNα may also enhance anti–PD-1 therapeutic efficiency in HCC by regulating glycolysis. To verify this hypothesis, we first evaluated the glycolytic capacities of tumors and CD8+ T cells via a Seahorse extracellular acidification rate (ECAR) assay, which is a standard way to measure glycolysis in cells (38). The results showed that ECAR was significantly elevated in HCC cells derived from the groups that received anti–PD-1 treatment alone compared with those from the control groups in both C57BL/6J and BALB/c mouse models; however, ECAR was dramatically decreased after the addition of IFNα. Importantly, ECAR was the lowest in HCC cells derived from mice treated with the combination therapy when compared with either treatment alone or vehicle control treatment (Fig. 4D; Supplementary Fig. S4E). In contrast, ECAR was significantly decreased in CD8+ T cells derived from mice treated with anti–PD-1 treatment alone but increased after IFNα single reagent and was highest in CD8+ T cells from mice treated with anti–PD-1 and IFNα in combination in both C57BL/6J and BALB/c mouse models (Fig. 4E; Supplementary Fig. S4F). Accordingly, the microenvironmental glucose concentration was significantly decreased after anti–PD-1 treatment, but this effect was dramatically reversed by IFNα (Fig. 4F; Supplementary Fig. S4G). The proportions of IFNγ+ and CD27+ cells derived from both mouse models were also highest when mice were treated with both anti–PD-1 and IFNα (Fig. 4G and H; Supplementary Fig. S4H and S4I). These results indicated that HCC cells with enhanced HIF1α signaling activity and glycolytic potential survive; however, IFNα addition could restrain glycolysis in HCC cells, thus fostering activation of CD8+ T cells. To further exclude a direct effect of IFNα on CD8+ T cells, we also treated CD8+ T cells separated from the spleens of C57BL/6J mice with 500 IU/mL of IFNα, followed by functional assays. Flow cytometry assays demonstrated that IFNα treatment alone failed to increase CD27+ proportion (Supplementary Fig. S4J), proliferation (Supplementary Fig. S4K), or IFNγ secretion of CD8+ T cells (Supplementary Fig. S4L). Taken together, these data indicated that IFNα could modulate glucose competition between tumor cells and CD8+ T cells but not directly act on T cells, which might rely on HIF1α signaling in tumor cells.

Next, we aimed to determine whether HIF1α signaling drives the metabolic restriction of effector CD8+ T cells and impairment of anti–PD-1 treatment efficacy. First, we established an in vitro coculture system with syngeneic peripheral blood mononuclear cells (PBMC) derived from C57BL/6J or BALB/c mice and HCC cells in the presence of KC7F2 (a specific HIF1α inhibitor; ref. 39) or not (Fig. 4I). Immunoblot assays confirmed that HIF1α expression was extensively inhibited by KC7F2 in Hepa1-6 and H22 cells isolated from C57BL/6J or BALB/c mouse models, respectively (Supplementary Fig. S4M). Anti–PD-1 treatment resulted in a dramatically increased ECAR in both Hepa1-6 and H22 cells, and the effect was ameliorated by the HIF1α antagonist (Fig. 4J; Supplementary Fig. S4N). The ECAR of cocultured CD8+ T cells decreased afterward and was lowest when anti–PD-1 was used alone. In contrast, the combination of the HIF1α antagonist and anti–PD-1 antibody sustained the ECAR of cocultured CD8+ T cells at the highest level, which was observed in both coculture models (Fig. 4K; Supplementary Fig. S4O). Further experiments indicated that CD8+ T cells cocultured with HIF1α antagonist–pretreated Hepa1-6 or H22 cells supplemented with anti–PD-1 had the highest proportions of CD27+ and IFNγ+ cells (Fig. 4L and M; Supplementary Fig. S4P and S4Q). To further verify these in vitro results, we established Hif1α-KD Hepa1-6 cells to generate orthotopic HCC models in C57BL/6J mice and treated the mice with anti–PD-1. Tumor volume was assessed by b-mode ultrasound tomography (Fig. 4N). The results indicated that Hif1α KD exerted synergistic effects with anti–PD-1 treatment to inhibit tumor growth (Fig. 4O). Moreover, silencing Hif1α expression in HCC cells established a high-glucose microenvironment and exerted synergistic effects with anti–PD-1 treatment, as evidenced by this approach producing the highest microenvironmental glucose concentration (Fig. 4P). In concordance with the ex vivo glucose results, infiltrating CD8+ T cells from the Hif1α-KD group showed a significantly higher proportion of CD27+ cells after anti–PD-1 treatment (Fig. 4Q).

Subsequent in vitro cytotoxicity experiments were performed to verify whether IFNα promotes cytotoxic activities at the cellular level. In many studies, a CFSE killing assay has been widely used to detect the in vitro killing activity of cytotoxic T cells (40, 41). OT-I splenocytes were cocultured with EL4 cells in vitro, followed by different concentrations of IFNα treatment. Results showed that CD27+CD8+ T cells were also significantly increased in a dose-dependent manner in response to IFNα treatment (Supplementary Fig. S5A). In addition, combination treatment with IFNα and anti–PD-1 significantly improved T-cell cytotoxic activity (Supplementary Fig. S5B). The effects of the glucose concentration on the killing capacity of CD8+ T cells were also verified. Compared with those in the low-glucose (5 mmol/L) group, OT-I CTLs in the high-glucose (25 mmol/L) group showed a stronger killing ability with obvious concentration-dependent effects (Supplementary Fig. S5C).

Together, our accumulated results suggest that IFNα implementation can effectively enhance anti–PD-1 treatment efficacy in HCC by inhibiting HIF1α signaling in tumor cells while enhancing HIF1α signaling and cytotoxic activity in CD8+ T cells.

FosB Is the Key Regulator of HIF1α Signaling Activation in Surviving HCC Cells after Anti–PD-1 Treatment

To elucidate the molecular mechanism by which HIF1α signaling is regulated in this process, we analyzed the differentially expressed genes in tumor samples from orthotopic HCC mice treated either with the anti–PD-1 antibody alone or with the combination of the anti–PD-1 antibody and IFNα. Venn diagram analysis identified seven overlapping transcripts with a negative fold change, which were ranked by P value, suggesting the expressions of these genes were significantly downregulated upon combination therapy with the anti–PD-1 antibody and IFNα compared with anti–PD-1 antibody monotherapy. Interestingly, FosB, which conventionally interacts with c-Jun to promote the DNA-binding activity of the AP-1 complex (42), showed the most significant difference in expression (Fig. 5A). Immunoblot assays confirmed that FosB expression was lower in Hepa1-6 or H22 derived from C57BL/6J and BALB/c mouse models treated with anti–PD-1 and IFNα than in those derived from mice that received anti–PD-1 single reagent (Fig. 5B; Supplementary Fig. S5D). As a classic oncogene, FosB is an important component of the transcription factor AP-1, which has been reported to be significantly implicated in accelerating HCC progression (43, 44). Considering that AP-1 is reported to interact with HIF1α and enhance HIF1α target transcription, we first validated the role of FosB in regulating glycolytic enzymes in HCC cells. We found that silencing FosB expression greatly decreased the expression of PKM2 and LDHA, two major rate-limiting enzymes involved in glycolysis, in Hepa1-6 and H22 cells transfected with FosB shRNA compared with those transfected with control shRNA (Fig. 5C; Supplementary Fig. S5E). Furthermore, coimmunoprecipitation (co-IP) assays demonstrated that FosB could physically interact with HIF1α (Fig. 5D and E) in Hepa1-6 cells. Next, the effect of FosB on HIF1α transcriptional targets was assessed by chromatin immunoprecipitation (ChIP)-qPCR assays. The results indicated that silencing FosB expression drastically decreased the occupation frequency of HIF1α on the promoters of its glycolysis-associated targets, such as Pfkl, Hk1, Pkm2, and Ldha; however, this inhibition effect was abolished when IFNα was applied (Fig. 5FI). More­over, cotransfection of FosB and HIF1α dramatically boosted glycolysis-related enzyme expression as well as the ECAR, whereas HIF1α transfection alone produced only moderate alterations (Supplementary Fig. S5F and S5G).

Figure 5.

IFNα restrains HIF1α signaling by inhibiting FosB transcription in an IRF1-dependent manner. A, Left, heat map demonstrating differentially expressed transcripts between tumor cells derived from model mice that received anti–PD-1 alone or IFNα combined with anti–PD-1 (left). Right, Venn diagram showing seven overlapping genes according to the fold change (FC; left) and P value (right) of the anti–PD-1 group versus the anti–PD-1 and IFNα combination group. B, The expression of FosB in tumor cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments was detected by immunoblot assays. Anti–PD-1: 10 mg/kg, IFNα: 1 × 104 IU. C, Impact of FosB silencing on PKM2 and LDHA expression in Hepa1-6 cells, as evaluated by immunoblot assays. D, Co-IP assays confirmed that HIF1α could physically interact with FosB in surviving Hepa1-6 cells derived from C57BL/6J mouse models that had received anti–PD-1 treatment (10 mg/kg). E, Co-IP assays confirmed that FosB could physically interact with HIF1α in surviving Hepa1-6 cells derived from C57BL/6J mouse models that had received anti–PD-1 treatment (10 mg/kg). F–I, Occupation frequency of HIF1α on the gene region of Pfkl (F), Hk1 (G), Pkm2 (H), and Ldha (I) detected by ChIP-qPCR assays. J and K, Occupation frequency of POLII (J) and H3K9ac (K) on the promoter of FosB after IFNα treatment (500 IU/mL). L, Expression of IRF1 in Hepa1-6 and H22 cells derived from the indicated C57BL/6J or BALB/c mouse models, as determined by immunoblot assays. M, The impact of IFNα on the expression of IRF1 in whole-cell lysates and nuclear fractions of Hepa1-6 cells was evaluated by nuclear–cytoplasmic fractionation followed by immunoblot assays. WCL, whole-cell lysis. IFNα: 0 IU/mL, 50 IU/mL, 100 IU/mL, or 500 IU/mL. N, The impact of IFNα on the expression of HK1, PFKL, LDHA, and PKM2 in Hepa1–6 cells after Irf1 KD. IFNα: 500 IU/mL. The results are shown as the mean ± SD for experiments performed in triplicate. Each experiment was repeated three times, and the statistical significance of differences between groups was determined by an unpaired Student t test. *, P < 0.05; **, P < 0.01; ns, not significant.

Figure 5.

IFNα restrains HIF1α signaling by inhibiting FosB transcription in an IRF1-dependent manner. A, Left, heat map demonstrating differentially expressed transcripts between tumor cells derived from model mice that received anti–PD-1 alone or IFNα combined with anti–PD-1 (left). Right, Venn diagram showing seven overlapping genes according to the fold change (FC; left) and P value (right) of the anti–PD-1 group versus the anti–PD-1 and IFNα combination group. B, The expression of FosB in tumor cells derived from indicated Hepa1-6 tumors isolated from C57BL/6J mouse models that received the indicated treatments was detected by immunoblot assays. Anti–PD-1: 10 mg/kg, IFNα: 1 × 104 IU. C, Impact of FosB silencing on PKM2 and LDHA expression in Hepa1-6 cells, as evaluated by immunoblot assays. D, Co-IP assays confirmed that HIF1α could physically interact with FosB in surviving Hepa1-6 cells derived from C57BL/6J mouse models that had received anti–PD-1 treatment (10 mg/kg). E, Co-IP assays confirmed that FosB could physically interact with HIF1α in surviving Hepa1-6 cells derived from C57BL/6J mouse models that had received anti–PD-1 treatment (10 mg/kg). F–I, Occupation frequency of HIF1α on the gene region of Pfkl (F), Hk1 (G), Pkm2 (H), and Ldha (I) detected by ChIP-qPCR assays. J and K, Occupation frequency of POLII (J) and H3K9ac (K) on the promoter of FosB after IFNα treatment (500 IU/mL). L, Expression of IRF1 in Hepa1-6 and H22 cells derived from the indicated C57BL/6J or BALB/c mouse models, as determined by immunoblot assays. M, The impact of IFNα on the expression of IRF1 in whole-cell lysates and nuclear fractions of Hepa1-6 cells was evaluated by nuclear–cytoplasmic fractionation followed by immunoblot assays. WCL, whole-cell lysis. IFNα: 0 IU/mL, 50 IU/mL, 100 IU/mL, or 500 IU/mL. N, The impact of IFNα on the expression of HK1, PFKL, LDHA, and PKM2 in Hepa1–6 cells after Irf1 KD. IFNα: 500 IU/mL. The results are shown as the mean ± SD for experiments performed in triplicate. Each experiment was repeated three times, and the statistical significance of differences between groups was determined by an unpaired Student t test. *, P < 0.05; **, P < 0.01; ns, not significant.

Close modal

Our above data imply that FosB might trigger downstream glycolytic targets of HIF1α transcription by enhancing HIF1α binding affinity for its target genes.

IFNa Suppresses FosB Expression in an IRF1-Dependent Manner

Considering that FosB mRNA expression was downregulated after IFNα treatment according to the RNA-seq results, IFNα might hinder FosB transcription. Interestingly, ChIP-PCR assays demonstrated reduced occupation of the FosB promoter by Pol-II after IFNα treatment (Fig. 5J). Moreover, decreased levels of H3K9ac in the promoter region of FosB were also observed (Fig. 5K), indicating that IFNα treatment could result in chromatin condensation in the FosB promoter region and inhibition of transcription initiation complex binding. A previous study revealed that IRF1, a key regulator of IFNα signaling, can counteract the pro-oncogenic function of FosB (45), suggesting a potential role for IRF1 in IFNα-regulated FosB expression. To verify this, we first evaluated IRF1 expression in HCC tissues from mice that received distinct treatments as described above by conducting immunoblot analysis. The results showed that IRF1 expression was greatly increased in HCC tissues after IFNα treatment, and the highest level was observed with anti–PD-1 treatment combined with IFNα administration (Fig. 5L). We also treated HCC cells with IFNα in vitro and found that IRF1 expression could be induced in a time-dependent manner, whereas FosB expression showed gradual downregulation (Supplementary Fig. S5H and S5I). Importantly, IFNα treatment induced increasing nuclear accumulation of IRF1, a critical intracellular translocation process for IRF1 function, in a dose-dependent manner (Fig. 5M; Supplementary Fig. S5J). In addition, IRF1 depletion abolished the inhibitory effects of IFNα on glycolytic enzyme expression (Fig. 5N). Together, these data imply that IFNα inhibits FosB expression by inducing IRF1-dependent transcriptional suppression.

A High-Glucose Microenvironment Promotes Cd27 Transcription via mTOR-Induced FOXM1 Expression

Previous studies have revealed the vital role of mTOR signaling in sensing glucose in the microenvironment and modulating glycolysis in T cells (46). As glycolysis has been found to be a critical biological process for T-cell activation and antitumor cytotoxic functions (47), we hypothesized that the high-glucose microenvironment caused by the IFNα-induced inhibition of glycolysis in tumor cells may trigger mTOR signaling activation in CD8+ T cells to render them more cytotoxic toward HCC cells. Indeed, CD8+ T cells cultured in high-glucose medium exhibited higher levels of p-mTOR, HIF1α, PKM2, and LDHA than those cultured in low-glucose medium, which could be reversed with rapamycin, a specific inhibitor of mTOR signaling (Fig. 6A), indicating the essential role of mTOR signaling in regulating CD27 expression. We further deployed HCC PDO models and confirmed that the proportion of the CD27+CD8+ population among total CD8+ T cells was decreased when TILs were pretreated with rapamycin compared with the control group (Supplementary Fig. S6A). Collectively, these data suggest that mTOR sig­naling might contribute to increased CD27+CD8+ T cells resulting from IFNα and anti–PD-1 antibody combination therapy.

Figure 6.

A high-glucose microenvironment triggers CD27 transcription by activating the mTOR–FOXM1 axis. A, Immunoblot analysis of p-mTOR, mTOR, HIF1α, PKM2, LDHA, and CD27 in CD8+ T cells derived from PBMCs isolated from C57BL/6J mice cultured in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) medium with or without 100 nmol/L rapamycin (Rapa). B, Immunoblot analysis of p-mTOR, mTOR, HIF1α, PKM2, LDHA, and CD27 expression in CD8+ T cells derived from Hepa1-6 cells isolated from C57BL/6J mouse models that were treated with IgG, 10 mg/kg anti–PD-1, 1 × 104 IU IFNα, or combination therapy. C, Venn diagram demonstrating 12 overlapping candidates according to the SPP database and single-cell sequencing data. The fold changes (FC) and P values of these 12 candidates are shown in a heat map. TF, transcription factor. D, The FOXM1 expression levels of CD8+ T cells derived from the indicated model mice were determined by immunoblot assays. anti–PD-1: 10 mg/kg; IFNα: 1 × 104 IU. E, CD8+ T cells derived from PBMCs isolated from C57BL/6 mice were cultured in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) medium with or without rapamycin (100 nmol/L). Afterward, FOXM1 expression levels were detected via immunoblot assays. F, CD8+ T cells derived from PBMCs isolated from C57BL/6 mice were cultured in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) medium with or without a HIF1α antagonist (10 μmol/L). Afterward, FOXM1 expression levels were detected via immunoblot assays. G, CD8+ T cells derived from indicated Hepa1-6 tumors received Foxm1 KD (left) or overexpression (right), and FOXM1, p-mTOR, mTOR, CD27, and PCNA expression levels were determined. Anti–PD-1: 10 mg/kg; IFNα: 1 × 104 IU. HIF1αi, HIF1α inhibitor. EV, empty vector; OE, overexpression. H, The TCGA database (left) revealed a positive correlation between FOXM1 and CD27 expression in HCC. Integrated single-cell sequencing (SC-seq) data (right) confirmed that FOXM1 and CD27 were positively correlated in CD27+CD8+ T cells in HCC. The results are shown as the means ± SD for experiments performed in triplicate. Each experiment was repeated three times, and the statistical significance of differences between groups was determined by an unpaired Student t test. Pearson correlation analysis was used to describe the correlation between variables.

Figure 6.

A high-glucose microenvironment triggers CD27 transcription by activating the mTOR–FOXM1 axis. A, Immunoblot analysis of p-mTOR, mTOR, HIF1α, PKM2, LDHA, and CD27 in CD8+ T cells derived from PBMCs isolated from C57BL/6J mice cultured in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) medium with or without 100 nmol/L rapamycin (Rapa). B, Immunoblot analysis of p-mTOR, mTOR, HIF1α, PKM2, LDHA, and CD27 expression in CD8+ T cells derived from Hepa1-6 cells isolated from C57BL/6J mouse models that were treated with IgG, 10 mg/kg anti–PD-1, 1 × 104 IU IFNα, or combination therapy. C, Venn diagram demonstrating 12 overlapping candidates according to the SPP database and single-cell sequencing data. The fold changes (FC) and P values of these 12 candidates are shown in a heat map. TF, transcription factor. D, The FOXM1 expression levels of CD8+ T cells derived from the indicated model mice were determined by immunoblot assays. anti–PD-1: 10 mg/kg; IFNα: 1 × 104 IU. E, CD8+ T cells derived from PBMCs isolated from C57BL/6 mice were cultured in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) medium with or without rapamycin (100 nmol/L). Afterward, FOXM1 expression levels were detected via immunoblot assays. F, CD8+ T cells derived from PBMCs isolated from C57BL/6 mice were cultured in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) medium with or without a HIF1α antagonist (10 μmol/L). Afterward, FOXM1 expression levels were detected via immunoblot assays. G, CD8+ T cells derived from indicated Hepa1-6 tumors received Foxm1 KD (left) or overexpression (right), and FOXM1, p-mTOR, mTOR, CD27, and PCNA expression levels were determined. Anti–PD-1: 10 mg/kg; IFNα: 1 × 104 IU. HIF1αi, HIF1α inhibitor. EV, empty vector; OE, overexpression. H, The TCGA database (left) revealed a positive correlation between FOXM1 and CD27 expression in HCC. Integrated single-cell sequencing (SC-seq) data (right) confirmed that FOXM1 and CD27 were positively correlated in CD27+CD8+ T cells in HCC. The results are shown as the means ± SD for experiments performed in triplicate. Each experiment was repeated three times, and the statistical significance of differences between groups was determined by an unpaired Student t test. Pearson correlation analysis was used to describe the correlation between variables.

Close modal

CD27 expression was greatly enhanced by IFNα and anti–PD-1 combination therapy, accompanied with dramatically increased p-mTOR, HIF1α, PKM2, and LDHA expression (Fig. 6B), further strengthening the necessity of mTOR sig­naling in CD27 expression in mouse models. We proceeded to examine the potential mechanism underlying how mTOR signaling regulated CD27 expression. To further identify upstream transcription factors that regulate Cd27 expression, we mapped our integrated single-cell data with potential transcription factors of CD27 in human leukocytes predicted by the Signaling Pathways Project (SPP) database (48). A total of 2,490 differential transcripts were identified between CD27hi and CD27lo CD8+ T cells. Furthermore, 57 of these transcripts were predicted to be transcription factors by the SPP database. Venn diagram analysis identified 12 overlapping genes in CD27hi cells, which were ranked by P value. We observed that FOXM1, a member of the Fox transcription factor family (49), exhibited the greatest statistical significance and a noticeable fold change (Fig. 6C). To further confirm the bioinformatic results, we overexpressed Foxm1 via retrovirus infection in CD8+ T cells, followed by flow cytometry assay. We found that Foxm1 overexpression could not only significantly increase the proportion of CD27+CD8+ T cells (Supplementary Fig. S6B) but also induce CD8+ T-cell activation, as evidenced by increased IFNγ+ and IL2+ CD8+ T proportions (Supplementary Fig. S6C and S6D). On the contrary, silencing Foxm1 expression in CD8+ T cells via siRNAs achieved the opposite results (Supplementary Fig. S6E–S6G). In summary, these results suggest that FOXM1 is critical for CD8+ T-cell activation and FOXM1 might regulate CD27 expression in CD8+ T cells.

Interestingly, IFNα treatment of model mice enhanced FOXM1 expression in CD8+ T cells, and CD8+ T cells derived from mice treated with both IFNα and anti–PD-1 exhibited the highest level of FOXM1 expression (Fig. 6D). Moreover, in vitro experiments indicated that high-glucose medium could promote FOXM1 expression in the vehicle-treated group. However, both rapamycin and the HIF1α inhibitor could abolish this effect (Fig. 6E and F). To verify the vital role of FOXM1 in regulating CD27, we used a lentivirus to knock down Foxm1 expression in infiltrating CD8+ T cells derived from mice treated with IFNα and the anti–PD-1 antibody (Fig. 6G). We found that CD27 and PCNA expression was greatly downregulated; however, the p-mTOR level showed no obvious alteration, indicating that FOXM1 is downstream of mTOR. In contrast, as determined by immunoblot analysis, overexpression of FOXM1 in infiltrating CD8+ T cells derived from mice treated with anti–PD-1 therapy alone resulted in dramatic elevations in CD27 and PCNA expression without an alteration in the p-mTOR level (Fig. 6G).

To validate the correlation between FOXM1 and CD27 in clinical HCC specimens, TCGA analysis of HCC was conducted, and the data showed a significant positive correlation between CD27 and FOXM1 expression (Fig. 6H). In addition, our integrated single-cell data also showed a significant positive correlation between CD27 and FOXM1 expression in CD27+FOXM1+CD8+ T cells in HCC (Fig. 6H). Together, these data confirmed that a high-glucose microenvironment could trigger mTOR signaling activation, resulting in FOXM1 overexpression and subsequent Cd27 transcription. The combination of IFNα and anti–PD-1 showed the most significant capacity to enhance FOXM1 expression, subsequently resulting in the highest CD27 expression in HCC.

CD27+CD8+ T Cells Are Associated with the Response to PD-1–Based Immunotherapy and Survival in HCC

We observed that high expression of CD8 or CD27 indicated a significantly better prognosis in patients with HCC than did corresponding low expression, as evidenced by longer progression-free survival (PFS) and OS in data from Zhongshan Hospital (Fig. 7A). Moreover, combined analysis of CD8 and CD27 was also performed with tumor microarray chips. We classified patients with HCC into three subgroups: (i) low CD8 and low CD27 (n = 31), (ii) high CD8 and high CD27 (n = 38), and (iii) low CD8 and high CD27 or high CD27 and low CD8 (n = 111). We found that OS was remarkably shorter in group 1 than in group 2 (P < 0.001; Fig. 7A). Likewise, PFS was significantly shorter in group 1 than in group 2 (P < 0.001; Fig. 7A). We also validated our findings above and demonstrated that combined analysis of CD8 and CD27 provided a relatively powerful tool to predict the prognosis of patients with HCC using TCGA data (Fig. 7B). By using multicolor immunofluorescence, we also found that CD27+CD8+ infiltrating TILs correlated with the responses of HCC patients treated with PD-1–based immunotherapy (Fig. 7C). Moreover, analysis of single-cell sequencing data for TILs in patients with melanoma (GSE120575) and bladder cancer (GSE145281) treated with an anti–PD-1 antibody revealed that CD27+CD8+ subsets were significantly enriched in responders compared with nonresponders (Fig. 7D).

Figure 7.

The combination of CD27 and CD8 achieved satisfactory power for predicting prognosis in multiple cancer types treated with anti–PD-1–based immunotherapy. A, The prognostic prediction performances of CD27, CD8, and their combination for OS and PFS were assessed by Kaplan–Meier curve analysis for data from Zhongshan Hospital. B, The prognostic prediction performances of CD27, CD8, and their combination for OS and PFS were assessed by Kaplan–Meier curve analysis using a TCGA data set. C, Multiplex immunofluorescence staining images illustrate responders with high-expression levels of both CD8 and CD27 in formalin-fixed, paraffin-embedded samples taken before anti–PD-1–based immunotherapy (n = 19). Quantitative analysis of the densities of CD8 and CD27 in intratumoral regions between responders and nonresponders. Scale bar: 50 µm. D, Single-cell sequencing data revealed that responders had high expression of both CD8 and CD27 in a melanoma cohort and bladder cancer cohort. E, Illustration of the proposed working model. IFNα inhibits glucose metabolism in HCC tumor cells while increasing T-cell glycolysis, thus improving the cytotoxic capacities of T cells by promoting the PD-1 blockade–induced immune response. ab, antibody. The results are shown as the means ± SD, and the statistical significance of differences between groups was determined by an unpaired Student t test. ***, P < 0.001.

Figure 7.

The combination of CD27 and CD8 achieved satisfactory power for predicting prognosis in multiple cancer types treated with anti–PD-1–based immunotherapy. A, The prognostic prediction performances of CD27, CD8, and their combination for OS and PFS were assessed by Kaplan–Meier curve analysis for data from Zhongshan Hospital. B, The prognostic prediction performances of CD27, CD8, and their combination for OS and PFS were assessed by Kaplan–Meier curve analysis using a TCGA data set. C, Multiplex immunofluorescence staining images illustrate responders with high-expression levels of both CD8 and CD27 in formalin-fixed, paraffin-embedded samples taken before anti–PD-1–based immunotherapy (n = 19). Quantitative analysis of the densities of CD8 and CD27 in intratumoral regions between responders and nonresponders. Scale bar: 50 µm. D, Single-cell sequencing data revealed that responders had high expression of both CD8 and CD27 in a melanoma cohort and bladder cancer cohort. E, Illustration of the proposed working model. IFNα inhibits glucose metabolism in HCC tumor cells while increasing T-cell glycolysis, thus improving the cytotoxic capacities of T cells by promoting the PD-1 blockade–induced immune response. ab, antibody. The results are shown as the means ± SD, and the statistical significance of differences between groups was determined by an unpaired Student t test. ***, P < 0.001.

Close modal

Together, these data confirmed that CD27+CD8+ T cells were associated with the survival of patients with HCC. Moreover, CD27+CD8+ T cells also correlated with the survival of HCC patients treated with PD1 blockade–based immunotherapy.

Anti–PD-1 antibody–based immunotherapy has become a promising cancer treatment strategy with durable clinical benefits in a wide range of cancer types (50). However, not all patients respond to anti–PD-1 monotherapy, and patients with HCC are often unresponsive (2, 3). Here, we report that combination therapy comprising IFNα administration and PD-1 blockade resulted in significant tumor repression in murine HCC models and patients with HCC. We found that IFNα induced IRF1 expression in HCC cells, thereby inactivating FosB transcription, which further attenuated the transcriptional activity of HIF1α and subsequently led to downregulation of glycolysis-related gene expression in HCC cells. Sequentially, combination IFNα and anti–PD-1 treatment remodeled the high-glucose microenvironment, which is essential for the activation of T cells and promoted T-cell costimulatory molecule (CD27) transcription via mTOR-induced FOXM1 expression in T cells (Fig. 7E).

It has been recently reported that some conventional reagents used in clinical practice, including metformin and cyclophosphamide, are able to synergize with PD-1 blockade and improve the ICB response (51, 52). As a major advantage, repurposing conventional drugs for a new use accelerates clinical application without new drug development. IFNα has been used to treat HCC for over 20 years, but its clinical benefit is limited, largely due to severe systemic side effects resulting from the regular dose used for treatment (53). These observations indicated that optimizing the dose and formulation of IFNα is essential to improve the therapeutic effects of IFNα. In our experiments, we found that 1 × 104 IU of IFNα was sufficient to induce tumor regression in immunocompetent mice but not in immunodeficient mice without a significant change in body weight. These observations highlight for the first time that a suitable dose of IFNα is sufficient to inhibit tumor progression mainly through immune modulation rather than directly inducing tumor cell apoptosis (21). These findings are striking and provide a rationale for evaluating the effect of anti–PD-1 therapy on tumors exposed to IFNα. Our clinical data further provided early encouraging evidence for IFNα-based combination therapy, highlighting the potential of IFNα as a conventional drug in new applications for HCC treatment.

A “switch” from oxidative phosphorylation to glycolysis is a hallmark of T-cell activation, and this process is critical for meeting the metabolic demands of T-cell proliferation and activation (54). PD-1 signaling reprograms and inhibits glycolysis in T cells, and restricting glycolysis in tumor cells preserves the effector functions of T cells and can augment the response rate for ICB (55, 56). Interestingly, it has also been demonstrated that increased tumor glycolysis contributes to resistance to immunotherapy (57). Indeed, there is competition for glucose in the TME that can directly regulate T-cell function and ICB efficiency (58). In the current work, we were surprised to find that PD-1 blockade was able to upregulate glycolysis-related gene expression in a HIF1α-dependent manner. Strikingly, we found a previously unrevealed function of IFNα. Namely, IFNα was able to suppress tumor glycolysis by restraining expression of HIF1α-targeted master glycolytic transcripts including PKM2 and LDHA. Intriguingly, the suppressive effects of IFNα were preferentially limited in the tumor cell, evidenced by the fact that IFNα lost its suppressive effect when Ifnar1, the specific receptor of IFNα, was silenced in HCC cells. According to the scRNA-seq data of the Human Protein Atlas project, IFNAR1 showed relatively low expression on T cells (ref. 59; http://www.proteinatlas.org), and more importantly, we found that IFNAR1 expression was also higher in HCC cells when compared with infiltrating T cells according to single-cell sequencing data. Therefore, it was rational that HCC cells responded toward IFNα priorly, resulting in the selective effects of IFNα on HCC cells over T cells in HCC. Together, we uncovered a novel mechanism by which IFNα could indirectly reprogram glucose metabolism toward glycolysis in tumor-infiltrating CD8+ T cells by limiting tumor-imposed glucose competition, which increased glucose uptake by tumor-infiltrating CD8+ T cells and restored TIL function.

CD27 has been considered an activation marker of CD8+ T cells. We found that tumor-infiltrating CD27hiCD8+ T cells express a higher level of activation-related genes, including CD28, IL2RA, IFNG, and GZMB, than those in CD27loCD8+ T cells using single-cell sequencing data of tumor-infiltrating CD8+ T cells from patients with HCC. Moreover, we knocked down Cd27 in activated CD8+ T cells and found reduced secretion of effector cytokines including IFNγ, TNFα, and GZMB, supporting that the expression of CD27 was essential for CD8+ T-cell activation. We identified CD27+CD8+ T cells as the most enriched subtype after combination treatment with IFNα and an anti–PD-1 antibody. The proportion of CD27+CD8+ T cells might serve as a predictive biomarker for PD-1 therapy. CD27, which binds to its ligand, CD70, plays a critical role in T-cell activation, and agonistic anti-CD27 antibodies have shown great anticancer efficacy because CD27 signaling induction can enhance the proliferation and differentiation of effector and memory T cells (60). However, the transcriptional control of CD27 is poorly understood. Here, we report that IFNα-mediated increases in glucose uptake and glycolysis in T cells increased the levels of phosphorylated mTOR, which subsequently activated expression of a key transcription factor, FOXM1, to promote Cd27 transcription. Notably, it came to our attention that the expression of PDCD1 (which encodes the PD-1 protein) was also preferentially expressed in CD27hiCD8+ T cells according to both public single-cell sequencing data and human samples. These data suggested that although CD27+CD8+ T cells induced by IFNα treatment acted as major effector T cells to attack tumor cells, their effector function was, at the same time, limited by feedback-high expression of PD-1. Therefore, anti–PD-1 treatment is essential to liberate the immunologic effect of CD27+CD8+ T cells. Collectively, our results provide secondary evidence explaining the synergistic effect of IFNα and anti–PD-1 antibody combination treatment. The clinical data on tumor regression in 15 HCC patients treated with IFNα combined with anti–PD-1 indicated enrichment of CD27+CD8+ T cells, which will help establish the translational potential and clinical applicability of this work. These findings could also deepen our understanding of the mechanism influencing the outcome of PD-1 blockade in patients with HCC, the potential factors related to treatment efficacy, or the factors related to selecting the right patient population for treatment. Addressing the translational and clinical potential of this work would advance the field and has potential immediate relevance to patient care. Compared with previously published data (27), our research explored the broader applicability and/or potential limitations of this therapy.

How metabolic changes in the TIME affect the development and maintenance of a favorable immune response to ICB remains unclear. We identified the role of IFNα in enhancing anti–PD-1 efficacy by suppressing HIF1α-induced glycolysis in HCC and found that IFNα suppresses FosB in an IRF1-dependent manner. We first found that IFNα could restrain the glycolytic capacity of tumor cells to enhance microenvironmental glucose levels, resulting in CD27+CD8+ T-cell infiltration. This result suggests how the metabolic function of tumor cells contributes to disease outcomes. Mechanistically, we also found that a high-glucose microenvironment could trigger CD27 expression in CD8+ T cells by activating the mTOR–FOXM1 axis. The mechanism will help to identify potential biomarkers to stratify patients who may respond to IFNα and anti–PD-1 combination treatment, and understanding the molecular mechanism will be of interest. The combination of IFNα and an anti–PD-1 antibody improves immunotherapeutic efficacy in patients through multiple mechanisms. We demonstrate a novel mechanism through which IFNα can reshape the HCC TME to enhance glucose metabolism.

Although our study indicated that IFNα combined with anti–PD-1 could be of therapeutic value in HCC, additional clinical studies in larger patient cohorts (with or without hepatitis etiology), as well as additional transgenic mouse models of HCC, are needed to further validate this hypothesis. In addition, unintended side effects should be carefully and systematically evaluated. Importantly, our work provides a potential application of IFNα and anti–PD-1 combination therapy for the treatment of other additional cancer types, as tumor glycolysis is a hallmark of cancer and a critical barrier to successful immunotherapy in several types of cancer models, including melanoma, breast cancer, and lung cancer models. Our work also highlights the clinical value of tumor-infiltrating CD27+CD8+ T cells as a biomarker for stratifying patients for anti–PD-1 treatment in tumors other than HCC. However, these applications still need to be further investigated.

In conclusion, we found that the combination of IFNα and anti–PD-1–based immunotherapy exhibited promising anticancer effects in 15 patients with HCC. Our work reveals a novel mechanism of the synergistic efficacy of the combination of IFNα and an anti–PD-1 antibody in HCC. The combination therapy greatly induced CD27+CD8+ T-cell infiltration, remodeled the TIME, and subsequently regressed HCC tumors. Mechanistically, IFNα corrected tumor-imposed glucose competition in the HCC microenvironment and restored the effector function of CD8+ T cells by increasing CD27 transcription. The clinical applicability of these data could potentially advance HCC therapy. Our data also imply the potential for broader applicability of IFNα with other ICBs in addition to PD-1 blocking and for evaluating IFNα and anti–PD-1 combination therapy in other cancer types. In summary, our data provide the initial clinical evidence and mechanistic insight on a novel combination strategy to improve the clinical outcome of anti–PD-1 antibody–based immunotherapy in HCC.

Patients and Specimens

A tissue microarray composed of HCC samples was obtained from Zhongshan Hospital, Fudan University, Shanghai, China (n = 180). Biopsies were also obtained from patients with unresectable HCC (n = 19) treated with PD-1–based immunotherapy to evaluate the correlation between CD27+CD8+ TILs and drug response by multicolor IHC analysis. Radiographic assessment of response by independent radiology review was performed. Tumor response for patients was assessed by modified RECIST (mRECIST; ref. 61). IFNα is approved by the FDA for the treatment of chronic hepatitis B. Evidence supports that IFNα directly and indirectly modulates immune responses and may add to PD-1 blockade in enhancing immune and clinical responses to solid tumors (27). In addition, several studies indeed have proven that IFNα may be an effective treatment and can prolong survival in patients with HCC (62, 63), which prompted us to observe the efficacy of PD-1 blockade (200 mg, every 2/3 week, intravenous) combined with pegylated-interferon alfa-2b (3 μg/kg, every week, subcutaneous). Peripheral blood samples were collected from patients (n = 8) before/after combination treatment to measure lymphocyte levels using flow cytometry based on the prospective clinical trial (NCT04639284). In addition, a total of 15 consecutive patients treated with IFNα combined with anti–PD-1–based immunotherapy were included in the efficacy observation. Clinicopathologic information was summarized (Supplementary Table S1). These patients were enrolled under Institutional Review Board (IRB)–approved protocols (Approval No. B2020-177R).

These studies were conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization Guidelines for Good Clinical Practice. All patients signed written informed consent prior to having any study procedures performed.

Cell Lines and Cell Culture

HEK293T (RRID: CVCL_0063), Hepa1-6 (RRID: CVCL_0327), and H22 (RRID: CVCL_H613) cell lines were kindly provided by Dr. Xun Huang at Lingang Laboratory, Shanghai, China. HEK293T and Hepa1-6 cells were cultured in DMEM containing 10% FBS (Gibco; #10099141C) and 100 U/L penicillin/streptomycin (Gibco; #15140122) at 37°C and 5% CO2 in an incubator. H22 cells were cultured in RPMI 1640 (Gibco; #61870036) supplemented with 10% FBS (Gibco; #10099141C) and 100 U/L penicillin/streptomycin (Gibco; #15140122) at 37°C and 5% CO2 in an incubator. Both of the cell lines were routinely authenticated and were free of Mycoplasma contamination. The EL4 (RRID:CVCL_0255) cell line was purchased from the ATCC and cultured in DMEM containing 10% FBS (Gibco; #10099141C) and 100 U/L penicillin/streptomycin (Gibco; #15140122) at 37°C and 5% CO2. Cell lines were authenticated using STR profiling and used at low passage numbers. Infiltrating CD8+ T cells were cultured in RPMI 1640 medium (Gibco; #61870036) supplemented with 10% FBS (Gibco; #10099141C), 1% penicillin/streptomycin (Gibco; #15140122), 5 μg/mL SIINFEKL peptide (Sangon Biotech; #T510212), and 20 ng/mL mIL2 (R&D Systems; #402-ML) at 37°C and 5% CO2. Additionally, OVA-specific CD8+ CTLs were generated by incubating OT-I mouse splenocytes in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) RPMI 1640 medium for 7 days. On the other hand, HCC cells derived from orthotopic models were cultured in DMEM (Gibco; #11965092) supplemented with 10% FBS (Gibco; #10099141C) and 1% penicillin/streptomycin (Gibco; #15140122) in a 37°C and 5% CO2 incubator.

Animal Studies

All animal care and experimental protocols performed were approved by the Institutional Animal Care and Use Committee of Zhongshan Hospital, Fudan University. To establish a Myc-induced spontaneous HCC model, 50 μg pT3-EF1a-c-myc (RRID: Addgene_92046) and 10 μg Sleeping Beauty SB100x transposase-encoding plasmid (RRID: Addgene_34879) per mouse dissolved in 2 mL of 0.9% NaCl solution were injected into C57BL/6J mice through tail-vein injection within 3 to 5 seconds. For orthotopic HCC models, 6-week-old male C57BL/6J mice (Charles River Company) were injected subcutaneously in the right flank with 5 × 106 Hepa1-6 cells in 100 μL PBS and 6-week-old male BALB/c mice (Charles River Company) were injected subcutaneously in the right flank with 5 × 106 H22 tumor cells in 100 μL PBS. Two weeks after cell implantation, the tumors derived from Hepa1-6 or H22 tumor cells were dissected and cut into small cubes (1 mm3) under aseptic conditions. Single cubes of equal volume were implanted into the right lobe of the liver parenchyma of 6-week-old C57BL/6J mice or BALB/c mice. Ifnar1-deficient HCC cells were used to establish an orthotopic HCC model using C57BL/6J mice. NCG mice (GemPharmatech) were also used to establish an orthotopic HCC model using Hepa1-6 cells following the aforementioned schedule. Three days after implantation, mice bearing Hepa1-6 or H22 orthotopic tumors were randomized into the indicated groups. For combination therapy, C57BL/6J or BALB/c tumor–bearing mice were treated intraperitoneally with 1 × 104 IU mIFNα (Miltenyi Biotec; #130-093-130) and/or 10 mg/kg anti–PD-1 monoclonal antibody (Bio X Cell, BE0146, RRID: AB_10949053) or isotype control monoclonal antibody (Bio X Cell, BE0089, RRID: AB_1107769) following the schedule shown in Fig. 2A. Briefly, the anti–PD-1 antibody was administered 3 times per week for 4 weeks, and mIFNα was given daily for 4 weeks. For the in vivo T-cell depletion studies, the indicated mice were administered anti-CD8α (Bio X Cell, BE0061, RRID: AB_1125541), anti-CD4 (Bio X Cell, BE0003-1, RRID: AB_1107636), or both the anti-CD8α and anti-CD4 antibodies [250 μg in 100 μL PBS (intraperitoneally)] following the schedule shown in Fig. 3A. For mIFNα monotherapy studies, NCG tumor-bearing mice were injected intraperitoneally with 1 × 104 IU mIFNα or 0.9% NaCl as a control. Tumor growth was monitored daily by a Preclinical Vevo 3100 micro-ultrasound imaging platform (VisualSonics) and measured every 3 to 5 days. B-mode, or brightness mode, imaging was used to acquire 3D images of an area of interest and for identification of liver tumors. Images were quantified for tumor volume using VevoLAB analysis software (VisualSonics). All mice used in this study were housed under specific pathogen–free conditions in the animal center of Zhongshan Hospital. Mouse body weight was measured daily. Mice were routinely monitored to observe the effects of tumor growth and treatment on normal activity, food and water consumption, weight gain/loss, and other features. All experiments were conducted to reduce the number of mice as much as possible and alleviate their suffering. All studies that employed anesthesia or euthanasia methods were consistent with the commonly accepted norms of veterinary best practices (e.g., CO2). At the end of the indicated therapy, mice were sacrificed, and livers bearing tumors were dissected. Tumor volume was calculated using the formula length (mm) × width2 (mm) × 0.5.

HCC PDO and TIL Coculture

For assaying the effects of rapamycin on IFNα plus anti–PD-1 treatment in HCC, an HCC PDO and autologous intratumoral immune cell coculture model was established according to published methods (32, 33) with optimization. Briefly, primary HCC patient resection tumors were mechanically minced and dissociated into 1 mm3 fragment; diluted in Advanced DMEM/F-12 (Thermo Fisher Scientific; #12634010) with 0.1% collagenase, Type 4 (Worthington; #LS004188), 0.05% hyaluronidase (Sigma; #H3506), and 0.01% deoxyribonuclease (Sigma; #DN25); and shaken on a horizontal platform for 30 minutes at 37°C. The homogenate was filtered through a 100-μm filter and then a 40-μm filter, pelleted, and washed in 1× red blood cell lysis buffer (BD; #555899). The spheroids between 40 and 100 μm were then resuspended in the classic human liver organoid isolation medium for 3 days (64). Cells less than 40 μm were isolated for TILs by a Ficoll density method (65). TILs were cultured in ImmunoCult-XF T-cell Expansion Medium (STEMCELL; #10981) with ImmunoCult Human CD3/CD28 T-cell Activator (STEMCELL; #10971) and 30 ng/mL IL2 (R&D Systems; #202-IL-050) with or without 200 ng/mL rapamycin for 3 days. On day 3, HCC organoids and autologous activated TILs were directly cocultured at a 5:1 ratio in Matrigel (Corning; #356231) in a 24-well plate and overlayed with human liver organoid isolation medium with 1 × 104 IU/mL IFNα (Miltenyi Biotec; #130-093-876) plus 100 μg/mL anti–PD-1 (Bio X Cell; cat. #BE0188, RRID: AB_10950318) after incubation in a 37°C and 5% CO2 culture incubator for 10 minutes. After 72 hours of coculture, the specific killing efficiency of T cells and CD27+CD8+ population were evaluated by flow cytometry to explore whether rapamycin could blockade the effects of IFNα treatment. The following antibodies were used for flow cytometry staining: CD45 (PerCP cy5.5, BioLegend; cat. #982318, RRID: AB_2888786), CD8 (APC-A750, BioLegend; cat. #344722, RRID: AB_2075388), CD27 (PE, Thermo Fisher Scientific; cat. #MA5-17905, RRID: AB_2539289), PD-1 (BV421, BioLegend; cat. #329920, RRID: AB_10960742).

CyTOF Sample Preparation

Freshly harvested tumors were treated with the Mouse Tumor Dissociation Kit (Miltenyi Biotec; #130-096-730) according to the manufacturer's instructions. Briefly, dissociated tumor samples were filtered through a 70-μm strainer and centrifuged with 36% Percoll reagent to remove cell debris. ACK lysis buffer was then utilized to lyse erythrocytes. The cells were resuspended in 100 μL Living Cell Staining Mix (250 nmol/L cisplatin in PBS) and incubated for 5 minutes on ice. The samples were blocked and stained for 30 minutes with a surface antibody panel developed in-house, followed by fixation overnight. For intracellular marker staining, permeabilization buffer (eBioscience) was utilized following the manufacturer's instructions. Cells were then washed, resuspended, and stored at 4°C before acquisition on a Helios instrument (Fluidigm). Antibody clones and suppliers are listed in Supplementary Table S2.

CyTOF Data Analysis

Mass cytometry data were randomized and normalized before processing as previously described (66). A doublet filtering scheme was used on mass-tagged barcodes for differing samples (67). Generally, the gating strategy was as follows: cells, live cells, singlets, and valid immune cells (CD45+). Separate t-distributed stochastic neighbor embedding (t-SNE) plots and heat maps were developed for all CD45+ cells to specifically characterize the T-cell clusters. X-shift was applied to the T-cell compartment defined in the first X-shift analysis. MHC-II, B220, CD21, IgM, IgD, CD11c, CD19, F4/80, CD115, iNOS, Arginase I, CD49b, CD49a, CD23, and CD11b were removed from the analysis to exclude markers that were not expressed on T cells and reduce the likelihood of adding noise during cluster generation.

Immunoblot and Coimmunoprecipitation

Cultured cells or mouse tumor tissue proteins were dispersed in RIPA Lysis and Extraction Buffer (Thermo Fisher Scientific; #89901) supplemented with 1/100 Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific; #78446) for 30 minutes. All the processes were performed on ice. For mouse tumor tissue samples, the tissues were first homogenized for 15 minutes. Cell or tissue lysates were centrifuged at 12,000 rpm for 15 minutes at 4°C. Supernatants were collected and evaluated by a BCA protein concentration assay (Beyotime; #P0010). Then, 5× SDS loading buffer was added to the protein samples, and the samples were boiled at 95°C for 5 minutes. A total of 50 μg protein samples was loaded onto a 10% SDS-PAGE gel, separated by electrophoresis, and then transferred onto a polyvinylidene fluoride membrane (Millipore). After transfer, membranes were blocked with TBST containing 5% skim milk for 2 hours at room temperature. The membranes were then incubated with the indicated primary antibody specific for PKM2 (Cell Signaling Technology; cat. #4053, RRID: AB_1904096, 1:1000), LDHA (Cell Signaling Technology; cat. #3582, RRID:AB_2066887, 1:1,000), HK1 (Cell Signaling Technology; cat. #2024, RRID: AB_2116996, 1:1000), HK2 (Cell Signaling Technology; cat #2867, RRID: AB_2232946, 1:1000), FosB (Cell Signaling Technology; cat. #2251, RRID: AB_2106903, 1:1,000), β-actin (Cell Signaling Technology; cat. #4970, RRID: AB_2223172, 1:1,000), histone H3 (Cell Signaling Technology; cat. #4499, RRID: AB_10544537), IRF1 (Cell Signaling Technology; cat. #8478, RRID: AB_10949108, 1:1,000), HIF1α (Cell Signaling Technology; cat #36169, RRID: AB_2799095, 1:1,000), PCNA (Cell Signaling Technology; cat. #13110, RRID: AB_2636979, 1:1,000), CD27 (R&D Systems; cat. #MAB574, RRID: AB_2073420, 1:500), mTOR (Cell Signaling Technology; cat. #2972, RRID: AB_330978, 1:1,000), p-mTOR (Cell Signaling Technology; cat. #5536, RRID: AB_10691552, 1:1,000), FOXM1 (Proteintech; cat. #13147-1-AP, RRID: AB_2106213, 1:500), IFNAR1 (Abcam; cat. #ab124764, RRID: AB_10972855, 1:800), cleaved caspase-3 (Cell Signaling Technology; cat. #9661, RRID: AB_2341188, 1:1,000), or PFKL (Abcam; cat. #ab181064, 1:1,000) at 4°C overnight. On the second day, the membranes were washed with TBST 3 times (each for 5 minutes) and then incubated with horseradish peroxidase–conjugated secondary antibodies for 2 hours at room temperature. After that, membranes were washed with TBST 3 times (each for 10 minutes), and reactive bands were visualized using ECL substrate (Tanon).

For cytoplasmic and nuclear extract preparation, nuclear and cytoplasmic extraction reagents (Thermo Fisher Scientific; #78833) were used according to the manufacturer's instructions. β-Actin and histone H3 were used as internal references for whole-cell extracts and nuclear extracts, respectively.

For co-IP, indicated cells were lysed with NP-40 buffer (Beyotime; #P0013F) containing 1/100 protease and phosphatase inhibitor cocktail (Thermo Fisher Scientific; #78446). Cell lysates were incubated with co-IP–grade antibodies with rotation at 4°C overnight. On the second day, antibodies were pulled down with Protein A Magnetic Beads (Cell Signaling Technology; #73778) at room temperature with rotation for 20 minutes. After washing with lysis buffer five times, the supernatant was boiled at 95°C for 10 minutes, followed by immunoblot analysis.

RT-PCR

Total RNA was extracted from cultured cells using an RNeasy Mini Kit (Qiagen; #74106) and used as a template to generate cDNA with the QuantiTect Reverse Transcription Kit (Qiagen; #205313). Quantitative RT-PCR was performed using FastStart Universal SYBR Green Master Mix (Roche) on a LightCycler 480 384-well plate (Roche) and analyzed using LightCycler 480 software v1.5 (Roche).

Multiplex Immunofluorescence Staining

For multiplex staining, we followed the Opal protocol staining method with the following reagents: an anti-CD27 antibody (Abcam; #ab214041, 1:500) with subsequent visualization using fluorescein-cyanine 3 (Cy5; 1:50), an anti-CD8 antibody (Cell Signaling Technology; cat. #98941, RRID: AB_2756376, 1:500) with visualization using Cy3 (1:50), and DAPI (1:2,000) for subsequent visualization of nuclei. All sections were coverslipped using VECTASHIELD Hardset Antifade Mounting Medium (Vector Laboratories; #H-1400). For multispectral analysis of the data, a detailed methodology described previously (68) was followed. Each of the individually stained sections was used to establish the spectral library of fluorophores required for the multispectral analysis. The slides were scanned using a Vectra slide scanner (PerkinElmer) under fluorescence conditions. For each marker, the mean fluorescence intensity per case was then determined as the basis from which positive cells could be established. Finally, the colocalization algorithm was used to determine the percentage of CD27 and CD8 colocalization.

Immunochemistry Analysis

A paraffin-embedded HCC tissue microarray was constructed, and IHC analysis was performed as described previously. Briefly, patient HCC specimens were fixed with 4% paraformaldehyde and washed with 75% ethanol. The samples were then deparaffinized and hydrated, and endogenous peroxidase activity was blocked. Antigens were retrieved in EDTA buffer by microwaving for 15 minutes, followed by incubation with 5% BSA for 30 minutes at room temperature. The antibodies used for IHC staining were anti-CD27 (Abcam; cat. #ab131254, RRID: AB_11155136, 1:100), anti-CD8α (Cell Signaling Technology; cat. #85336, RRID: AB_2800052, 1:100), anti-Foxp3 (Abcam; cat. #ab20034, RRID: AB_445284, 1:500), and anti-CD206 (Abcam; cat. #ab64693, RRID: AB_1523910, 1:100). For spontaneous HCC model staining, the antibodies were anti–HNF-4α (Abcam; cat. #ab181604, RRID: AB_2890918, 1:800) and anti–CK-19 (Abcam; cat. #ab52625, RRID: AB_2281020, 1:400). For immunostaining evaluation of CD27 and CD8, the tissue microarrays were assessed by two independent pathologists blinded to the clinical data using a light microscope (200×, Olympus). CD27+ or CD8+ cells in each 1-mm-diameter cylinder were counted and are shown as the mean value of triplicates (cells/spot).

RNA Transfection

Gene expression KD was achieved by using a shRNA-mediated silencing approach as previously described. To generate stable silenced cell lines, shRNA lentiviral vectors targeting Hif1α, FosB, Irf1, or Ifnar1 were transfected into murine HCC cell lines with corresponding control vectors. The sequences of the shRNAs were as follows: Hif1α, 5′-CCGGTATGCACTTTGTCGCTATTAACTCGAGTTAATAGCGACAAAGTGCATATTTTTG-3′; FosB-1, 5′-CCGGCAACGGTCACCGCAATCACAACTCGAGTTGTGATTGCGGTGACCGTT

GTTTTTTG-3′; FosB-2, 5′-CCGGGCCAGGGTCAACATCCGCTAACTCGAGTTAGCGGATGT

TGACCCTGGCTTTTTG-3′; Irf1, 5′-CCGGGTTGCATAGAGAGGGTCTTATCTCGAGATAAG

ACCCTCTCTATGCAACTTTTTG-3′; and Ifnar1, 5′-GATCCGAATGAGGTTGATCCGTTTATCTCGAGATAAACGGATCAACCTCATTCTTTTTT-3′. shRNA lentiviral vectors targeting Foxm1 were transfected into CD8+ T cells to silence FOXM1 expression with the corresponding control vectors. The shRNA sequence was as follows: shFoxm1-1: 5′-CCGGACTTCCTATTCAGTCCATTAACTCGAGTTAATG GACTGAATAGGAAGTTTTTTG-3′; shFoxm1-2: 5′-CCGGGCTATCCAGTGAAGGAATAGCCTCGAGGCTATTCCTTCACTGGATAGCTTTTTG-3′. Meanwhile, for siRNA KD, 4 × 106 cells/mL CD8+ T cells were cultured in a 24-well plate for 12 hours before transfection. Diluting 2 μL of Lipofectamine 2000 (Thermo Fisher Scientific; #11668019) with 25 μL of Opti-MEM, and diluting 120 pmol of siRNA with 25 μL of Opti-MEM. Next, the diluted Lipofectamine 2000 and diluted siRNA were mixed and incubated for 20 minutes. Nontargeting siRNA was used as negative control. The mRNA expression level of Foxm1 was measured by qPCR after 48 hours. The siRNA sequence was as follows: sense, 5′-GCAAAUUUCCAGCCGGAAUTT-3′; antisense: 5′-AUUCCGGCUGGAAAUUUGCTT-3′. For Foxm1 overexpression, murine Foxm1 cDNA was amplified and cloned into the lentiviral vector pHBLV (Hanbio Biotechnology), a lentiviral vector encoding the ZsGreen cDNA downstream of the EF1 promoter. To prepare lentiviral particles, the lentiviral vector, psPAX2, and pMD2G helper plasmid were cotransfected into HEK293T cells according to the manufacturer's protocol (Hanbio Biotechnology). CD8+ T cells were infected by Foxm1 overexpression lentiviruses according to the manufacturer's protocol using Polybrene (Hanbio Biotechnology). A lentiviral vector that expressed only ZsGreen (pHBLV ZsGreen) was produced as a negative control. Positive cell clones were selected by adding puromycin to the culture medium for 7 days, and antibiotic-resistant cells were identified as successfully transfected clones, which were used for further investigations.

ChIP

In total, 5 × 105 indicated cells were cross-linked with 1% formaldehyde for 15 minutes at room temperature, quenched with 125 mmol/L glycine, and lysed with SDS buffer, and the nuclear extracts were sonicated. The sonicated lysates were precleared with normal IgG and then immunoprecipitated with antibodies against POL2 (Santa Cruz Biotechnology; cat. #sc-47701, RRID: AB_677353, 1:20), H3K9Ac (Cell Signaling Technology; cat. #9649, RRID: AB_823528, 1:25), or HIF1α (Cell Signaling Technology; cat. #36169, RRID: AB_2799095, 1:50) or control IgG (rabbit, Cell Signaling Technology; cat. #3900, RRID: AB_1550038, 1:100; mouse, Cell Signaling Technology; cat. #53484, RRID: AB_2799435, 1:100) overnight at 4°C. On the second day, the lysates were incubated with ChIP-grade protein G agarose beads for 2 hours at room temperature. After washing with low-salt buffer (3 times, each for 5 minutes at 4°C with rotation) and high-salt buffer (5 minutes at 4°C with rotation), the chromatin was eluted from the protein/DNA complexes for 30 minutes at 65°C, and the DNA was purified with DNA purification columns and collection tubes. The purified DNA was then analyzed by qRT-PCR. The ChIP primers used in the present study were designed as follows: forward 5′-CTGCCCATAGTGTGACCCTG-3′, reverse 5′-TGGCTAATTGCGTCACAGGA-3′ for verifying POLII occupation of the mouse FosB promoter region; forward 5′-ATGTGCTCCCTCCCTGATCT-3′, reverse 5′-GTGTGGGTTCCTACCTGCTC-3′ for verifying H3K9ac occupation of the mouse FosB promoter region; forward 5′-GGCTGTGCACCCTCAACTAT-3′, reverse 5′-TGCTTCCTGTCCCTTGGATG-3′ for verifying HIF1α occupation of the mouse Pfkl gene region; forward 5′- CAAACTGGAGAACGCGAACG-3′, reverse 5′-CTCTCTTTCCACGCCCACTT-3′ for verifying HIF1α occupation of the mouse Hk1 gene region; forward 5′-CTATCCTCGCCACCCTGTTC-3′, reverse 5′-AGGGCACCGAATTATGAGGG-3′ for verifying HIF1α occupation of the mouse Pkm2 gene region; and forward 5′-CACTCTTGCAGGGACATCGT-3′, reverse 5′-GGAACCCACGTGTAGGCTG-3′ for verifying HIF1α occupation of the mouse Ldha gene region.

Flow Cytometry and Cytokine/Surface Staining

For the isolation of human tumor-infiltrating T cells, fresh human HCC tumor samples were collected and were cut into small pieces. Minced sample pieces were transferred to a gentleMACS C Tube (Miltenyi Biotec; #130-093-237) containing 5 mL digestion enzyme mix prepared according to the manufacturer's instructions (Human Tumor Dissociation Kit; Miltenyi Biotec, #130-095-929). CD45+ cells were isolated by CD45 (TIL) human microbeads (Miltenyi Biotec; #130-118-780). Mouse CD8+ T cells were isolated by the EasySep Mouse CD8+ T-cell Isolation Kit (STEMCELL; #19853). In vitro isolation of PBMCs was done using Ficoll-Paque PLUS (GE Life; #17144002). For surface staining, the following antibodies were used for flow cytometry staining: CD45-PerCP-Cy5.5 (Thermo Fisher Scientific; cat. #45-0451-82, RRID: AB_1107002), CD45-BV421 (BioLegend; cat. #103134, RRID: AB_2562559), CD3-FITC (Thermo Fisher Scientific; cat. # 11-0037-42, RRID: AB_2016669), CD4-APC (Thermo Fisher Scientific; cat. #17-0049-42, RRID: AB_1272048), CD4-APC (Thermo Fisher Scientific; cat. #17-0041-82, RRID: AB_469320), CD8-APC-eFlour780 (Thermo Fisher Scientific; cat. #47-0081-82, RRID: AB_1272185), CD8-APC-eFlour780 (Thermo Fisher Scientific; cat. #47-0088-42, RRID: AB_1272046), CD8-PerCP-Cyanine5.5 (Thermo Fisher Scientific; cat. #45-0081-82, RRID: AB_1107004), CD27-PE (Thermo Fisher Scientific; cat. #MA5-17905, RRID: AB_2539289), CD27-APC (Thermo Fisher Scientific; cat. #17-0271-82, RRID: AB_469370), PD-1-eFlour450 (Thermo Fisher Scientific; cat. #48-9969-42, RRID: AB_2762470), CD45RA-BV421 (BioLegend; cat. #304130, RRID: AB_10965547), CCR7-APC (BioLegend; cat. #353214, RRID: AB_10917387), CD56-APC (BioLegend; cat. #362504, RRID: AB_2563913), CD16-PE (BioLegend; cat. #302008, RRID: AB_314208), PD-1-FITC (Thermo Fisher Scientific; cat. #11-9985-82, RRID: AB_465472), TIM-3-APC (Thermo Fisher Scientific; cat. #17-3109-42, RRID: AB_1963622), and TIM-3-PerCP-Cy5.5 (BioLegend; cat. #119718, RRID: AB_2571935). For intracellular cytokine staining, T cells were stimulated after the indicated treatment with a cocktail containing 50 ng/mL PMA (Sigma; #P8139), 1 μg/mL ionomycin (Sigma; #407951), and 1 μg/mL Golgi­Stop (BD; #554724) for 4 hours. Activated T cells were stained with Live/Dead Fixable Violet (Invitrogen; #L34963) at room temperature for 30 minutes, and then fixed and permeabilized (BD Biosciences; #554722). After washing with Perm/Wash buffer (BD Biosciences; cat. #554723, RRID: AB_2869011), cells were stained with the following antibodies: IFNγ-PE (Thermo Fisher Scientific; cat. #12-7311-82, RRID: AB_466193), Granzyme B-eFlour450 (Thermo Fisher Scientific; cat. #48-8898-82, RRID: AB_11149362), Granzyme B-FITC (Thermo Fisher Scientific; cat. #11-8898-82, RRID: AB_10733414), TNFα-APC (BioLegend; cat. #506308, RRID: AB_315429), IL2-APC (Thermo Fisher Scientific; cat. #17-7021-82, RRID: AB_469490), and TCF-1-Alexa Flour647 (BioLegend; cat. #655204, RRID: AB_2566620). For the OT-I T-cell cytotoxicity assay, after the indicated treatment, OT-I CTLs were collected and resuspended in PBS for surface staining. For the CFSE T-cell proliferation assay, separated CD8+ T cells were incubated with 1 μmol/L CFSE (eBioscience; #65-0850-84) for 30 minutes. After being neutralized with FBS and washed with PBS three times, CFSE-labeled T cells were used for further assessment. Flow cytometric data were acquired on a Beckman CytoFlex S and analyzed using FlowJo V10 software (RRID: SCR_008520).

T-cell Activation and Cd27 KD

For in vitro CD8+ T-cell activation, CD8+ T cells were magnetically enriched (BioLegend; #480035) from the spleen and lymph nodes of 6-week-old male wild-type C57BL/6J mice. Cells were plated in 96-well plates precoated with 2 μg/mL anti-mouse CD3 plus 2 μg/mL anti-mouse CD28 at a density of 5 × 105/well in T-cell culture medium. Twenty-four hours later, Cd27-specific siRNA prepared with OPTI-MEM (Thermo Fisher Scientific; #31985088) and RNAiMAX Transfection Reagent (Thermo Fisher Scientific; #13778150) was added to the culture medium at a final concentration of 5 μmol/L according to the manufacturer's protocol. The cells were incubated for another 48 hours, stimulated as described above, and evaluated by flow cytometry to detect the indicated cytokines and other cell markers. Sequences that specifically target murine Cd27 were sense-5′-CCAGCAUGUUUCUUAUCUUTT-3′ and antisense-5′-AAGAUAAGAAACAUGCUGGAG-3′.

Single-Cell Transcriptomic Analysis

Single-cell transcriptomic data were extracted from the Gene Expression Omnibus [(GEO) RRID: SCR_005012, accession code GSE140228 and GSE125449], containing data from 19 HCC patients generated with the smart-seq2 and 10X Genomics platforms. These data sets were integrated using Seurat's CCA to remove potential batch effects. For other collected scRNA-seq data sets (accession codes GSE120575 and GES145281), we applied the same filtering steps to 10X Genomics data. Data processing and analysis were performed using Seurat (version 4.0, RRID: SCR_007322).

Principal Component Analysis, t-SNE, and Uniform Manifold Approximation and Projection Analysis

All cells that had low-quality unique molecular identifier sequences were removed. For the remaining cells, the similarity and variability were summarized by principal component analysis. The expression trend of cell genes was proportional to the sample distance. For visualization, the dimensionality of each data set was further reduced using either Barnes-Hut t-SNE or uniform manifold approximation and projection analysis from the Seurat toolkit with the top 30 principal components with default settings. CD8+ T cells were reanalyzed to identify subclusters and were contrasted using the Seurat FindMarkers function to identify conserved marker genes for these subsets.

Glucose and ECAR Assays

For the glucose assay, the supernatant of tumor and T cells was collected, and the glucose concentration was assessed (GemPharmatech; ref. 58). For ECAR, mouse orthotopic tumor cells or tumor-infiltrated CD8+ T cells harvested at different time points after the indicated treatment were plated in nonbuffered RPMI 1640 culture medium with 25 mmol/L glucose. In some experiments, cells were pretreated with an mTOR inhibitor or a HIF1α inhibitor for another 24 hours. The real-time ECAR was measured on an XF96 extracellular flux analyzer (Seahorse Bioscience).

RNA Isolation and Library Preparation

The TRIzol method was adopted to extract total RNA according to the manufacturer's protocol. A NanoDrop 2000 spectrophotometer (Thermo Scientific, RRID: SCR_018042) was applied for RNA purity measurement and quantification, and an Agilent 2100 bioanalyzer (Agilent Technologies, RRID: SCR_019389) was applied for RNA integrity assessment. Sequencing libraries were generated using the TruSeq Stranded mRNA LT Sample Prep Kit (Illumina) following the manufacturer's recommendations.

OT-I T-cell Cytotoxicity Assay

OVA-specific CD8+ CTLs were generated by incubating splenocytes from OT-I mice with 5 μg/mL SIINFEKL peptide (Sangon Biotech; #T510212) and 20 ng/mL mIL2 (R&D Systems; #402-ML) in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) RPMI 1640 medium for 7 days.

Prepared OT-I CTLs were treated with IFNα at different concentrations in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) RPMI 1640 medium for 48 hours. Then EL4 cells were harvested and resuspended at 1 × 106 cells/mL. EL4 cells were pulsed with medium containing a vehicle control or 10 μg/mL SIINFEKL peptide for 2 hours at 37°C and then labeled with 0.5 μmol/L or 5 μmol/L CFSE (Thermo Fisher Scientific; #C34554), respectively, for 10 minutes at 37°C. EL4 cells were cocultured with OT-I CTLs at different effector:target ratios in low-glucose (5 mmol/L) or high-glucose (25 mmol/L) RPMI 1640 medium for 12 hours. After incubation, the percentage of specific killing was determined by flow cytometric analysis. Specific killing (%) = [1 − (sample ratio/negative control ratio)]. Sample ratio = [CFSEhi/CFSElo] value of each sample cocultured with CD8+ T cells. Negative control ratio = [CFSEhi/CFSElo] value of EL4 cells not cultured with CD8+ T cells.

RNA-seq and Analysis of Differentially Expressed Genes

Prepared libraries were sequenced on an Illumina HiSeq X Ten platform (RRID: SCR_016385). Raw sequence reads were first aligned to the mouse UniGene transcriptome with Bowtie (v1.0.0, RRID: SCR_005476) to estimate the insert fragment size and the SD needed by TopHat2 (RRID: SCR_013035) to align the reads to the genome. Then, TopHat2 (RRID: SCR_013035) was used to align the reads to the reference mouse genome (GRCm38) with the aligning parameter bowtie1 and Ensembl-annotated transcripts (version 77) as a guide reference. Uniquely mapped reads were used to quantify gene expression by Cufflinks (v2.1.1, RRID: SCR_014597). The abundances of transcripts (including mRNAs, pseudogenes, noncoding RNAs, and other predicted RNAs) were calculated and normalized in fragments per kilobase of transcript per million mapped reads, as described above, from the raw RNA-seq data and used for GSEA (Broad Institute, RRID: SCR_003199). Differential gene expression evaluation was analyzed using DESeq2 software (v1.30.0, RRID: SCR_015687; ref. 69).

Statistical Analysis

R (4.0.3) software was used for data analysis, and statistical significance was determined using a t test. P < 0.05 was considered statistically significant. Pearson correlation analysis was used to describe the correlation between variables. For analysis of the TCGA data set, a Kaplan–Meier curve was constructed to compare the OS and PFS rates of two groups. The log-rank P value and hazard ratio were calculated. For single-cell analysis, comparison of two groups was performed using the nonparametric Wilcoxon rank sum test with Bonferroni correction.

Data Availability

The TCGA data set, including liver hepatocellular carcinoma, was downloaded from cBioPortal (ref. 70; http://www.cbioportal.org/; RRID: SCR_014555). The full RNA-seq data set has been uploaded to the GEO repository (accession code GSE179352). None of the flow-cytometric data or IHC data supporting the current study have been deposited in a public repository because of privacy and ethical restrictions but are available from the corresponding author on request.

Code Availability

Codes supporting the findings of this study have been deposited at https://github.com/zyangx/IFN_Sens_PD1/.

H. Sun reports personal fees from Eisai, MSD, Hengrui, TopAlliance, Beigene, AstraZeneca, Bayer, Roche, and Zelgen, and grants and personal fees from Innovent during the conduct of the study. A.X. Zhu reports personal fees from I-Mab, Lilly, Merck, Exelixis, Roche, Eisai, and Bayer outside the submitted work. Y. Xu reports grants from the National Natural Science Foundation of China, the National Science and Technology Major Project, and the Science and Technology Commission of Shanghai Municipality during the conduct of the study. J. Fan reports grants from the National Natural Science Foundation of China, the National Science and Technology Major Project, and the Science and Technology Commission of Shanghai Municipality during the conduct of the study. No disclosures were reported by the other authors.

B. Hu: Conceptualization, resources, data curation, formal analysis, supervision, investigation, methodology, writing–original draft, project administration. M. Yu: Conceptualization, resources, data curation, formal analysis, supervision, investigation, methodology, writing–original draft, project administration. X. Ma: Conceptualization, resources, data curation, formal analysis, supervision, investigation, methodology, writing–original draft, project administration. J. Sun: Conceptualization, resources, data curation, formal analysis, supervision, investigation, methodology, writing–original draft, project administration. C. Liu: Data curation, software, methodology. C. Wang: Software, validation, visualization, methodology. S. Wu: Data curation, validation, investigation, methodology. P. Fu: Formal analysis, validation, visualization, methodology. Z. Yang: Software, validation, visualization. Y. He: Software, validation, visualization. Y. Zhu: Methodology. C. Huang: Supervision, writing–review and editing. X. Yang: Supervision, writing–review and editing. Y. Shi: Supervision, writing–review and editing. S. Qiu: Supervision, writing–review and editing. H. Sun: Supervision, writing–review and editing. A.X. Zhu: Supervision, writing–review and editing. J. Zhou: Supervision, writing–review and editing. Y. Xu: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing. D. Zhu: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing. J. Fan: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing.

We thank Xun Huang at Lingang Laboratory, Shanghai, China, for providing cell lines. We thank Yan-Yan Nie at the Shanghai Laboratory Animal Research Center for providing technical assistance with animal studies. We thank Ruo-Yu Shi at the department of Anatomical Pathology, Singapore General Hospital, for providing pathology diagnosis. We also thank Merdobio Corporation (Shanghai, China) for providing technical assistance with PDO-TIL model construction. This research was supported by Research Projects from the Science and Technology Commission of Shanghai Municipality (grant 21JC1410100, to J. Fan; grant 22140900600, to B. Hu), the National Natural Science Foundation of China (no. 81772551, to J. Fan; no. 81802364, to B. Hu; no. 82073272, to Y. Xu; no. 82073881 and 81872895, to D. Zhu; no. 82002618, to X. Ma; no. 81871929 and 82072667, to C. Huang), and the National Science and Technology Major Project (no. 2018ZX10723204-006-002; to Y. Xu).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Sung
H
,
Ferlay
J
,
Siegel
RL
,
Laversanne
M
,
Soerjomataram
I
,
Jemal
A
, et al
.
Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
2021
;
71
:
209
49
.
2.
El-Khoueiry
AB
,
Sangro
B
,
Yau
T
,
Crocenzi
TS
,
Kudo
M
,
Hsu
C
, et al
.
Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial
.
Lancet
2017
;
389
:
2492
502
.
3.
Yau
T
,
Kang
YK
,
Kim
TY
,
El-Khoueiry
AB
,
Hsu
C
.
Nivolumab (NIVO) + ipilimumab (IPI) combination therapy in patients (pts) with advanced hepatocellular carcinoma (aHCC): results from CheckMate 040
.
J Clin Oncol
2019
;
37
:
4012
.
4.
Sangro
B
,
Gomez-Martin
C
,
de la Mata
M
,
Iñarrairaegui
M
,
Garralda
E
,
Barrera
P
, et al
.
A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C
.
J Hepatol
2013
;
59
:
81
8
.
5.
Qin
S
,
Ren
Z
,
Meng
Z
,
Chen
Z
,
Chai
X
,
Xiong
J
, et al
.
Camrelizumab in patients with previously treated advanced hepatocellular carcinoma: a multicentre, open-label, parallel-group, randomised, phase 2 trial
.
Lancet Oncol
2020
;
21
:
571
80
.
6.
Murciano-Goroff
YR
,
Warner
AB
,
Wolchok
JD
.
The future of cancer immunotherapy: microenvironment-targeting combinations
.
Cell Res
2020
;
30
:
507
19
.
7.
Liberti
MV
,
Locasale
JW
.
The Warburg effect: how does it benefit cancer cells?
Trends Biochem Sci
2016
;
41
:
211
8
.
8.
Leone
RD
,
Powell
JD
.
Metabolism of immune cells in cancer
.
Nat Rev Cancer
2020
;
20
:
516
31
.
9.
Nagao
A
,
Kobayashi
M
,
Koyasu
S
,
Chow
CCT
,
Harada
H
.
HIF-1-dependent reprogramming of glucose metabolic pathway of cancer cells and its therapeutic significance
.
Int J Mol Sci
2019
;
20
:
238
.
10.
Semenza
GL
.
HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations
.
J Clin Invest
2013
;
123
:
3664
71
.
11.
Noman
MZ
,
Desantis
G
,
Janji
B
,
Hasmim
M
,
Karray
S
,
Dessen
P
, et al
.
PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation
.
J Exp Med
2014
;
211
:
781
90
.
12.
Dai
X
,
Pi
G
,
Yang
SL
,
Chen
GG
,
Liu
LP
,
Dong
HH
.
Association of PD-L1 and HIF-1α coexpression with poor prognosis in hepatocellular carcinoma
.
Transl Oncol
2018
;
11
:
559
66
.
13.
Yin
Z
,
Bai
L
,
Li
W
,
Zeng
T
,
Tian
H
,
Cui
J
.
Targeting T cell metabolism in the tumor microenvironment: an anti-cancer therapeutic strategy
.
J Exp Clin Cancer Res
2019
;
38
:
403
.
14.
Palazon
A
,
Tyrakis
PA
,
Macias
D
,
Veliça
P
,
Rundqvist
H
,
Fitzpatrick
S
, et al
.
An HIF-1α/VEGF-A axis in cytotoxic T cells regulates tumor progression
.
Cancer Cell
2017
;
32
:
669
83
.
15.
Kishton
RJ
,
Sukumar
M
,
Restifo
NP
.
Metabolic regulation of T cell longevity and function in tumor immunotherapy
.
Cell Metab
2017
;
26
:
94
109
.
16.
Yu
YR
,
Ho
PC
.
Sculpting tumor microenvironment with immune system: from immunometabolism to immunoediting
.
Clin Exp Immunol
2019
;
197
:
153
60
.
17.
Sugiura
A
,
Rathmell
JC
.
Metabolic barriers to T cell function in tumors
.
J Immunol
2018
;
200
:
400
7
.
18.
Lim
S
,
Phillips
JB
,
Madeira da Silva
L
,
Zhou
M
,
Fodstad
O
,
Owen
LB
, et al
.
Interplay between immune checkpoint proteins and cellular metabolism
.
Cancer Res
2017
;
77
:
1245
9
.
19.
Ivashkiv
LB
,
Donlin
LT
.
Regulation of type I interferon responses
.
Nat Rev Immunol
2014
;
14
:
36
49
.
20.
Seto
WK
,
Lo
YR
,
Pawlotsky
JM
,
Yuen
MF
.
Chronic hepatitis B virus infection
.
Lancet
2018
;
392
:
2313
24
.
21.
Herzer
K
,
Hofmann
TG
,
Teufel
A
,
Schimanski
CC
,
Moehler
M
,
Kanzler
S
, et al
.
IFN-alpha-induced apoptosis in hepatocellular carcinoma involves promyelocytic leukemia protein and TRAIL independently of p53
.
Cancer Res
2009
;
69
:
855
62
.
22.
Hobeika
AC
,
Subramaniam
PS
,
Johnson
HM
.
IFNalpha induces the expression of the cyclin-dependent kinase inhibitor p21 in human prostate cancer cells
.
Oncogene
1997
;
14
:
1165
70
.
23.
Cheon
H
,
Borden
EC
,
Stark
GR
.
Interferons and their stimulated genes in the tumor microenvironment
.
Semin Oncol
2014
;
41
:
156
73
.
24.
Di Franco
S
,
Turdo
A
,
Todaro
M
,
Stassi
G
.
Role of type I and II interferons in colorectal cancer and melanoma
.
Front Immunol
2017
;
8
:
878
.
25.
Terawaki
S
,
Chikuma
S
,
Shibayama
S
,
Hayashi
T
,
Yoshida
T
,
Okazaki
T
, et al
.
IFN-α directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity
.
J Immunol
2011
;
186
:
2772
9
.
26.
Liang
Y
,
Tang
H
,
Guo
J
,
Qiu
X
,
Yang
Z
,
Ren
Z
, et al
.
Targeting IFNα to tumor by anti-PD-L1 creates feedforward antitumor responses to overcome checkpoint blockade resistance
.
Nat Commun
2018
;
9
:
4586
.
27.
Davar
D
,
Wang
H
,
Chauvin
JM
,
Pagliano
O
,
Fourcade
JJ
,
Ka
M
, et al
.
Phase Ib/II study of pembrolizumab and pegylated-interferon Alfa-2b in advanced melanoma
.
J Clin Oncol
2018
;
36
:
JCO1800632
.
28.
Ma
L
,
Wang
L
,
Khatib
SA
,
Chang
CW
,
Heinrich
S
,
Dominguez
DA
, et al
.
Single-cell atlas of tumor cell evolution in response to therapy in hepatocellular carcinoma and intrahepatic cholangiocarcinoma
.
J Hepatol
2021
;
75
:
1397
408
.
29.
Tumeh
PC
,
Harview
CL
,
Yearley
JH
,
Shintaku
IP
,
Taylor
EJ
,
Robert
L
, et al
.
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
2014
;
515
:
568
71
.
30.
Fehlings
M
,
Simoni
Y
,
Penny
HL
,
Becht
E
,
Loh
CY
,
Gubin
MM
, et al
.
Checkpoint blockade immunotherapy reshapes the high-dimensional phenotypic heterogeneity of murine intratumoural neoantigen-specific CD8(+) T cells
.
Nat Commun
2017
;
8
:
562
.
31.
Gubin
MM
,
Zhang
X
,
Schuster
H
,
Caron
E
,
Ward
JP
,
Noguchi
T
, et al
.
Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens
.
Nature
2014
;
515
:
577
81
.
32.
Koikawa
K
,
Kibe
S
,
Suizu
F
,
Sekino
N
,
Kim
N
,
Manz
TD
, et al
.
Targeting Pin1 renders pancreatic cancer eradicable by synergizing with immunochemotherapy
.
Cell
2021
;
184
:
4753
71
.
33.
Kong
JCH
,
Guerra
GR
,
Millen
RM
,
Roth
S
,
Xu
H
,
Neeson
PJ
, et al
.
Tumor-infiltrating lymphocyte function predicts response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer
.
JCO Precis Oncol
2018
;
2
:
1
15
.
34.
Zhang
Q
,
He
Y
,
Luo
N
,
Patel
SJ
,
Han
Y
,
Gao
R
, et al
.
Landscape and dynamics of single immune cells in hepatocellular carcinoma
.
Cell
2019
;
179
:
829
45
.
35.
Butler
A
,
Hoffman
P
,
Smibert
P
,
Papalexi
E
,
Satija
R
.
Integrating single-cell transcriptomic data across different conditions, technologies, and species
.
Nat Biotechnol
2018
;
36
:
411
20
.
36.
Aran
D
,
Sirota
M
,
Butte
AJ
.
Systematic pan-cancer analysis of tumour purity
.
Nat Commun
2015
;
6
:
8971
.
37.
Lim
AR
,
Rathmell
WK
,
Rathmell
JC
.
The tumor microenvironment as a metabolic barrier to effector T cells and immunotherapy
.
eLife
2020
;
9
:
e55185
.
38.
TeSlaa
T
,
Teitell
MA
.
Techniques to monitor glycolysis
.
Methods Enzymol
2014
;
542
:
91
114
.
39.
Narita
T
,
Yin
S
,
Gelin
CF
,
Moreno
CS
,
Yepes
M
,
Nicolaou
KC
, et al
.
Identification of a novel small molecule HIF-1alpha translation inhibitor
.
Clin Cancer Res
2009
;
15
:
6128
36
.
40.
Taylor
A
,
Harker
JA
,
Chanthong
K
,
Stevenson
PG
,
Zuniga
EI
,
Rudd
CE
.
Glycogen synthase kinase 3 inactivation drives T-bet-mediated downregulation of co-receptor PD-1 to enhance CD8(+) cytolytic T cell responses
.
Immunity
2016
;
44
:
274
86
.
41.
Gropper
Y
,
Feferman
T
,
Shalit
T
,
Salame
TM
,
Porat
Z
,
Shakhar
G
.
Culturing CTLs under hypoxic conditions enhances their cytolysis and improves their anti-tumor function
.
Cell Rep
2017
;
20
:
2547
55
.
42.
Milde-Langosch
K
.
The Fos family of transcription factors and their role in tumourigenesis
.
Eur J Cancer
2005
;
41
:
2449
61
.
43.
Trierweiler
C
,
Hockenjos
B
,
Zatloukal
K
,
Thimme
R
,
Blum
HE
,
Wagner
EF
, et al
.
The transcription factor c-JUN/AP-1 promotes HBV-related liver tumorigenesis in mice
.
Cell Death Differ
2016
;
23
:
576
82
.
44.
Xu
W
,
Zhang
X
,
Wu
JL
,
Fu
L
,
Liu
K
,
Liu
D
, et al
.
O-GlcNAc transferase promotes fatty liver-associated liver cancer through inducing palmitic acid and activating endoplasmic reticulum stress
.
J Hepatol
2017
;
67
:
310
20
.
45.
Tanaka
N
,
Ishihara
M
,
Taniguchi
T
.
Suppression of c-myc or fosB-induced cell transformation by the transcription factor IRF-1
.
Cancer Lett
1994
;
83
:
191
6
.
46.
Chi
H
.
Regulation and function of mTOR signalling in T cell fate decisions
.
Nat Rev Immunol
2012
;
12
:
325
38
.
47.
Man
K
,
Kallies
A
.
Synchronizing transcriptional control of T cell metabolism and function
.
Nat Rev Immunol
2015
;
15
:
574
84
.
48.
Ochsner
SA
,
Abraham
D
,
Martin
K
,
Ding
W
,
McOwiti
A
,
Kankanamge
W
, et al
.
The Signaling Pathways Project, an integrated ‘omics knowledgebase for mammalian cellular signaling pathways
.
Sci Data
2019
;
6
:
252
.
49.
Nandi
D
,
Cheema
PS
,
Jaiswal
N
,
Nag
A
.
FoxM1: repurposing an oncogene as a biomarker
.
Semin Cancer Biol
2018
;
52
:
74
84
.
50.
Gong
J
,
Chehrazi-Raffle
A
,
Reddi
S
,
Salgia
R
.
Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations
.
J Immunother Cancer
2018
;
6
:
8
.
51.
Scharping
NE
,
Menk
AV
,
Whetstone
RD
,
Zeng
X
,
Delgoffe
GM
.
Efficacy of PD-1 blockade is potentiated by metformin-induced reduction of tumor hypoxia
.
Cancer Immunol Res
2017
;
5
:
9
16
.
52.
Lossos
C
,
Liu
Y
,
Kolb
KE
,
Christie
AL
,
Van Scoyk
A
,
Prakadan
SM
, et al
.
Mechanisms of lymphoma clearance induced by high-dose alkylating agents
.
Cancer Discov
2019
;
9
:
944
61
.
53.
Uka
K
,
Aikata
H
,
Takaki
S
,
Miki
D
,
Kawaoka
T
,
Jeong
SC
, et al
.
Pretreatment predictor of response, time to progression, and survival to intraarterial 5-fluorouracil/interferon combination therapy in patients with advanced hepatocellular carcinoma
.
J Gastroenterol
2007
;
42
:
845
53
.
54.
Chang
CH
,
Curtis
JD
,
Maggi
LB
Jr
,
Faubert
B
,
Villarino
AV
,
O'Sullivan
D
, et al
.
Posttranscriptional control of T cell effector function by aerobic glycolysis
.
Cell
2013
;
153
:
1239
51
.
55.
Renner
K
,
Bruss
C
,
Schnell
A
,
Koehl
G
,
Becker
HM
,
Fante
M
, et al
.
Restricting glycolysis preserves T cell effector functions and augments checkpoint therapy
.
Cell Rep
2019
;
29
:
135
50
.
56.
Patsoukis
N
,
Bardhan
K
,
Chatterjee
P
,
Sari
D
,
Liu
B
,
Bell
LN
, et al
.
PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation
.
Nat Commun
2015
;
6
:
6692
.
57.
Cascone
T
,
McKenzie
JA
,
Mbofung
RM
,
Punt
S
,
Wang
Z
,
Xu
C
, et al
.
Increased tumor glycolysis characterizes immune resistance to adoptive T cell therapy
.
Cell Metab
2018
;
27
:
977
87
.
58.
Chang
CH
,
Qiu
J
,
O'Sullivan
D
,
Buck
MD
,
Noguchi
T
,
Curtis
JD
, et al
.
Metabolic competition in the tumor microenvironment is a driver of cancer progression
.
Cell
2015
;
162
:
1229
41
.
59.
Uhlén
M
,
Fagerberg
L
,
Hallström
BM
,
Lindskog
C
,
Oksvold
P
,
Mardinoglu
A
, et al
.
Proteomics; tissue-based map of the human proteome
.
Science
2015
;
347
:
1260419
.
60.
Buchan
SL
,
Fallatah
M
,
Thirdborough
SM
,
Taraban
VY
,
Rogel
A
,
Thomas
LJ
, et al
.
PD-1 blockade and CD27 stimulation activate distinct transcriptional programs that synergize for CD8(+) T-cell-driven antitumor immunity
.
Clin Cancer Res
2018
;
24
:
2383
94
.
61.
Llovet
JM
,
Lencioni
R
.
mRECIST for HCC: performance and novel refinements
.
J Hepatol
2020
;
72
:
288
306
.
62.
Ji
J
,
Shi
J
,
Budhu
A
,
Yu
Z
,
Forgues
M
,
Roessler
S
, et al
.
MicroRNA expression, survival, and response to interferon in liver cancer
.
N Engl J Med
2009
;
361
:
1437
47
.
63.
Patt
YZ
,
Hassan
MM
,
Lozano
RD
,
Brown
TD
,
Vauthey
JN
,
Curley
SA
, et al
.
Phase II trial of systemic continuous fluorouracil and subcutaneous recombinant interferon Alfa-2b for treatment of hepatocellular carcinoma
.
J Clin Oncol
2003
;
21
:
421
7
.
64.
Broutier
L
,
Mastrogiovanni
G
,
Verstegen
MM
,
Francies
HE
,
Gavarró
LM
,
Bradshaw
CR
, et al
.
Human primary liver cancer-derived organoid cultures for disease modeling and drug screening
.
Nat Med
2017
;
23
:
1424
35
.
65.
Tan
YS
,
Lei
YL
.
Isolation of tumor-infiltrating lymphocytes by Ficoll-Paque density gradient centrifugation
.
Methods Mol Biol
2019
;
1960
:
93
9
.
66.
Zhang
Q
,
Lou
Y
,
Yang
J
,
Wang
J
,
Feng
J
,
Zhao
Y
, et al
.
Integrated multiomic analysis reveals comprehensive tumour heterogeneity and novel immunophenotypic classification in hepatocellular carcinomas
.
Gut
2019
;
68
:
2019
31
.
67.
Zunder
ER
,
Finck
R
,
Behbehani
GK
,
Amir el
AD
,
Krishnaswamy
S
,
Gonzalez
VD
, et al
.
Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm
.
Nat Protoc
2015
;
10
:
316
33
.
68.
Stack
EC
,
Wang
C
,
Roman
KA
,
Hoyt
CC
.
Multiplexed immunohistochemistry, imaging, and quantitation: a review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis
.
Methods
2014
;
70
:
46
58
.
69.
Love
MI
,
Huber
W
,
Anders
S
.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
.
70.
Cerami
E
,
Gao
J
,
Dogrusoz
U
,
Gross
BE
,
Sumer
SO
,
Aksoy
BA
, et al
.
The cBio Cancer Genomics Portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
2012
;
2
:
401
4
.

Supplementary data