Immune cell infiltration patterns of TIME
We applied the ssGSEA method to assess the infiltration abundance of 24 immune cell types for 1,821 HCC samples. The correlation between these immune cells was presented in Fig. S1A. It was observed that several pairs strongly correlated with immune cells, such as T cell-cytotoxic cells, B cell-T cells and macrophage-immature DC cells. Subsequently, we performed a consensus cluster analysis, in which all HCC samples were initially grouped into different k (k = 2-9) clusters. The CDF curves of the consensus score and PAC value suggested that the optimal division was achieved when k = 3 (Fig. 2A-C). The same result was obtained from NbClust (Fig. S1B). The three clusters of samples were separated from each other on the two-dimensional principle component plot (Fig. 1D). Thus, based on the infiltration profiles of 24 immune cells in TIME, 1,821 HCC samples were finally classified into three TIME phenotypes (TIME-1 = 721, TIME-2 = 530, TIME-3 = 570). As shown in Fig. 1E-F, TIME-1 presented as an immune deficiency phenotype due to the lowest infiltration in almost all immune cells. On the contrary, it was found that TIME-3 had a significantly higher infiltration level in the majority of immune cells, especially adaptive immune cells (e.g. CD8+T cells and B cells), suggesting that TIME-3 was associated with immune activation and superior cytotoxic potential. TIME-2 was in an intermediate status of immune infiltration between TIME-1 and TIME-3, and was characterized by higher infiltration immunosuppressive cells that contain Treg and TH17. In addition, it was also observed that there was a richer infiltration in some innate immune cells that contain DC and NK cells in TIME-2. Two other different algorithms were further applied: CIBERSORT and MCP-counter. The results shared a consistent immune infiltration pattern with the ssGSEA method in HCC (Figs. S1C–S1D).
In order to ensure the reproducibility and robustness of the TIME phenotypes derived from the GEO cohort, we further conducted the IGP statistical method to validate the TIME phenotypes in the TCGA cohort. The three phenotypes were highly consistent between the discovery and validation cohorts, with the corresponding IGP values at 95.6%, 93.3% and 94.7%, respectively, and the three phenotypes were deemed to be of high-quality due to the statistically significance (all, P<0.001). The immune cells infiltration patterns in the TCGA cohort exhibited a very similar pattern of immune infiltration to the GEO cohort (Figs. S1E–S1F). Furthermore, the NbClust also indicated that the three clusters configuration was “optimal” in the TCGA cohort (Fig. S1G).
Specific functional pathways of each TIME phenotype
We further explored the specific functional status and biological mechanisms of each phenotype in the GEO cohort (Fig. 2G and Table S2). TIME-1 was prominently enriched in pathways, such as Myc targets, G2M checkpoint, and DNA repair. These pathways were remarkably associated with MKI67, and thereby with proliferation (Fig. 2H). Furthermore, it was observed that the immune-relevant pathways were significantly downregulated in TIME-1 (Table S2). Combined with the lack of immune cell infiltration, we inferred that TIME-1 may present an immune deficiency phenotype. On the contrary, TIME-3 enriched intensive pathways related to immune activation, and these pathways had a remarkably positive association with the immune score assessed through the ESTIMATE algorithm [27] (Fig. 2I), suggesting that TIME-3 may exhibit a state of immune activation. Notably, TIME-2 was significantly upregulated in metabolic-relevant pathways. It was observed that there was significantly negative correlation between the immune score and these specifically activated pathways in TIME-2 (Fig. 2I). Moreover, TIME-2 was rich in immunosuppressive cells (e.g. TH17 cell and Treg), which is known from the previous descriptions. Hence, it was concluded that TIME-2 may present as an immune-suppressed phenotype. The KEGG results was in accordance with the above (Fig. 2J and Table S3), and similar results were achieved in the TCGA cohort (Fig. S2A–S2B and Table S4-S5). Overall, we identified three TIME phenotypes in HCC showing significantly different immune cell infiltration and biological functions, respectively. TIME-1 was categorized as an immune-deficiency phenotype, characterized by immune cell depletion and proliferation; TIME-2 was categorized as an immune-suppressed phenotype, characterized by being in immunosuppressive states; TIME-3 was categorized as an immune-activated phenotype, characterized by abundant leukocytes infiltration and immune activation.
The clinical value of TIME phenotypes
We explored the prognostic value in the two independent cohorts (TCGA-LIHC and NCI cohort), which contained the complete overall survival (OS) and relapse free survival (RFS) information. The Kaplan-Meier analysis of both OS and RFS exhibited that HCC patients had an increasingly favorable prognosis from TIME-1 to TIME-3 (Figs. 3A-D). Furthermore, we assessed the sensitivity to sorafenib in TIME phenotypes by the pRRophetic package. TIME-1 was found to be more sensitive to sorafenib than the other phenotypes (Fig. 3E and Fig. S3A). Notably, TIME-1 exhibited the highest expression in sorafenib related target genes (Fig. 3F). Therefore, this suggested that patients in TIME-1 may benefit from sorafenib the most. As formerly mentioned, TIME-3 had higher levels of immune cell infiltration abundance (e.g. CD8+ T cells). Hence, we speculated that patients in TIME-3 might be more responsive to immunotherapy. The TIDE algorithm was applied to infer the response to immunotherapy. As respected, TIME-3 had a higher response rate than the other phenotypes (Fisher’s exact test: P=0.009) in the GEO cohort (Fig. 3G), and consistent results was found in the TCGA cohort (Fisher’s exact test: P=0.048) (Fig. S3B). We also utilized the submap algorithm to compare the similarity of the expression profiles between the three TIME phenotypes and 47 previous melanoma patients with detailed immunotherapeutic information, and revealed that patients in TIME-3 were more responsive to anti-PD1 treatment (Bonferroni corrected P=0.008)[28] (Fig. 3H). The submap analysis on the TCGA cohort also achieved similar results (Fig. S3C).
Potential extrinsic immune escape mechanism
To further research the regulatory mechanisms of the TIME phenotypes, we focused on the TCGA cohort, which possessed multiple omics data and comprehensive clinical data.
We firstly investigated the extrinsic immune escape mechanisms. Previous studies indicated that extrinsic immune escape may include three major aspects: absence of leukocytes, presence of immunosuppressive cells, and release of abundant immunosuppressive cytokines [29, 30]. As described above, TIME-1 was characterized by deficient immune cell infiltration and then lack of immune mediated elimination. TIME-2 was characterized by higher levels of immunosuppressive cells (e.g. TH17 cell and Treg; Figs. S4A–S4B), which indicated a role of immunosuppressive cells in immune escape. In addition, TIME-2 lacked immune active cells (e.g. CD8+T cells). Therefore, it was speculated that TIME-1 and TIME-2 probably reflect the absence of recruitment or activation of innate immune cells, inducing failure of adaptive anti-tumor immune responses. The low expression of molecules in TIME-1 and TIME-2, such as AIM2, TLR7 and TLR8, was potentially involved in priming of innate immunity, which further confirmed our speculation (Fig. 4A). TIME-3 was characterized by the presence of abundant innate and adaptive immune cells. In addition, TIME-3 had a higher expression of both immunostimulatory and immunoinhibitory cytokines, while these cytokines were all relatively low in TIME-1 and TIME-2 (Fig. S4C). These results implied that high concentrations of immunoinhibitory cytokines might contribute to the immune escape in TIME-3. It was noteworthy that the differential expression of cytokines in these three phenotypes could not be explained by the CNV and mutation frequency (all, P>0.05; Table S6). Overall, our analysis revealed that the extrinsic immune escape mechanisms of three phenotypes were lack of tumor-infiltrating leukocytes, increased immunosuppressive cells, and rich in immunoinhibitory cytokines, respectively.
Potential intrinsic immune escape mechanism
We further explored the potential intrinsic immune escape mechanisms in two major facets: tumor immunogenicity and the expression level of immune checkpoint molecules [31]. First, a series of elements associated with tumor immunogenicity were estimated: genomic instability degree, neoantigen burden, genomic viral content, CTA level, and tumor antigen presentation competence [32]. The first four elements were the main source of tumor-specific antigens. It was found that the genomic instability degree presented a decreasing trend from TIME-1 to TIME-3 (HRD, AS and MSI; all, P<0.05; Figs. 4B-D). Similarly, TIME-3 had a lower TMB, when compared to TIME-2 (P<0.05, Fig. 4E). In terms of genomic viral content, TIME-3 had more HBV read counts than TIME-1 (mean viral read counts, 44.365 vs. 20.907, P<0.05; Fig. S4D), in contrast to the HCV read counts (mean viral read counts, 12.931 vs. 16.111, P<0.1; Fig. S4E). Of note, the neoantigen burden was relatively lower in TIME-3, although the statistical difference among the three phenotypes in SNV or indel neoantigens was not reached (Figs. S4F–S4G). There was also not distinct variation among the CTAs overall expression of the three phenotypes (Fig. S4H). Overall, these above indicators had little difference in the TIME phenotypes. We further investigated the tumor antigen presentation capacity of the three phenotypes, and observed that TIME-3 had the highest APS and MHC-related molecules expression level, as opposed to TIME-1 (all, P<0.01; Fig. S4I and Fig. 4F), which was consistent with the CYT value and BCR/TCR diversity (all, P<0.05; Figs. 4G-I and Figs. S4J-S4K). This indicated that defective tumor antigen presentation capacity may be an intrinsic immune escape mechanism for TIME-1.
Subsequently, the genomic alterations of 62 immunomodulators were further summarized within the three TIME phenotypes (Fig. 4F). It was found that TIME-3 had higher costimulatory and coinhibitory molecules than the other phenotypes. This suggested that TIME-3 may overexpress the immune checkpoint molecules (such as CTLA4, CD274 and PDCD1; all BH-adjusted P<0.001) to evade the immune elimination after immune activation. All somatic mutations and CNVs did not significantly differ among the three phenotypes, and most of immunomodulators exhibited rare somatic mutations and CNVs, which indicated that the mutations and CNVs in the immunomodulators had little effect on TIME. Of note, DNA methylation negatively regulated many immunomodulators, such as CD27, CD226 and TNFSF8, implying epigenetic silencing. The associations were shown between miRNAs and immunomodulators for each TIME phenotype (BH corrected P-value <0.05; Fig. 4J), such as miR-17 negative correlated with CD274 and PDCD1LG2. It was also observed that the three phenotypes shared a common TNFSF4 negative regulator: miR-204. Compared with the mutation and CNV, methylation modification and miRNA sponges played leading roles in regulating the immunomodulators, indicating a new perspective for the development of immune checkpoint inhibitors.
Genomic alterations of the three TIME phenotypes
The investigators separately determined the SMGs among the three phenotypes using MutSigCV (Fig. 5A). All SMGs had mutation rates greater than 5% in three phenotypes. Among these three TIME phenotypes, the common SMGs (including TP53, CTNNB1 and ALB) had the top three significant MutSigCV q-value and frequent mutation rates, indicating that the mutation of TP53, CTNNB1 and ALB was broad in HCC. Additionally, the three phenotypes also displayed distinct SMGs, such as RB1, ACVR2A and CREB3L3 were SMGs of the three phenotypes, respectively. Besides, two newly identified SMGs, namely, BRD7 and RASA1, were classified as tumor suppressor genes, and these were associated with chromosome remodeling and cell proliferation[33, 34]. Based on the NMF, we isolated the mutation signatures of each phenotype. The age-related mutational processes (spontaneous deamination of 5-methylcytosine for signature 1 and unknow aetiology for signature 5) were prevalent in three phenotypes (Fig. 5B). TIME-1 had the least proportion than the other phenotypes (Fig. 5C). In addition, the three phenotypes also shared a common mutation profile (signature 22) associated with exposures to aristolochic acid. This may be associated with high-proportioned Asian patients in three phenotypes (Fig. S5A), and the aristolochic acid was mainly derived from herbal drugs of traditional Asian medicine[35]. Notably, signature 24, which represented the mutational pattern related with aflatoxin, was identified only in TIME-1, and possessed the maximum proportion (40.4%) (Fig. 5C). This implied that HCC patients in TIME-1 were more likely to be exposed to aflatoxin.
Tumor ploidy was estimated by ABSOLUTE, suggesting that a larger scale of HCC presented genome doubling, and that the doubling pattern was more frequent in TIME-1 compared with the other phenotypes (P=0.018, Fig. S5B). As shown in Fig. 4D-F, the FGA, FGG and FGL in TIME-1 were significantly higher than the other phenotypes, which might promote the cell proliferation and immune escape[36]. We further applied GISTIC 2.0 to delineate the significant focal copy number alterations of each phenotype (Fig. 5G and Table S7). CNVs that were recurrent in TIME-1 contained the focal amplification of 8q24.21 (MYC, ANXA13) and 13q34 (CDC16, TFDP1), and the focal loss of 14q22.1 (SAV1). Recurring focal arms CNVs in TIME-2 included the only amplification of 6p21.1 (VEGFA), and the loss of 13q13.3 (CCNV1). The genes on the focal loss arms of TIME-1 and TIME-2 were mainly associated with chemokines and cytokine through the GO annotation. Hence, the loss of these genes may contribute to the low immune infiltration of TIME-1 and TIME-2. TIME-3 exhibited the focal amplification that involved 8q24.12 (MTBP) and the focal deletion that involved 5q13.2 (TERT) and 10q23.31 (PTEN). These phenotype-special CNVs may play a crucial role in the biological features of the three phenotypes.
Furthermore, the combination of mutation and CNV data revealed the frequent alterations in different pathways (Fig. 5H). It was found that some genome alterations distributed evenly in these three phenotypes, such as TP53, CTNNB1 and ALB. In addition to these common alterations, we also observed the diverse alteration patterns in the pathways among three phenotypes. In TIME-1, cell cycle regulatory factor CDKN2A mutated in 12% of the cases. In TIME-2, VEGFA and its downstream genes PIC3KA and PTEN were frequently altered, and all of which were known to activate the PI3K pathway. APOB consumes an abundant cellular energy to facilitate very low-density lipoprotein (VLDL) secretion, and we found that APOB was frequently altered in 19% of the cases for TIME-2. Overall, the diverse genomic alteration preferences in three phenotypes might contribute to shape the TIME, and lead to differences in immune cell infiltration.
Methylation modification and regulation of the TIME phenotypes
As tumor cells divide, the loss of global methylation levels (GMLs) can result in chromosomal instability, and affect immune cell infiltration [37-39]. The GMLs in TIME-3 was significantly higher than the other phenotypes (P<0.001, Fig. 6A). Furthermore, there was a positive correlation between tumor-infiltrating CD8+ T cells and GMLs (Fig. 6B). In contrast, the decrease in GML might promote tumor cell proliferation (Fig. 6C). Subsequently, we identified 31, 25 and 39 ESGs in TIME-1, TIME-2 and TIME-3, respectively (Table S8 and Figs. 6D-E). It was noted that CPS1, FURIN and PHYHD1 were shared by the three phenotypes. As a liver-specific enzyme, CPS1 can facilitate cell division by instituting the pyrimidine synthesis pathway[40]. In TIME-1, FOXD4 was marked as epigenetically silenced in 85% of the cases, and its methylation silencing may lead to immune system dysfunction and tumor proliferation[41] (Fig. 6F). Tumor suppressors L1TD1 and PARP6, also the specific ESGs of TIME-1, and their methylation may promote tumor proliferation and low immune infiltration [42, 43] (Figs. 6G-H). SEMA3B belonged to the ESG of TIME-2, and its methylation can accelerate the progression of HCC[44]. FER1L is an another ESG of TIME-2, which can activate the PI3K/AKT pathway, further leading to the formation of the immunosuppressive microenvironment[45]. These results suggested that ESGs exhibit diversity among the three phenotypes, which may be involved in shaping the TIME, and these specific ESGs may also be potential therapeutic targets.
A robust prognostic and immunotherapy signature: TIME index
We identified 98 phenotype-related DEGs (Table S9). These genes, which significantly varied within the three phenotypes, possibly contributed to form the heterogenous TIME of HCC. In addition, many genes have been reported to be critical in immune response, such as CD27, CD8A, GZMA and IL7R [46-49]. The GO and KEGG annotation also displayed intensive immune phenotypes (Figs. S6A-S6B, Table S10-S11). Based on these DEGs, the ssGSEA was performed to obtain the TI of each patient. The TI presented a gradual increase from TIME1 to TIME3 (Figs. 7A-B). HCC patients with low TI were mainly distributed in TIME-1 (Fig. 7C). According to the optimal cut-off determined by the survminer package, we classified HCC patients into high and low TI groups. As expected, patients in the high TI group exhibited a tendency to better outcomes in the two independent cohort (Log-rank P<0.001, Figs. 7D-E). Besides, the multivariable Cox regression revealed that the TI was an independently prognostic factor in HCC (P=0.018, Table S12). We further assessed the TI of 10,121 patients that involved 33 differing types of cancers. Obvious TI diversity was observed in different cancers (Fig. 7F), which indicated that heterogeneous immune infiltration existed not only within the tumor, but also between tumors. The survival analysis for pancancer indicated that patients with a higher TI had a better prognosis (Log-rank P<0.001, Fig. 7G) and that the TI could independently affect the prognosis in the multivariable Cox regression (P=0.006, Table S12). Furthermore, it was found that 24 of 33 cancers presented a statistical significance in the Kaplan-Meier analysis (Fig. S7), and univariate Cox’s regression indicated that the TI was a protective factor in many different tumors (Fig. 7H).
We further explored the predictive ability of the TI for immunotherapy. In order to determine the predictive power, we also computed the response prediction of 11 other known biomarkers. The area under the ROC curve (AUC) was used as the quality metric of prediction. We found that the TI exhibited robust predictions in the six cohorts (Fig. 7I). Especially in the Riaz et al. cohort, the TI reached the highest prediction accuracy with AUC = 0.955. It was also observed that although the AUC value of TI in the Liu et al. cohort was less than 0.7, this was still the highest, when compared to the other markers. In addition, the TI displayed the highest mean AUC value compared with the other biomarkers (Fig. 7J). Hence, the present work strongly suggests that the TI was a potential and robust biomarker for the prognosis and clinical response assessment of immunotherapy.
In addition, to advance clinical application, we developed a R package termed “TIME” in the GitHub website (https://github.com/Zaoqu-Liu/TIME). The pipeline could classify single patient into one of these three subtypes and calculated the TI.