Pantethine therapy reduces MCA205 growth in immunocompetent mice
We implanted the MCA205 (MCA) sarcoma cells in immunocompetent mice to test whether a systemic administration of pantethine (Pant) would compensate for the lack of Vnn1 in the tumor mass (18). Daily Pant therapy was started on established tumors at day 10 post cell engraftment (Fig. 1A). Growth reduction occurred within a few days post injection and persisted during the treatment period without leading to a complete tumor regression (Fig. 1B). A similar result was obtained by co-injecting PanSH and cysteamine (CEA), the products of pantetheinase activity (FigS1A-B). Importantly, when Pant was administered to a Nude mouse bearing MCA tumor, no growth inhibition was detected (Fig. 1C). This result suggested that Pant effect might directly or indirectly regulate immune responses. Since Pant enhances mitochondrial activity (11), we compared its effect to that of dichloroacetate (DCA), a compound that increases pyruvate uptake in mitochondria via PDK inhibition (19) and has previously been shown to exert anti-tumor activity (20, 21). As shown in Fig. 1D, Pant and DCA comparably reduced MCA tumor growth without synergy. We then performed a global metabolomics analysis of tumors from mice treated with vehicle PBS, Pant, DCA or combined drugs. Tumors harvested on day 28 (D28) showed that the global metabolic changes were not superimposable (Fig. 1E). Pant enhanced the level of metabolites associated with stress response, amino acids and nucleotide metabolism whereas DCA enhanced carbohydrate metabolism and mitochondrial activity. Interestingly, their association had a significant effect on the production of mitochondrial energetic metabolites and biogenic amines, a phenotype confirmed by nuclear magnetic resonance analysis (Fig S1C). We then focused our metabolic analysis on in vitro grown or ex vivo isolated cancer cells. In vitro, addition of Pant slightly enhanced the oxygen consumption (OCR) and extracellular acidification rates (ECAR) measured by a Seahorse analysis (Fig. 1F), and more significantly the production of mitochondrial ROS (mtROS) (Fig. 1G). In vivo, from D24 to D28, the mitochondrial polarization over mass ratio, scored by flow cytometry using the MitoTracker Deep Red (MDR) and MitoTracker Green (MG) probes, respectively, increased similarly between PBS and Pant samples (Fig. 1H-S1D) (22). Since alterations in mitochondrial metabolism can contribute to the reinforcement of a Warburg effect, we tested lactate production in tumor masses (Fig. 1I) and quantified by qRT-PCR the expression level of transcripts associated with an increased glycolytic flow on isolated cancer cells (Fig. 1J). The results showed that neither lactate nor transcript levels were modified by Pant. Altogether, Pant administration modulates metabolic cues in the tumor but does not fully rescue mitochondrial activity nor reduce the Warburg phenotype associated to cancer cell growth. These findings suggest that, in the MCA model, Pant might rather impact the activity of tumor-associated cells such as immunocytes.
Pantethine enhances myeloid signatures associated with antigen presentation and type 1 immunity
We performed a follow-up of immune cell infiltration at different stages of MCA tumor development using flow cytometry. As shown in Fig. 2A, CD11b+ myeloid cells predominated, with a myeloid/lymphoid cell ratio evolving progressively towards an enrichment in lymphocytes after one week of Pant administration. In Pant-treated mice, the proportion of tumor-infiltrating monocytes on day 20 (D20) was reduced whereas that of tumor-associated macrophages (TAMs) was increased (Fig. 2B, C). The latter subset was significantly enriched in MHCIIhigh cells (Fig. 2D), previously identified as owing potential anti-tumor functions (23).
To further dissect myeloid cell heterogeneity and maturation stages, we performed a single-cell RNA sequencing analysis of CD45+ tumor-infiltrating cells at D20 and D28 of tumor progression. We focused on the three main metaclusters corresponding to myeloid, lymphoid and antigen-presenting cells (APC), the cluster “others” being invariant in our experimental conditions (FigS2A). The myeloid metacluster was further split in 11 clusters (Fig. 2E) based on the preferential expression of cell-specific markers (Fig. 2F) defining neutrophils (Ly6g, cluster 8), monocytes (Ly6c2, cluster 7), TAM subsets (Adgre1, Cd64, clusters 2, 4–6, 10, 11), proliferating cells (mKi67, cluster 9) or cell signatures linked with activation or polarization pathways (cluster 11, TNF/NF-κB; cluster 10 and 2 for M1- versus M2-like phenotypes) (Fig. 2F, S2C,D). Transitional stages of TAM maturation were annotated mono-TAM A to C (clusters 4–6) as described (24). Interestingly, the density plots clearly distinguished untreated from Pant-treated samples (FigS2B) and showed an enhanced transition of mono-TAM A into mono-TAM C and M1-like TAMs in D20 to D28 Pant samples (Fig. 2G and S2B). Furthermore, Pant samples expressed higher levels of MhcII, Cd74, Cxcl9 transcripts linked to antigen presentation and lymphocyte chemoattraction pathways. In contrast, control tumors overexpressed Arg1 and Nnip3, Pgam1, Ldha, Ddit4 associated with M2 polarization and tumor hypoxia (25) (Volcano plot in Fig. 2H for D20 and maintained till D28 as shown in FigS2C). Within the APC metacluster (FigS3A), 7 clusters were defined and further characterized by differential gene expression (FigS3B). Among them, the proliferating (D20) and IFNγ-responsive monocyte-derived (D28) DC subsets were enriched in Pant samples whereas control PBS samples contained more cDC2 cells (Fig. 2I-S3C). Membrane MHCII expression by various APC was higher in Pant samples (FigS3D). This observation was confirmed by pathway enrichment analysis using Geneset Enrichment Analysis (GSEA) (Table 1) and Kyoto Encyclopedia of Genes and Genomes pathways (KEGG, FigS2D). As shown in Table 1, from D20 to D28 Pant samples overexpress transcripts related to the IFNγ response and antigen presentation process in myeloid and dendritic cells. On D28, these cells show reduced expression of genes linked with oxidative phosphorylation (OXPHOS) and aerobic respiration. Interestingly, an immunofluorescence analysis of tumor sections stained for MHCII to detect antigen presenting cells and CD8 documented 2 to 3-fold more MHCII+ / CD8+ cell contacts (Fig. 2J, S3E). In some discrete myeloid subsets, chemokine signaling was highlighted (Table 1). To identify dominant chemokine profiles at the protein level, we performed a proteome analysis (Fig. 2K) and a cytometry beads assay (CBA) (FigS3F). Control PBS samples showed an overrepresentation of proteins associated with tissue clearance (pentraxin), extra cellular matrix (MMP9, Serpin) and endothelial cell reorganization (angiopoietin, VEGF, endostatin). Pant-treated samples contained higher levels of cytokines or chemokines associated with type 1 immunity. This included CCL2 or CXCL9 (26, 27) (Fig. 2J) involved in monocyte (via CCR2) or effector lymphocyte (via CXCR3) recruitment, IL2p40 (28, 29) or Flt3-L (30), associated with M1 or DC maturation (Fig. 2J). Within each cluster, we did not observe major changes in the overall expression of different genes previously associated with anti or pro-tumor function (FigS3G). Altogether, Pant samples are enriched in APC subsets highlighting IFNγ responsiveness, enhanced antigen presentation and chemoattraction potential.
Pantethine enhances the development of effectors within adaptive lymphocytes
We used the CD8+/Treg cell ratio as a global indicator of anti-tumor lymphoid responses and observed a progressive enrichment in CD8+ over Treg CD4+ T cells after D17 till D28 (Fig. 3A). We took advantage of MCA cells expressing the OVA antigen to track tumor-specific CD8+ cell responses using a SIINFEKL tetramer. On D20, quantification of lymphocytes showed that Pant therapy led to a 2-fold increase in the number of NK/ILC, CD4+ and effector CD8+ T cells, including OVA-specific CD8+ T cells in the tumor and the draining lymph nodes (TDLNs) (Fig. 3B,C). In contrast, the proportion of immune cells remained globally unchanged by Pant administration in lymph nodes or spleen, at steady state or in a tumor context (FigS4A). This phenotype was comforted as CD8+ T cells tended to express higher levels of effector molecules such as GZMB, PRF1 or TNFα (Fig. 3D). The analysis of the lymphoid metacluster derived from the single-cell RNA sequencing analysis (FigS5A-C) confirmed the cytometry results. On D20, total CD4+ and more dramatically CD8+ T cells were increased to the expense of Tregs and activated memory CD8+ cells (Fig. 3E and S5A-C). Furthermore, CD8+ T cells showed an enriched representation of pathways associated with IFNγ response, cytotoxic function, chemokine production and to a lesser extent PD1 signaling (Fig. 3F). On D28, NK/ILC or Treg signatures predominated in Pant or PBS samples, respectively. We completed this analysis by quantifying cell surface expression of checkpoint inhibitors on various T lymphocyte subsets on D28 (Fig. 3G). In Pant treated samples, two-fold less CD4+ but not CD8+ T cells expressed CTLA4, PD1 or TIM3 molecules. Since exhaustion is associated with mitochondrial alterations, we monitored various parameters reflecting mitochondrial respiration and depolarization among lymphocytes. In D20 Pant tumors, a Seahorse analysis performed on purified CD8+ lymphocytes showed increased levels of OCR and ECAR witnessing a higher energetic state (Fig. 3H). We then quantified mitochondrial depolarization defined by the MDR/MG ratio on CD4+ and CD8+ T cells at different time points. Whereas no difference was observed in both cell subsets on D20, this ratio was lowered in D28 CD8+ T cells from Pant samples (Fig. 3I). Altogether, our results show that Pant therapy induces a more robust lymphocyte response affecting preferentially effector cell subsets. We thus scored the number of genes co-expressed by each cell and associated with a cytotoxic program. Although this analysis was limited by the total amount of cells, CD8+ T cells derived from Pant samples expressed a higher number of cytokine or cytotoxicity-associated genes, an argument in favor of their polyfunctionality (FigS5D). Furthermore, we used a global score recapitulating the expression level of genesets involved in activation or exhaustion programs in CD8+ lymphocytes. PBS and Pant samples were comparable for each cell subset, suggesting that Pant administration modifies the proportion and not the transcriptional state of these cells (FigS5E). While the relative number of total NK/ILC1 was increased in the cytometry analysis, their proportion within the lymphoid metacluster was reduced in Pant samples. Interestingly, by projecting NK versus ILC1 signatures (FigS5F) and applying single cell velocity analysis of the NK/ILC cluster (Fig. 3J), we observed an enhanced dynamic conversion of NK into ILC1 cells in control samples, whereas Pant therapy tended to reinforce NK cell signatures (31, 32), showing by GSEA an enrichment in IFNγ response (Fig. 3K).
Pantethine therapeutic efficacy depends on IFNγ, cDC1 and tumor-infiltrating CD8 + T cells
Efficacy of cytotoxic CD8+ T lymphocytes requires antigen cross presentation by cDC1 cells and their ability to infiltrate a tumor, a program driven by type 1 cytokines. We performed an immunohistochemistry analysis of CD3 and CD8+ TILs on tumor sections. As shown in Fig. 4A (quantified in Fig. 4B), in control tumors were either in reduced numbers or confined to the periphery of the tumor. In contrast, TILs were more abundant and infiltrating in Pant samples, in agreement with the IFNγ-driven increased CXCL9 expression (33). Accordingly, MCA tumors grew more rapidly in IFNGR1-deficient mice where Pant rather enhanced tumor growth (Fig. 4A). This result reinforced the notion that Pant efficacy relies on IFNγ-driven programs in APC and lymphocytes (Table 1). To dissect Pant effect at the cellular level, we transferred OT1 cells in mice, treated them or not with Pant prior to immunization with OVA. We then scored the expansion of CTV-labeled CD8 OT1 lymphocytes in draining lymph nodes. In this context, no difference between PBS and Pant samples was detected (FigS6A,B), suggesting that a chronic inflammatory might be required for Pant effect. We thus injected OVA-expressing MCA cells, extracted CD11c+ DC from TDLNs (FigS3D) and co-cultured them in vitro with CFSE-labeled OT-1 CD8+ T cells in the presence or absence of OVA (Fig. 6SC,D). Again, both cell numbers and CD8+ T cell proliferation index were comparable under the two conditions. To evaluate the contribution of cDC1 cells in vivo, we tested Pant therapy on MCA tumors grafted in control versus Xcr1-deficient mice that lack cDC1. As shown in Fig. 4B, tumor progression was uncontrollable in the absence of cDC1 cells, confirming the importance of this cell subset in the development of anti-tumor immunity (34). Furthermore, although Pant could reduce tumor growth in control mice, it slightly enhanced tumor growth in the absence of cDC1 cells. This showed that the therapeutic effect of Pant depended on the presence of cDC1 cells.
To prove the contribution of CD8+ T cells to Pant efficacy, we injected tumor-bearing mice with a depleting anti-CD8 mAb from D10 every 3 days onwards. As shown in Fig. 4C (and depletion controls in Fig. 6E,F), the depletion of CD8+ T cells boosted tumor growth in control mice as expected. In Pant-treated mice, this effect was also detected but tumor growth did not reach the level observed in control mice. This suggested that CD8+ T cells mediated anti-tumor effect might have been initiated before D10 or that Pant might have affected the function of other anti-tumor cells. Indeed, we observed an augmentation of NK1.1+ cells at an early stage (D20) of tumor progression and these cells may participate to tumor control (Fig. 3E) (32). We thus tested whether NK1.1+ cells might also contribute to Pant effect. Administration of a depleting anti-NK1.1 mAb had a modest enhancing effect on tumor growth after D22 (Fig. 4D). However, anti-tumor Pant effect persisted at the same level under both conditions. Altogether, these results show that Pant boosts type 1 immunity and that its anti-tumor effect depends critically on the presence of an IFNγ / cDC1 / CD8+ T cell pathway. Since this pathway is also required for the control of viral infection, we used a model of cutaneous HSV-1 infection in which the virus load and the proportion of virus-specific T cells could be monitored. The virus load was equivalent under both conditions showing that Pant does not have a significant impact on its replication in vivo (Fig. 4E). As seen in Fig. 4F, the proportion of central memory and effector CD8+ T cells in DLNs was enhanced 2-fold by Pant treatment, including virus-specific CD8+ T cells. This confirmed that Pant exerted its beneficial effect in the context of type 1-driven tissue inflammation.
Potential and limits of Pant therapy
Sarcoma cell lines show variable levels of susceptibility to immune checkpoint inhibitors (ICI) (35). As shown in Fig. 5A-B, administration of an anti-CTLA4 or anti-PD1 mAbs inhibited MCA growth with variable efficacy and combination with Pant modestly accentuated the inhibition. We then tested an osteosarcoma cell line in which ICI therapy alone or combined with doxorubicine had no effect. Addition of Pant treatment did not change the behavior of this highly resistant tumor (Fig. 5C) or the organization of the immune infiltrate (FigS7A-B). Nevertheless, the combined therapy enhanced the extent of necrosis (FigS7A-B). In the B16F10 melanoma model, known to be sensitive to anti PD1 therapy (36), Pant inhibited B16F10 growth to a level comparable to that obtained with MCA tumors and its effect was equivalent to that of an anti-PD1 mAb or their combination (Fig. 5D). Given the role of IFNγ in the development of resistance to immunotherapy (37), we administered an inhibitory anti-IFNγ antibody during the late phase where resistance to immunotherapy might develop. As seen in Fig. 5E, neutralization of IFNγ boosted tumor growth in control animals suggesting that it participated to the development of anti-tumor immunity. In Pant-treated mice, this boosting effect was absent suggesting that Pant therapy might have imprinted earlier so that it could not be reversed by IFNγ neutralization at later time points. In agreement with an early imprinting effect, we found that administration of Pant from D1 to D10 also inhibited growth (Fig. 5F).
High VNN1 expression in STS correlates with higher immune response and better prognosis
Since Pant therapy mimics VNN1 function (11), we first queried the TCGA database (including 206 soft tissue sarcoma STS samples) searching for VNN1-coexpressed gene modules. VNN1 expression matched with MSigDB hallmarks associated with IFNγ / inflammatory responses (FigS8A). Using a Cibersort deconvolution method to link VNN1 expression with the presence of immunocyte subsets, we found a significant correlation between high VNN1 expression and the presence of immune cells that do not express VNN1 such as M1/M2 macrophages and T cells (including CD8+) transcripts (FigS8B). This suggested that VNN1 expression within the tumor mass positively correlated with the presence of immunocytes with anti-tumor potential. To broaden this analysis, we searched for correlations between VNN1 mRNA expression and clinicopathological and immune variables in our large cohort of 1377 clinical STS samples (Fig S6C) collected during the surgery of non-metastatic primary tumors. Their characteristics are summarized in FigS6D. VNN1 expression was heterogeneous across the cohort with a range of intensities over 20 units in log2 scale (Fig. 7A), allowing the search for correlations with tumor variables. VNN1 expression, assessed as discrete variable (high vs. low), correlated (FigS8D) with patient’s age at diagnosis pathological tumor grade, and tumor site: as compared to the “VNN1-low” class, the “VNN1-high” class included older patients (p = 3.78E-08), more grade 1 tumors (p = 5.22E-06), and more extremity and less superficial trunk tumor sites (p = 5.20E-03). No significant correlation was found with patient’s sex, pathological tumor size, and tumor depth. Then, we analyzed the correlation of VNN1 classes with immune variables (Fig. 7B). First, the probability of activation of IFNα, IFNγ and STAT3 immune pathways was higher in the “VNN1-high” class than in the “VNN1-low” class (p < 0.05). Second, we compared the composition and functional orientation of tumor-infiltrated immune cells between VNN1 classes using the 24 Bindea’s immune cell types defined as the immunome. “VNN1-high” tumors displayed a higher infiltrate concerning 18 immune cell types (p < 0.05) including B cells, T cells, Tem cells, Th1 cells, TFH cells, Th17 cells, CD8+ T cells, γδ T cells, cytotoxic cells, CD56dim NK cells, dendritic cells (DC, interstitial DC, activated DC and plasmacytoid DC), eosinophils, macrophages, mast cells, and neutrophils. By contrast, “VNN1-low” tumors displayed a higher infiltrate in two immune cell types, NK cells and CD56bright NK cells (p < 0.05). Third, “VNN1-high” samples displayed higher ICR score (p = 1.81E-05) and immune cytolytic activity score (p = 3.13E-29), reflects of an antitumor cytotoxic immune response, than “VNN1-low” samples, and higher scores for signatures associated with response to ICI: tumor inflammation signature (TIS) (p = 1.58E-33) and tertiary lymphoid structure (TLS) (p = 1.56E-36). Finally, “VNN1-high” samples displayed higher TILs scores using lymphoid-alone, myeloid-alone and combined signatures (p < 0.05), and higher Antigen-Processing Molecules (APMs) score (p = 7.55E-18). Next, we searched for correlation of VNN1 classes with the clinical outcome. The patients with “VNN1-high” samples displayed longer MFS than those with “VNN1-low” samples (Fig. 7C with respective 5-year MFS equal to 66% (95%CI 60–73) vs. 61% (95%CI 54–68) (p = 3.01E-02, Fig. 7D). In univariate prognostic analysis (Fig. 7D), high VNN1 expression was associated with longer metastasis-free survival (p = 3.08E-02), as was the pathological grade 1 (p = 8.46E-03). In multivariate prognostic analysis, the VNN1 status tended to remain significant (Hazards Ratio HR = 0.64, 95%CI 0.41-1.00, p = 0.051) (Fig. 7D). Altogether, these results showed heterogeneous VNN1 expression levels in STS, and correlations with immune variables and clinical outcome. Of note, such prognostic value of VNN1 expression was not observed in a cohort of 326 patients with bone sarcoma (p = 0.397), nor in the subcohort of 94 patients with osteosarcoma (p = 0.229; FigS9).