Graphical abstract graphic file with name fx1.jpg [45]Open in a new tab Highlights * • PTCL molecular subtypes reveal distinct genetic and transcriptional features * • Four molecular subtypes exhibit potential vulnerabilities to targeted agents * • Four microenvironment subtypes reflect different immune communities __________________________________________________________________ Huang et al. integrate the genomic and transcriptomic data from 221 patients of peripheral T cell lymphoma to establish 4 molecular and microenvironment subtypes with distinct features and therapeutic responses. These findings suggest potential precision medicine approaches that target both tumors and the microenvironment in this hard-to-treat disease. Introduction Peripheral T cell lymphoma (PTCL), comprising ∼15%–20% of all aggressive non-Hodgkin’s lymphomas, is characterized by malignant proliferation of post-thymic lymphocytes and heterogeneous clinicopathological manifestations.[46]^1 The most frequent subtypes include PTCL not otherwise specified (PTCL-NOS; ∼35%), angioimmunoblastic T cell lymphoma (AITL; ∼15%–42%), and anaplastic large cell lymphoma (ALCL; ∼15%), which can be further divided into anaplastic lymphoma kinase (ALK) fusion-positive ALCL (ALK+ ALCL) and ALK fusion-negative ALCL (ALK− ALCL).[47]^2^,[48]^3 Except for ALK+ ALCL, PTCL exhibits a poor response to standard anthracycline-based cyclophosphamide, doxorubicin, vincristine, and prednisolone (CHOP) chemotherapy, resulting in a 5-year survival of less than 30% for most pathological subtypes.[49]^4 Consequently, effective targeted therapies are an urgent need for this lethal disease, which has long been impeded by the dearth of knowledge of the genetic landscape of PTCL. Previous studies have suggested a complex pathobiology underlying the heterogeneity of PTCL.[50]^3 Various intrinsic culprits dysregulate essential pathways in mature T cells, thus promoting malignant transformation.[51]^5 Frequent gene mutations associated with epigenome (e.g., TET2, DNMT3A, and IDH2) and T cell receptor (TCR) signaling (e.g., RHOA) have been detected in AITL, accompanied by activation of the TCR and phosphatidylinositol 3-kinase (PI3K)-AKT pathways.[52]^6^,[53]^7^,[54]^8^,[55]^9 In PTCL-NOS, histone-modifying genes, mainly on methylation and acetylation, are commonly observed.[56]^10 ALK+ ALCL is defined by the NPM1-ALK fusion protein, along with an increased expression of genes related to hypoxia-inducible factor alpha (HIF1-α) and interleukin-10 (IL-10),[57]^11 while ALK− ALCL represents a pathologically distinct but genetically heterogeneous group of disease, all expressing three genes: TNFRSF8, BATF3, and TMOD1.[58]^12 As for the cell of origin (COO), gene expression profiling disclosed a significant enrichment of the T follicular helper (Tfh) cell signature in AITL.[59]^13 In PTCL-NOS, two major molecular subgroups have been delineated, PTCL-TBX21 and PTCL-GATA3, characterized by high expression of Th1 and Th2 cell differentiation regulators, respectively.[60]^11 More recently, a Tfh-originating PTCL-NOS subtype has been identified that is highly similar to AITL[61]^5 and harbors frequent Tfh-relevant mutations.[62]^14^,[63]^15 In addition to intrinsic defects, growing evidence suggests that extrinsic stimuli by cellular and non-cellular components in the lymphoma microenvironment (LME) are highly paramount for tumor cell behaviors and clinical manifestations in PTCL.[64]^3 In AITL, gene expression profiling unveiled an enrichment of B cells and plasma cells as well as a complex cytokine milieu involved in various immune processes, indicating a complicated immunologic network.[65]^11^,[66]^16 High expression of IL-4, IL-6, and IL-21 is associated with the expansion of B cells and plasma cells, likely leading to hypergammaglobulinemia and autoimmune phenomena. Accumulation of tumor-associated macrophages and overexpression ofvascular endothelial growth factor (VEGF), transforming growth factor β (TGF-β), and IL-10 are associated with immunosuppression and frequent Epstein-Barr virus (EBV) infection. Clinically, B cell association is significantly correlated with favorable outcomes, while high VEGF expression is relevant to tumor progression in AITL.[67]^17 In PTCL-NOS, TBX21 expression presents a Th1-driven subset, which features abundant immune cell infiltration, especially macrophage polarization, thereby accounting for unfavorable outcomes.[68]^18^,[69]^19^,[70]^20 In contrast, a minimally inflamed microenvironment was detected in the Th2-originating PTCL-GATA3 subset, which has an even worse prognosis.[71]^20 Another study revealed a microenvironment classification in PTCL-NOS based on gene expression signatures of tumor-infiltrating immune cells, in which the subtype exhibiting both B cell and dentritic cell (DC) signatures shows better prognosis, while the subtype with neither signature represents the worst.[72]^21 Elevated expression of immune checkpoint genes, including PD-L1 and IDO1, was observed in macrophage-enriched cases, implying potential use of immune checkpoint inhibitors (ICIs) in this subtype. Despite the previous efforts, a lack of large-scale RNA sequencing (RNA-seq) in PTCL put limitations on the discovery of potential biomarkers for genetic subtyping and targeted therapies. In this study, by integrating whole-exome sequencing (WES), targeted sequencing, and RNA-seq on 221 PTCL patients, we aimed to portray a comprehensive genomic landscape of mutations as well as establish a molecular stratification based on genetic signatures, biological alterations, and therapeutic responses, proposing four distinct molecular subgroups of PTCL. Additionally, we defined a microenvironment stratification derived from gene expression footprints of infiltrating immune cells and stromal components, which also reflected distinct biological features and clinical properties. This is a multiomics study exploiting RNA-seq data on a large cohort of PTCL, which revealed a series of cancerous as well as microenvironment biomarkers, providing a rationale for personalized treatment of PTCL patients. Results Unique molecular subtypes of PTCL defined by genetic landscape Based on WES data of 101 PTCL patients, we detected a total of 36,625 somatic mutations. Among all mutation genes, 82 candidate genes (SNVs and insertions or deletions [indels]) with predicted functional alterations in tumorigenesis were selected to initiate a gene panel for deep targeted capture sequencing in an additional cohort of 120 PTCL patients. With a combined overview of the results from WES and targeted sequencing, 1,412 non-silent somatic mutations were discovered in 82 genes, which included 1,065 missense, 96 nonsense, 229 indel, and 22 splice-site mutations, showing a significantly high frequency of missense mutations ([73]Figure S1A). Among these 82 genes, an average of 6 mutations were detected in each sample, ranging from 1 to 20 ([74]Figure S1B). SNVs in PTCL were dominated by C>T and T>C transitions, analogous to the somatic SNV spectrum in other cancers ([75]Figure S1C; [76]Table S1). Besides, copy number aberrations (CNAs) analysis was conducted in WES samples, and the results showed that copy number gains/amplifications or losses/deletions were relatively common ([77]Figures S1D and S1E). Consistent with previous studies, the loss of significant genes, including TP53, CDKN2A, and PTEN, was frequently observed ([78]Figure S1F).[79]^5^,[80]^16 With the comprehensive list of mutations in PTCL, 40 genes were designated as recurrently mutated, as they were detected in more than 5% of all cases ([81]Figure 1A). The most frequently mutated genes included TET2 (49%), RHOA (27%), KMT2C (20%), DNMT3A (16%), and KMT2D (12%). Involved pathways included DNA methylation (e.g., TET2, DNMT3A, and IDH2), TCR signaling (e.g., RHOA, CARD11, and FYN), PI3K-AKT signaling (e.g., VAV1, PI3KR1, and ITPR3), histone modification (e.g., KMT2C, KMT2D, CREBBP, and EP300), p53 signaling (e.g., TP53, ATM, and JMY), Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling (e.g., PTPN13 and JAK3), immune surveillance (e.g., HLA-A and HLA-B), chromatin remodeling (e.g., ARID1A and ARID1B), NOTCH signaling (e.g., NOTCH1 and NOTCH3), and tumor suppression (e.g., MGA and CIC). Of note, all patients were covered by mutations of these 10 categories, indicating a complicated pathogenic role of intrinsic defects in PTCL ([82]Table S2). Analysis of pairwise relationships across the recurrent genetic lesions uncovered a series of co-occurring and mutually exclusive mutations ([83]Figure S1G). We discovered strong co-alteration tendencies in Tfh-related genes, including TET2, RHOA, DNMT3A, and IDH2, as well as in the immune-related genes PTPN13 and HLA-B. KMT2C and KMT2D mutations were found mutually exclusive of the aforementioned alterations, providing a hint about subtype-defining genes for categorization. We then performed principal-component analysis (PCA) of the mutation data and identified 3 major molecular subtypes in PTCL associated with genetic alterations, including T1 (60 cases, characterized by TET2 and RHOA mutations), T2 (49 cases, characterized by TET2 without RHOA mutations), and T3 (112 cases, characterized by other mutations with wild-type TET2 and RHOA) ([84]Figure 1B). Moreover, AITL and PTCL-NOS patients could be separated into 3 subtypes by PCA ([85]Figures S2A and S2B). The number of ALK+ ALCL and ALK− ALCL patients was limited to be further subclassified. Figure 1. [86]Figure 1 [87]Open in a new tab Genomic landscape and mutational features of PTCL (n = 221) (A) Landscape of somatic mutations detected by WES (n = 101) and targeted sequencing (n = 120) in 221 patients with PTCL. Mutations in 82 candidate genes (SNVs and indels) with predicted functional alterations in tumorigenesis were ranked by prevalence in all cases. The numbers of mutational burdens in each patient are indicated at the top, and the prevalence of each mutation is depicted on the right. Pathological subtypes and IHC makers (BCL6, CD10, PD-1, and CXCL13) are also shown. (B) PCA of the genetic data and projection of representative clusters. Projection of mutational status along with the first two collective principal components (PC1 and PC2) divided 221 patients into 3 molecular subtypes. The individual cases are colored by subtype, and a 95% confidence ellipse is depicted to define each subtype. (C) Evolutionary tree plot of the unsupervised hierarchical clustering within the T3 subtype, revealing 2 subtypes: T3.1 and T3.2. (D) Bar chart overview of the defining alterations significantly different between T3.1 and T3.2. (E) Frequencies of common genetic mutations across 4 molecular subtypes. The T1 subtype has been extensively investigated due to its distinctive co-existence of TET2 and RHOA mutations. In addition, we explored an increased frequency of other mutations regarding DNA methylation and the TCR signaling pathway, such as DNMT3A, IDH2, and CARD11. On the other hand, the T2 subtype was found to bear frequent mutations in the PI3K-AKT signaling pathway, such as TSC2 and VAV1. The largest PTCL subtype T3, accounting for 51% of our cohort, featured heterogeneous genetic alterations that participated in a complex of biological functions. Subsequently, further stratification was carried out, utilizing unsupervised hierarchical clustering, and 2 subtypes, T3.1 (71 cases) and T3.2 (41 cases), were discovered according to their discrete genetic traits and gene functions ([88]Figure 1C). To explore the defining alterations that distinguished T3.1 and T3.2, we compared the mutational loads between the 2 subtypes ([89]Figure 1D). T3.1 presented a significant enrichment in mutations involving histone modification, including methylation (KMT2C, 31%; KMT2D, 21%; and KDM6B, 17%) and acetylation (YEATS2, 13%, and CREBBP, 7%). Similarly, compared with T1 and T2, T3.1 also showed distinctively high frequencies in these mutations, thereby determining its mutational feature as histone modification alterations. T3.2 favored immune-related mutations, such as PTPN13 (17%), HLA-B (12%), and HLA-A (7%), within both T3 and the whole cohort ([90]Figure 1E). In addition, some relatively common gene mutations were evenly distributed in each subtype, especially ARID1A and ARID1B (switch defective/sucrose non-fermentable chromatin remodeling complex, SWI/SNF). It has been reported that ARID1A and ARID1B participated in multiple biological processes, involving methyltransferase activity, the PI3K-AKT pathway, p53 signaling, and immune responsiveness, which may play a general role in the pathogenesis of different PTCL subtypes.[91]^22 In summary, PTCL was classified into 4 subgroups according to the mutational status of RHOA, TET2, histone-modifying, and immune-related genes ([92]Figures 1E and [93]S2C). PTCL molecular subtypes associated with distinct clinicopathologic manifestations Molecular subtypes differed significantly in pathological distribution ([94]Figure 2A). It is well known that TET2, RHOA, DNMT3A, and IDH2, which are recognized as Tfh-associated genetic alterations, are frequently altered in Tfh-originating AITL and PTCL-Tfh entities, and affect 73%, 50%, 24%, and 18% of AITL in our cohort, respectively ([95]Figure S3A).[96]^1^,[97]^23 Accordingly, T1 and T2 were mostly composed of AITL, taking up 85% and 49% of the subtype, respectively, followed by PTCL-NOS (13% in T1 and 37% in T2). T3.1 was heterogeneous and included AITL, PTCL-NOS, ALK+ ALCL, and ALK− ALCL cases, accounting for 23%, 34%, 23%, and 21%, respectively. That is, histone-modifying gene alterations were frequently discovered regardless of pathological classification. T3.2 also represented a subgroup with pathological heterogeneity, thus indicating an essential role of immune-related mutations across different pathological subtypes. Figure 2. [98]Figure 2 [99]Open in a new tab Clinical features of the molecular subtypes in PTCL (n = 221) (A) Sankey plot of the distribution of pathological categories within each molecular subtype (left) and correlations between molecular subtypes and pathological categories (right). (B) IPI (0–2 vs. 3–5) according to molecular subtypes. (C) EBER (positive vs. negative) by in situ hybridization according to molecular subtypes (n = 171). (D) Kaplan-Meier curves of progression-free survival (PFS) and overall survival (OS) according to molecular subtypes. The clinicopathological characteristics of the patients are listed in [100]Table S3. Among 4 pathological subtypes, ALK+ ALCL patients showed the highest percentage of low-risk international prognostic index (IPI) ([101]Figure S3B). It is worth noting that T1 presented the lowest percentage of low-risk IPI patients ([102]Figure 2B). As for Epstein-Barr virus-encoded RNA (EBER) expression, EBV infection was detected in 67% of AITL and 38% of PTCL-NOS, consistent with previous studies ([103]Figure S3B).[104]^24 Interestingly, the T1 subtype exhibited a remarkably higher proportion of EBER positivity, implying a potential association of TET2 and RHOA mutations with EBV infection in PTCL ([105]Figure 2C). With a median follow-up of 43.5 months (range, 0.4–123.0 months), ALK+ ALCL patients had better progression-free survival (PFS) and overall survival (OS) than those with PTCL-NOS, AITL, and ALK− ALCL, while the other 3 subtypes shared a similar unfavorable prognosis ([106]Figure S3C). Besides, we segregated the 71 PTCL-NOS patients into PTCL-Tfh, PTCL-GATA3, PTCL-TBX21, and unclassified[107]^15 ([108]Figure S3D). Most patients of T1 and T2 subtypes were defined as PTCL-Tfh. Consistent with previous studies,[109]^11^,[110]^15 PTCL-GATA3 patients present with the worst prognosis compared with others ([111]Figure S3E). According to molecular subtypes, all 4 molecular subtypes showed inferior PFS and OS. The predicted 3-year OS rate for the T1, T2, T3.1, and T3.2 subtypes was 41.3%, 55.5%, 54.8%, and 41.8%, respectively ([112]Figure 2D). PTCL molecular subtypes associated with divergent transcriptional signatures Next, we investigated RNA-seq data to profile biological features in different molecular subtypes. We performed differential gene expression (DGE) analysis, contrasting each subtype to all others, and revealed that the 4 molecular subtypes differed in gene expression pattern ([113]Figure 3A). As revealed by gene set enrichment analysis (GSEA) ([114]Figure 3B), the TCR signaling pathway was the most enriched gene set in T1, followed by the PI3K-AKT, chemokine, JAK-STAT, ERBB, Hippo, VEGF, mitogen-activated protein kinase (MAPK), and mammalian target of rapamycin (MTOR) signaling pathways. In T2, the PI3K-AKT signaling pathway was most significantly activated, with the TCR and MAPK signaling pathways slightly enriched. Conversely, T3.1 was involved in other cellular processes, including cell cycle checkpoints, DNA replication, and translation, while T3.2 featured NOTCH-, interferon (IFN)-α/β-, and VEGF-related genes. The signaling pathways enriched in each subtype and the corresponding genomic alterations are summarized in [115]Figures 3C and 3D. T1 was characterized by enrichment of the TCR signaling pathway, in accordance with RHOA mutation. In comparison, T2 exhibited a peculiar enrichment in the PI3K-AKT signaling pathway both transcriptionally and genetically, with minor enrichment in the TCR signaling pathway, probably due to the absence of RHOA mutation. On the other hand, T3.1 harbored genetic alterations involving the histone modification, chromatin modeling, tumor suppressor, and p53 pathways, consistent with enrichment of cognate pathways, including translation and cell cycle checkpoints. Finally, T3.2 exhibited a genetic profile of immune surveillance defects, accompanied by enrichment of the IFN-α/β and VEGF signaling pathways. Figure 3. [116]Figure 3 [117]Open in a new tab PTCL molecular subtypes associated with divergent transcriptomic signatures (A) Heatmap showing gene expression profiles across 4 molecular subtypes. (B) Dot plot representing hallmarks of enrichment scores for each molecular subtype compared with all other patients by gene set enrichment analysis (GSEA). Yellow represents upregulated pathways and blue downregulated pathways. (C) Density distribution of oncogenic pathways according to molecular subtypes. The x axis shows the rank metric score, and the y axis shows the running enrichment score by GSEA. (D) Driver mutations affecting functional pathways of PTCL are selected and summarized for comparison between T1, T2, T3.1, and T3.2. Each gene box includes the mutation frequency and regulatory status of activation or disruption. Synergy of targeted agents and exploration of potential monotherapy in T1 and T2 subtypes To better understand the features of the T1 and T2 subtypes, both harboring TET2 mutations, we compared gene expression with respect to oncogenic pathways between the 2 subtypes. As expected, the T1 subtype showed enrichment of the TCR signaling pathway as well as chemokines, NOTCH, VEGF, and negative regulation of PI3K-AKT compared with the T2 subtype ([118]Figure 4A). As the TCR and PI3K-AKT signaling pathways may be potential therapeutic targets for the T1 and T2 subtypes, respectively ([119]Figure 4B), we explored the combination of the hypomethylating agent 5-azacytidine, which can target TET2 mutation,[120]^25 with the multikinase inhibitor dasatinib, which can inhibit TCR signaling,[121]^26 in the T1 subtype and with the PI3K inhibitor duvelisib in the T2 subtype. In vitro, TET2^mutRHOA^mut Jurkat and H9 cells were treated with different concentrations of 5-azacytidine and dasatinib for 48 h. The majority of the data points were found in the area <1 on the combination index (CI) curve, denoting synergistic interactions ([122]Figures 4C and [123]S4A). To clarify the molecular basis for synergy, RNA-seq of TET2^mutRHOA^mut Jurkat cells, treated with 5-azacytidine and dasatinib alone or in combination, was performed. The Venn diagram identified a specific pattern of 1,551 genes altered by the combination group but not by the untreated or the single-agent group ([124]Figure S4B). Combined treatment led to significant downregulation of multiple signaling pathways associated with cancer, especially the TCR signaling pathway ([125]Figures 4D and [126]S4C). Furthermore, preclinical zebrafish patient-derived xenograft (zPDX) models bearing TET2 and RHOA mutations present longer survival in the combination group than the untreated and the single-agent group ([127]Figure 4E). Meanwhile, TET2^mutRHOA^wt Jurkat and H9 cells were treated with different concentrations of 5-azacytidine and duvelisib for 48 h. A synergistic inhibitory effect was found in the combination group ([128]Figures 4F and [129]S4D), accompanied by a specific pattern of 1,487 genes, as well as downregulation of PI3K-AKT and inactivation of IRF7 through the TLR signaling pathways ([130]Figures 4G, [131]S4E, and S4F). In zPDX models, combined treatment also resulted in increased survival of those carrying TET2 mutations ([132]Figure 4H). Figure 4. [133]Figure 4 [134]Open in a new tab Synergy of targeted agents and exploration of potential monotherapy in T1 and T2 subtypes (A) Pathway enrichment analysis in T1 and T2 according to the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome databases. (B) GSEA enriched differentially expressed genes of TCR signaling pathway and negative regulation of the PI3K-AKT network in T1 compared with T2. (C) Combination index (CI) curve calculated by Compusyn software in TET2^mutRHOA^mut Jurkat cells treated with 5-azacytidine (Aza) and dasatinib (Dasa) for 48 h. (D) Pathway analysis of differentially downregulated genes in each group upon treatment with Aza and/or Dasa. (E) Survival of zebrafish patient-derived xenografts (zPDXs) upon Aza and/or Dasa treatment alone or in combination (n = 11 for each group). (F) CI curve calculated by Compusyn software in TET2^mut Jurkat cells treated with Aza and duvelisib (Duv). (G) Pathway analysis of differentially downregulated genes in each group upon treatment with Aza and/or Duv. (H) Survival of zPDX upon Aza and/or Duv treatment alone or in combination (n = 11 for each group). (I) The ratios of cell viability of TET2^mutRHOA^mut Jurkat cells and TET2^WTRHOA^WT cells after treatment with 7,316 compounds at 0.4 μM for 72 h (left). Compounds that specially suppressed the growth of TET2^mutRHOA^mut Jurkat cells are listed on the right panel. (J) Cell viability of TET2^mutRHOA^mut Jurkat cells after treatment with different concentrations of KPT-9274 for 48 h. ∗p < 0.05 compared with TET2^WTRHOA^WT cells. Data are presented as the mean ± SD (n = 3). (K) Cell viability ratio of TET2^mutRHOA^WT Jurkat cells compared with TET2^WTRHOA^WT cells after treatment with 7,316 compounds at 0.4 μM for 72 h (left). Compounds that specially suppressed the growth of TET2^WTRHOA^WT Jurkat cells are listed on the right. (L) Cell viability of TET2^mutRHOA^wt Jurkat cells after treatment with different concentrations of LY-364947 for 48 h. ∗p < 0.05 compared with TET2^WTRHOA^WT cells. Data are presented as the mean ± SD (n = 3). In search of reagents selectively targeting TET2^mutRHOA^mut and TET2^mutRHOA^wt cells, we examined cell growth of Jurkat cells exposed to 7,316 targeted agents ([135]Table S4). A number of potent inhibitors of the growth of TET2^mutRHOA^mut Jurkat cells, but not TET2^WTRHOA^WT cells, were identified by a drug sensitivity screen. The most effective agents are depicted in [136]Figure 4I. The selective suppressive function of KPT-9274, a dual inhibitor of PAK4 and nicotinamide phosphoribosyltransferase (NAMPT), decreased the growth of both TET2^mutRHOA^mut Jurkat and H9 cells ([137]Figures 4J and [138]S4G). Similar results were obtained when screening TET2^mutRHOA^WT Jurkat cells ([139]Figure 4K). The efficacy of LY-364947, a TGF-β1 receptor I (TβRI) inhibitor, was observed and validated in TET2^mutRHOA^WT Jurkat and H9 cells ([140]Figures 4L and [141]S4H). These findings suggest the potential of KPT-9274 and LY-364947 as a monotherapy for the T1 and T2 subtypes, respectively. Synergy of targeted agents and exploration of potential monotherapy in T3.1 and T3.2 subtypes Next, we evaluated differentially expressed genes between the T3.1 and T3.2 subtypes. As demonstrated by GSEA, higher expression of genes associated with DNA replication, translation, cell cycle checkpoints, and p53 signaling pathways were found in the T3.1 subtype, while VEGF, TGF-β, NOTCH, WNT, and MAPK signaling pathways were in the T3.2 subtype ([142]Figure 5A). Significant activation of DNA replication and VEGF signaling pathways is illustrated in [143]Figure 5B. As the T3.1 subtype was characterized by histone-modifying gene mutations, we examined the combination of the histone deacetylase inhibitor chidamide with decitabine in KMT2C^mut Jurkat and H9 cells and observed synergy function compared with KMT2C^WT cells ([144]Figures 5C and [145]S4I). The Venn diagram identified a specific pattern of 2,168 genes altered by the combination group, but not by the untreated or the single-agent group ([146]Figure S4J). Genes associated with cytokine-cytokine receptor interaction were enriched in the combination group as well as the MAPK signaling pathway, in agreement with our previous study of KMT2D mutations ([147]Figures 5D and [148]S4K).[149]^10 zPDX models of the T3.1 subtype showed longer survival in the combination group than in the untreated or the single-agent group ([150]Figure 5E). Meanwhile, Ki-67 expression was evaluated to test the inhibitory activity of the anti-angiogenic agent apatinib and PD-1 inhibitor nivolumab in PTPN13^mut Jurkat and H9 cells when co-cultured with peripheral blood mononuclear cells (PBMCs) at a ratio of 1:5. Apatinib and nivolumab exhibited an elevated anti-tumor activity in PTPN13^mut Jurkat and H9 cells ([151]Figures 5F and [152]S4L). As revealed by multicolor flow cytometry for immunophenotyping in the co-culture system of Jurkat cells and PBMCs, the combination group significantly decreased GATA3 expression of CD3+/CD4+ Th2 cells in the co-culture system of PTPN13^mut Jurkat cells compared with the untreated group, while the inhibitory effect was not observed in PTPN13^WT cells ([153]Figure 5G). Accordingly, upregulation of GATA3 was observed in the T3.2 subtype using RNA-seq data of PTCL patients ([154]Figure 5H). Figure 5. [155]Figure 5 [156]Open in a new tab Synergy of targeted agents and exploration of potential monotherapy in T3.1 and T3.2 subtypes (A) Pathway enrichment analysis in T3.1 and T3.2 according to the KEGG and Reactome databases. (B) GSEA enriched differentially expressed genes of DNA replication in T3.1 and the VEGF signaling pathway in T3.2. (C) CI curve calculated by Compusyn software in KMT2C^mut Jurkat cells treated with chidamide (Chid) and decitabine (Deci) for 48 h. (D) Pathway analysis of differentially downregulated genes in each group upon treatment with Chid and/or Deci. (E) Survival of zPDX upon Chid and/or Deci treatment alone or in combination (n = 11 for each group). (F) Ki-67 positivity of Jurkat cells transfected with PTPN13^WT and PTPN13^mut upon nivolumab (Nivo) and apatinib (Apa) treatment after 72 h. Peripheral blood mononuclear cells (PBMCs) and Jurkat cells were co-cultured at a ratio of 5:1. Nivo was at a concentration of 10 μg/mL, and Apa was at 10 μM. Data are presented as the mean ± SD (n = 3). (G) Multiflow cytometry analysis of Th2 cell markers (CD4 and GATA3) in PBMCs, co-cultured with PTPN13^mut Jurkat cells with or without Nivo (10 μg/mL) and Apa treatment for 72 h. Data are presented as the mean ± SD (n = 3). (H) Normalized mRNA expression of GATA3 across 4 molecular subtypes as revealed by RNA-seq. (I) Cell viability ratio of KMT2C^mut Jurkat cells and KMT2C^WT cells after treatment with 7,316 compounds at 0.4 μM for 72 h (left). Compounds that specially suppressed the growth of KMT2C^mut Jurkat cells are listed on the right. (J) Cell viability of KMT2C^mut Jurkat cells after treatment with different concentrations of KDM4D-IN-1 for 48 h. ∗p < 0.05 compared with KMT2C^WT cells. Data are presented as the mean ± SD (n = 3). (K) Cell viability ratio of PTPN13^mut Jurkat cells and PTPN13^WT cells after treatment with 7,316 compounds at 0.4 μM for 72 h (left). Compounds that specially suppressed the growth of PTPN13^mut Jurkat cells are listed on the right. (L) Cell viability of PTPN13^mut Jurkat cells after treatment with different concentrations of anlotinib for 48 h. ∗p < 0.05 compared with PTPN13^WT cells. Data are presented as the mean ± SD (n = 3). We also investigated cellular vulnerabilities to monotherapy induced by KMT2C and PTPN13 mutations, respectively, using a drug sensitivity screen. The selective sensitivity of KMT2C mutation to the KDM4D inhibitor KDM4D-IN-1 was validated by KMT2C^mut Jurkat and H9 cells ([157]Figures 5I, 5J, and [158]S4M), and that of PTPN13 mutation to the VEGFR2 inhibitor anlotinib was validated by PTPN13^mut Jurkat and H9 cells ([159]Figures 5K, 5L, and [160]S4N). Unique LME subtypes of PTCL defined by gene expression profiling Besides the molecular alterations of tumor cells in PTCL, their interactions with LME play a pivotal role in tumor biology and clinical behavior as well.[161]^3^,[162]^21 To define the diversity in the composition of LME in PTCL, we conducted unsupervised hierarchical clustering of RNA-seq data based on a list of essential immune profiling genes ([163]Table S5) and identified 4 distinct subtypes associated with immune infiltration and stromal components: LME1 (39.2%), LME2 (19.9%), LME3 (11.8%), and LME4 (29.0%) ([164]Figure S5A). To validate our classification scheme, we utilized the statistical power conferred by the external gene expression profiles of 396 PTCL patients provided in published literature (Published Data: [165]https://github.com/emacgene/PTCL/blob/master/Supplementary_Data.Rm d)[166]^16 ([167]Table S6), and 4 LME clusters were identified with similar proportions: LME1 (36.6%), LME2 (15.7%), LME3 (12.1%), and LME4 (35.6%) ([168]Figure S5B). To further explore specific characteristics of different LME subtypes, we developed 25 functional gene expression signatures covering various aspects of the PTCL microenvironment based on previous studies, including tumor cells of origin (e.g., Tfh, Th1, Th2, and Th17 cells), immune cells (e.g., B cells, plasma cells, regulatory T [Treg] cells, and M1 and M2 macrophages), stroma-related cellular and non-cellular components (e.g., lymphatic endothelial cells [LECs], vascular endothelial cells [VECs], cancer-associated fibroblasts [CAFs], fibroblastic reticular cells [FRCs], and extracellular matrix [ECM]), and LME-dependent signaling and transcriptional pathways (e.g., B cell traffic, TNF, immune checkpoints, IFN-α/β, VEGF, and HIF) ([169]Table S7). To reconstruct the general composition and biological features, the values of single-sample GSEA (ssGSEA) were compared among 4 LME subtypes ([170]Figure 6A). According to the combinations of signatures within each subtype, they were termed “Tfh like” for the unique enrichment of Tfh cells as its tumor COO in LME1; “inflammatory” due to the aggregation of immune cells (e.g., Treg cells and M1 macrophages) and the extensive activation of various immune-infiltration signaling pathways (e.g., VEGF, IFN-α/β, and immune checkpoints) in LME2; “mesenchymal,” considering the abundance of M2 macrophages, stromal cells, and ECM in LME3; and finally “depleted” as LME4 was featured by an overall low expression of most microenvironment signatures compared with other subtypes. Moreover, all samples were examined for tumor content, and the results were comparable with previous studies ([171]Figure S5C).[172]^5^,[173]^15 No significant difference was detected between different subtypes based on pathological and molecular classification. However, when comparing the tumor content of the 4 LME subtypes, we observed remarkable differences. The depleted subtype showed significantly higher purity, consistent with the scarcity of immune- or stroma-related signatures and strong cell proliferation in this subtype. The inflammatory and mesenchymal subtypes revealed lower tumor content since they both featured abundant LME components. LME subtypes revealed an uneven pathological distribution ([174]Figure 6B). The Tfh-like subtype was mainly composed of AITL (53.4%) and PTCL-NOS (37.0%), corresponding to AITL and Tfh-originating PTCL-Tfh entities.[175]^5^,[176]^15 The major components of the inflammatory subtype were also AITL (56.8%) and PTCL-NOS (29.7%). Of note, most of mesenchymal subtype were ALK+ ALCL with a Th17-cell and stromal enrichment signature. The depleted subtype showed no specific inclination. We also performed ssGSEA analysis on the published gene expression data, and all immune-related signatures of each LME subtype were validated by the external cohort ([177]Figure 6C). In addition, despite the different proportions of pathological subtypes in the external cohort, LME subtypes shared similar pathologic distributions with our data ([178]Figure S5D). The Tfh-like and inflammatory subtypes mainly contained AITL (61.4% and 25.8%) and PTCL-NOS (32.4% and 59.7%); most ALK+ ALCL were classified into the mesenchymal (42.9%) and depleted (44.6%) subtypes. As for the association of LME subtypes with molecular subtypes, we observed a correlation between Tfh-like and T2 subtypes, in accordance with the association of TET2 mutations with Tfh and B cell markers in AITL and PTCL-NOS.[179]^27^,[180]^28^,[181]^29^,[182]^30 The inflammatory subtype showed the closest association with the T1 subtype, which was characterized by RHOA mutations. The mesenchymal subtype, enriched in M2 macrophages and Th17-originating tumor cells, both of which are associated with histone-modifying mutations,[183]^31^,[184]^32 was mainly composed of the T3.1 subtype ([185]Figure 6D). Figure 6. [186]Figure 6 [187]Open in a new tab Immune clusters of PTCL revealed distinct gene expression-based lymphoma microenvironment (LME) subtypes (A) Heatmap of the activity scores of 25 functional gene expression signatures denoting four major LME clusters termed Tfh like, inflammatory, mesenchymal, and depleted. Tfh, follicular T helper; LEC, lymphatic endothelial cell; VEC, vascular endothelial cell; CAF, cancer-associated fibroblast; FRC, fibroblastic reticular cell; ECM, extracellular matrix; TNF, TNF signaling pathway; IS cytokines, immunosuppressive cytokines; IFNab, IFN-α/β signaling pathway; VEGF, VEGF signaling pathway; HIF, HIF signaling pathway; TGFb, TGF-β signaling pathway. (B) The proportion of pathological category distributions of four LME subtypes. The number of cases in each subtype is shown. (C) Heatmap of the activity scores of 25 functional gene expression signatures from external GEP of 396 PTCL patients, denoting four major LME clusters termed Tfh like, inflammatory, mesenchymal, and depleted. (D) Relationships and phi coefficients between LME subtypes and molecular subtypes. (E) PFS and OS curves of 186 PTCL patients according to LME subtype. Both PFS and OS in our cohort indicated a better prognosis of the mesenchymal subtype compared with other subtypes ([188]Figure 6E). Similarly, OS of the mesenchymal subtype in the external cohort also presented a significantly better prognosis, confirming its association with favorable outcomes ([189]Figure S5E). Moreover, among PTCL-NOS patients, PTCL-GATA3 mostly belonged to the depleted subtype with a worse prognosis, and PTCL-Tfh was associated with the Tfh-like subtype with a better prognosis ([190]Figure S5F). Distinct immune communities dissected by LME subtypes To better profile the immune nature of each LME subtype, a precise overview of cellular abundance and relative population within each LME subtype was calculated using Cibersort. Moreover, the reproducibility of the LME subtypes was validated in the external cohort, showing comparable community enrichment profiles in each LME subtype ([191]Figures 7 and [192]S6). The Tfh-like subtype revealed a Tfh origin of lymphoma cells ([193]Figures 7A and [194]S6A), along with remarkably higher proportions of B cells and plasma cells ([195]Figures 7B, 7C, and [196]S6B). The inflammatory subtype mainly originated from Th1 or Th2 cells ([197]Figures 7D and [198]S6C) with enrichment of Treg cells and M1 macrophages ([199]Figures 7E, 7F, and [200]S6D). The mesenchymal subtype mainly originated from Th17 cells ([201]Figures 7G and [202]S6E) with enrichment of M2 macrophages and extensive stromal components, encompassing LECs, VECs, CAFs, FRCs, and ECM ([203]Figures 7H, 7I, and [204]S6F). As for the depleted subtype, it was characterized by the minimal presence of most LME components. Figure 7. [205]Figure 7 [206]Open in a new tab Dissection of LME subtypes uncovered unique immune communities (A) Significant enrichment of Tfh cells in Tfh-like LME, obtained by Cibersort. (B) Significant enrichment of B cells and plasma cells in Tfh-like LME, obtained by Cibersort and single-sample GSEA (ssGSEA), respectively. (C) Schematic of selected features of Tfh-like LME. (D) Significant enrichment of Th1 and Th2 cells in inflammatory LME obtained by ssGSEA. (E) Significant enrichment of Treg cells and M1 macrophages in inflammatory LME, obtained by ssGSEA and Cibersort, respectively. (F) Schematic of selected features of inflammatory LME. (G) Significant enrichment of Th17 cells in mesenchymal LME obtained by ssGSEA. (H) Significant enrichment of M2 macrophages, LECs, VECs, CAFs, FRCs, and ECM in mesenchymal LME, obtained by Cibersort or ssGSEA. (I) Schematic of selected features of mesenchymal LME. (J) Normalized mRNA expression of CRBN, TIGIT, and CD52 in Tfh-like LME vs. other LMEs as revealed by RNA-seq. (K) Normalized mRNA expression of PD-L1, LAG3, and IDO1 in inflammatory LME vs. other LMEs as revealed by RNA-seq and normalized mRNA expression of TIM-3 and CD30 in inflammatory LME vs. other LMEs as revealed by RNA-seq. As for possible immunotherapeutic targets, expression levels of Cereblon (CRBN), T-cell immunoglobulin and ITIM domain (TIGIT), and CD52 were higher in the Tfh-like subtype than in other LME subtypes, suggesting potential therapeutic vulnerabilities ([207]Figures 7J and [208]S6G). Increased expression of various immune checkpoint genes was observed in the inflammatory subtype, such as PD-L1, LAG3, and IDO1 ([209]Figures 7K and [210]S6H), and TIM-3 in the mesenchymal subtype ([211]Figure 7L). Moreover, enrichment of CD30 expression was discovered in the mesenchymal subtype, indicating potential targeted therapy with an anti-CD30 antibody ([212]Figures 7L and [213]S6I). Therefore, the immunosuppressive condition may effectively contribute to the reconstruction of anti-tumor immunity and subsequent tumor growth inhibition in each LME subtype. Discussion Despite the revolutionary introduction of targeted therapy and immunotherapy into lymphoma treatment, CHOP-based chemotherapy protocols remain an “unsatisfactory” standard of care in PTCL. Here we reported an integrated genomic and transcriptomic analysis of 221 PTCL patients, representing a large set of DNA- and RNA-seq profiling to understand the molecular and microenvironment characterization of PTCL and, more importantly, to provide a basis for the selection of more effective targeted and immunotherapeutic approaches in PTCL. Our study identified distinct molecular subtypes varying from genomic alterations and transcriptional signatures to clinical features. The T1 subtype was defined as the coexistence of TET2 and RHOA mutations. TET2 is a dioxygenase that functions within the DNA demethylation process.[214]^33 RHOA is a GTPase essential for multiple T cell processes, including cell proliferation and modulation of TCR signaling.[215]^30^,[216]^34 It has been reported that RHOA mutation together with TET2 loss induces Tfh cell specification and Tfh-originating lymphomagenesis, including AITL and PTCL-Tfh.[217]^35 As RHOA functions as an essential GTPase in TCR signaling, we identified an enrichment of TCR-related gene expression in the T1 subtype. Due to the coexistence of TCR-related (RHOA, CARD11, and FYN) or DNA methylation-related (TET2, IDH2, and DNMT3A) mutations, we evaluated co-targeting therapy with the hypomethylating agent 5-azacytidine and the multikinase inhibitor dasatinib and exploited their synergistic function in TET2^mutRHOA^mut cells, which has been reported recently in a clinical trial for relapsed/refractory AITL patients.[218]^26 Interestingly, the drug sensitivity screen showed a potential monotherapy, KPT-9274, a dual inhibitor of PAK4 and NAMPT, to selectively suppress the growth of TET2^mutRHOA^mut cells. The T2 subtype was characterized by TET2 mutation with wild-type RHOA and mainly consisted of AITL and PTCL-NOS patients. Mutations in PI3K-AKT-associated genes were enriched in T2, such as TSC2, ITPR3, and PIK3R1, in accordance with activation of the PI3K-AKT pathway. The dysregulation of PI3K-AKT has been reported to drive the pathogenesis of AITL and PTCL-NOS and is linked to poor clinical outcomes.[219]^36^,[220]^37 Subsequently, we proposed and confirmed that a combination of agents co-targeting DNA methylation and PI3K, 5-azacytidine and duvelisib, exerted a synergistic impact on TET2^mutRHOA^WT cells. Besides, the TβRI inhibitor LY-364947 was identified as a potential monotherapy for the T2 subtype. The T3.1 subtype mainly presented alterations in histone modification, particularly KMT2C and KMT2D. So far, inactivating histone methyltransferase KMT2C and KMT2D mutations have been identified in PTCL, including PTCL-NOS, PTCL-Tfh, and ALCL.[221]^38^,[222]^39^,[223]^40 Pathologically, the T3.1 subtype was enriched in ALCL, which was also associated with epigenetic modifier mutations.[224]^31 The dysfunction in histone modifiers led to enrichment in DNA replication and cell cycle checkpoint pathways. Combined treatment with two epigenetic agents, chidamide and decitabine as well as the monotherapy KDM4D-IN-1 both exerted potential inhibition of KMT2C^mut tumor cells. The T3.2 subtype was enriched for mutations in immune-related genes, especially HLA-A, HLA-B, and PTPN13, sharing similar features of a PTCL-NOS subtype with immune surveillance alterations.[225]^5 The VEGF pathway was upregulated in T3.2, together with immune-related mutations, explaining why an anti-PD-1 antibody and VEGF inhibitor as well as anlotinib, a tyrosine kinase inhibitor targeting the VEGF receptor,[226]^41 may contribute to favorable efficacy in the T3.2 subtype. In the second approach, we obtained microenvironment evidence and established distinct immune-related LME subtypes in PTCL, simultaneously integrating malignant T cell populations and microenvironment components into the prognosis and treatment of the disease. The Tfh-like subtype resembled a Tfh-originated lymphoma and was mainly composed of AITL and PTCL-NOS patients.[227]^13^,[228]^42 The high frequency of RHOA, TET2, DNMT3A, and IDH2 mutations in this subtype explained the differentiation of Tfh cells and Tfh-related lymphomagenesis. Cereblon, the direct binding target for immunomodulatory agents, was significantly overexpressed in this subtype, which may indicate a better response to lenalidomide-containing regimens. Moreover, increased expression of immune checkpoint TIGIT and the cell marker CD52 renders the Tfh-like subtype sensitive to TIGIT inhibition and the CD52-antibody alemtuzumab, respectively. The inflammatory subtype represented a microenvironment mediated by M1 macrophages and Treg cells. Pro-inflammatory M1 macrophages may contribute to the differentiation of neoplastic T cells, Th1 and Th2, and suggest a certain degree of anti-lymphoma immunity. However, Treg cell-associated immunosuppressive LME has been reported in different PTCL pathological entities and is associated with angiogenesis and tumor aggressiveness.[229]^3^,[230]^43 Gene expression profiling unveiled significant enrichment of immune therapy-associated and angiogenesis pathways, including immune checkpoints, immunosuppressive cytokines, IFN-α/β, and the VEGF signaling pathway. Therapeutically, PD-L1, IDO1, and LAG3 were increasingly expressed, implying more potential efficacy of ICIs for these targets, while VEGF pathway activation suggested a favorable response to anti-angiogenesis agents. The mesenchymal subtype featured M2 macrophage infiltration as well as abundant cellular and non-cellular stromal components, including LECs, VECs, CAFs, FRCs, and ECM, together shaping an immunosuppressive microenvironment. The transcriptional signature was identified as upregulation of ECM remodeling, VEGF, HIF, and TGF-β pathways. As essential tumor-promoting cells, M2 macrophages and CAFs play immunosuppressive roles by directly modulating immune cell activities with cytokines and chemokines (e.g., VEGF, IL-10, and TGF-β) and indirectly mediating ECM accumulation.[231]^44 In addition, TGF-β represents the master chemokine regulator in mesenchymal LME and, notably, induces Th17 cell differentiation.[232]^45 This subtype showed special preference for ALK+ ALCL, resulting in a dominant COO of Th17 and favorable clinical outcomes. Interestingly, recent advances have attributed the dynamic process of epithelial-mesenchymal transition to epigenetic modification, including histone methylation and acetylation,[233]^46 consistent with the high frequency of epigenetic modification-related mutations in mesenchymal subtypes. TIM-3 overexpression provides an opportunity for ICI treatment in the mesenchymal subtype. This LME subtype was also characterized by a high CD30 positivity rate. Thus, the therapeutic activity of CD30-targeted agents is potentiated in relevant patients and has already been recommended as first-line therapy in CD30+ PTCL.[234]^47 The depleted subtype presented low microenvironment-derived signatures, featuring heterogenic COO and a lack of typical pathological preference. Consistent with the high tumor content, this subtype showed enrichment in cell proliferation signatures. A similar LME subtype with non-significant immune components has been reported in diffuse large B cell lymphoma and Burkitt lymphoma.[235]^48 Notably, despite the overexpression of GATA3, immunohistochemistry (IHC) and gene expression profile (GEP) approaches have characterized the microenvironment of the PTCL-GATA3 subgroup as “minimally inflamed” due to the limited amount of various immune cells, comparable with our depleted LME signature.[236]^20 Enrichment of available immunotherapy markers is not detected in this subtype; thus, effective therapies warrant further investigation. In conclusion, assisted by large-scale tumor biopsies and system biology studies, genomic and transcriptomic characterization were accomplished for categorizing PTCL into distinct molecular and microenvironment subtypes. Future study is necessary to elucidate the potential synergy of targeted therapy and immunotherapy, realizing curable PTCL in the era of precision medicine. Limitations of the study Although we did not identify novel gene mutations in PTCL, we provided a simple and practical subtyping method suitable for clinical application. Meanwhile, due to the long-term lack of suitable cell and animal models in the basic research field of PTCL, we failed to obtain the most appropriate preclinical research model. We tried to overcome this limitation by maximizing the clinical utility of the scheme. A multicenter, randomized, phase 2 trial within each molecular subtype is currently ongoing (ClinicalTrials.gov: [237]NCT05675813). Finally, our LME subtyping is based on transcriptome sequencing, which requires further validation by pathological immunohistochemistry. STAR★Methods Key resources table REAGENT or RESOURCE SOURCE IDENTIFIER Antibodies __________________________________________________________________ Alexa Fluor® 647 Mouse anti-Ki-67 BD Cat# 561126; RRID: [238]AB_10611874 BUV395 Mouse Anti-Human CD45 BD Cat# 563792; RRID: [239]AB_2869519 High Parameter Custom BV570 Conjugate Anti-Human CD4 BD Cat# 624298; special order High Parameter Custom BB660-P2 Conjugate Anti-Human T-Bet BD Cat# 624295; special order High Parameter Custom BV605 Conjugate Anti-Human RORγt BD Cat# 624290; special order BUV496 Mouse Anti-Human CD3 BD Cat# 612940; RRID: [240]AB_2870222 BV510 Mouse Anti-Human CD8 BD Cat# 563919; RRID:[241]AB_2722546 PE-Cy™7 Mouse anti-GATA3 BD Cat# 560405; RRID:[242]AB_1645544 __________________________________________________________________ Biological samples __________________________________________________________________ Human PTCL tumor tissues/peripheral blood This paper N/A __________________________________________________________________ Chemicals, peptides, and recombinant proteins __________________________________________________________________ 5′-azacytidine (Aza) Selleck S1782 Dasatinib (Dasa) Selleck S1021 Duvelisib (Duv) Selleck S7028 Chidamide (Chid) Shenzhen Chipscreen N/A Decitabine (Deci) Selleck S1200 Nivolumab (anti-PD-1) Selleck A2002 Apatinib (Apa) Hengrui N/A KPT-9274 Selleck S8444 LY-364947 Selleck S2805 KDM4D-IN-1 Selleck S1059 Anlotinib Selleck S8726 __________________________________________________________________ Critical commercial assays __________________________________________________________________ Wizard® Genomic DNA Purification Kit Promega A1120 DNeasy Blood & Tissue Kit Qiagen 69504 RNeasy Mini Kit Qiagen 74106 Qubit dsDNA HS Assay Kit Invitrogen [243]Q32854 Qubit RNA HS Assay Kit Invitrogen [244]Q32852 Qubit Assay Tube Invitrogen [245]Q32856 Covaris microTube Covaris 520045 Agilent High Sensitivity DNA Kit Agilent 5067–4626 Agilent DNA 1000 Kit Agilent 5067–1504 Agencourt® AMPure® XP Kit Beckman Coulter A63881 Dynabeads MyOne Streptavidin T1 Invitrogen 65602 SureSelect XT Human All Exon v6 Agilent 5190–8863 SureSelect XT Reagent Kit, Illumina (ILM) platforms Agilent G9611A Herculase II Fusion DNA Polymerase Agilent 600677 TargetSeq One Kit iGeneTech TPB1-i96 Enzyme Plus Library Prep Kit iGeneTech TLP13-i96 VAHTS Total RNA-seq (HMR) Library Prep Kit for Illumina vazyme NR603 NovaSeq 6000 S4 Reagent Kit v1.5 (300 cycles) Illumina 20028312 100% Ethanol, molecular biology grade Sigma E7023 1X Low TE Buffer (10 mM Tris-HCl, pH 8.0, 0.1 mM EDTA) Life Technologies 12090–015 KAPA RNA HyperPrep Kit with RiboErase KAPA KK8560 Nuclease-free Water Ambion AM9930 RosetteSep Human T Cells StemCell 15021 lipofectamine 2000 Invitrogen 11668019 Transcription Factor Buffer Set BD 652574 CellTiter-Glo Luminescent Cell Viability Assay Promega G7572 __________________________________________________________________ Deposited data __________________________________________________________________ Raw sequencing data This paper GSA-Human database (HRA004246) __________________________________________________________________ Experimental models: Cell lines __________________________________________________________________ Jurkat ATCC TIB-152 H9 ATCC HTB-176 HEK-293T ATCC CRL-11268 __________________________________________________________________ Experimental models: Organisms/strains __________________________________________________________________ Zebrafish: Tuebingen (wild type) China Zebrafish Resource Center CZ3 __________________________________________________________________ Recombinant DNA __________________________________________________________________ plasmids pCV186/Cherry/Puro-RHOA^wt This paper NA Plasmids pCV186/Cherry/Puro- RHOA^G17V This paper NA Plasmids pGV655/GFP/Puro-TET2^wt This paper NA Plasmids pGV655/GFP/Puro-TET2^H1380Y This paper NA Plasmids pGV655/GFP/Puro-KMT2C^wt This paper NA Plasmids pGV655/GFP/Puro- KMT2C^N4686S This paper NA Plasmids pGV655/GFP/Puro-PTPN13^wt This paper NA Plasmids pGV655/GFP/Puro- PTPN13^D2176N This paper NA __________________________________________________________________ Software and algorithms __________________________________________________________________ FastQC NA [246]http://www.bioinformatics.babraham.ac.uk/projects/fastqc/ Trimmomatic Bolger et al., 2014[247]^49 [248]https://www.bioinformatics.babraham.ac.uk/projects/fastqc/ BWA Li and Durbin, 2009[249]^50 [250]http://bio-bwa.sourceforge.net/ Genome Analysis Toolkit McKenna et al., 2010[251]^51 [252]https://software.broadinstitute.org/gatk/ ANNOVAR Wang et al., 2010[253]^52 [254]http://annovar.openbioinformatics.org/en/latest/ STAR NA [255]https://github.com/alexdobin/STAR Integrative Genomics Viewer Thorvaldsdóttir et al., 2013[256]^53 [257]http://software.broadinstitute.org/software/igv/ Picard Broad Institute [258]http://broadinstitute.github.io/picard/ BEDtools Quinlan, 2014[259]^54 [260]https://bedtools.readthedocs.io/en/latest/ HTSeq Anders et al., 2015[261]^55 [262]https://htseq.readthedocs.io/ DESeq2 Love et al., 2014[263]^56 [264]https://bioconductor.org/packages/DESeq2/ Samtools Li et al., 2009[265]^57 [266]http://samtools.sourceforge.net/ CNVkit Talevich et al., 2016[267]^58 [268]https://cnvkit.readthedocs.io/en/stable/pipeline.html Genomic Identification of Significant Targets in Cancer (GISTIC 2.0) Mermel et al., 2011[269]^59 NA Maftools Mayakonda et al., 2018[270]^60 [271]https://bioconductor.org/packages/release/bioc/html/maftools.html Limma (v3⋅38⋅3) NA [272]http://bioconductor.riken.jp/packages/3.0/bioc/vignettes/limma/ins t/doc/intro.pdf ggplot2 (v3.5.5) NA [273]https://ggplot2-book.org/ ggbiplot NA [274]http://github.com/vqv/ggbiplot Pheatmap v1.0.12 NA [275]https://bioconductor.org/packages/release/bioc/manuals/heatmaps/ma n/heatmaps.pdf Gene set enrichment analysis (GSEA) Subramanian et al., 2005[276]^61 [277]http://www.broad.mit.edu/gsea/ Cibersort Newman et al., 2015[278]^62 [279]https://cibersort.stanford.edu GSVA package v1.40.1 Hänzelmann et al., 2013[280]^63 [281]http://www.sagebase.org HISAT2 software (v2.0.4) Kim et al., 2015[282]^64 [283]https://daehwankimlab.github.io/hisat2/ GFOLD (generalized fold change) algorithm Feng et al., 2012[284]^65 [285]https://zhanglab.tongji.edu.cn/softwares/GFOLD/index.html Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.8 NA [286]https://david.ncifcrf.gov Calcusyn software program Chou, 2006[287]^66 NA [288]Open in a new tab Resource availability Lead contact Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Wei-Li Zhao (zhao.weili@yahoo.com). Materials availability All unique/stable reagents generated in this study are available from the [289]lead contact with a completed Materials Transfer Agreement. Data and code availability * • All sequencing data (including WES and RNA-seq) have been deposited in the GSA-Human database and can be accessed through the following link: [290]https://bigd.big.ac.cn/gsa-human/browse/HRA004246. It is available by contacting the [291]lead contact, Wei-Li Zhao (zhao.weili@yahoo.com), upon request. * • The paper does not generate original code. We utilized publicly available software in all the analyses. These are listed with appropriate citations in the methods. * • Any additional information required to reanalyze the data reported in this work paper is available from the [292]lead contact upon request. Experimental model and study participant details Patient information We collected 221 frozen tumor samples of PTCL patients. Histological diagnoses were established according to the World Health Organization (WHO) classification and reviewed by two independent pathologists (C.-F. Wang and B.-SH. Ou-Yang). All the patients received 6 cycles of CHOP/CHOP-like regimen, consisting of 103 AITL (47%), 71 PTCL-NOS (32%), 24 ALK+ALCL (11%), and 23 ALK-ALCL (10%) patients. Two or more TFH makers (including BCL6, CD10, PD-1, and CXCL13) were evaluated by IHC in most of AITL and PTCL-NOS ([293]Figure 1A). These samples were subjected to WES (n = 101, including 46 AITL, 39 PTCL-NOS, 12 ALK+ALCL, and 4 ALK-ALCL) or targeted sequencing (n = 120, including 57 AITL, 32 PTCL-NOS, 12 ALK+ALCL, and 19 ALK-ALCL), and RNA-seq (n = 186). The study was approved by the Institutional Review Boards of all centers, and informed consent was obtained in accordance with the Declaration of Helsinki. Zebrafish patient-derived xenografts (zPDX) Tumor tissues of three PTCL patients from T1, T2, and T3.1 subtypes were used to establish zPDX models, which were obtained from Shanghai Ruijin Hospital with written informed consent. Tumor cells were stained with Dil (Vybrant; Molecular Probes, Invitrogen) and then carefully injected into the perivitelline space of each anesthetized larva from wild-type Tuebingen zebrafish. In each subtype, successful injected xenografts were chosen and were randomly distributed in four groups: the untreated, the single-agent, and the combination group (3μM 5-azacytidine with 0.1 μM dasatinib for T1 subtype, 3μM 5-azacytidine with 5μM duvelisib for T2 subtype, and 3μM chidamide with 3μM decitabine for T3.1 subtype for three consecutive days). In the following 10 days, the death event of zebrafish was recorded. The zebrafish study was approved by the Ethics Committee of Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine and the methods were carried out in accordance with the approved guidelines. Method details Whole exome sequencing Genomic DNA was extracted from frozen tumor samples of 101 patients and peripheral blood of 21 patients randomly selected from 101 patients, using Wizard Genomic DNA Purification Kit (Promega, Wisconsin-Madison, USA) according to the manufacturer’s instructions. A SureSelect XT Human All Exon v6 was used for capturing exome regions of these samples, which were sequenced on NovaSeq 6000 platform with 150bp paired-end strategy. Raw sequencing data were assessed by FastQC (version 1.11.4), and then processed by Trimmomatic (version 3.6) to remove sequencing adapters and low-quality reads that the joint sequence fragments of the 3′ end and low-quality fragments with Q value <25 and fragments with <35 bp. All sequence data was processed through the Broad Institute’s data processing pipeline to prepare read alignments for analysis. Next, reads were aligned to the Human Genome Reference Consortium build 37 (GRCh37) using BWA (version 0.7.17-r1188), and the quality scores were recalibrated using the TableRecalibration tool from the Genome Analysis Toolkit (GATK) (version 4.1.4.0). Variant detection and analysis were performed by the Broad Institute’s Cancer Genome Analysis infrastructure program Mutect2 and mutations were annotated using the Annovar software (version 2017-07-17). The mean depth of each sample was 97.9× (range 66.6–159.1×). A total of 82 genes were selected for further study based on two criteria: (i) genes with recurrent mutations (>3%) and/or (ii) genes with relevance to oncogenesis of PTCL ([294]Table S2). Sanger sequencing was used to confirm these somatic mutations in all WES samples. CNAs analysis for WES samples were inferred by CNVkit using the batch pipeline recommended, and significant focal CNAs across all samples were identified by Genomic Identification of Significant Targets in Cancer (GISTIC, version 2.0) with default parameters. Targeted sequencing Genomic DNA was extracted from frozen or paraffin tumor samples of 120 PTCL patients using a DNeasy Blood & Tissue Kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. Targeted capture sequencing for 82 genes selected from WES data was performed using established Illumina protocols on novaseq6000 platform. Multiplexed libraries of tagged amplicons from PTCL tumor samples were generated by Shanghai Yuanqi Bio-Pharmaceutical Multiplex-PCR Amplification System. RNA sequencing of tumor samples Total RNA was extracted from frozen tumor samples of 186 PTCL patients using RNeasy Mini Kit (Qiagen, Hilden, Germany). RNA size, concentration, and integrity were verified using Agilent 2100 System (Agilent). RNA libraries were constructed by using VAHTS total RNA-seq (HMR) library prep kit according to the manufacturer’s instructions, and sequenced on Illumina NovaSeq 6000 System. Paired-end reads were harvested from Illumina NovaSeq 6000 System, and the quality were controlled by Q30. After 3′ adaptor-trimming and low-quality reads removing by Trimmomatic software (v0.36), the high-quality trimmed reads were aligned to the reference genome (UCSC hg19) guided by the Ensembl GFF gene annotation file with STAR software (v2.5.3a). R package limma (v3⋅38⋅3) was used to normalize raw reads and obtain differentially expressed genes. GSEA was performed using the BROAD Institute GSEA software ([295]http://www.broad.mit.edu/gsea/) referring to Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome databases.[296]^48^,[297]^67 LME heterogeneity analysis In our RNA-seq dataset, we first calculated the log2(TPM+1) matrix based on the expression of 152 immune profiling genes extracted from LM22 signature gene list ([298]Table S5). The selected genes represented characteristic and exclusive markers in various immune components, covering all 22 major leukocyte subsets and activation state.[299]^68 Hierarchical clustering of the matrix was performed using the R package pheatmap (v1.0.12) using ward.D2 as linkage. To determine the optimal number of clusters, we utilized the silhouette analysis of KMeans by the cluster R package. The number of 4 clusters was identified as a better pick than more numerous clusters, and the 4 clusters were identified by the cutree function. In the validation cohort, we clustered the gene expression data using 149 genes that were presented in all clustered datasets ([300]Table S5). Cibersort is a deconvolution algorithm that predict the proportion of different cell types from bulk RNA-seq data based on a list of 547 reference gene expression values, using support vector regression.[301]^62 We used the Cibersort algorithm to calculate percentages of 22 immune cell types from the normalized RNA-seq data. Next, we curated 25 gene expression signatures, reflecting various immune cells, stromal cells, noncellular substances based on published gene sets, as well as pathways based on KEGG and Reactome databases ([302]Table S7). Gene set analysis was carried out using the GSVA Bioconductor package v1.40.1. We then obtained a score of each sample, representing the enrichment of a gene set using gene expression data, including RNA-seq data and gene expression profile. Cell transfection For RHOA^wt, RHOA^mut, TET2^wt, TET2^mut, KMT2C^wt, KMT2C^mut, PTPN13^wt, and PTPN13^mut transfection, purified plasmids pCV186/Cherry/Puro-RHOA^wt, pCV186/Cherry/Puro-RHOA^G17V, pGV655/GFP/Puro-TET2^wt, pGV655/GFP/Puro-TET2^H1380Y, pGV655/GFP/Puro-KMT2C^wt, pGV655/GFP/Puro-KMT2C^N4686S, pGV655/GFP/Puro-PTPN13^wt, and pGV655/GFP/Puro-PTPN13^D2176N were transfected into package HEK-293T cells using lipofectamine 2000 (11668019, Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s protocol. The supernatant fraction of HEK-293T cell cultures was condensed to a viral concentration of approximately 2 × 10^8 units/ml. Then, the lentiviral particles were incubated with Jurkat and H9 cells for 72h with addition of polybrene (8 μg/mL). The stably transduced clones were selected by green and/or red fluorescence protein using flow cytometry or puromycin treatment for two weeks. RNA sequencing of cell lines TET2^mutRHOA^mut, TET2^mutRHOA^wt, and KMT2C^mut Jurkat cells were treated with different agents. RNA size, concentration, and integrity were verified using Agilent 4200 TapeStation System (Agilent). RNA libraries were constructed by using KAPA RNA HyperPrep Kit with RiboErase (HMR, KAPA) according to the manufacturer’s instructions. Libraries were sequenced on Illumina NovaSeq 6000 System. Paired-end reads were harvested from Illumina NovaSeq 6000 System, and were quality controlled by Q30. After 3′adaptor-trimming and low-quality reads removing by cutadapt software (v1.9), the high-quality trimmed reads were aligned to the reference genome (UCSC hg19) guided by the Ensembl GFF gene annotation file with hisat2 software (v2.0.4). The GFOLD (generalized fold change) algorithm was used to find biologically meaningful rankings of differentially expressed genes from RNA-seq data of cell lines.[303]^65 DEGs were then analyzed by the Database for Annotation, Visualization and Integrated Discovery (DAVID) and were enriched in KEGG pathways. Isobolographic analysis To determine the synergistic effect of targeted agent combination, including 5-azacytidine with dasatinib, 5-azacytidine with duvelisib, and chidamide with decitabine, combination index (CI) method described by Chou and Talalay[304]^66 was applied for automated analysis by the Calcusyn software program (Biosoft, Cambridge, UK). When at least 80% of CI values for a combination were <1, the drug combination was considered to be synergistic, while CI values = 1, and >1, the drug combination was considered to be additive effect, and antagonism, respectively. T-lymphoma cell co-culture and multi-color flow cytometry Co-culture of PTPN13^wt or PTPN13^mut cells with PBMCs was conducted at 1:5 ratio, treated with anti-PD-1 antibody nivolumab and/or apatinib for 72h. All cells were maintained in RPMI-1640 medium supplemented with 10% heat-inactivated fetal bovine serum and 1% penicillin/streptomycin. Then multi-color flow cytometry was carried out to assess the immune receptor expression of immune cells. CD45, CD3, CD4, CD8, T-Bet, RORγt, FoxP3, and GATA3 antibodies were used for cell labeling. Ki-67 were used to assess tumor cell proliferation. Flow cytometry data were collected by a FACS Calibur cytometer (BD Biosciences) and analyzed by FlowJo software. Drug library screen TET2^wtRHOA^wt, TET2^mutRHOA^mut, TET2^wt, TET2^mut, KMT2C^wt, KMT2C^mut, PTPN13^wt, and PTPN13^mut Jurkat cells were used for screening assays. Cells were seeded in 384-well plates and then treated with the drug at a concentration of 0.4 μM (SCADS Inhibitor Kit, including 7315 compounds, [305]Table S4). Cell viability was assessed after 72h using the CellTiter-Glo Luminescent Cell Viability Assay (Promega, G7572). Luminescence was measured using an Envision Multi-label plate reader (PerkinElmer), and the luminescence reading was used to determine the cell viability relative to that of cells treated with solvent (DMSO). Candidate compounds were considered if their viability in TET2^mutRHOA^mut, TET2^mut, KMT2C^mut, or PTPN13^mut Jurkat cells was less than 50%, and that in TET2^wtRHOA^wt, TET2^wt, KMT2C^wt, or PTPN13^wt Jurkat cells was more than 80%.[306]^69 Quantification and statistical analysis Data were calculated as the mean ± standard deviation (SD) from three separate experiments. The Student’s t test was used to compare two normally distributed groups and the Mann-Whitney U test to compare two groups which did not conform to normal distribution. PFS was calculated from the date when treatment began to the date when the disease progression was recognized or the date of the last follow-up. OS was measured from the date of diagnosis to the date of death or the last follow-up. The Kaplan-Meier method was used to estimate survival functions were using and the log rank test to assess differences in PFS and OS between different subtypes. The data analysis for this manuscript was conducted using Statistical Package for the Social Sciences (SPSS, v24.0), GraphPad Prism 8 software or R software. p < 0.05 was considered statistically significant. Acknowledgments