The composition of the intestinal microbiota regulates susceptibility to immune checkpoint inhibitor-induced colitis
To understand how immune checkpoint perturbation impacts the colonic immune system, we administered anti-CTLA-4/anti-PD-1 combination therapy to wild type (WT) Balb/C mice. Although checkpoint inhibition alone triggered systemic immune activation with induction of splenomegaly, there was no change in colonic mass of treated with immune checkpoint inhibitors (CPIs) (Figs. 1A and B). Since intestinal microbiota influences colitis susceptibility in cancer patients treated with CPI 21, 22, 23, 24, we investigated whether altering the intestinal microbiota impacted colitis susceptibility. To test this hypothesis, we transplanted the caecal microbiota harvested from TRUC mice, that are colonized with a pro-inflammatory microbiota 25, 26 to WT mice. Although faecal microbiota transplantation (FMT) by itself did not induce colitis, subsequent challenge with combination anti-CTLA-4/anti-PD-1 therapy did (Figs. 1A and B). We observed increased colon mass and failure to gain weight gain in recipients of combination anti-CTLA-4/anti-PD-1 and FMT (Figs. 1A-C). Histological assessment of the colon demonstrated relatively mild changes in mice receiving combination CPI and FMT, including although features consistent with reports of CPI-colitis in patients, with increased lymphocytes in the colonic lamina propria, increased intraepithelial lymphocytes and increased epithelial apoptosis (Fig. 1D and Supplementary Figs. 1A and B). Colonic lymphocyte expansion was corroborated by flow cytometry with increased infiltration with both CD4+ and CD8+ T cells in the lamina propria of mice treated with combination anti-CTLA-4/anti-PD-1 and FMT (Figs. 1E-G).
Neutrophil accumulation in the colon is a key feature of CPI-colitis 27, therefore, we also evaluated the colonic myeloid compartment. There was also significant accumulation of CD11b+ Gr-1high neutrophils and Ly6Chigh MHC class II− inflammatory monocytes in the colon of mice treated with FMT and CPI (Figs. 1H and I and Supplementary Figs. 2A-C). Disease could also be induced in C57BL/6 mice (Supplementary Fig. 2D), indicating that this phenomenon was mouse strain independent.
Changes in the intestinal microbiota induced by FMT were recorded using 16S rRNA amplicon sequencing. There was no significant change in α-diversity following FMT (Shannon index 2.19 in control vs 2.40 in FMT group P=0.24, Supplementary Fig. 3A); however, β-diversity was significantly different between control and FMT groups (P=0.038, Supplementary Fig. 3B). At phylum level, there were no differences between control mice or FMT recipients (Supplementary Fig. 3C). However, at family level, Rikenellaceae, Prevotellaceae and Desulfovibrionaceae were more abundant in the FMT samples compared to untreated controls, while Tannerellaceae and Muribaculaceae were significantly reduced (Supplementary Fig. 3D). At genus level, Alistipes and Rikenella RC9 gut group were significantly more abundant in FMT samples. By contrast, members of the Parabacteroides and Muribaculaceae genera were significantly more abundant in control samples (Supplementary Fig. 3E).
Transcriptomic profiling demonstrates colonic epithelial dysfunction, interferon signalling and cytotoxicity in CPI-colitis
To investigate the immunopathology of this model of CPI-induced colitis, we analysed gene expression changes in the distal colon using bulk RNA-sequencing. There were only minor transcriptional changes observed in mice treated with CPI only (Fig. 2A). However, following FMT, transcriptomic variation was also moderate, although we did observe significant induction (FDR < 0.05) of genes involved in humoral immunity, such as the activation-induced cytidine deaminase gene (Aicda), which plays a critical role in somatic hypermutation and class switching in B cells activated in response to microbial challenge 28, and over-expression of immunoglobulin chains, including selective variable regions (Ighg2b, Igkv8-21, Igkv4-61) (Supplementary Fig. 4A). Biological pathway analysis (IPA, QIAGEN) identified three causal networks associated with the gene expression changes induced by FMT that were merged into a single network, additionally identifying IFNγ and TNFα as key nodes (Supplementary Fig. 4B).
Transcriptomic changes were more pronounced in FMT and combination CPI treated mice, with a greater number of differentially expressed genes (DEGs) and a greater magnitude of the differences, including 385 transcripts significantly affected (FDR<0.05), of which 258 showed at least doubled expression levels and 22 at least halved (Fig. 2A and B). The most significantly upregulated transcripts encoded proteins involved in epithelial barrier function, extracellular matrix regulation and anti-microbial responses. The most prominent upregulated transcripts included multiple members of the late cornified envelope (LCE) gene cluster and keratin family genes (Fig. 2C). Their expression is dysregulated in barrier surface diseases in response to tissue injury and infection 29, 30, 31. Expression of other genes involved in epithelial function, including defensins, aquaporins and were also dysregulated, as is observed in conventional IBD 32. Other upregulated genes included interferon stimulated genes (e.g. Nos2, Isg15, Ifit2, Gbp3 and Gbp7), molecules involved in cell-mediated cytotoxicity (Gzma, Gzmb, Tnf), proteases and their inhibitors (Ctsc, Srgn, Serpina12, Serpina3c) and molecules involved in antigen processing/presentation (Cd74 and multiple MHC molecules) (Figs. 2B and C).
Biological pathway analysis identified significant enrichment of biological processes, such as interferon signalling, T cell exhaustion, dendritic cell maturation, death receptor signalling and activation of phospholipase C and p38 MAP kinase in CPI-colitis (Fig. 2D). Causally affected processes (Downstream Effects Analysis, IPA, QIAGEN) predicted to be significantly activated (z-score>2, FDR<0.05) in CPI-colitis included systemic autoimmune syndrome, anti-microbial response, activation of leukocytes, cytolysis, and activation of cytotoxic T cells (Fig. 2E). Causal network analysis of the most enriched pathway (systemic autoimmune syndrome) identified a complex interplay of cytokines, chemokines, antigen processing/presenting molecules, cytotoxicity molecules, myeloid molecules, and transcription factors (Supplementary Fig. 5A).
We found multiple predicted upstream regulators of the molecular changes observed in the colon in CPI-colitis, including cytokines (IFNγ, IL1β, IL6, IL21, IL27 and TNFα), microbial products/TLR agonists (LPS, TLR3, TLR4, CpG, MyD88) and transcription factors (NFκB, CEBPB, NFATC2) (Fig. 2F). Immunosuppressive drugs, including etanercept (an anti-TNFα agent), sirolimus, and the immunosuppressive cytokines IL10 and SOCS1 were predicted inhibitors of the gene expression changes observed in CPI-colitis. The cytokine predicted to be most highly activated in CPI-colitis was IFNγ (z-score= 6.6, FDR=2.06x10−23) (Fig. 2F). Network analysis of its interactions with the DEGs identified a range of biological processes regulated, including antigen presentation, oxidative stress, chemokine induction, JAK/STAT signalling, and proteasome activation (Supplementary Fig. 6A).
To determine whether this model of CPI-colitis mirrored aspects of human disease, we evaluated the similarity between the transcriptional differences observed in our model and those reported in cancer patients developing combination CPI-colitis 33. Consistent with our model recapitulating gene expression changes in human CPI-induced colitis, gene set enrichment analysis (GSEA) demonstrated that the mouse homologs of the most significantly up-regulated genes probed in patients were among the most over-expressed ones in our model (Supplementary Fig. 7A).
High resolution single cell transcriptomics reveals colonic lymphocyte remodelling and emergence of polyfunctional, cytolytic lymphocyte responses in CPI-induced colitis
To further probe immune mechanisms of CPI-induced colitis, we performed single cell RNA-sequencing (scRNA-seq) from FACS purified live CD45+ lymphocytes from the colons of mice with CPI-colitis and control mice. Cluster analysis demonstrated shifts in the proportional abundance of colonic lymphocyte populations, including expansion of T cell clusters 1, 4, 6, 24, B cell clusters 3 and 8, and ILC cluster 22 (Figs. 3A-D, Supplementary Fig. 8A).
Analysis of the main lymphocyte lineages identified remodelling of the transcriptome in T cells, B cells and ILCs (Figs. 3B-D). There was significant upregulation (Bonferroni corrected P-value <0.001) of Ifng, Gzmb, Gzma, Nkg7, and the chemokines Ccl3, Ccl4 and Ccl5 in T cell compartment in CPI-induced colitis (Fig. 3B). In T cells, Canonical Pathway Analysis (IPA, QIAGEN) identified significant enrichment of biological processes including TH1 and TH2 activation, cytotoxicity, and T cell exhaustion in CPI-colitis (Fig. 3B). In B cells, there was upregulation of transcripts involved in protein synthesis and endoplasmic reticulum stress, such as Calr, Hspd1, Hspa5, in keeping with B cells adopting a secretory state in CPI-induced colitis (Fig. 3C). In B cell clusters the most upregulated biological pathways were antigen presentation pathways, and pathways linked to immune-mediated inflammatory diseases, including SLE and GvHD (Fig. 3C). In ILCs there was also upregulation of cytotoxicity molecules, such as Gzma, Gzmb, Gzmc and Prf1, and the cytokines/chemokines Il22 and Ccl3 (Fig. 3D). In ILCs, crosstalk between NK cells and DCs, and cytotoxicity signalling pathways were enriched (Fig. 3D).
Cluster-specific analysis of lymphocytes in CPI-colitis demonstrated expression of Ifng across multiple T cell clusters, including Cd4+ clusters 4, 6 and 9, Cd8+ clusters 24 and 25, and ILC clusters 16 and 22 (Fig. 3E). These clusters also expressed high levels of cytotoxicity molecules (Gzma, Gzmb, Prf1 and Nkg7), chemokines (Ccl3, Ccl4 and Ccl5), both subunits of the IL12 receptor heterodimer (Il12rb1 and Il12rb2), and Cxcr6, Tnfrsf9 and Stat1 (Fig. 3E and Supplementary Fig. 8B).
IFNγ producing CD4 + and CD8+ T cell populations co-express multiple pro-inflammatory cytokines and cytotoxicity molecules in CPI-colitis
Given the prominent IFNγ footprint observed in CPI-colitis, we further probed the transcriptome of Ifng-expressing lymphocytes in CPI-colitis in comparison with their Ifng-expressing counterparts in control mice. As well as being expanded in CPI-colitis, Ifng-expressing lymphocytes co-expressed Gzmb (Fig. 4A and B). In comparison with Ifng-expressing CD4+ T cells in control mice, the top 20 most highly upregulated transcripts in Ifng-expressing CD4+ T cells in CPI colitis, included chemokines (Ccl3, Ccl4, Ccl5, Ccl9), other cytokines (Il22, Il13, Il17f, Il10, Spp1), and cytotoxicity molecules (Gzma, Gzmb, Prf1, Nkg7) (Fig. 4C). In CPI colitis, Ifng-expressing CD8+ T cells upregulated a similar pattern of transcripts, with increased expression of other cytotoxicity molecules, chemokines, and cytokines among the most highly upregulated 20 transcripts (albeit with a more limited repertoire of cytokines than CD4+ T cells, Fig. 4C). The most highly upregulated transcript in Ifng-expressing ILCs in CPI-colitis was Il22, and cytotoxicity molecules were again among the most highly expressed transcripts in these cells (Supplementary Fig. 9A).
To determine whether these changes were observed at protein level, we performed flow cytometry. An increased proportion of colonic T cells co-expressing IFNγ and cytotoxicity molecules, such as granzyme B and perforin was observed in CPI-colitis, especially in CD4+ and CD8+ T cells (Figs. 4D and E, Supplementary Fig. 9B and C), and to a lesser extent CD3− IL-7R+ cells (Supplementary Fig. 9D).
We investigated which biological pathways were enriched in Ifng-expressing lymphocytes using the Canonical pathways tool (Ingenuity). The top 20 most enriched, overlapping canonical pathways in Ifng-expressing CD4+ T cells in CPI colitis in comparison with Ifng-expressing lymphocytes in control mice included TCR and co-stimulatory molecule activation, effector lineage activation (Th1, Th2 and Th17 pathways), coronavirus signalling pathways and metabolic reprogramming, including oxidative phosphorylation (Fig. 4F and Supplementary Fig. 10A). Other important pathways that were significantly activated in these cells included ICOS-ICOSL signalling in Helper T cells (z-score =2.53, P<3.3 x10−5), OX40 signalling pathway (z-score =1.89, P<3.3 x10−5) cross talk between DCs and NK cells (Z-score 2.53, P<0.0013) and the IL23 signalling pathway (z-score=2.45, P<0.032). There was a very similar pattern of Canonical Pathways activated in Ifng-expressing CD8+ T cells in CPI colitis in comparison with Ifng-expressing lymphocytes in control mice (Supplementary Fig. 10B).
To determine whether IFNγ was functionally important in CPI colitis we administered neutralizing anti-IFNγ monoclonal antibodies, or isotype matched control antibodies, to mice during induction of CPI-colitis. Antibodies were administered simultaneously at the same time as combination CPI at weeks 0, 1 and 2. In keeping with IFNγ playing an important role in CPI colitis, neutralization of this cytokine significantly reduced colon mass in anti-IFNγ treated animals (Fig. 4G).
IFNγ producing lymphocytes have increased expression of co-stimulatory and immune checkpoint molecules
To further analyse the phenotype of IFNγ producing lymphocytes in CPI-colitis, we examined their expression of co-stimulatory molecules, co-inhibitory molecules, chemokine receptors and gut homing integrins. In both CD4+ and CD8+ T cells, there was an increase expression of co-stimulatory molecules in IFNγ producing lymphocytes in CPI-colitis. In CD4+ T cells this included classical co-stimulatory molecules, such as Cd28 and Cd2, and numerous members of the TNF receptor superfamily, including Tnfrsf4, Tnfrsf18 and Tnfrsf9 (Fig. 5A) There was a similar pattern of co-stimulatory molecule upregulation in CD8+ T cells, although CD27 was the most upregulated receptor in these cells (Fig. 5A). In CPI-colitis, IFNγ producing lymphocytes also upregulated co-inhibitory receptors. In CD4+ T cells, the most upregulated checkpoints molecules, included Ctla4 (fold change = 2.2, FDR=7.0x10−46), Hacvr2 (Tim-3, fold change 1.8, FDR=3.7 x10−58) and Lag3 (fold change = 1.7, FDR=1.2 x10−38). A broadly similar pattern was observed in CD8+ T cells with Hacvr2 (fold change 2.6, FDR=1.8 x10−61) and Lag3 (fold change = 2.5, FDR=6.7 x10−27) being the most upregulate checkpoint molecules (Fig. 5A and Supplementary Fig. 11A). These data imply that both CD4+ and CD8+ IFNγ producing lymphocytes are very tightly regulated in CPI-colitis.
To evaluate the functional role of these activated lymphocytes in CPI-colitis we took advantage of antibodies that deplete CD90 expressing cells. Although CD90 is often regarded as a pan-T cell marker, it is also a potent co-stimulatory molecule, and its ligation imparts a potent activation signal to T cells 34. Like other activation markers, the expression of CD90 increased in Ifng expressing CD4+ and CD8+ T cells (Fig. 5A). Similar findings were observed at protein level, with upregulation of CD90 in both CD4+ and CD8+ T cells after induction of CPI-colitis (Fig. 5B and C). Consistent with a more activated phenotype, cells with the highest expression of CD90 produced more cytokine, including a population of dual IFNγ and TNFα producing cells, in both CD4+ and CD8+ T cells (Fig. 5D and Supplementary Fig. 12A). Administration of depleting anti-CD90 antibodies significantly reduced the number of CD3+ CD90+ T cells in the colon following induction of CPI colitis and significantly attenuated disease, with reduced colon mass and abrogation of excessive neutrophil recruitment (Figs. 5E-H).
To further evaluate the phenotype of Ifng-expressing lymphocytes in CPI colitis, we compared chemokine and gut homing receptors in cells in comparison with lymphocytes that do not express Ifng. In both CD4+ cells and CD8+ T cells, the most upregulated chemokine receptor was Cxcr6 (6.3-fold, FDR=2.1x10−105 in CD4+ T cells and 2.2-fold, FDR, and 3.6-fold, 1.1x10−19 in CD8+ T cells), and in the case of CD8+ T cells, Cxcr6 was the only upregulated chemokine receptor (Fig. 5I and Supplementary Figs. 13A and B). Gut homing receptors, including Itga4 and Itgb7 that encode the classical gut homing integrin heterodimer α4β7 was downregulated in both Ifng-expressing CD4+ and CD8+ lymphocytes. These data maybe consistent with the hypothesis that tissue resident cells, or cell arising from tissue resident cells, are key drivers of CPI colitis rather than newly recruited T cells, especially since CXCR6 was the most highly upregulated chemokine receptor in both CD4+ and CD8+ lymphocytes. Notably, markers of tissue resident memory lymphocytes, including CD103 and CD69 were not upregulated in Ifng-expressing lymphocytes. To determine whether de novo recruitment of lymphocytes is required to initiate colitis, we administered anti-integrin α4β7 mAbs to mice prior to, and during, induction of CPI-colitis. Although anti-integrin α4β7 mAbs substantially reduced the number of integrin α4β7 expressing T cells in the colon and mesenteric lymph nodes (Fig. 5J), it failed to prevent or even reduce the severity of CPI-colitis (Figs. 5K and L). These data are consistent with an important pathogenic role of CXCR6+ lymphocytes likely arising from tissue resident memory cells, rather than lymphocyte populations that have been newly recruited to the gut.
IL23 blockade suppresses IFNγ-producing CD4+ colonic T cells and attenuates the development of CPI-colitis.
There is a major unmet need to identify therapeutically tractable mediators in CPI colitis, therefore, we conducted an Enhanced Causal Network Analysis (IPA) to identify upstream mediators responsible for activating the pattern of gene expression that we observed in Ifng expressing lymphocytes in CPI colitis. Analysis of cytokines predicted to activate Ifng expressing CD4+ T cells in CPI colitis included IL2 (z-score 4.7, P<1.0x10−27), IL7 (z-score 3.8, P<9.8x10−19), IL15 (z-score 3.4, P<5.2x10−34), IL18 (z-score 2.0, P<4.0x10−25) and IL23A (z-score 2.6, P<2.1x10−31) (Fig. 6A, Supplementary Table 1). Other predicted regulators included transcriptional regulators (TBX21, NFATC2, STAT3, IRF6 and IRF9) and transmembrane receptors, including co-stimulatory molecules (CD244), cytotoxicity receptors (NKG2D), CD69 and the complement receptor CD46 (Fig. 6A, Supplementary Table 1).
From a therapeutic perspective targeting cytokines predicted to activate these pathogenic cells is an attractive strategy, as many neutralizing monoclonal antibodies have been, or are currently, in clinical development for other immune-mediated inflammatory diseases. IL23 is an especially attractive target in CPI-colitis with multiple reagents is advanced development to treat conventional inflammatory bowel diseases. To determine whether IL23 is functionally important in CPI-colitis, we administered mAbs that neutralize p19 subunit of the IL23 heterodimer, or isotype matched control antibody at the same time as combination immunotherapy. IL23 blockade reduced the number of IFNγ producing CD4+ T cells in the colon in CPI-colitis and especially IFNγ/TNFα co-producing cells (Figs. 6B and C). The proportion of cytokine producing CD8+ T cells were unaffected by the blockade of IL23 (Supplementary Figs. 14A and B). IL23 blockade attenuated key disease features, including significantly reduced colon mass and reduced recruitment of colonic neutrophils (Figs. 6D and E). These data were corroborated by inducing CPI-colitis in Il23−/− mice, with reduced colon mass and reduced numbers of infiltrating neutrophils in comparison with WT littermate control mice (Figs. 6F and G). Furthermore, CD4+ T cells also had reduced number of IFNγ/TNFα co-producing cells (Fig. 6H and I). Proportions of CD8+ T cells were again unaffected by the genetic ablation of Il23 (Supplementary Figs. 14C and D). Together these data are consistent with IL23 playing an important role regulating the evolution of pathogenic effector CD4+ T cells in CPI-colitis and identify IL23 as a potentially important therapeutic target in CPI-colitis.
To investigate whether IL23 could be a viable target in human CPI-induced colitis we interrogated a gene expression in the colon from a recently published dataset of patients developing CPI-colitis following treatment with combination immunotherapy. Consistent with our analyses there was significant upregulation of cytotoxicity molecules (PRF1, GZMB, GZMA) and interferon-responsive genes (CXCL10) (Fig. 6J). There was also significant upregulation of IL23 and its key signalling molecule STAT3, indicating that IL23 is likely to be a viable therapeutic target in CPI-colitis.
We constructed a regulatory network predicted to be activated by IL23 based on downstream gene expression changes in Ifng expressing CD4+ T cells, which identified activation of transcription factors (STAT3, NFATC1), MAP kinase pathways (ERK1/2, p38 MAPK), signalling molecules (Phospholipase C gamma 2) and other kinases (PRKCD, Syk) implicated in inflammatory diseases (Fig. 6K). Together our data support the rationale for therapeutic targeting of IFNγ producing, polyfunctional T cells in CPI colitis, including neutralization of their upstream regulators, such as IL23.