Graphical abstract graphic file with name fx1.jpg [45]Open in a new tab Highlights * • apCAFs are associated with unfavorable outcomes in NSCLC patients receiving NCIT * • apCAFs’ expansion is triggered by IFN-γ via the JAK1/2-STAT1 pathway * • apCAFs promote the formation of FOXP1^+Tregs through the PD-L2-RGMB axis * • Targeting apCAFs by the blockade of the PD-L2-RGMB axis improves immunotherapy efficacy __________________________________________________________________ Cao et al. reveal that antigen-presenting cancer-associated fibroblasts (apCAFs) hinder NCIT effectiveness in NSCLC. IFN-γ induces apCAF expansion, which promotes FOXP1^+Treg formation through the PD-L2-RGMB axis. Targeting this pathway enhances immunotherapy outcomes, offering a potential therapeutic strategy. Introduction Neoadjuvant therapy has revolutionized perioperative regimens for cancer patients.[46]^1^,[47]^2^,[48]^3 One notable example is neoadjuvant chemoimmunotherapy (NCIT), which was associated with significantly greater event-free survival than neoadjuvant chemotherapy (NCT) alone.[49]^3 The efficacy of neoadjuvant therapy is primarily assessed by the major pathological response (MPR) rate, defined as less than 10% residual viable tumor in the resected specimen.[50]^4 Non-small cell lung cancer (NSCLC) was the first cancer type reported to benefit from neoadjuvant immune checkpoint inhibitors (neo-ICIs).[51]^5 Meanwhile, NCIT has become the dominant therapeutic approach in the neoadjuvant setting, extending its use in clinical practice due to its promising clinical response rates compared to either NCT[52]^6 or neo-ICIs alone.[53]^7 The NADIM-2 phase 2 randomized trial highlighted that the pathologically complete regression (pCR) rate improved to 37% in the nivolumab plus chemotherapy group compared to 7% with chemotherapy alone.[54]^6 Furthermore, the KEYNOTE-671[55]^6 and AEGEAN[56]^8 trials indicated that neoadjuvant anti-PD1/L1 regimens plus chemotherapy confer greater benefits compared to NCT alone.[57]^9^,[58]^10 Despite these promising results, the non-MPR rate remains high, reaching approximately 60% in the CheckMate 816 trial.[59]^11 Furthermore, the LCMC3 trial reported a higher non-MPR rate of 80%, with 9% experiencing progressive disease (PD). However, the specific mechanisms responsible for non-MPR remain largely unknown.[60]^12 This clinical evidence underscores the need to develop new strategies to overcome resistance to NCIT. Cancer-associated fibroblasts (CAFs) are the predominant type of stromal cells in the tumor microenvironment (TME).[61]^13 CAFs can induce immunosuppressive effects by interacting with T cells[62]^14 and tumor-associated macrophages,[63]^15 among others. Numerous studies have demonstrated their tumor-promoting behaviors. Recently, a few studies exploring their role in immunotherapy resistance have emerged. Liang et al. unveiled that autophagy-enhanced CAFs could compromise immunotherapy by up-regulating PD-L1.[64]^14 Similarly, Lin et al. identified biglycan-secreting CAFs as a barrier to anti-PD1 efficacy.[65]^16 However, the role of specific CAF subtypes in inducing anti-PD1 resistance in NSCLC, particularly in the context of NCIT, remains unexplored. In this study, using single-cell RNA sequencing (scRNA-seq), digital spatial profiling (DSP) spatial sequencing, and bulk RNA sequencing (RNA-seq) of patients’ tumor samples during NCIT, we identified that antigen-presenting cancer-associated fibroblasts (apCAFs) are associated with NCIT non-MPR in NSCLC patients. apCAFs are a newly identified CAF subcluster characterized by major histocompatibility complex (MHC)II expression.[66]^17 Brekken et al. reported apCAFs in the pancreatic ductal adenocarcinoma (PDAC) TME and confirmed that apCAFs could induce regulatory T cell (Treg) expansion in ex vivo assays, although the exact mechanism remained unclear.[67]^17 Jiang et al. further reported that apCAF accumulation also depended on the interleukin (IL)-1-IL-1R2 signaling pathway in Tregs.[68]^18 However, their specific function in NSCLC, particularly in the context of NCIT, was largely unknown. Through our previously developed cell-communication analysis pipeline[69]^19 and a 3D ex vivo co-culture platform,[70]^20 we discovered that the expansion of apCAFs, which relied upon anti-PD1, could promote FOXP1^+ Treg accumulation via the PD-L2-repulsive guidance molecule b (RGMB) axis, thereby dampening anti-PD1 efficacy in NSCLC, in turn. Mechanistically, we discovered that anti-PD1 could augment apCAF expansion dependent on interferon (IFN)-γ, which stimulated the JAK1/2-STAT1-IFI6/27 axis, ultimately leading to the upregulation of MHCII and PD-L2. PD-L2, another PD1 ligand, showed limited efficacy with anti-PD-L2 therapies for a long time.[71]^21 Sharpe et al. recently found RGMB as a new key PD-L2 partner, and combining anti-RGMB with anti-PD1 may overcome microbiome-mediated immunotherapy resistance.[72]^22 However, the specific cell cluster responsible for RGMB expression remained unknown. In our study, we identified Tregs as the major contributors to RGMB expression in the TME. Additionally, we found that apCAFs were the primary source of PD-L2 expression. Utilizing murine tumor models and organoid platforms, we confirmed that, in apCAF-enriched lung tumors, targeting the PD-L2-RGMB axis could significantly overcome anti-PD1 resistance. This finding not only provides additional therapeutic options for addressing NCIT resistance but also opens potential application opportunities for anti-PD-L2 regimens in clinical settings. Results Antigen-presenting CAFs specifically enriched in the TME of post-non-MPR The TME is a complex ecosystem composed of tumor, immune, and stromal cells. Recently, Ye et al. identified fibroblasts as the dominant stromal cells subtype.[73]^23 Moreover, several studies have demonstrated the influence of fibroblasts on immunotherapy.[74]^16^,[75]^24 Coincidentally, we uncovered that higher expression of the CAF signature was significantly correlated with worse progression-free survival in a public NSCLC cohort treated with immune checkpoint inhibitors (ICIs) as well[76]^25 ([77]Figure S1A). To deeply explore fibroblasts in NCIT, we conducted scRNA-seq on tumor specimens from 10 NSCLC patients and DSP spatial sequencing on specimens from 6 patients, either before or after NCIT ([78]Figure 1A). Smooth-muscle actin (SMA) was a typical marker of CAFs, and we therefore selected SMA^+ cells within the TME for DSP spatial sequencing ([79]Figure 1B). To comprehensively analyze the phenotypic changes within different TME components during NCIT treatments, we collected, sorted, and cultured primary CAF cell lines (37 patients), tumor-infiltrating lymphocytes (TILs), and organoids (11 patients). CAF cell lines were cultured in a 3D manner in the non-immunogenic hydrogel that was developed by us,[80]^19^,[81]^20 followed by various readouts ([82]Figure 1A). Figure 1. [83]Figure 1 [84]Open in a new tab apCAFs accumulated in TME of post-non-MPR (A) Experimental design and the patient cohort. The respective experimental details referred to here are in the STAR Methods. (B) Three representative immunohistochemistry (IHC) pictures of fibrotic “hotspots” in non-small cell lung cancer (NSCLC) TME sequenced by DSP spatial sequencing. (C) Uniform manifold approximation and projection (UMAP) plot of 6,987 CAF cells colored by clusters in our NCIT cohort. “apCAF” is encircled by a dashed line. (D) Scatterplot showing gene expression change for post-non-MPR versus pre-non-MPR (x axis) against the post-MPR versus pre-MPR (y axis, left) or post-non-MPR versus post-MPR (y axis, right) in all CAFs. Genes significantly upregulated in post-non-MPR versus pre-non-MPR while downregulated in post-MPR versus pre-MPR (left) or upregulated in post-non-MPR versus post-MPR (right) are marked in blue. Genes belonging to apCAF signature are bolded. Genes marked by red are well-established markers for apCAFs according to the literature. (E) Tumor weights of LLC tumors. n = 5 mice per group, representative of three independent experiments. p value is calculated using unpaired Student’s t test. Error bars show the mean and SEM. (F) Representative flow cytometric scatterplots of apCAFs within PDTF from non-MPR and MPR patients before or after anti-PD1 (left), with quantification on the right. In the barplots, apCAF means PDPN^+HLA-DR^+ cells (gated from CAF). Data are representative of three different experiments. p value is calculated using paired Student’s t test. (G) Experimental design of the PDTF platform. (H) Representative flow cytometric scatterplots of the transwell co-culture systems. Quantification barplot is on the right. Data are representative of three different experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. During the scRNA-seq analysis, we used COL1A1 and COL3A1 as exclusive markers for CAFs and excluded contamination by other cell types using digital flow cytometry sorting (fluorescence-activated cell sorting [FACS]) ([85]Figures S1B–S1E). In total, we identified 6,987 CAF cells and categorized them into 6 distinct subtypes ([86]Figure 1C). Differential gene (DEG) analysis revealed unique expression patterns across these subtypes ([87]Figures S1F and S1G). To investigate the potential changes within different CAF subtypes during NCIT, we conducted DEG analysis systematically in all CAFs, comparing specimens before (pre-non-MPR and pre-MPR) and after NCIT (post-non-MPR and post-MPR). Interestingly, we found that the signature genes of apCAFs[88]^17 were significantly upregulated in post-non-MPR samples compared to both pre-non-MPR and post-MPR samples ([89]Figure 1D). Conversely, these genes were downregulated in post-MPR samples compared to pre-MPR samples ([90]Figure 1D). To validate this, we utilized another NSCLC NCIT dataset (GEO: [91]GSE207422 ).[92]^26 Major subclusters identified in GEO: [93]GSE207422 were well-mapped to the CAF subtypes in our dataset ([94]Figures S1H and S1I). Due to the limited MPR patient cells in GEO: [95]GSE207422 (73/1,063 CAFs), we focused on non-MPR patients. Besides, the fraction of apCAFs was larger in post-non-MPR samples compared to pre-non-MPR samples ([96]Figure S1J). Additionally, the expression levels of CD74, histocompatibility complex, class II, DR alpha (HLA-DRA) and histocompatibility complex, class II, DR belta 1 (HLA-DRB1) were higher in post-non-MPR samples compared to pre-non-MPR samples ([97]Figure S1K). However, whether the fraction of apCAFs was indeed upregulated in post-non-MPR samples remained unclear. We thus collected an additional NCIT cohort consisting of 14 samples for validation ([98]Figure S2A). By integrating the in-house supplementary dataset and GEO: [99]GSE207422 with our discovery dataset, we gathered an adequate sample size and found that apCAFs were the only subcluster to exhibit significant enrichment in post-non-MPR samples ([100]Figures S2A–S2C). Moreover, Luo et al. proposed that apCAFs likely originate from macrophages, and we further corroborated their findings through trajectory analysis, demonstrating that apCAFs indeed share transcriptional similarities with macrophages ([101]Figures S2D–S2G). During single-cell analysis, subcluster “contamination” can occur, although it is rare. We double-checked the expression of typical CAF markers on apCAFs, including Podoplanin (PDPN), COL1A1, and platelet-derived growth factor receptor alpha (PDGFRA) ([102]Figure S3A), confirming that apCAFs are indeed a distinct subcluster of CAFs. Additionally, we conducted bulk RNA-seq on primary CAF cell lines derived from patients’ tumors ([103]Figure 1A). Using morphological analysis and quantitative reverse-transcription PCR (RT-qPCR), we ensured the purity of the CAFs ([104]Figure S3B). Among these, there were 4 cell lines each for post-non-MPR and post-MPR. We created a signature by intersecting characteristic genes of apCAFs from the integrated single-cell dataset, naming it the “apCAF NCIT signature.” Indeed, the apCAF NCIT signature was significantly upregulated in post-non-MPR compared to post-MPR ([105]Figures S3C and S3D). Moreover, the apCAF NCIT signature also showed exclusive enrichment in post-non-MPR specimens within SMA^+ CAFs as revealed by DSP spatial sequencing ([106]Figures S3E and S3F). In summary, through multi-omics analysis, we identified a specific CAF subcluster, apCAFs, as a potential indicator of post-non-MPR. apCAFs expanded during immunotherapy and dampened anti-PD1 efficacy Although our in silico analysis suggested that apCAFs might have expanded in post-non-MPR samples, its direct influence on immunotherapy efficacy and the factors responsible for its expansion remained unclear. We first conducted multiplexed immunofluorescence (mIF) to profile the distribution pattern of apCAFs. Notably, apCAFs were enriched in non-MPR samples compared to MPR samples. Moreover, after NCIT, the number of apCAFs was upregulated in post-non-MPR compared to pre-non-MPR. ([107]Figure S3G). Next, we constructed an orthotopic Lewis lung carcinoma (LLC) lung tumor model in C57BL/6 mice and sorted apCAFs and non-apCAFs from the tumors on day 27. These CAFs were mixed with LLC cells, followed by the construction of subcutaneous tumor models ([108]Figure S3H).[109]^27 We noticed that tumor growth curves were unaffected by non-apCAFs ([110]Figures 1E, [111]S3I, and S3J). Besides, apCAFs alone, without anti-PD1 intervention, did not exhibit pro-tumorigenic effects ([112]Figures 1E and [113]S3I). On the contrary, apCAFs significantly accelerated tumor growth during anti-PD1 regimens ([114]Figures 1E, [115]S3I, and S3J). Additionally, the granzyme B (GZMB) degranulation of CD8^+ TILs was dampened by apCAFs as well ([116]Figure S3K). This indicated that apCAFs could directly dampen the efficacy of anti-PD1 therapy. Nevertheless, LLC is considered insensitive to anti-PD1 therapy, and, in our study, anti-PD1 treatment indeed failed to induce significant tumor shrinkage ([117]Figures 1E and [118]S3I). Therefore, we utilized two additional cell lines, SJT1601 and Kras^G12DTp53^−^/^− (KP), both of which have been reported to respond to anti-PD1 therapy. Using the same methodology as in the LLC models, we demonstrated that, in anti-PD1-sensitive tumor models, apCAFs significantly induced immunotherapy resistance ([119]Figure S4). In contrast, in non-apCAF settings, tumor growth remained largely unaffected, regardless of anti-PD1 treatment ([120]Figure S4). However, in vivo models could not provide insights into the driving force behind apCAF expansion during NCIT. We then questioned whether apCAFs could be stimulated during anti-PD1 treatments. To explore this, we adopted an ex vivo 3D platform naming as patient-derived tumor fragments (PDTFs).[121]^28 PDTFs were encapsulated in the same hydrogel used in the 3D culture of CAFs, and multiplexed flow cytometry (mFC) was conducted on them after 48 h of anti-PD1 treatment ([122]Figures 1F–1H). We found that post-non-MPR samples had the largest fraction of apCAFs at baseline ([123]Figures 1F and [124]S5A). Furthermore, anti-PD1 treatment significantly enhanced apCAF expansion in both pre-non-MPR and post-non-MPR samples, but not in MPR samples ([125]Figures 1F and [126]S5A). To determine whether direct contact or soluble factors led to apCAF marker upregulation, we first sorted CAFs and non-CAFs and then used the transwell assay to profile changes in apCAFs ([127]Figures 1G and 1H). Besides, non-CAFs could stimulate apCAF accumulation in a non-contact manner ([128]Figure 1H). Additionally, supernatants from non-CAFs had the same effect ([129]Figures 1F–1H). Moreover, the non-MPR-specific expansion of apCAFs was also effectively recapitulated in CAFs cultured with supernatants from non-CAFs ([130]Figure S5B). In summary, we concluded that anti-PD1 directly stimulated apCAF expansion in non-MPR samples, and apCAFs served as a direct cause of resistance to immunotherapy. Anti-PD1 stimulated apCAF expansion dependent upon overexpressed IFN-γ in non-MPR Although we discovered that apCAF accumulation in post-non-MPR samples was dependent on anti-PD1 treatment, the specific pathway responsible remained unclear. Re-analysis of scRNA-seq data revealed that IFN receptors, such as interferon alpha and beta receptor subunit 2 (IFNAR2) (for type I IFN), interferon-gamma receptor 1/2 (IFNGR1/2) (for type II IFN), and IL-10RB (for type III IFN), were significantly upregulated in apCAFs compared to non-apCAFs ([131]Figure 2A). Pathway enrichment analysis confirmed that IFN-related pathways were upregulated in apCAFs ([132]Figure 2B), verified by gene set enrichment analysis (GSEA) in the NCIT dataset GEO: [133]GSE207422 ([134]Figure 2C). Additionally, in the previously described bulk RNA-seq of CAFs and DSP spatial sequencing data, the IFN-γ pathway also showed significant enhancement in post-non-MPR samples ([135]Figures 2D and 2E). Figure 2. [136]Figure 2 [137]Open in a new tab Anti-PD1-stimulated apCAF expansion is dependent upon IFN-γ (A) IFN-related gene expression in apCAFs or other CAFs in our own NSCLC cohort. (B) Heatmap showing scaled gene sets scores in apCAFs and non-apCAFs in our own NSCLC cohort. (C) GSEA plot for REACTOME_INTERFERON_GAMMA_ SIGNALING in apCAFs in the GEO: [138]GSE207422 cohort. (D and E) Scores of REACTOME_INTERFERON_GAMMA_ SIGNALING in our bulk RNA-seq of CAFs (D) and our DSP dataset (E). p value is calculated using paired Student’s t test. (F) The concentration of interferons (IFNs) in supernatants of PDTF. (G) Experimental design of the PDTF and 2D and 3D culture. (H) The frequency of HLA-DR^+CAFs under the conditions of PDTF and 2D and 3D culture (G). (I) Representative ridge plots of HLA-DR^+CAFs with the treatment of IFNs under the condition of 2D and 3D culture (upper) and quantification barplot (lower). (J) Representative immunocytochemistry (ICC) pictures of HLA-DR^+CAFs with different treatments. Scale bar, 50 μm. (K) Representative ICC pictures of HLA-DR^+CAFs with different treatments. Scale bar, 50 μm. (L) Representative flow cytometric scatterplots of CAFs with different treatments (left) and the quantification barplot (right). For (F), (H), (I), and (L), data are representative of three different experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. To verify the in silico findings, we collected supernatants from PDTF culture for ELISA profiling ([139]Figures 2F and 2G). Indeed, all three types of IFN, including IFN-α/β, IFN-γ, and IFN-λ, showed upregulation in the post-non-MPR PDTF supernatants ([140]Figure 2F). We then transitioned from ex vivo to in vitro assays. We used the previously described human primary CAF biobank for downstream experiments ([141]Figure 2G). We found that traditional 2D culture settings largely lost HLA-DRA expression compared to PDTF at the baseline time point. Meanwhile, 3D culture of CAFs could recover HLA-DRA expression ([142]Figure 2H). Previous reports have also noted that CAFs lose MHCII molecule expression in 2D settings,[143]^29 emphasizing the necessity of our 3D culture system. Regardless of the culture method, IFN-γ, but not IFN-α/β or IFN-λ, was the major IFN responsible for apCAF expansion, as confirmed by immunocytochemistry (ICC) ([144]Figures 2I and 2J). As previously mentioned, we showed that anti-PD1 could enhance apCAF expansion. Using an IFN-γ neutralizing antibody (emapalumab), we further confirmed that IFN-γ was necessary for the stimulation of apCAFs by anti-PD1 ([145]Figures 2K and 2L). Supplementation with emapalumab largely blocked the upregulated apCAF fraction following anti-PD1 treatment, as corroborated by ICC ([146]Figures 2K and 2L). To further investigate whether IFN-γ contributes to apCAF expansion in vivo, we generated primary apCAFs with stable interferon-gamma receptor (IFNGR) knockdown (KD), negative control knockdown (NCKD) group and utilized the previously described co-transplantation models for validation ([147]Figure S5C). Compared to NCKD, KD apCAFs exhibited reduced expansion under anti-PD1 treatment ([148]Figure S5I). Moreover, KD largely abolished apCAF-mediated resistance to anti-PD1 in vivo ([149]Figures S5E–S5H), indicating that the IFN-γ-IFNGR axis plays a critical role in anti-PD1-induced apCAF expansion. Although we concluded that the anti-PD1-IFN-γ axis was responsible for the expansion of apCAFs and that apCAFs served as important mediators of anti-PD1 resistance, the widespread expression of IFN-γ and IFNGR1/2 posed challenges for developing drugs targeting the “origin” of apCAFs due to the risk of off-target effects. Therefore, after addressing the questions of “what are apCAFs” and “where do they come from,” we set out to investigate “where do they go,” aiming to identify “targetable” axes centered around apCAFs. Spatial co-localization of apCAFs with Tregs indicated their potential interactions Using CellChat analysis, we found that CAFs exhibited the strongest cell-cell communication among major cell types within the TME ([150]Figure S5J). We hypothesize that apCAFs contribute to ICI resistance through interactions with other cell types. TILs, the primary ICI responders, showed stronger interaction with apCAFs than non-apCAFs ([151]Figure S5K), including CD8 TILs and Tregs. We previously proposed that non-CAF-secreted IFN-γ was responsible for apCAF expansion. Here, we identified a certain CD8 subtype, BAG3^+ T cells, as the most probable source of IFN-γ ([152]Figures S5M–S5S). Additionally, compared to other known interaction partners of Tregs, such as macrophages and tumor cells, apCAFs exhibited even more interactions ([153]Figure S5L). Brekken et al. found that apCAFs from mice orthotopic PDAC tumors could lead to Tregs formation in in vitro assays.[154]^17 However, such immune-modulating effects have not been reported in other tumors or in in vivo scenarios. We hypothesized that apCAFs might also induce Tregs formation in the NSCLC TME. First, we tested if apCAFs were closely located near Tregs. Using mIF, we found that Tregs were more prevalent in post-non-MPR compared to pre- or post-MPR ([155]Figure S6A) and were in direct contact with antigen-presenting cells (APCs) (CD74^+ cells) ([156]Figure 3A), which are indispensable for Tregs differentiation.[157]^30 While apCAFs are considered non-classic APCs,[158]^17 the number of Tregs was positively correlated with the number of apCAFs ([159]Figures 3B and 3C), which is not the case for non-apCAFs ([160]Figure S6B). In post-non-MPR samples, the distances between Tregs and apCAFs were much shorter ([161]Figures 3D and [162]S6C), which is not observed in non-apCAFs either ([163]Figure S6D). Although the number of APCs within a 25 μm radius around Tregs showed no difference between post-non-MPR and post-MPR ([164]Figure 3E), the number of apCAFs and the ratios of apCAFs to APCs were significantly higher ([165]Figures 3F and 3G). Thus, among all APCs, apCAFs likely played a more important role in Treg formation in post-non-MPR compared to post-MPR or pre. Figure 3. [166]Figure 3 [167]Open in a new tab The spatial proximity of apCAFs and Tregs suggests potential communications between them (A) Representative multiplexed IF (mIF) pictures of pre, MPR, non-MPR lung tumors, showing the spatial distributions of APCs (CD74^+) and Tregs (FOXP3^+). Arrows indicate the interaction between Treg and apCAFs. For each group, at least 4 patients were included. Scale bar, 50 μm. (B) Representative mIF pictures showing the distributions of apCAFs (COL1A1^+CD74^+) and Treg (FOXP3^+). For each group, at least 4 patients were included. Arrows indicate the spatial distribution of Treg. Scale bar, 60 μm. (C) Scatterplot showing the relationships between the number of Tregs and apCAFs in (B), colored by groups. Only visions with comparable DAPI^+ cells were selected for calculation. Each dot represents a vision. For each group, at least 4 patients were included; for each patient, at least 3 visions with Tregs were selected randomly. The correlation coefficient is calculated by Pearson, and p value is determined by two-sided linear regression t test. (D) Violin plot showing the shortest distance of Treg to apCAFs/APC in (B). The shortest distance was determined by the distance between the apCAFs closest to a specific Treg. Only visions with comparable DAPI^+ cells were selected for calculation. Each dot represents a Treg cell. For each group, at least 4 patients were included; for each patient, at least 3 visions with Tregs were selected randomly; for each vision, every Treg was designated a shortest distance value. Significance was determined using unpaired Student’s t test. (E–G) (E) Boxplots showing the numbers of APCs within 25 μm radius centered around Tregs in (A). Only visions with comparable DAPI^+ cells were selected for calculation. (F) Boxplots showing the numbers of apCAFs within 25 μm radius centered around Treg in (B). Data are representative of three different experiments. (G) Boxplots showing the numbers of apCAFs/APCs within 25 μm radius centered around each Treg in (B). For (E)–(G), each dot represents a Treg cell. For each group, at least 4 patients were included; for each patient, at least 3 visions with Tregs were selected randomly; for each vision, at least one Treg was adopted to calculate the value. p value is calculated using unpaired Student’s t test. (H) UMAP plot of 9,363 CD4^+T cells colored by clusters in our NSCLC cohort. (I) UMAP plot of 13,893 CD4^+T cells colored by clusters in the GEO: [168]GSE207422 cohort. (J) Heatmap showing the correlations between CD4^+T cells in our own CAF cohort and GEO: [169]GSE207422 calculated by SingleR. (K) The proportions of CD4^+T cell clusters. Significance was determined using unpaired Student’s t test, and the absence of significance indicates no significant difference. ∗p < 0.05, ∗∗p < 0.01. (L) The proportions of IL-1R1_Treg in different efficacy groups from the GEO: [170]GSE207422 cohort. p value is calculated by unpaired Student’s t test. (M) Network graphs representing the relative interaction strength between CAFs and CD4^+ T cells (Treg and non-Treg). (N) Network graphs representing the interaction between CAFs (apCAFs and non-apCAFs) and CD4^+ T cells. Edge strength represents the interaction strength calculated by CellChat. (O) Expression of FOXP3 and IL-1R1 in Treg and Tconv in GEO: [171]GSE253540 datasets (left). Expression of FOXP3 and IL-1R1 in Treg, Tconv, and CD8^+T in PBMC of the healthy donors or NSCLC patients from GEO: [172]GSE211044 datasets (middle, right). p value is calculated using unpaired Student’s t test. (P) Monocle trajectory inference for IL-1R1_Treg, LEF1_Treg, ANXA1_CD4, and CCR7_CD4, colored by cell states. Analysis was conducted on our own CAF scRNA-seq data. (Q) Proportion of cell state in each cluster. The advent of scRNA-seq has revealed significant heterogeneity in TME-infiltrated Tregs.[173]^31 Ley et al. identified a cytotoxic Tregs subset (exTreg) that promotes pro-inflammatory responses rather than suppressing them.[174]^32 Therefore, a detailed analysis of Tregs is necessary to identify the specific subtypes interacting with apCAFs. We closely examined the CD4^+ TILs and found that, among the seven well-distinguished subclusters ([175]Figures 3H, [176]S6E, and S6F), five were also present in the CD4^+ TILs from GEO: [177]GSE207422 ,[178]^26 showing excellent similarity ([179]Figures 3I, 3J, and [180]S6G). We then found that IL-1R1_Tregs were exclusively enriched in non-MPR both in our dataset and in GEO: [181]GSE207422 ([182]Figures 3K, 3L, and [183]S6H). In contrast, another Tregs subtype, LEF1_Tregs, showed no significant changes in either dataset ([184]Figures 3K and [185]S6H). Therefore, we concluded that IL-1R1_Tregs, not LEF1_Tregs, is an important marker of non-MPR. Nevertheless, the potential interactions between IL-1R1_Tregs and apCAFs remained unclear. Using our previously developed cell-communication pipeline, we found that apCAFs were the only subset showing enhanced communication with Tregs compared to non-Tregs ([186]Figure 3M). Furthermore, apCAFs demonstrated stronger communication with IL-1R1_Tregs compared to non-apCAFs ([187]Figure 3N), indicating potential interactions. IL-1R1_Tregs is a newly discovered suppressive CD4^+ subset.[188]^33 We uncovered, using publicly available datasets,[189]^34^,[190]^35 that IL-1R1 is specifically expressed on Tregs from NSCLC patients’ PBMC both in vitro and ex vivo ([191]Figure 3O). To distinguish IL-1R1_Tregs from LEF1_Tregs, we conducted pseudotime analysis and found that LEF1_Tregs is a transitional subcluster along the differentiation trajectory from naive CD4^+ T cells (ANXA1_CD4 and CCR7_CD4) to IL-1R1_Tregs ([192]Figures 3P–3Q). Interestingly, IL-1R1_Tregs was predominant in state 2, which showed higher expression of exhaustion markers ([193]Figures 3Q, [194]S6I–S6K, [195]S7A, and S7B). However, IL-1R1 expression on Tregs was undetectable in our LLC mice models and Prlic et al.’s head and neck squamous cell carcinoma (HNSCC) mouse models,[196]^33 likely due to potential intratumoral microbiome colonization. This dilemma remains unresolved, prompting us to consider surrogate markers for IL-1R1_Tregs through other multimodal analyses to better investigate apCAF-Tregs relationships in vivo. apCAF-stimulated Tregs were characterized by FOXP1 expression To identify markers equivalent to IL-1R1, we used single-cell regulatory network inference and clustering (SCENIC) analysis to pinpoint intrinsic transcription factors (TFs) driving IL-1R1_Tregs differentiation ([197]Figure S7C). Notably, as cells transitioned from state 3 (dominated by naive CD4^+ TILs) to state 2 (dominated by IL-1R1_Tregs), FOXP1 activity increased ([198]Figure 4A). Additionally, FOXP1 was more active in IL-1R1_Tregs compared to LEF1_Tregs ([199]Figure 4B) and showed significantly higher expression in post-non-MPR samples ([200]Figure 4C). Since FOXP1 can be reliably detected in mouse LLC models, we used FOXP1^+Tregs as a surrogate for IL-1R1_Tregs. Figure 4. [201]Figure 4 [202]Open in a new tab FOXP1^+Tregs specifically induced by apCAFs (A) SCENIC-inferred TF activity of FOXP1, FOXP3, and BATF along the pseudotime trajectory. (B) SCENIC-inferred TF activity of FOXP1 in IL-1R1_Treg and LEF1_Treg. p value is calculated by exact binomial test. (C) Scatterplot showing gene expression change for post-non-MPR versus pre-non-MPR (x axis) against the post-MPR versus pre-MPR (y axis, left) or post-non-MPR versus post-MPR (y axis, right) in Treg. Genes significantly upregulated in post-non-MPR versus pre-non-MPR while downregulated in post-MPR versus pre-MPR (left) or upregulated in post-non-MPR versus post-MPR (right) are marked in blue. FOXP1 is bolded and highlighted. (D) Expression of FOXP1 among immune cells from TCGA lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD) datasets. p values are calculated using Wilcoxon test. (E) Expression of FOXP1 in different cell types in GEO: [203]GSE253540 datasets. p value is calculated using unpaired Student’s t test. (F) The coherence of apCAFs and IL-1R1_Treg in ST datasets. Spots are colored by the average expression of their markers relatively, selected from single-cell dataset. (G) The correlation between fraction of apCAFs and IL-1R1_Treg (brown) or LEF1_Treg (gray) in spots from different ST datasets. The correlation coefficient is calculated by Pearson, and p value is determined by two-sided linear regression t test. (H) Expression heatmaps (mean values) of genes from the IL-1R1_Treg and stem CD4^+ T cells signature in murine intratumoral lung CD4^+ T cells, purified from Col1a2 Cre^ER+I-Ab^fl/fl versus I-Ab^fl/fl mice and analyzed by bulk RNA-seq from GEO: [204]GSE164659 datasets. (I) Representative flow cytometric scatterplots of Treg in mice tumors, and the quantification of them, corresponding to the experiments in [205]Figure 1E. Data are representative of three independent experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (J) Representative flow cytometric scatterplots of GZMB^+CD8^+ T cells and the quantification of them. Data are representative of three different experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (K and L) Representative ridge plots of Treg induced by apCAFs or non-apCAFs, with the treatment of anti-PD1, and the quantification. For (I)–(L), data are representative of three different experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. FOXP1 is crucial for FOXP3-mediated expression of CTLA-4 and CD25 on Tregs.[206]^36^,[207]^37^,[208]^38 Deleting FOXP1 in FOXP3^+ Tregs leads to spontaneous inflammatory diseases in mice.[209]^38 However, the biological roles of FOXP1^+ Tregs in tumor immunology remain largely unknown. First, we examined whether FOXP1 is exclusively expressed on CD4^+ TILs in the NSCLC TME. Deconvolution analysis of The Cancer Genome Atlas (TCGA) data showed that FOXP1 indeed has the highest expression in CD4^+ TILs ([210]Figures 4D and [211]S7D). Furthermore, Tregs exhibited significantly higher FOXP1 expression compared to conventional T cell (Tconv) or CD8^+ T cells in NSCLC PBMC ([212]Figure 4E). We then investigated the spatial relationship between FOXP1^+ Tregs and apCAFs. Using publicly available spatial transcriptomics datasets[213]^39^,[214]^40^,[215]^41^,[216]^42 (ST-data), we found that the spatial distribution of apCAFs coincided with FOXP1^+ Tregs ([217]Figure 4F). Additionally, FOXP1^+ Treg fractions were more correlated with apCAF fractions in ST-data spots compared to LEF1^+ Tregs ([218]Figure 4G). We thus hypothesized that apCAFs might induce the expansion of FOXP1^+Tregs. Besides, compared to CD4^+ TILs from apCAF-knockout mice (Col1a2 CreER^+I-Ab^fl/fl), CD4^+ TILs from the control group (I-Ab^fl/fl) showed upregulation of FOXP1_Tregs signature genes (Foxp3, Il1rl1, Il1rl2, etc.) and diminished expression of stem CD4^+ T cells signature genes (Tcf7, Il7r, Gzmk, etc.) ([219]Figure 4H), suggesting that apCAFs might directly stimulate FOXP1^+ Treg expansion in vivo. In co-transplantation models, the TME of apCAF-enriched tumors also exhibited significantly more FOXP1^+ Tregs, as well as a higher overall number of Tregs ([220]Figures 4I and [221]S7E). Tregs sorted from the TME of apCAF-enriched tumors (termed “FOXP1high Tregs”) showed the strongest suppressive function on the cytotoxic behavior of CD8^+ TILs ex vivo ([222]Figures 4J and [223]S7F). We then co-cultured apCAFs or non-apCAFs sorted from patient-derived CAFs with corresponding autologous naive CD4^+ TILs. Additionally, Tregs, including FOXP1^+Tregs, exhibited substantial expansion in the apCAF co-culture groups, regardless of anti-PD1 stimulation ([224]Figures 4K and 4L). Taken together, we identified that FOXP1^+Tregs could well-represent IL-1R1^+Tregs and uncovered that apCAFs could directly stimulate FOXP1^+Treg expansion, both in vivo and in vitro. Anti-PD1 induced exclusive expression of PD-L2 on apCAFs relying upon IFN-γ The primary objective of our investigation into the interaction between apCAFs and FOXP1^+ Tregs was to identify “targetable” axes to counteract apCAFs-mediated NCIT resistance. We first examined whether neutralizing HLA-DRA expression on apCAFs could inhibit Tregs differentiation. Besides, αMHCII only partially reduced the effect of apCAFs on Tregs and FOXP1^+ Tregs, indicating that additional mechanisms were involved ([225]Figure S8A). To explore potential mediators beyond MHCII, we conducted Virtual Inference of Protein-activity by Enriched Regulon (VIPER) analysis, a protein-activity quantification algorithm,[226]^43 on our single-cell data of CAFs. The re-clustering of CAFs based on protein activity was highly consistent with transcriptomic data ([227]Figures 5A and 5B). Crucially, VIPER-derived apCAFs uniquely expressed PD-L1 and PD-L2 ([228]Figures 5A–5C). This finding was significant as Akbari et al. recently demonstrated that PD-L2 is crucial for Tregs maintenance and stability.[229]^44 Thus, beyond MHCII/T cell receptor signaling, the co-inhibitory axis of PD-L1/2 may also contribute to apCAF-mediated Tregs expansion. Despite the low RNA levels of PD-L1/2 and the sparse nature of single-cell transcriptomics, which limited the identification of PD-L1/L2 on apCAFs, VIPER analysis successfully highlighted their presence. mFC analysis confirmed that apCAFs, rather than non-apCAFs, were indeed the primary CAF sub-cluster expressing PD-L1/L2 ([230]Figure 5E). Figure 5. [231]Figure 5 [232]Open in a new tab PD-L2 and HLA-DRA expression on apCAFs both under control of IFN-γ (A) UMAP plot of CAFs colored by clusters in our NSCLC cohort based on the VIPER algorithm. (B) Heatmap showing certain proteins’ activity in apCAFs and non-apCAFs, calculated by VIPER. (C) Feature plots of PDCD1LG2 and CD274 protein activity based on the VIPER metrics. (D) Expression of PDCD1LG2 among immune cells from TCGA LUSC and LUAD datasets. p values are calculated using Wilcoxon test. (E–G) (E) Representative flow cytometric histograms of expression of PD-L1 and PD-L2 in apCAFs and non-apCAFs, with quantification on the right. (F) PD1, PD-L1, and PD-L2 expression in tumor cells (A549), immune cells (PBMCs), and CAFs assessed by RT-qPCR. (G) mRNA expression level of genes quantified by RT-qPCR for organoids, TILs, or CAFs with treatment of anti-PD1 or its isotype antibody. For (E)–(G), data are representative of three independent experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (H) Representative flow cytometric scatterplots of PDTF from non-MPR and MPR patients before or after anti-PD1 (left), with quantification in (I). Data are representative of three independent experiments. (I) Frequency of PD-L2^+ apCAFs in PDTF. Data are representative of three different experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (J) Representative immunocytochemistry (ICC) pictures of HLA-DR^+CAFs/PD-L2^+CAFs with the supplement of IFN-γ, emapalumab, fludarabine, or ruxolitinib. Data are representative of three independent experiments. Scale bar, 50 μm. (K) Heatmap showing the mRNA expression level assessed by RT-qPCR. Each specific column means a specific patient’s CAF cell line. For every gene listed in the apCAFs and PD-L1/2 signatures, the difference between any two treatment groups is significant, determined by two-way ANOVA test. (L) The protein expression levels of PD-L2 and HLA-DR in patients-derived CAFs measured by WB (left) and the integrated density of them measured by ImageJ. Data of three representative patients are posted here. p values are determined using unpaired Student’s t test. We next questioned the biological value of PD-L1/2^+ CAFs. First, in TCGA lung cancer data, CAFs contribute comparable PD-L2 expression in the TME as macrophages, while this is not the case for PD-L1 ([233]Figures 5D and [234]S8B–S8D). Additionally, we found that CAFs manifested the highest PD-L1/2 expression compared to tumor cells and immune cells ([235]Figure 5F). We then conducted RT-qPCR upon FACS-sorted organoids, CAFs, and TILs from the same individual. While anti-PD1 diminished PD-L1 expression on TILs, it significantly upregulated HLA-DRA and PD-L2 expression on both organoids and CAFs, with the latter showing a much greater increase ([236]Figure 5G). Taken together, we suggested that CAFs were pivotal contributors to PD-L1/2 expression in the TME during anti-PD1 treatments. Theoretically, anti-PD1 blocks PD1/L1-mediated effects, leading us to hypothesize that, in the case of NCIT resistance, PD-L2 might be responsible. TCGA analysis confirmed that PD-L1 expression is relatively low in CAFs ([237]Figure S8D), and anti-PD1 treatment did not further up-regulate PD-L1 expression on CAFs ([238]Figure 5G). Therefore, we focused on PD-L2 expression fluctuations in apCAFs using the previously mentioned PDTF obtained during NCIT. Additionally, PD-L2^+ apCAFs were significantly enriched in post-non-MPR specimens ([239]Figures 5H and 5I). After anti-PD1 treatments ex vivo, their fractions increased further in both pre- and post-non-MPR ([240]Figures 5H and 5I). As mentioned earlier, PD-L2 is exclusively expressed by apCAFs, whose expansion depends on IFN-γ. Since IFN-γ was reported to be responsible for PD-L2 expression on tumor cells, we investigated whether this was also the case for CAFs.[241]^45 Additionally, IFN-γ significantly upregulated HLA-DRA and PD-L2 expression on CAFs ([242]Figure 5K). Neutralization of IFN-γ, knockdown of IFNGR, or inhibition of JAK1/2 or STAT1 largely rescued these effects ([243]Figures 5K and [244]S5D). Notably, neither other CAF subsets such as inflammatory CAFs (iCAFs) and myofibroblast CAFs (myCAFs) nor co-stimulatory molecules (CD80/86) were controlled by the IFN-γ signaling pathways in CAFs ([245]Figure 5K). Western blot (WB), ICC, and mFC analyses of patient-derived CAFs cell lines further corroborated our RT-qPCR findings ([246]Figures 5J, 5L, and [247]S8E). We further used a human fibroblast cell line (HFL1) and a murine fibroblast (MF) to double-check our results, which remained consistent ([248]Figures S8F and S8G). Altogether, we concluded that anti-PD1 stimulated PD-L2^+apCAFs expansion in an IFN-γ-dependent manner, offering perspectives on apCAFs-Tregs interactions. Reprogramming apCAFs and anti-PD-L2 (clone 3.2) overcame anti-PD1 resistance No detailed research has been conducted on the specific regulatory axis responsible for PD-L2^+apCAFs until now. We discovered that IFN-γ can stimulate PD-L2^+apCAFs via the JAK1/2-STAT1-IFI6/27 pathway ([249]Figure 6A). Through WB and RT-qPCR analyses of patient-derived CAF cell lines, we confirmed that IFN-γ directly upregulated the phosphorylation of the JAK1/2-STAT1 pathway ([250]Figures 6B and 6C). Ruxolitinib, a selective JAK1/2 inhibitor, significantly inhibited the total amount of JAK1/2 and STAT1 protein expression, as well as their phosphorylation ([251]Figures 6B and 6C). Similarly, fludarabine, a STAT1 activation inhibitor, showed the same effects ([252]Figures 6B and 6C). Additionally, emapalumab, an IFN-γ neutralization antibody, reduced JAK1/2-STAT1-IFI6/27 expression at the RNA level to some extent ([253]Figure 6C). We further verified these findings in the HFL1 cell line ([254]Figures S9A and S9B). We previously identified IFI6/27 as markers of apCAFs through multi-omics analysis, which were traditionally considered downstream responders to IFN-α/β-mediated STAT1 activation.[255]^46 However, few studies have reported their correlation with IFN-γ signaling. Using RT-qPCR and ICC, we confirmed that JAK1/2-STAT1 also controls the expression of IFI6/27 ([256]Figures 6C and 6D). On the other hand, since the JAK3-STAT3 axis is also under IFNs’ control and receptors for IFNα/β were exclusively enriched in apCAFs, we questioned whether there could be bypass activation of STAT3 or IFNα/β receptors signaling pathways in PD-L2^+apCAFs. However, neither IFNAR1/2 inhibitors (anifrolumab and IFNAR-IN-1) nor the STAT3 inhibitor (NSC74859) could reverse the effects of IFN-γ ([257]Figures S9C and S9D). Finally, inhibition of the IFN-γ-JAK1/2-STAT1 pathway significantly downregulated PD-L2 and HLA-DRA expression on CAFs ([258]Figures 5J–5L). In general, we uncovered that the IFN-γ-JAK1/2-STAT1-IFI6/27 axis was responsible for PD-L2^+apCAF expansion. Figure 6. [259]Figure 6 [260]Open in a new tab Disruption of the JAK1/2-STAT1-IFI6/27-PD-L2 axis in apCAFs to compromise FOXP1^+Tregs (A) Diagram of mechanism of IFN-γ on apCAFs. (B) The protein expression levels of JAK-STAT pathway. Data are representative of three independent experiments. (C) Heatmap showing the mRNA expression level for genes from JAK-STAT pathway assessed by RT-qPCR. Each specific column means a specific patient’s CAF cell line. For every gene, the difference between any two treatment groups is significant, determined by two-way ANOVA test. (D) Representative ICC pictures of IFI6/27, p-STAT1, and p-JAK1 pathway. Scale bar, 50 μm. (E) Tumor growth curves: tumor cells + apCAFs + anti-PD1 (light blue), tumor cells (light gray), tumor + apCAFs (gray), tumor + anti-PD1 (navy blue), tumor + apCAFs + anti-PD-L2 (pink), and tumor + apCAFs + anti-PD1 + anti-PD-L2 (magenta). Individual and mean tumor volume over time. n = 5 mice per group, representative of three independent experiments. (F) The tumor photos of mice in (E). n = 4 mice per group. (G) Representative flow cytometric scatterplots of FOXP1^+Tregs in mice tumors and the quantification. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (H and I) Representative ridge plots. For (D)–(I), data are representative of three independent experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. Aforementioned work led us to identify interventions targeting apCAFs. Considering methods targeting the IFN-γ-JAK1/2-STAT1-IFI6/27 axis may not be cost-effective or easily translatable ([261]Figures S10A–S10H), we next investigated whether anti-PD-L2 (clone 3.2) could overcome anti-PD1 resistance in an apCAF-enriched TME. Besides, anti-PD-L2 significantly decelerated tumor growth even in the presence of apCAFs ([262]Figures 6E, 6F, [263]S11A–S11C, and S11F–S11H). Furthermore, anti-PD-L2, or the combination of anti-PD-L2 and anti-PD1, largely rescued the expansion of FOXP1^+Tregs and total Tregs ([264]Figures 6G, [265]S11D, and S11I), resulting in reactivation of CD8^+ TILs ([266]Figures S10H, [267]S11E, and S11J). Interestingly, in ex vivo co-culture assays, although αMHCII could diminish Treg expansion to some extent, it had limited effects on FOXP1^+ Tregs ([268]Figures 6H and 6I). In contrast, the addition of anti-PD-L2 further reduced Treg formation and significantly inhibited FOXP1^+Treg expansion, indicating that FOXP1^+Tregs might be specifically induced by the PD-L2 signaling pathway ([269]Figures 6H and 6I). Moreover, both in vivo and in vitro, anti-PD-L2 combined with anti-PD1 restored the anti-tumor capacity of CD8^+ TILs, which was originally dampened by the apCAF-FOXP1^+Tregs axis. Altogether, we concluded that, since a considerable amount of PD-L2 was attributed to apCAFs, apCAF-enriched TME resistant to anti-PD1 could significantly benefit from the addition of anti-PD-L2 (clone 3.2). The PD-L2-RGMB axis was indispensable for apCAFs-FOXP1^+Tregs interaction Recently, Sharpe et al. identified RGMB as another indispensable binding partner of PD-L2.[270]^22 They found that TILs from mice transplanted with gut microbiome from non-responders to anti-PD1 had higher expression of RGMB. The clone 3.2 of anti-PD-L2 blocks both PD-L2-RGMB and PD1-PD-L2 interactions, thus synergizing with anti-PD1.[271]^22 We also found that, in an apCAF-enriched TME, anti-PD-L2 (clone 3.2) could significantly reverse resistance to anti-PD1. Since these effects were accompanied by a reduction in FOXP1^+Tregs, we hypothesized that RGMB might be involved in apCAFs-FOXP1^+Tregs communication. Currently, the differences in RGMB expression among major clusters of TILs remain unclear. Interestingly, we found that CD4^+ TILs exhibited higher RGMB expression compared to B cells, CD8^+ T cells, or natural killer cells in the NSCLC TME ([272]Figure 7D; [273]Figure S12A). Additionally, Tregs showed significantly higher RGMB levels compared to Tconv and CD8^+ TILs ([274]Figure 7B). This indicates that Tregs are the major contributors to RGMB expression among TILs. Next, our analysis of publicly available RNA-seq data of FOXP1^KO Tregs not only confirmed that FOXP1 is essential for FOXP3 but also revealed that FOXP1 might positively regulate RGMB ([275]Figure 7C). We verified these findings through mFC analysis, which showed that CD4^+ TILs indeed exhibited significantly higher RGMB expression ([276]Figure 7E). Although apCAFs did not up-regulate RGMB expression on FOXP1^+Tregs ([277]Figure 7G), RGMB expression on FOXP1^+Tregs was notably higher than that on FOXP1^−Tregs ([278]Figure 7F). Furthermore, anti-PD1 treatment increased the fraction of RGMB^+Tregs ([279]Figure 7G). Finally, we observed increased chromatin accessibility of RGMB, IL-1R1, and FOXP1 in Tregs compared to CD8^+ T cells or Tconv, highlighting their potential influence on Treg formation[280]^34 ([281]Figures 7H and [282]S12B). Figure 7. [283]Figure 7 [284]Open in a new tab Blockade of PD-L2-RGMB interaction to overcome anti-PD1 resistance both in vivo and in PDTF platforms (A) Diagram of interaction between apCAFs and Treg. (B) Expression of RGMB in GEO: [285]GSE253540 datasets (left). (C) Expression of Foxp3 and RGMB in aTreg (activated Tregs) and rTreg (rested Tregs) either with knockout (KO) of FOXP1 or not in the merged GEO: [286]GSE118595 and GEO: [287]GSE121251 datasets. (D) Expression of RGMB among immune cells from TCGA and LUAD datasets. p values are calculated using Wilcoxon test. (E) Expression of RGMB in CD4^+T cells and CD8^+T cells. Data are representative of three independent experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (F) Expression of RGMB in FOXP1^+ and FOXP1^− Tregs. Data are representative of three independent experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (G) Representative ridge plots of RGMB^+ cells (upper) and quantification (lower). Data are representative of three independent experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (H) Re-analyzation results for Foxp1, Il1r1, and RGMB in Treg, Tconv, or CD8^+ T cells in the GEO: [288]GSE211062 assay for transposase-accessible chromatin by sequencing (ATAC-seq) dataset. (I) Tumor growth curves: tumor + anti-PD1 (gray), tumor apCAFs + anti-PD1 (orange), tumor cells + apCAFs + anti-PD-L2 + anti-RGMB (blue), and tumor cells + apCAFs + anti-PD-L2 + anti-RGMB (light blue). Individual and mean tumor volume over time. n = 5 mice per group, representative of three independent experiments. (J) The tumor photos of mice in (I). n = 4 mice per group. (K) The tumor weight of mice in (I). n = 4 mice per group. (L) Representative flow cytometric scatterplots of FOXP3^+Treg and FOXP1^+FOXP3^+Treg in mice tumors in (I) (left) and the quantification of them (right). Data are representative of three different experiments. p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. (M and N) Representative flow cytometric scatterplots of GZMB^+CD8^+T cells (M) and FOXP1^+FOXP3^+Tregs (N) in PDTF (left) and the quantification of them (right). For (B)–(C), p value is calculated using unpaired Student’s t test. For (I)–(K), data are representative of three independent experiments, p values are determined using unpaired Student’s t test, and error bars show the mean and SEM. For (L)–(N), data are representative of three independent experiments, and p values are determined using unpaired Student’s t test and error bars show the mean and SEM. In the apCAF-enriched TME, anti-RGMB could also overcome resistance to anti-PD1 ([289]Figures 7I–7K). When combined with anti-PD1 or anti-PD-L2 (clone 3.2), anti-RGMB significantly reduced tumor growth and volumes ([290]Figures 7I–7K, [291]S13A–S13C, and S13F–S13H), as well as the fractions of FOXP1^+Tregs and total Tregs ([292]Figures 7L, [293]S12C, [294]S13D, and S13I). Besides, either in vivo or in the PDTF platforms, the addition of anti-RGMB enormously increased GZMB secretion by CD8^+ TILs ([295]Figures 7M, [296]S13E, and S13J). Although the shrinkage of FOXP1^+Tregs under anti-RGMB was not significant in vitro (Figure 7N), likely due to the absence of the gut microbiome, we could still conclude that anti-RGMB together with anti-PD1/L2 managed to successfully overcome apCAF-mediated resistance to anti-PD1. Discussion NCIT has provided a valuable opportunity to investigate the mechanisms underlying immunotherapy resistance. Accordingly, we conducted a multi-omics analysis of NSCLC patient samples treated with NCIT. Our focus on CAFs led to the identification of apCAFs as key mediators of the resistance to NCIT. Although apCAFs had been previously identified, the mechanisms of their formation remained poorly understood. In PDAC, apCAFs were thought to differentiate from mesothelial cells.[297]^17 Later, Jiang et al. demonstrated that IL-1R2^+Tregs could maintain apCAFs by inhibiting IL-1-IL-1R1 signaling in colon cancers.[298]^18 However, our single-cell data of NSCLC revealed comparable IL-1R1 expression on both Tregs and apCAFs, suggesting that additional factors are involved. Using 3D culture settings,[299]^20 we identified IFN-γ as another crucial mediator of apCAF expansion. Furthermore, we clarify that the JAK1/2-STAT1-IFI6/27 axis is the primary pathway driving apCAF expansion. This finding significantly advances the understanding of apCAFs. IFNs have traditionally been considered crucial to anti-tumor immunity.[300]^47 On the contrary, we found that IFN-γ can reduce NCIT efficacy by specifically stimulating apCAFs. This discovery stemmed from observing increased IFNs in post-non-MPR samples, which might seem counterintuitive since IFNs are typically considered markers of “hot” tumors. However, recent studies demonstrate that “hot” tumors are actually more susceptible to acquired immunotherapy resistance.[301]^48 For example, IFN-γ can promote Yes-associated protein (YAP) phase separation in cancer cells,[302]^49 leading to resistance to anti-PD1. What is more, Hellmann et al. found persistent IFN-γ signaling in the TME of stage IV NSCLC patients with acquired anti-PD1 resistance.[303]^50 Our research further confirms that the IFN-γ-apCAF axis contributes to NCIT resistance in stage I/II NSCLC as well. Both our study and Hellmann et al.’s[304]^50 support the idea that a persistently inflamed, rather than immune-desert, TME might drive acquired immunotherapy resistance. We further identified a potential mechanism of IFN-γ-mediated immunotherapy resistance: the exclusive expression of PD-L2 on apCAFs. PD-L2^+ CAFs manifested cancer-type-specific, context-dependent roles. PD-L2 expressed by CD29^+FAP^+ CAFs mediate Treg retention in breast cancer,[305]^51 while PD-L2^+CAFs accelerate CD8^+ T cells’ apoptosis in melanoma.[306]^52 However, the relationship between apCAFs and PD-L2 in NSCLC was still unclear. We revealed that apCAFs, not other CAF subtypes, are the primary source of PD-L2 in the NSCLC TME. Although anti-PD1 blocked the PD1/PD-L2 axis, the PD-L2/RGMB axis emerged to dampen anti-PD1 efficacy. Sharpe et al. reported that RGMB on T cells contributes to T cell anergy.[307]^22 We further confirmed that FOXP1^+Tregs are the main contributors to RGMB within TILs. FOXP1 is indispensable for FOXP3 chromatin accessibility and Treg homeostasis.[308]^36^,[309]^37^,[310]^38 However, the role of FOXP1 in Tregs within the context of NCIT was previously unknown. We discovered that FOXP1 acts as a TF marker for a post-non-MPR-responsive Treg subcluster, IL-1R1^+Tregs, and may positively regulate RGMB expression on Tregs. Interestingly, as previously reported, FOXP1 could facilitate CTLA-4 expression on Tregs,[311]^38 and RGMB binding enhanced its suppressive activity.[312]^53 Taken together, we believed that, as the “executioner” of apCAF-mediated resistance to NCIT, RGMB^+FOXP1^+Tregs warrant further investigation. Lastly, our research highlights pivotal translational aspects and future research directions. First, in co-culture assays, we found that inhibiting the JAK1/2-STAT1 pathway in CAFs not only reduced PD-L2 expression but also restored CD80/86, thereby reversing FOXP1^+ Treg expansion through co-stimulatory signaling. This suggests another potential strategy to counter apCAF-mediated resistance, though further in vivo validation is needed. For instance, the JAK1/2 inhibitor ruxolitinib has shown synergy with nivolumab in phase 1/2 trials for NSCLC and Hodgkin’s lymphoma.[313]^54^,[314]^55 Thus, whether JAK1/2 inhibitors may also be able to reprogram apCAFs in vivo warrants further investigation. Finally, we also observed enrichment of apCAFs in pre-non-MPR, suggesting additional mechanisms for apCAF residency before anti-PD1 therapies. Given that apCAF expansion is IFN-γ dependent, we hypothesized that certain subclusters enriched in pre-non-MPR with high expression of IFN-γ might contribute to it and identified BAG3^+CD8^+TILs as a key source. Therefore, further investigation into the interplay between apCAFs and BAG3^+CD8^+TILs will be of great significance to further target apCAFs. Limitations of the study The central mechanism we identified—apCAFs driving immunotherapy resistance via FOXP1^+ Treg induction—requires further contextualization within broader immune cell interactions. The role of apCAFs as non-professional APCs in modulating other immune cells, such as CD8^+T cells and dendritic cells (DCs), remains unclear. Therefore, future studies using advanced approaches, such as lineage-specific genetically engineered models, are essential to elucidate the cell-cell communication between apCAFs and immune cells beyond Tregs. These insights could uncover novel therapeutic targets beyond the PD-L2/RGMB axis and further enhance our understanding of apCAF-mediated resistance to neo-ICIs. Resource availability Lead contact Further information and requests for resources and regents should be directed to and will be fulfilled by the lead contact, Dr. Ziming Li (liziming1980@shsmu.edu.cn). Materials availability This study did not generate any new unique reagents, plasmids, or mouse lines. All unique/stable reagents used in this study are available from the [315]lead contact with a completed Materials Transfer Agreement. Data and code availability * • All data generated or analyzed during this study are included in this manuscript (and its supplementary information files). Raw human sequencing data including RNA-seq, DSP spatial sequencing, and scRNA-seq in this paper will be shared by the [316]lead contact upon request. This paper also analyzes existing, publicly available data (deposited data of STAR Methods). Sequencing datasets generated in this article without human information are available publicly (deposited data of [317]key resources table). * • scRNA-seq datasets were deposited into the Zenodo Database: [318]https://zenodo.org/records/14249268. * • DSP spatial sequencing datasets were deposited into the Zenodo Database: [319]https://zenodo.org/records/14249266. * • Bulk RNA-seq of CAF cell lines were deposited into the Zenodo Database: [320]https://zenodo.org/records/14249260. * • This study did not generate any original code. * • Any additional information required to reanalyze the data reported in this paper is available from the [321]lead contact upon request. Acknowledgments