TLR agonists polarize interferon responses in conjunction with dendritic cell vaccination in malignant glioma: a … – Nature.com

Patient characteristics and safety

A total of 23 patients with resection-eligible WHO grade III or IV glioma were enrolled and randomized between September 2010 and August 2014. All patients received ATL-DC vaccination as an initial series of 3 biweekly bilateral upper extremity injections of 2.5x10e6 ATL-DCs followed by up to 7 booster injections at 4-month intervals. Randomization allocated nine into the adjuvant TLR-7/8 agonist (resiquimod, 0.2% gel, 3M, applied to ATL-DC injection site days 0, 2, 4 post-DC injection) group, nine into the adjuvant TLR-3 agonist (poly-ICLC, 20mcg/kg IM, Oncovir, upper extremity, at time of DC injection) group, and five to the adjuvant placebo arm where patients received either carrier gel without resiquimod or IM saline injection. (Fig.1A, Supplementary Fig.1). All patients were followed for clinical evaluations, toxicity, survival, imaging changes, as well as in-depth systemic immune monitoring. Baseline patient characteristics are presented and segregated by treatment group in Table1 (see also Supplementary Data1). The median age was 45.3 (range 26.272.8) years, and 57% of the enrolled patients were male. Patients were enrolled prior to the 2016 update to the WHO classification of central nervous system tumors; 65% (n=15) had histopathological diagnoses of WHO Grade IV glioblastoma (now consistent with IDH wild-type glioblastoma), while 35% (n=8) of the patients were WHO Grade III (all of which would now classify as IDH-mutant astrocytoma or oligodendroglioma). Fifty-two percent (n=12) of patients were treated following recurrence, while 48% (n=11) were treated in the newly diagnosed setting. All patients were treated following surgical resection and standard-of-care treatment. The molecular characteristics of the patient tumors are outlined in Table1. Overall, MGMT methylation was seen in 35% (n=8), IDH mutations were observed in 35% (n=8, all grade III), and EGFR amplification was seen in 44% (n=10, all glioblastoma) of patients, consistent with the heterogenous population of malignant glioma patients. There were no statistically significant differences in age, sex, Karnofsky performance status, MGMT methylation status, pre- or postsurgery enhancing tumor volume, nor steroid administration at enrollment. No statistically significant differences were observed between the molecular characteristics, although the number of patients in each treatment group was small.

A Timeline of PBMC acquisition and analysis using CyTOF and/or RNAseq. V = vaccine, D = Day. (Figure created with the help of BioRender). B Schematic of differential gene expression analysis performed on pre-treatment and post-treatment PBMCs of indicated treatment groups. Differentially expressed genes (DEGs) in TLR agonist-treated groups are compared against their changes in the placebo group to identify DEGs specific to the TLR-agonist groups. C, D Enriched gene set terms in Gene Ontology Biological Process (C) or ARCHS4 TF Coexp (D) datasets that significantly overlap with the union of DEGs from ATL-DC + poly-ICLC and ATL-DC + resiquimod groups (P values, FDR-adjusted, two-sided fisher exact test). E Differential gene expression (pre vs. post-treatment fold change, in log2) of representative antigen presentation and IFN-related genes across treatment groups (P values, two-sided Welch t test). F Gene set enrichment score differences (pre vs. post-treatment, delta GSVA score) of representative IFN-related genesets across treatment groups (P values, two-sided Welch t test). G Heatmap of single-sample, gene set enrichment scores (GSVA) of type I and type II interferon genesets in pre-treatment, ATL-DC + placebo, ATL-DC+poly-ICLC and ATL-DC+resiquimod samples. The number of sample pairs analyzed in panels E and F are: ATL-DC+placebo, 5 pairs; ATL-DC+poly-ICLC, 8 pairs; ATL-DC+resiquimod, 8 pairs. The rectangular box in each boxplot represents the interquartile range (IQR), spanning from the first quartile (25th percentile, bottom of box) to the third quartile (75th percentile top of box). Inside the box, the median (50th percentile) is marked. The whiskers (shown as lines extending from the box) extend to the largest and smallest non-outlier values within 1.5 times the IQR, while outliers lie beyond the whiskers.

Overall, the addition of a TLR agonist-induced only Grade 1-2 treatment-related adverse events (TRAEs), and all adverse events reported resolved without further treatment or hospitalization (Table2). The most common TRAEs were rash (39%), fever (35%), and fatigue (26%; see Table2), and were more common in patients treated with resiquimod and poly-ICLC. 88.9% of patients who received resiquimod reported a temporary localized, cutaneous rash that resolved without further treatment. Other observed adverse events were not uncommon in the setting of postoperative central nervous system (CNS) tumor treatment. However, no serious adverse events (Grade 3-4) attributable to the treatment were observed. As such, the addition of a TLR agonist to ATL-DC vaccination in malignant glioma patients was found to be safe and tolerable.

The primary endpoint of this clinical trial was to evaluate systemic immune response changes induced by ATL-DC vaccination with and without TLR agonist administration. As such, we collected PBMCs at baseline (pre-treatment), one day after the vaccination (on treatment), and then following the completion of the treatment cycle (post-treatment) of each patient (Fig.1A). We aimed to understand how the adjuvant administration of TLR agonists modified the immune response in comparison with ATL-DC vaccination alone (placebo control).

We first performed paired bulk RNA-seq on patient-matched, pre-treatment and post-treatment PBMC samples that passed QC (see sample list in Supplementary Data1C). For each gene, we computed the difference between its expression in the pre- and post-samples of patients in each treatment group: ATL-DC+placebo (n=5 pairs); ATL-DC+poly-ICLC (n=8 pairs); ATL-DC+resiquimod (n=8 pairs); for brevity, we refer to them as placebo, poly-ICLC and resiquimod, respectively. To identify expression changes specific to the TLR agonist groups, we identified genes whose average upregulation in the TLR agonist pairs (poly-ICLC or resiquimod) were at least two-fold higher than the placebo pairs (Fig.1B, Supplementary Data2A, see Methods).

Genes upregulated in the TLR agonist groups were involved in antigen processing and were enriched with known interferon-stimulated genes (ISGs) (Fig.1CE, Supplementary Data2B, C). This observation was also confirmed by per-sample gene set enrichment analysis, where the TLR agonist-treated groups displayed higher enrichment of both type I and II interferon downstream gene sets compared to ATL-DC/placebo (Fig.1F, Supplementary Data2D, E). PBMC samples with higher absolute enrichment scores of interferon gene sets were dominated by post-treatment samples from both grade III and IV glioma patients in the TLR-agonist-treated groups (Fig.1G). The two TLR agonist-treated groups showed a largely similar trend in treatment-induced gene expression changes, which included a measurable increase in the expression of ISGs in the peripheral blood of malignant glioma patients. However, we noted that the resiquimod group had a more heterogenous response, which resulted in a lower degree of statistical significance compared to that of the poly-ICLC group.

We performed CyTOF on PBMC timepoints with a 27-marker heavy metal antibody-conjugated panel for 20 of the 23 patients where sufficient material was available (placebo, n=4 pairs; poly-ICLC, n=9 pairs; resiquimod, n=7 pairs; see Supplementary Data1C, 3A, 3B). The panel was selected to be able to broadly characterize different immune cell types, activation/effector, memory, and exhaustion phenotypes, with a bias towards T-cell relevant markers. The different immune cell type populations were visualized by the uniform manifold approximation and projection (UMAP) method (Fig.2A), which we broadly assigned to seven different major immune populations based off the normalized heatmap marker expression (Fig.2B).

A A UMAP projection of the pre- and post-treatment PBMC sample pairs from twenty patients (placebo, n=4 pairs; poly-ICLC, n=9 pairs; resiquimod, n=7 pairs). Clustering was performed with a random sampling of 5,000 cells from each patient. B Heatmap of normalized expression of all 27 cell markers within cell populations identified in the patient PBMCs. C, D Normalized expression of indicated markers in monocyte (C), or T cell populations (D) within the PBMC samples of patients from indicated treatment groups. P values, two-sided Wilcoxon rank sum test. E, UMAP projection of the PBMC-derived single cells (n=99,590). The immune subset associated with each cluster is inferred based on the clusters differentially expressed transcripts. Canonical markers of known immune subsets are shown. F, G Heatmaps showing the union of recurrent DEGs computed between ATL-DC treated samples (combined with placebo, resiquimod or poly-ICLC) and pre-treatment samples in the myeloid populations (F) or lymphocyte populations (G). Shown in the heatmaps are the log fold change values of the DEGs in each cell population grouped by their treatment groups. The number of sample pairs analyzed in C and D are: ATL-DC+placebo, 4 pairs; ATL-DC+poly-ICLC, 9 pairs; ATL-DC+resiquimod, 7 pairs. The rectangular box in each boxplot represents the interquartile range (IQR), spanning from the first quartile (25th percentile, bottom of box) to the third quartile (75th percentile the top of box). Inside the box, the median (50th percentile) is marked. The whiskers (shown as lines extending from the box) extend to the largest and smallest non-outlier values within 1.5 times the IQR, while outliers lie beyond the whiskers.

After 3 cycles of treatment, the post-treatment samples of patients in the TLR agonist groups showed a significant increase in the proportion of proliferating Ki67+CD14+ classical monocytes (Fig.2C, Supplementary Data3C). Such findings were corroborated by the increased monocyte fraction and CD14 transcript expression after ATL-DC+TLR agonist-treated samples (Supplementary Fig.2A, B, Supplementary Data3D). ATL-DC+TLR agonist treatment induced PD-1 expression in CD4 T cell population and increased the T-cell normalized expression of PDCD1 (the transcript that encodes PD-1 protein) and TCF7 (a marker of progenitor-like T cells) (Fig.2D, Supplementary Fig.2C). Moreover, expression of markers associated with irreversible T cell exhaustion, such as CD38 and CD3933,34, were also significantly reduced after ATL-DC+TLR agonist treatment (Fig.2D, Supplementary Fig.2D). Increased expression of PD-1 and decreased expression of CD38 and CD39 suggest the addition of the TLR agonists led to enhanced systemic T cell activity and cellular fitness in the patient.

To delineate the changes induced by ATL-DC and TLR agonist treatment in discrete peripheral blood immune cell subsets, we performed single-cell RNA-seq on selected patients at baseline and then following the completion of therapy. We analyzed two representative sample pairs from each cohort (placebo, poly-ICLC, and resiquimod) (Supplementary Data1C, 3E). We identified a total of twelve clusters from the total PBMC immune cell population and annotated these clusters based on differentially expressed gene markers in each cluster. From the initial clustering, we were able to identify multiple populations of CD4+ and CD8+ T cells, two populations of NK cells, three monocytic cell populations, B cell, and dendritic cells (type 2 conventional dendritic cells (cDC2) and plasmacytoid dendritic cells (pDCs), in accordance with the previous characterization of these cell types in peripheral blood (Fig.2E and Supplementary Fig.2E, F).

Differential gene expression analysis across the different lymphoid and myeloid populations revealed concordant upregulation of known ISGs (e.g. IFI6/35/44L, ISG15/20, IFIT3, IFITM1/3, GBP1/5, MX1, STAT1, and CXCL10) and antigen presentation-related proteasomes (PSMB9 and PSME2) in both TLR agonist sample pairs. The magnitude of induction was weaker in the paired PBMC samples obtained from the resiquimod group compared to the poly-ICLC group (Fig.2F, G).

Thus, our combination of high dimensional proteomics, bulk and single-cell RNAseq demonstrates how adjuvant TLR administration in conjunction with ATL-DC reproducibly increases the proportion of canonical CD14+ monocytes within the systemic blood circulation. This TLR agonist administration was also associated with enhanced T cell activity, coupled with decreased expression of CD38 and CD39 and their downstream T cell-suppressive adenosine pathway33,34,35. ATL-DC+TLR agonist-driven induction of ISGs across lymphoid and myeloid populations identified in our scRNAseq analysis corroborated our bulk transcriptomic analysis. Given the consistent changes observed with TLR agonist administration, we examined whether these systemic measurements correlated the observed progression-free and overall-survival differences between these patient populations to speculate on their contribution.

Median follow-up of patients treated on this clinical trial was 2.2 years after surgery, although the long-term survivors have now been followed for over 10 years. Median progression-free survival (PFS) was 8.1 months; and median overall survival (OS) was 26.6 months. Although this clinical trial was not designed or powered to detect effects of these treatments on survival between the treatment groups, there were noticeable differences in median survival between the treatments groups for both OS (placebo: 7.7 months, poly-ICLC: 52.5 months, and resiquimod: 16.7 months; log-rank P=0.017) and PFS (placebo: 5.5 months, poly-ICLC: 31.4 months and resiquimod: 8.1 months; log-rank P=0.0012) (Fig.3A). Because the trial included patients with both grade III and IV tumors, we stratified our analysis based on tumor grade. When we analyzed only the grade IV (GBM) patients, we observed a trend towards improved PFS (log-rank P=0.068) and OS (P not significant) (Fig.3B). Interestingly, for the IDH mutant/Grade III cohort, all four patients that received ATL-DC + poly-ICLC treatment are still alive at the data cutoff date (three of the patients have survival > 120 months and one > 112 months), and they have significantly longer OS and PFS compared to the other (n=4) grade III patients who received ATL-DC + resiquimod or ATL-DC alone where median OS was 15.73 months (Fig.3C).

AC Progression-free survival (PFS, top) and overall survival (OS, bottom) of all patients (A), patient subset with GBM (B), or grade III glioma (C) in indicated treatment groups. P values, log-rank test. D, E, Multivariate Cox proportional hazards analysis assessing the hazard ratios of tumor progression in TLR agonist treatment groups against placebo in all patients (D) or GBM subset (E) after adjusting for other clinical covariates (Tx_Group=treatment group, RecurNum=number of recurrences prior to ATL-DC treatment). In the forest plot, the squares are the hazard ratio (HR) estimates, the error bars are 95% confidence interval (CI) of the HR, the P value of each covariate is based on its Wald statistics, the P values are not adjusted. In D, the sample distribution in each covariate is Tx_Group: placebo=5, poly-ICLC=9, resiquimod=9; Grade: III=8, IV=15; MGMT_methylation: True=8, False=15. In E, Tx_Group: placebo=4, poly-ICLC=5, resiquimod=6. F, MR-computed volumes of post-treatment, recurrent tumors in indicated treatment groups. Treatment groups: Placebo (n=5), Resiquimod (n=8); Poly ICLC (n=9). P values, unpaired, two-sided Wilcoxon rank sum test.

We performed multivariate Cox proportional hazard (PH) analysis, adjusting for clinical variables that are significantly correlated with OS or PFS as a single variable (tumor grade, MGMT methylation status, and number of recurrences). Our analysis confirmed that patients in the poly-ICLC and resiquimod treatment groups had a lower risk of progression that was independent of grade, MGMT methylation, and number of recurrences (Fig.3D). Risk of death was significantly lower in the poly-ICLC group, while the resiquimod group showed a similar trend that was not statistically significant (Supplementary Fig.3A). In the GBM patient subset, TLR agonist treatment also significantly lowered risk of recurrence, but not risk of death (Fig.3E, Supplementary Fig.3B).

To determine whether this treatment directly impacted tumor volume, MR imaging was performed, and contrast-enhancing tumor volume was quantified over time. We noted that the rate of tumor volume increase over time in the ATL-DC/placebo treatment cohort was higher than in the ATL-DC/resiquimod treatment (p=0.022) and the ATL-DC/poly-ICLC treatment groups (P<0.001; Fig.3F). Anecdotally, we observed an increased T2/FLAIR MRI signal after completion of the vaccine series in two of the four long-term survivors who received ATL-DC/poly-ICLC (Supplementary Fig.3C, D), although such findings are potentially confounded by prior radiation therapy, and thus we cannot ascribe such changes solely to the vaccine/TLR agonist intervention. However, this increased post-vaccination T2/FLAIR on MRI was not seen in patients who did not receive poly-ICLC (not shown).

Finally, we asked if the magnitude of interferon pathway induction by the adjuvant TLR agonist treatment directly correlated with OS or PFS. This could allow for the use of an interferon activity score as a biomarker for productive anti-tumor immune responses following ATL-DC immunotherapy. To this end, we stratified the patients by the median GSVA score of the HALLMARK INTERFERON GAMMA RESPONSE gene set in post-treatment PBMC samples. We confirmed that patients whose post-treatment samples displayed higher interferon gene set scores (median) had longer OS and PFS than those with lower scores (Fig.4A, Supplementary Fig.4A). Separate analyses on the grade IV (GBM) and grade III glioma patients showed a concordant trend but with a lower degree of statistical significance; this was likely caused by the small sample sizes. Notably, multivariate Cox PH analysis strongly suggested that the interferon gene set score is a significant predictor of tumor recurrence (Fig.4B, C) and death (Supplementary Fig.4B), even after adjusting for other potentially confounding clinical variables. To ensure that the correlation is not specific to this single gene set, we confirmed that the gene set scores of other interferon gene sets after ATL-DC treatment are also positively correlated with the patients OS and PFS (Supplementary Data4A, B). Such findings can be confirmed in larger subsequent studies.

A Kaplan-Meier progression-free survival curves of all patients (left), GBM (center), and Grade III glioma subsets (right) stratified by their HALLMARK_INTERFERON_GAMMA_RESPONSE GSVA scores in their post-treatment PBMCs. P values, log-rank test. B, C Multivariate Cox proportional hazards analysis assessing hazard ratios of tumor progression in patients with high HALLMARK_INTERFERON_GAMMA_RESPONSE GSVA score in all patients (B) or GBM subset (C) after adjusting for other clinical covariates. In the forest plot, the squares are the hazard ratio (HR) estimates, the error bars are 95% confidence interval (CI) of the HR, the P value of each covariate is based on its Wald statistics, the P values are not adjusted. In B, the sample distribution in each covariate is GSVA score (post-Tx):

Taken together, these data suggest that the addition of TLR agonists to ATL-DC vaccination shifts towards an interferon-induced immune response in both lymphoid and myeloid cells. Poly-ICLC and resiquimod appear to upregulate similar ISGs but with different magnitude. Enhancing systemic ISG-signaling may reflect an environment more favorable towards the generation of an antitumor immune response and clinical effects.

View post:

TLR agonists polarize interferon responses in conjunction with dendritic cell vaccination in malignant glioma: a ... - Nature.com

Related Posts
Tags: