The impact of neoadjuvant chemotherapy on the tumor microenvironment in advanced high-grade serous carcinoma

Single-cell transcriptomic analysis in a patient with advanced stage high-grade serous fallopian tube carcinoma treated with neoadjuvant chemotherapy

The analytical scheme following the clinical course of a 71-year-old female patient with stage IIIC high-grade serous fallopian tube carcinoma is summarized in Fig. 1a. The staging and diagnosis of this patient were initially confirmed based on CT imaging plus laparoscopy to evaluate the feasibility of resection and obtain histological biopsy specimens (Fig. 1b, c). We confirmed that this patient was unlikely to be optimally cytoreduced by the assessment laparoscopy. NACT was a better initial treatment option. Then, we collected the pre-NACT tumor samples from the peritoneum and greater omentum, as well as a small volume of ascites (Fig. S1a). After undergoing two cycles of NACT regimens (carboplatin and paclitaxel), the patient received IDS. Dosing per cycle was shown as follows: (1) Cycle 1, carboplatin 535 mg (AUC 5) and paclitaxel 283 mg (175 mg/m2); (2) Cycle 2, carboplatin 503 mg (AUC 5) and paclitaxel 284 mg (175 mg/m2). We collected the post-NACT tumor samples from the colon serosa surface, greater omentum, and lesser omentum at the time of IDS (Fig. S1a). This patient was characterized as a high-grade serous fallopian tube carcinoma based on the H&E staining, exhibiting strong expression of PAX8, Ber-EP4, CA125, Ki-67, p16, WT1, and mutant p53 protein (Figs. 1c, d and S1b). Genetic risk evaluation showed that this patient did not have a BRCA1/2 mutation in germline or tumor DNA.

Fig. 1: Overview of the treatment schedule and characteristics of an advanced-stage high-grade serous fallopian tube carcinoma.figure 1

a Neoadjuvant chemotherapy treatment and sample acquisition for scRNA-seq. b CT imaging showing the clinical responses to NACT of the patient with high-grade serous fallopian tube carcinoma. c H&E staining of the pre-NACT and post-NACT samples. d IHC staining of PAX8, Ber-EP4, CA125, Ki-67, and p16 protein in pre-NACT and post-NACT tissue samples of the high-grade serous fallopian tube carcinoma.

Single cell RNA (scRNA) sequencing using the 10 x Genomics Chromium platform was performed on these matched longitudinal specimens from this patient to assess the cell compositions of the tumor environment in response to NACT. After stringent filtering, a total of 32,079 cells from the six samples, with 17,249 cells derived from the pre-NACT multisite tumor tissue samples and 14,830 cells derived from the post-NACT multisite tumor tissue samples, were retained for further analysis (Fig. 2a). The cell distribution of each sample was shown in Fig. S2. We conducted clustering to define 22 clusters that were visualized using uniform manifold approximation and projection (Fig. 2b and S3a). Copy number variation (CNV) analysis was used to distinguish malignant and non-malignant cells (Fig. 2c). These cells were assigned to seven distinct cell types using known marker genes (Figs. 2d, e and S3b): macrophages (marked with SPP1, APOE, C1QA, C1QB, and APOC1, and MS4A7); T cells (marked with CCL5, GZMA, NKG7, TRAC, GZMK, CD3D, CD3E, CD8A, and CD4); malignant epithelial cells (marked with KRT18, CLU, WFDC2, KRT8, KRT7, and EPCAM); stroma cells (marked with COL1A1, COL1A2, COL3A1, SPARCL1, DCN, and LUM); endothelial cells (marked with RAMP2, VWF, and CLDN5); smooth muscle cells (marked with RGS5 and MGP); and B cells (marked with IGLC2, CD79A, IGHM, MS4A1, and IGKC). Although the proportion of each cell type varied greatly by sample, we found that epithelial cells were enriched in the post-NACT samples, whereas T cells were relatively enriched in the pre-NACT samples (Fig. 2f). Furthermore, we applied single-cell regulatory network inference and clustering (SCENIC) to assess the transcription factors underlying the differences in the expression among different cell types. This identified a set of upregulated transcription factors, such as FHL2, FOXQ1, ID4, KLF5, MYC, NR2F6, OVOL2, PAX8, BARX2, SOX9 in epithelial cells, SOX4 in stromal cells, SOX7 and SOX18 in endothelial cells, ATF3, DDIT3, IRF8, JDP2, KLF10, NFATC1, NR1H3, PRDM1 and SNAI1 in macrophages, FOXP3, MEOX1 and MSX1 in T cells, and SPIB in B cells (Fig. 2g).

Fig. 2: scRNA-seq profiling of the pre-NACT and post-NACT high-grade serous fallopian tube carcinoma samples.figure 2

a UMAP plots showing cell groups by color in the pre-NACT and post-NACT sample groups. b UMAP plots showing pre-NACT and post-NACT cells, clustered and color-coded according to the group. c Box plots showing the CNV signals for each cell type. d UMAP plots showing the cell types by color. e Heatmap showing the top marker genes in each cell type. f Histogram indicating the proportion of cell types in each analyzed sample. g Heatmap showing the expression regulation by transcription factors in each cell type, as estimated using SCENIC. UMAP uniform manifold approximation and projection.

Distinct features and adaptive clonal evolution of malignant cells in response to NACT

To define the major subpopulation structure of the malignant epithelial cells, we performed MNN clustering on our scRNA-seq data, identifying 11 main subclusters with a panel of specific marker genes (Fig. 3a, b). We compared each subcluster before and after NACT (Fig. 3c). However, we did not find a new malignant subcluster that conferred a chemoresistant phenotype induced by NACT. All the subclusters were already present in the pre-NACT tumors. Hallmark analysis revealed that subclusters 0, 1, 2, and 5 were enriched in TGF-β signaling, WNT/β-catenin signaling, angiogenesis, and epithelial-mesenchymal transition (EMT) (Fig. 3d). Strikingly, we noticed that the vast majority of four subclusters were derived from post-NACT D, E and F tumor cells (Fig. 3e), suggesting that they might share a resistant genotype. We further observed that these cells from the pre-NACT tumors were primarily detected in ascites (Fig. 3e). The ascites also possessed more malignant epithelial cells compared with those in other tumor samples in the pre-NACT group (Fig. 3f). Moreover, enrichment of stem cells in subcluster 0, 1, 2 and 5 was also identified based on the cytoTRACE analysis (Fig. 3g). We showed that the drug resistance scoring was significantly increased in these subclusters (Fig. 3h). FOS and MYC were determined to be the underlying transcription factors contributing to drug resistance (Fig. 3i). Kaplan-Meier survival analysis showed that higher expression of FOS and MYC in HGSC data obtained from TCGA was associated with shorter OS times (Fig. 3j). Collectively, our findings suggest that chemoresistance may arise due to the selection and expansion of pre-existing subcluster malignant cells.

Fig. 3: The subtypes of epithelial cells in pre-NACT and post-NACT tumors.figure 3

a UMAP projections of subclustered epithelial cells, labeled in different colors. b Heatmap showing the expression of marker genes in each indicated cell subcluster. c UMAP projection showing pre-NACT and post-NACT cells, clustered and color-coded according to the group. d Heatmap indicating the primary hallmark pathways in each subcluster. e Histogram indicating the proportion of subcluster cells in each analyzed sample. f UMAP plots showing the malignant and non-malignant epithelial cells colored by red or blue in each tumor sample. g CytoTRACE analysis of epithelial cells in each subcluster. h UMAP plots showing distinct drug resistance scores in each subcluster. i Gene bubble plots showing different expression levels of drug resistant-related transcription factors in each subcluster. j Kaplan–Meier survival curves showing the association of FOS and MYC expression with overall survival in patient with HGSCs (from TCGA). Log-rank p values are shown.

To confirm the adaptive resistance hypothesis, we further conducted lineage tracing analysis using VarTrix (Fig. 4a). The phylogenetic trees showed four major subclones in malignant cells, including subclone 546, 319, 358, and 63 (Fig. 4a). Among these, analyses of HGSC cohorts from TCGA supported the enrichment of the top 56 marker genes associated with clone 63 cells as a significant indicator of adverse clinical outcomes in HGSC patients (Fig. 4b–d and Table S1). The expression of the gene signature was correlated with a significantly poorer clinical prognosis, including a shorter disease-free interval, progression-free interval, and OS time. Furthermore, the cell subcluster and tissue location distribution of all the clones was analyzed (Fig. 4e, f). The clusters 3 and 10, containing less than 50 epithelial clone cells, were excluded from the analysis. We found that the main clones 63, 546, 319, and 358 were persistent in both pre-NACT and post-NACT samples, and clone 63 was further enriched in the post-NACT samples (Fig. 4f), consistent with adaptive resistance. Of note, we observed that the primary pre-NACT tumor cells in the greater omentum were mostly eliminated in this clone. The tumor cells in the post-NACT greater omentum sample were primarily derived from pre-NACT ascites and the peritoneum tumor cells. In addition, we conducted evolutionary tree analysis based on the information of epithelial subclusters and tissue samples (Fig. 4g). We found that no matter which sample B cells (from pre-NACT greater omentum tissues) were evolved from in clones, they were convergent, whereas other samples had obviously mixed with each other, and there was no sample convergence. From the perspective of the composition of the clones, our present study supports the notion that the cells of post-NACT omentum tumors were not the same as pre-NACT omentum tumors, but more likely originated from the other pre-NACT samples. This may suggest a novel adaptive resistance theory for HGSC.

Fig. 4: Analysis of clonal evolution in high-grade serous fallopian tube carcinoma tissues following NACT treatment.figure 4

a Phylogenetic trees showing the analyses of the clonal dynamics calculated from each cell type, and the subpopulation-specific differences in each clone are indicated with color-coded bars. Kaplan–Meier survival curves for OS (b), DFI (c) and PFI (d) from TCGA HGSC data showing significant prognostic separation according to the clone 63 marker gene signatures from our scRNA-seq data. Log-rank p values are shown. e Clonal frequencies of the eight main clone cells are annotated in each epithelial subcluster. f The cell number distribution of the eight main clones in each tumor sample. g Evolutionary tree analysis showing the distribution of epithelial subclusters and corresponding tissue samples. DFI disease-free interval; PFI progression-free interval.

Fibroblasts from post-NACT tumors show functional alterations compared with fibroblasts from pre-NACT tumors

Further clustering in the fibroblast compartment gave rise to 3 cell subpopulations (Fig. S4a and b), of which one fibroblast cluster expressed high levels of ACTA2, POSTN, and HOPX, confirming their identity as myCAFs; another cluster expressed major MHC-II genes such as HLA-DRA, CD74, and HLA-DRB1, and was, therefore, termed them apCAFs; and a cluster expressed high levels of CFD, DPT, and CXCL12, and was considered to be inflammatory CAFs (iCAFs) (Fig. S4b). Interestingly, myCAFs and iCAFs were mainly enriched in post-NACT tissues, whereas apCAFs were mainly present in pre-NACT tissues (Fig. S4a). Moreover, apCAFs were related to response to IFN (Fig. S4c and S4d), while myCAFs indicated significant enrichment for WNT/β-catenin signaling (Fig. S4e).

Macrophage subclusters between pre-NACT and post-NACT tumors

Specific tumor-associated macrophage (TAM) subtypes have important impacts on ovarian cancer progression and therapy [27,28,29]. For instance, M2-like TAMs limit the effector function of CD8+T cells in metastatic HGSCs and associated with poor overall survival [27]. Thus, we studied the macrophage subclusters in our samples. According to the top differentially expressed genes and known marker genes, macrophage subclusters were designated as tissue-resident-, glycolysis related-, M1-, M2-, and cycling macrophages (Fig. S5a–c). Of note, M1 and M2 macrophages were mainly enriched in post-NACT tissues, whereas tissue-resident and glycolysis-related macrophages were mainly present in pre-NACT tissues (Fig. S5d).

Features of CD4+ Treg subtypes in response to NACT

The clustering of T/NK cells of the tumor environment revealed 5 main populations, including NK/NKT subtype (GNLY and TRDC), γδ T cells (GNLY, TRDC, and CD3D), CD4+CD8+ T cell subtype (CD3D, CD8A, and CD4), CD8+ T cells (CD3D and CD8A), and CD4+ T cells (CD3D and CD4) (Fig. 5a, b). The CD4+ and CD8+ T cell infiltration into the stroma and tumor epithelium was further determined by CD4 and CD8 IHC staining (Fig. S6).CD8+ T cells were further designated as CD8 TEM, CD8 TEMRA/TEFF, CD8 TRM, CD8 Naïve, CD8 IFN response, and CD8 Cycling cell subtypes, according to their marker genes (Fig. S7a, S7b). All these subtypes were shared across tumors and between pre-NACT and post-NACT samples (Fig. S7c). Of note, CD8 Naïve and CD8 IFN response cells were mainly enriched in pre-NACT samples (Fig. S7c).

Fig. 5: Distinct Treg subpopulations detected in pre-NACT and post-NACT tumors.figure 5

a UMAP plots showing the subtypes of T/NK cells, labeled in different colors. Subtype annotations are indicated in the Figure. b Violin plots showing selected marker genes in distinct T/NK cell subtypes. c UMAP plots showing the subtypes of CD4+ T cells from pre-NACT and post-NACT tumors. Each subcluster is color-coded. Subtype annotations are indicated in the Figure. d Heatmap depicting marker gene enrichment for each cell subtype of CD4+ T cells. e UMAP plots showing the color-coded cell groups of CD4+ T cells in response to NACT. f UMAP plots showing four main cell subtypes of Tregs, labeled in different colors. g Dot plots showing the expression levels of specific genes in each Treg subtype. h Metabolic pathway analysis showing the enrichment of Glycolysis/Gluconeogenesis and the citrate cycle in each Treg subtype. i KEGG pathway analysis showing the primary enriched pathways of IL2RAhi-CCL22+-Treg cells. j Cell communication analysis showing the overlapping relationship between specific pre-NACT and post-NACT tumor samples.

We then investigated the CD4+ T cell heterogeneity to determine their contribution to NACT responses, and identified 5 main subtypes of CD4+ T cells, including CD4 TEM/Th1-like, CD4 TEMRA/TEFF, CD4 Naïve, CD4 Treg, and CD4 Cycling cells (Fig. 5c, d). A high diversity of CD4+ T cells was observed between pre-NACT and post-NACT tumors (Fig. 5e). We found that the subclusters of Tregs displayed distinct enrichment in response to NACT (Figs. 5c, e). Thus, we re-clustered CD4 Tregs and further identified four states of Tregs, including IL2RA-high and CCL22 positive subtype (IL2RAhi-CCL22+-Treg), IL2RA-high subtype (IL2RAhi-Treg), Type 1 IFN positive subtype (Type1IFN-Treg), and IL2RA-Low subtype (IL2RAlo-Treg) (Fig. 5f), together consisting 34.3% of the tumor-infiltrating CD4+ T cells (Fig. 5c). These four Treg states were mainly distinguished by higher expression of known immune checkpoints IL2RA, TNFRSF4/9/18, and CD27 in IL2RAhi-Treg cells, and plus CCL22 in IL2RAhi- CCL22+-Treg cells (Figs. S8a and 5g). The gene features of each state are shown in Fig. 5g.

Cell metabolism is appreciated as a key regulator of T cell function and fate [30,31,32]. We noticed an extremely high expression of the glycolytic enzyme lactate dehydrogenase A (LDHA) in IL2RAhi-CCL22+-Tregs (Fig. S8b), which indicates LDHA might affect the function of Tregs. Consistently, previous studies reported that LDHA plays a key role in the altered glycolytic metabolism [33]. Therefore, we hypothesized that the glycolytic metabolism be important for those immune effector cells in the tumor. The metabolic features were further analyzed for those subclusters. As shown in Fig. 5h, when compared with the other three subtypes, Glycolysis/Gluconeogenesis was significantly enriched in IL2RAhi-CCL22+-Treg cells, while Citrate cycle was not notably increased, indicating an hypoxic environment with the high expression of LDHA. In addition, pathway analysis of the IL2RAhi-CCL22+-Treg cells also demonstrated that Glycolysis/Gluconeogenesis was the primarily enriched pathway (Fig. 5i). Interestingly, cell communication analysis indicated that the features of the post-NACT samples D, E, and F were more similar with pre-NACT sample A and C than with sample B (Fig. 5j), indicating that these Tregs were present in the pre-NACT tumors and were primarily derived from ascites and the peritoneum, and persisted in all residual tumor samples after NACT therapy.

Cell cross-talk between immune cells and tumor microenvironment

Next, we investigated the correlation between specific Treg subtypes and other cell types to explore the potential functional roles of Tregs. We first conducted cell communication analysis to determine which cell types could affect the enrichment of IL2RAhi-Tregs and IL2RAhi-CCL22+-Tregs (Fig. 6a). Interestingly, we observed that the IL2RAhi-CCL22+-Treg cells could recruit and enrich themselves through secreting the CCL22-CCR1 combination in all tumor samples. We next determined whether the two Treg subtypes could also affect other cell types in HGSC (Fig. 6b). Notably, we observed that these IL2RAhi-CCL22+-Treg cells from the post-NACT D, E, and F samples could express CD274 to suppress other CD4 and CD8 T cells through CD274-PDCD1 axis. Moreover, we found that the IL2RAhi-CCL22+-Tregs from pre-NACT sample A and post-NACT samples D, E, and F could secrete VEGFA to promote angiogenesis of endothelial cells via VEGFA-KDR and VEGFA-FLT1 signaling. In addition, both IL2RAhi-Treg and IL2RAhi-CCL22+-Treg cells could express PDGFA to promote the growth of CAFs through PDGFA-PDGFRA interaction.

Fig. 6: Analysis of cell communication and CD4+ Treg cell transition states in pre-NACT and post-NACT tumors.figure 6

a Dot plots showing chemokine-receptor communication between distinct cell types and Treg subtypes in the pre-NACT and post-NACT tumor microenvironment. b Dot plots showing ligand-receptor pair analysis of the interactions between IL2RAhi-CCL22+-Treg or IL2RAhi- Tregs and distinct cell types in the pre-NACT and post-NACT tumor microenvironment. c Heatmap showing the dynamic changes in gene expression along the pseudotime. The enriched pathways are labeled by colors. d Heatmap showing the dynamic changes in transcription factor expression along the pseudotime.

Pseudotime trajectory shows the characterization of distinct CD4+ Treg subtypes

We further explored the dynamic cell transitions by inferring the state trajectories using Monocle (Fig. 6c). This pseudotime analysis revealed that the IL2RAlo-Treg cells were present at the beginning of the trajectory path, whereas the IL2RAhi- CCL22+-Treg cells and Type1IFN-Treg cells were present at the two different terminal stages. One transition was determined to initiate with IL2RAlo-Tregs, through an intermediate IL2RAhi-Treg state characterized by enrichment of immune response, apoptosis, T cell activation, T cell co-stimulation, myeloid dendritic cell differentiation, and toll-like receptor signaling pathway, and finally reach the IL2RAhi- CCL22+-Treg stage, characterized by enrichment of NIK/NF-κB signaling, canonical glycolysis, glycolytic process, regulation of apoptosis, and the MAPK cascade (Fig. 6c). The main branch expression analysis modeling (BEAM) genes for IL2RAhi-CCL22+-Tregs were HAVCR2, CTLA4, TIGIT, TNFRSF4, TNFRSF9, and LAG3 (Fig. S9). The transcription factors enriched in each cell state were further analyzed. As shown in Fig. 6d, we revealed that STAT1, LEF1, IRF7, POLR2J3, and ETS1 contributed to the transition from IL2RAlo-Tregs to Type1IFN-Tregs, while ID3, FOSL2, NR4A1, ID2, REL, and VDR contributed to the transition from IL2RAlo-Tregs to IL2RAhi-CCL22+-Tregs. Collectively, our results suggest that manipulation of Tregs with high immune checkpoint characteristics might present a novel therapeutic strategy for both primary and chemo-resistant HGSCs.

留言 (0)

沒有登入
gif