Abstract Peripheral T cell lymphoma (PTCL) is a heterogeneous group of postthymic T cell neoplasms, with ~40% classified as PTCL–not otherwise specified (PTCL-NOS). PTCL-GATA3, a molecularly defined subtype, associated with T helper 2 (T[H]2)–like differentiation and poor prognosis, has frequent co-occurrence of TP53 loss/mutation and heterozygous PTEN loss. CD4+ T cell conditional mouse models with Trp53 mutation/deletion and Pten loss demonstrated mature T cell lymphomas (mTCLs) with T[H]2-like transcriptomic and immunophenotypic profiles. Molecular studies revealed that codeletion of Trp53/Pten induced T cell receptor and Janus kinase–signal transducer and activator of transcription signaling, promoting T[H]2 differentiation while inhibiting T[H]1 differentiation. These findings were validated by CRISPR editing of TP53/PTEN loss in human CD4+ T cells and mechanistically evaluated the p53 binding region in intron-3 of GATA3, resulting in transcriptional repression. Transcriptomic profiles of m-TCLs recapitulated human-PTCL-GATA3 transcriptome and distinguished PTCL-NOS subtypes. Preclinical assessment of m-TCLs with PI3Kγ/δ inhibitors significantly improved survival, supporting a therapeutic approach for the p53-aberrant PTCL-GATA3. __________________________________________________________________ TP53 and PTEN loss drives T[H]2 differentiation and T cell lymphomagenesis, highlighting increased sensitivity to PI3K inhibitors. INTRODUCTION Peripheral T cell lymphoma (PTCL) is a heterogeneous group of mature T cell malignancies ([84]1, [85]2), and patients with PTCL have inferior clinical outcomes compared with patients with B cell lymphoma, with a 5-year overall survival (OS) of <30% using current therapeutic regimens ([86]3). These lymphomas are difficult to diagnose, requiring a multiparameter approach that integrates clinical history, morphology, immunophenotypic findings, and genetic analysis to ensure an accurate diagnosis ([87]2, [88]4). About 30 to 50% of PTCLs are defined as PTCL-not otherwise specified (NOS) through diagnostic exclusion of the other World Health Organization (WHO)/International Consensus Classification (ICC) specified entities ([89]1, [90]2). Using multiomics approaches, we identified two major distinct biological and prognostic subtypes within PTCL-NOS ([91]5, [92]6) that are indicated in the current WHO/ICC classification of lymphoid neoplasms ([93]7). Thirty-five percent of PTCL-NOS cases are characterized by high GATA3 expression and its transcriptional target genes, designated as the PTCL-GATA3 subtype, and ~55% have high expression of TBX21 and its target genes, designated as the PTCL-TBX21 subtype. GATA3 and TBX21 are the master transcriptional regulators of T helper 2 (T[H]2) and T[H]1 cell differentiation, respectively, and regulate the expression of relevant cytokines ([94]8, [95]9). The biological and prognostic differences between PTCL-GATA3 and PTCL-TBX21 subtypes have been validated in other previous studies ([96]10, [97]11). PTCL-GATA3 and PTCL-TBX21 subtypes demonstrated distinctive oncogenic pathways, and the PTCL-GATA3 subtype showed recurrent aberrations in the p53 and PI3K pathways due to frequent del17p13 (TP53) and del10p23 (PTEN) ([98]12). Whereas del17p was associated with TP53 mutation or biallelic loss, del10p23 was primarily heterozygous and not associated with PTEN mutation. The significant association of the codeletions (i.e., PTEN and TP53) in PTCL-NOS with poor prognosis was noted in another study ([99]13). Although the canonical function of PTEN is to regulate PI3K activation through its phosphatase function, phosphatase-independent function was demonstrated in mice harboring a mutation causing loss of the PTEN C-terminal region ([100]14), resulting in a T cell acute lymphoblastic leukemia (T-ALL)–like disease. Similar PTEN C-terminal region mutations in patients with T-ALL were identified, resulting in truncation of the C2 domain without disrupting phosphatase activity ([101]15). PTEN promotes genomic stability via its direct interaction with TP53 ([102]16), and TP53 also directly regulates PTEN transcription ([103]17), thus having interdependent functions. Whereas mice with Pten deletions ([104]18) and Trp53 deletions ([105]19, [106]20) spontaneously develop T-ALL, mice with Trp53 deletion develop cytotoxic TCL with a CD1d-restricted natural killer (NK)/T cell of origin ([107]21) and a transcriptomic program similar to human hepatosplenic T cell lymphoma (HSTCL) and T cell prolymphocytic leukemia (T-PLL). The codeletion of TP53 and PTEN is an infrequent event in cancers but has been observed in advanced breast adenocarcinomas ([108]22), high-grade glioblastomas ([109]23), or aggressive prostate cancers ([110]24), and a murine model with codeletion demonstrated Myc-directed up-regulation of Phlpp2 to attenuate PI3K activity ([111]25). The co-occurring PTEN and TP53 functional loss (i.e., monoallelic PTEN loss with biallelic TP53 deletion/mutation) in PTCL-GATA3 is intriguing, but it is unclear how these two genetic events cooperate in T cell lymphoma pathogenesis, and the lack of authentic T cell or animal models to study PTCL-GATA3 pathogenesis is a major hurdle. Although the role of PI3K-AKT in thymic T cell selection, T cell activation ([112]26), effector T[H] cell function, polarization ([113]27), regulatory T cell (T[reg]) homeostasis, metabolic balance, and mitochondrial fitness ([114]28) has been explored, the functional relevance of PTEN in postthymic T cell neoplasms is unclear. TP53 regulates antigen-specific responses via interleukin-2R (IL-2R) up-regulation ([115]29), homeostasis by up-regulating pro-apoptotic SLAM-associated protein (SAP) ([116]30), cytolytic activity, stemness, and T[reg] differentiation ([117]31), but the understanding of its role in T[H] cell biology is limited. Here, we investigated the role of these alterations independently and cooperatively in mature T cell differentiation and lymphomagenesis by evaluating PTCL-NOS biospecimens and establishing mouse models with Trp53^R172H mutation or Trp53 deletion concurrent with Pten deletion in CD4+ T cells. Three models with Trp53 and Pten coaberration (Trp53^R172H;Pten^fl/+−, Trp53^R172H;Pten^fl/fl, and Trp53^fl/fl;Pten^fl/+) were highlighted, with the latter closely matching what is more frequently observed in PTCL-GATA3 cases. We also engineered normal human CD4+ T cells using a CRISPR-Cas9 methodology to delete TP53 or knock-in mutant TP53^R175H with PTEN deletion to investigate the human relevance of these genetic alterations. To enhance the translational relevance of these preclinical models, we compared the molecular features of mature T cell lymphomas (m-TCLs) with human PTCL (h-PTCL)–GATA3 and further investigated therapeutic interventions targeting codeleted m-TCLs. RESULTS TP53 and PTEN codeletion is associated with the PTCL-GATA3 subtype and poor clinical outcome Earlier genomic copy number (CN) analysis (gCNA) studies showed that frequent codeletion of TP53 (del17p13) and PTEN (del10p23) was associated with PTCL-GATA3 (26%) and a subset of ALK-ALCL (15%) but not in other PTCL entities ([118]12, [119]32). TP53 and PTEN deletions were confirmed by fluorescence in situ hybridization (FISH) in a subset of PTCL-NOS (n = 33) ([120]Fig. 1A). Combined CN loss (TP53 and PTEN) was associated with inferior OS (P < 0.05) in PTCL-GATA3 treated with chemotherapy (CHOP/CHOEP) ([121]Fig. 1B). Among the 32 PTCL-GATA3 cases analyzed in our previous study ([122]12), loss of TP53 was observed in 19 cases (59%) and PTEN loss in 11 cases (34%). Notably, nine of those cases (28%) exhibited codeletion of both tumor suppressors, indicating a substantial subset with dual gene loss (table S1). In contrast, among 30 PTCL-TBX21 cases, only two and one case showed TP53 or PTEN loss, respectively (6.7 and 3.3%), with no codeletions identified. This marked difference underscores the genomic divergence between PTCL-GATA3 and PTCL-TBX21, highlighting a potential molecular distinction between these subtypes. TP53 mutation co-occurred with del17p (5/20; 25%), mainly affecting the DNA binding domain, with the TP53^R175H variant being recurrent, whereas no PTEN mutations (0/20) were noted in PTCL-GATA3 ([123]Fig. 1C). PTEN and TP53 losses were significantly associated with down-regulated expression of their mRNAs ([124]Fig. 1D). Fig. 1. Evaluation of TP53 and PTEN codeletion in distinct PTCL-NOS subtype (i.e., PTCL-GATA3). [125]Fig. 1. [126]Open in a new tab (A) Confirmation of TP53 and PTEN aberration by FISH in representative PTCL-NOS cases (of n = 33 where FISH was performed). The results confirmed previous observations ([127]12, [128]32). (B) OS analysis of PTCL-GATA3 cases with available outcome data by PTEN and TP53 DNA CN (CNA) status, suggesting poor outcome associated with double loss. Two cases with PTEN only loss were excluded from the plot due to a low number. (C) Frequency plot of DNA CN gains and losses in PTCL-GATA3 affecting chromosomes 10 and 17 (top). Schematic of mutations identified affecting TP53 in PTCL-GATA3 (bottom). aa, amino acid. (D) mRNA expression of TP53 and PTEN genes in PTCL-GATA3 cases and association with gCNA of TP53 and PTEN showed significantly reduced mRNA expression with gCNA loss. (E) DEGs (P < 0.05; fold change > 2) between WT, del-TP53, del-PTEN, and TP53/PTEN codeleted in PTCL-GATA3 cases. Representative transcripts are noted. The median expression for four comparisons is shown on the right. TP53/PTEN codeleted cases showed significant enrichment of a proliferative and PI3K/AKT transcriptional program. (F) Bubble plot of GSEA data comparing the del-TP53, del-PTEN, and codeleted TP53/PTEN versus WT [false discovery rate (FDR) < 0.025] showing significant enrichment of MYC and STAT3 targets in codeleted cases. (G) Hematoxylin and eosin (H&E) and IHC staining for the noted biomarkers (i.e., GATA3, MYC, and Ki-67) in a representative case for TP53 and/or PTEN abnormality PTCL-GATA3 cases. (H) Validation of oncogenic MYC activation using IHC showed high MYC positivity in double loss cases, and GATA3 positivity was higher in TP53 loss cases. Statistical significance was determined using the Wilcoxon rank sum test. (I) Bubble plot of T cell subset proportions estimated by xCell ([129]35) in PTCL-GATA3, suggesting enrichment of T[H]2 immune cells in double deleted cases. NES, normalized enrichment score. Gene expression profiling (GEP) analysis of PTCL-GATA3 tumors with TP53/PTEN coaberrations (deletion/mutation) showed significant enrichment of a proliferative transcriptional program (i.e., transcriptional targets of MYC and E2F), transcripts regulating G[2]-M checkpoint, or PI3K-mTORC1 signaling ([130]Fig. 1, E and F). Single del17p (TP53) tumors showed similar GEP with lower enrichment, including transcriptional signatures associated with T[H] cell differentiation (IL-4–induced targets and STAT3 targets) ([131]33). In contrast, wild-type (WT) cases were significantly enriched in SRC-induced signatures or KRAS-dependent/addicted tumor survival signatures ([132]34). PTCL-GATA3 tumors with coaberrant (TP53 and PTEN) had higher MYC mRNA and MYC-target gene signatures, which were validated by immunohistochemistry (IHC) showing higher MYC and Ki-67 protein levels, the single TP53-del and WT cases had moderate and low MYC expression, respectively ([133]Fig. 1, G and H). PTCL-GATA3 cases with a TP53 loss showed higher GATA3 expression by IHC (P < 0.07), compared to TP53 WT cases. In general, MYC expression is higher in PTCL-GATA3 versus PTCL-TBX21 due to frequent 8q gain as shown previously ([134]12)[,] but codeleted tumors had even higher MYC expression. GEP deconvolution of immune-cell signatures using xCell ([135]35) indicated significant enrichment of T[H]2 cells and CD4+ T memory cells in tumors ([136]Fig. 1I). Overall, multiomics characterization of primary PTCL-GATA3 specimens indicated that MYC activation, T[H]2 differentiation, and inferior OS are associated with TP53/PTEN codeleted tumors. Trp53^R172 knock-in or Trp53 knockout and Pten knockout in murine CD4+ T cells lead to T cell malignancies Seven murine models, including WT (Cre−), Pten-heterozygous (Pten^Δ:Cd4-Cre^+/−;Pten^fl/+), Pten-homozygous (Pten^ΔΔ:Cd4-Cre^+/−;Pten^fl/fl), Trp53-mutant (Trp53^mut:Cd4-Cre^+/−;Trp53^LSL-R172H+/−), Trp53-mutant; Pten-heterozygous (Trp53^mutPten^Δ: Cd4-Cre^+/−; Trp53^LSL-R172H+/−;Pten^fl/+), and Trp53-mutant; Pten-homozygous (Trp53^mutPten^ΔΔ: Cd4-Cre^+/−;Trp53^LSL-R172H+/−; Pten^fl/fl) andTrp53-knockout; Pten-heterozygous (Trp53^ΔΔPten^Δ: Cd4-Cre^+/− Trp53^fl/fl;Pten^fl/+), were generated ([137]Fig. 2A). The Trp53^R172H mutation and Pten deletion were confirmed by polymerase chain reaction (PCR) of genomic DNA or from RNA sequencing (RNA-seq) and whole-exome sequencing (WES) of corresponding CD4+ T cells or murine tumors ([138]Fig. 2B). Trp53 deletion was confirmed by Transnetyx automated genotyping. The early onset of spontaneous lymphomas in double-mutant mice indicated that Pten loss accelerates tumor development, with a median OS of 10.5 weeks in Trp53^mutPten^ΔΔ (n = 9), 13.2 weeks in Pten^ΔΔ (n = 24), 22.4 weeks in Trp53^ΔΔPten^Δ (n = 7), 27.7 weeks in Trp53^mutPten^Δ (n = 33), and 49.7 weeks in Trp53^mut (n = 26), with several pathological manifestations including splenomegaly, enlarged thymus, or extranodal involvement (primarily in the liver) ([139]Fig. 2, C and D). Whereas Pten^ΔΔ showed complete lymphoma penetrance, no tumor was observed in Pten^Δ after 2 years of follow-up, in agreement with earlier findings ([140]36), but Trp53^mut provided accelerated and cooperative oncogenic stimulation in Pten^ΔΔ or Pten^Δ mice (P < 0.05). Trp53^ΔΔPten^Δ mice developed aggressive tumors affecting the spleen and liver (fig. S1A) and developed tumor masses in the subcutaneous tissue, and several succumbed to a fatal illness other than lymphoma. Fig. 2. Generation of conditional murine T cell lymphoma models. [141]Fig. 2. [142]Open in a new tab (A) Schematic of the characterization of different CD4+ T cell murine models and cross-breeding strategies of Cd4-Cre transgenic mice with Trp53^LSL-R172H+/−, Trp53^FL/FL, Pten^FL/FL designated as Pten^Δ, Pten^ΔΔ, Trp53^mutPten^ΔΔ, Trp53^mutPten^Δ, Trp53^mut, Trp53^mutPten^Δ, and Trp53^ΔΔPten^Δ. (B) Validation of Trp53 and Pten gene rearrangement in CD4+ T cells in murine models using standard PCR and SNV analysis in RNA-seq data. SNV, single nucleotide variant. (C) Necropsy of representative tumor-bearing mice with early onset in Pten^ΔΔ, Trp53^mutPten^ΔΔ, Trp53^ΔΔPten^Δ and Trp53^mut and late onset in Trp53^mut and Trp53^mutPten^Δ demonstrated splenomegaly and enlarged thymus, estimated by the corresponding weight analysis (bottom). (D) OS analysis of murine models. Kaplan-Meier curve analysis indicates that tumors with double loss have aggressive lymphoma and are associated with poor outcomes (Trp53^mutPten^ΔΔ, Trp53^ΔΔPten^Δ, and Pten^ΔΔ) compared to Trp53^mutPten^ΔΔ and Trp53^mut. (E) Morphologic and IHC characteristics in tumor-bearing mice. (F) Quantification of IHC staining of tumors or age-matched controls for CD3, CD4, CD8, and CD19, demonstrating a higher proportion of CD3, CD4, and CD8 splenocytes in tumor mice across most genotypes. (G) Clonality assessment using TCR repertoire analysis by RNA-seq in tumor-bearing mice compared to WT and preneoplastic mice. 4 wk mice, 4-week-old mice. (H) In silico analysis of subcellular fractions (using xCell) of the RNA-seq data indicated enrichment of the T[H]2 cellular profile in murine tumors affected by Trp53 or Pten aberration compared to WT mice. (I) Flow cytometry analysis and validation of T[H]2 immunophenotype of the murine tumors (gated on CD4+ T cells) indicated high GATA3 expression in the Trp53^ΔΔPten^Δ, Trp53^mutPten^Δ, and Pten^ΔΔ tumors compared to age-matched WT CD4+ T cells. The flow plots are one representative mouse, and the bar graph on the right shows the GATA3+/TBX21− percentage for three mice per genotype profiled. Whole-tissue sections or tissue microarrays (TMAs) with ~3 to 5 representative tumors for each genotype were immunoassayed for immunological and maturation biomarkers (i.e., CD3, CD4, CD8A, and TDT), and the pathological findings and immunophenotypic data are summarized in table S2. Tumors involved the thymus, spleen, and liver and had an atypical pattern of highly proliferative, large CD3^+ T cells that were single positive (SP; CD4^+ or CD8^+) or double positive (DP; CD4^+CD8^+) and TDT negative, indicating m-TCLs. Whereas CD4+ T cell neoplasms were dominant in Trp53^mutPten^ΔΔ and Pten^ΔΔ mice and had similar histological and immunophenotypic features as reported earlier ([143]37), Trp53^mutPten^Δ mice also developed CD8^+ T cell lymphomas ([144]Fig. 2, E and F). Trp53^ΔΔPten^Δ mice developed m-TCLs that were TDT negative, with four of seven mice indicating DP (CD4^+CD8^+) T cells and three of seven SP (CD4^+) T cells upon IHC examination. The clonal assessment with T cell receptor (TCR) repertoire analysis using RNA-seq or flow cytometry identified a major TCR-α or TCR-β clone in murine-TCL but not in age-matched WT CD4+ T cells, suggesting a restricted TCR repertoire and clonal progression ([145]Fig. 2G and fig. S2, A and B). In silico analysis of RNA-seq expression profiling of neoplastic cells suggested T[H]2-enriched gene expression in Trp53^mutPten^Δ and Trp53^ΔΔPten^Δ ([146]Fig. 2H). Flow cytometry analysis validated higher GATA3^+ CD4^+ T cell in Trp53^mutPten^Δ (29%), Trp53^ΔΔPten^Δ (70%), and Pten^ΔΔ (23%) versus age-matched WT cells (6.7%) ([147]Fig. 2I). Splenomegaly in tumor-bearing mice was associated with varied splenocyte subset cellular distribution compared to WT. Some Trp53^mutPten^Δ mice had decreased CD4+ T cells and an increase in CD8+ T cells compared to age-matched WT controls (fig. S1A), whereas corresponding IHC indicated infiltration of B cells in Trp53^mutPten^Δ ([148]Fig. 2E). Enlarged thymuses were also noted, and thymic cellular characterization demonstrated a significant increase in SP (CD4+) and reduced DP (CD4+CD8+) T cells in tumors with Pten^ΔΔ (i.e., Trp53^mutPten^ΔΔ or Pten^ΔΔ). A subset (two of seven) of Trp53^mut mice had reduced DP and increased double negative (DN) and SP (CD4 or CD8 markers) (fig. S1A), which may be due to depletion or negative selection of CD4+ thymocytes, as demonstrated in an earlier study ([149]38). The evaluation of the cellular proportions before lymphomagenesis at 6- and 10-week timepoints revealed no significant changes in either B cells, NK cells, myeloid cells, CD3^+ T cells, or CD4+/CD8+ T cell subpopulations in splenocytes (fig. S1B). Molecular characterization of murine lymphomas indicates a T[H]2-like transcriptomic program We performed RNA-seq analysis of CD4+ T cells isolated from 4-week-old mice before lymphoma development (termed as preneoplastic), neoplastic splenocytes, and age-matched WT cells from the above murine models ([150]Fig. 3A). Unsupervised principal components analysis (PCA) and hierarchical consensus clustering (HCC) indicated that neoplastic and preneoplastic CD4+ T cells cluster separately (fig. S2, C and D). GEP analysis of the CD4+ T cells with Pten^ΔΔ, Trp53^mutPten^ΔΔ or Trp53^ΔΔPten^Δ showed a higher number of differentially expressed transcripts compared to either Trp53^mutPten^Δ or Trp53^mut versus their corresponding age-matched WT CD4+ T cells, suggesting that complete loss of p53- or Pten-mediated signaling has a substantial impact on global transcriptional activity. Similar observations were noted in early-occurring m-TCLs compared to late-occurring m-TCLs (fig. S2E). In silico functional analysis [i.e., using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database] of the differentially expressed transcripts indicated distinctive and shared transcriptomic signatures associated with these aberrations in preneoplastic CD4^+ T cells including up-regulation of transcripts promoting cell cycle, TGF-β (transforming growth factor–β), or RAP1 (endocytosis) pathways and down-regulation of transcripts regulating extracellular matrix reorganization and cell adhesion ([151]Fig. 3B). CD4^+ T cells with Pten^ΔΔ, Trp53^mutPten^ΔΔ, Trp53^mutPten^Δ, and Trp53^ΔΔPten^Δ showed significant differential gene expression (DGE) of transcripts regulating pathways promoting T cell activation and differentiation [i.e., ROS (reactive oxygen species), TCR, JAK-STAT (Janus kinase–signal transducer and activator of transcription), NF-κB (nuclear factor κB), and T[H] cell differentiation] and WNT signaling in Trp53^ΔΔPten^Δ (see the gene list in [152]Fig. 3B). Gene Set Enrichment Analysis (GSEA) validated and demonstrated significant enrichment of RNA signatures associated with T cell activation, T[H] differentiation, STAT3, PI3K-responsive, and MYC-regulated transcriptome in Trp53^mutPten^ΔΔ, Trp53^mutPten^Δ, Pten^ΔΔ and Trp53^ΔΔPten^Δ ([153]Fig. 3C). We validated representative pathways including PI3K signaling, i.e., p-AKT (Ser^473), STAT3 activation (p-STAT3–Y705), and T cell activation [p-LCK (Y505) and p-ZAP70 (Y319)], at the protein level by using Western blot in another mouse cohort ([154]Fig. 3D). Fig. 3. RNA-seq analysis in neoplastic CD4+ T cells at the preneoplastic stage and neoplastic stage and validation at protein levels using either Western blot or flow cytometry in an independent murine cohort. [155]Fig. 3. [156]Open in a new tab (A) Schematic of the experimental design for functional characterization of preneoplastic CD4+ T cells and neoplastic cells isolated from mouse models. (B) RNA-seq analysis of the CD4+ T cells isolated from mouse models and heatmap depicting the DEGs versus WT before development of lymphoma (4 weeks). (C) Bubble plot of GSEA indicated perturbed pathways (T cell activation, T[H] differentiation, STAT3, PI3K-responsive, and MYC-regulated transcriptome) in preneoplastic CD4+ T cells compared to their WT counterpart. (D) Protein expression validation of key oncogenic genes including TCR, PI3K-AKT signaling, and STAT3 signaling by Western blot in an independent mouse cohort, indicating increases in TCR activation, STAT3 and PI3K/AKT signaling in mutant mice compared to WT. (E) Heatmap of the DEGs in tumors from the noted mouse models versus age-matched spleens. The preneoplastic cells are shown for comparison. Enriched pathways are indicated in the bubble plot on the left. (F) Bubble plot of select GSEA pathway enrichment analysis results of the neoplastic spleen compared to their WT age-matched counterpart. (G) IHC validation of p-STAT3 protein in tumor-bearing mice versus their corresponding WT controls. IHC quantification was performed on all biological replicates (n = 3) and shown as a bar chart. RNA-seq analysis of the m-TCLs versus their corresponding age-matched splenocytes indicated up-regulation of proliferation-promoting transcriptional signatures [e.g., cell cycle transition, DNA replication, RNA metabolism, and mammalian target of rapamycin (mTOR) pathway] and transcripts regulating metabolic reprogramming/thermogenesis, DNA damage, and aerobic mitochondrial metabolism, consistent with earlier findings ([157]39) ([158]Fig. 3, E and F). We evaluated transcriptome associated with oncogenic transition by performing DGE analysis between m-TCLs and their corresponding preneoplastic CD4+ T cells collected at an earlier timepoint (i.e., 4 weeks) and age-matched WT splenocytes. Whereas m-TCLs with Pten loss (i.e., Pten^ΔΔ, Trp53^mutPten^ΔΔ, Trp53^mutPten^Δ, and Trp53^ΔΔPten^Δ) showed the concordant genes/pathway promoting cell cycle, mitosis, and DNA repair, the m-TCLs from Trp53^mut showed dysregulations in the chemokine signaling (fig. S3). Specifically, transcripts regulating TCR, STAT3, and T[H]2 associated (GATA3) signaling were enriched in Trp53^mutPten^ΔΔ, Pten^ΔΔ, Trp53^mutPten^Δ, and Trp53^ΔΔPten^Δ m-TCLs and with down-regulation of IL-12 signaling, a major regulator of TBX21/T[H]1 signaling ([159]Fig. 3, E and F). Validation of the GATA3 protein expression was performed by flow cytometry ([160]Fig. 2I) and p-STAT3 by IHC in additional m-TCLs ([161]Fig. 3G). These findings in double-mutant m-TCLs show a T[H]2-like transcriptional program and immunophenotype, and STAT3 activation may induce GATA3 expression, as shown earlier ([162]33). We performed WES on several m-TCLs to uncover additional genetic drivers and revealed that four mice displayed gains or amplifications involving the Myc oncogene, in particular with Pten^ΔΔ. In addition, mutation of Stat2 (Pten^ΔΔ); Mybbp1a, DNA repair regulation (Trp53^mutPten^Δ); Bcl2l10, anti-apoptotic regulator (Trp53^mut), Dlx5, a Myc regulator (Trp53^mutPten^ΔΔ) and Pi3kr2, and PI3K modulator (Trp53^ΔΔPten^Δ) were identified, suggesting additional genes involved in Trp53/Pten-driven lymphomagenesis (fig. S2F). Trp53 and Pten abnormalities drive CD4+ T cell activation and T[H]2 differentiation ex vivo Proliferation, cell cycle, apoptosis, and plasticity of the CD4+ T cells isolated from splenocytes of each mouse genotype at 4 weeks were compared with those of WT cells under normal in vitro culture conditions (i.e., with/without α-CD28/CD3 and IL-2) ([163]Fig. 4A). Trp53^mutPten^Δ had a growth advantage compared to WT under normal conditions and reduced serum (5%) conditions, whereas the cells with other genotypes, including Trp53^mutPten^ΔΔ, showed reduced growth compared to WT ([164]Fig. 4, B and C). Under low (10 ng) or normal (20 ng) IL-2–only conditions, however, both Trp53^mutPten^Δ, Trp53^ΔΔPten^Δ, and Trp53^mutPten^ΔΔ CD4+ T cells survived and expanded without CD28/CD3 stimuli. Trp53^mutPten^ΔΔ cells demonstrated their growth responsiveness to elevated IL-2 levels, highlighting their reliance on IL-2, but not CD3+CD28 stimulation for proliferation. Pten^ΔΔ and Trp53^mutPten^ΔΔ proliferated the slowest and had increased apoptosis compared to other genotypes and WT ([165]Fig. 4C). Fig. 4. In vitro characterization of CD4+ T cells with Trp53 or Pten genetic lesions. [166]Fig. 4. [167]Open in a new tab (A) Schematic of the experimental designs for functional characterization of the CD4+ T cells with Trp53 or Pten loss. CD4+ T cells were isolated by negative selection from 4-week-old mice and cultured under various conditions to assess growth and survival characteristics. Four-week-old T cells were also transplanted into WT mice to assess their fitness in vivo. FBS, fetal bovine serum. (B) Proliferation of CD4+ T cells from the different mouse models under differing culture conditions, where Trp53^mutPten^Δ had a proliferative advantage under serum-starved conditions, and double-mutant cells had an advantage under IL-2–only conditions. (C) Apoptosis (annexin V and 7-AAD) was analyzed by flow cytometry. Pten loss was found to be associated with a higher apoptotic rate in these murine preneoplastic CD4+ T cells. 7-AAD, 7-aminoactinomycin D. (D) The steady-state engraftment of CD45.2+ cells was routinely detected in PBMCs obtained through maxillary bleeding 2 weeks postadoptive transfer. The right panel shows the flow cytometry profile of the CD45.2+ cells. (E) Schematic representation of experiments assessing T[H]1 or T[H]2 polarization in preneoplastic CD4+ T cells. (F) Representative Western blot of GATA3 and TBX21 in preneoplastic CD4+ T cells cultured under T[H]1 or T[H]2 conditions versus naïve conditions. Western blot quantification was performed on three biological replicates and shown as the bar graph on the right. 0 hr, 0 hours. (G) Representative flow cytometry analysis of Gata3 and Tbx21 in Trp53^mutPten^Δ, Trp53^ΔΔPten^Δ, and age-matched WT CD4+ T cells cultured in T[H]1- and T[H]2-polarizing media, suggesting a T[H]2-promoting immunophenotype in Trp53^mutPten^Δ. The bar graph depicts three biological replicates and shows GATA3+/TBX21− and GATA3−/TBX21+ for T[H]2 and T[H]1 conditions, respectively, compared to 0 hours. (H) Proliferation analysis of the preneoplastic CD4+ T cells under the T[H]1 or T[H]2 conditions indicates that Trp53^mutPten^Δ showed a survival advantage under both conditions compared to WT cells. To assess fitness under in vivo conditions, we performed adoptive transfer studies using CD4^+ T cells isolated from 4- to 5-week-old (preneoplastic) mice into 10-week-old recipient mice (postthymic development). Pten^ΔΔ or Trp53^mutPten^ΔΔ CD4+ T cells were able to engraft, survive, and proliferate for at least 12 weeks and outcompete recipient CD4+ T cells, whereas Trp53^mut and Trp53^mutPten^Δ donor CD4+ T cells spontaneously regressed in host mice ([168]Fig. 4D). Tumors occurred in mice engrafted with Pten^ΔΔ or Trp53^mutPten^ΔΔ CD4+ T cells at 3.4 months postengraftment, indicating that PI3K activation is required for engraftment fitness and is indicative of clonal expansion. The engrafted Pten^ΔΔ tumor-bearing mice show CD4+CD8+ T cell subpopulation in the thymus, spleen, and blood, whereas Trp53^mutPten^ΔΔ tumor-bearing mice showed a higher percentage of DP cells in the thymus and blood (fig. S1C). To assess the role of Trp53^mut and/or Pten deletion in T[H] cell differentiation, naïve CD4+ T cells were isolated from each murine genotype and stimulated under exogenous T[H]1- and T[H]2-polarizing conditions ([169]40) ([170]Fig. 4E). GATA3 protein expression increased substantially in Trp53^mut, Trp53^mutPten^Δ, and Trp53^mutPten^ΔΔ, indicating preferential polarization toward the T[H]2 phenotype compared to corresponding WT cells, a finding consistent with corresponding transcriptomic analysis ([171]Fig. 4, F and G). Furthermore, flow cytometry analysis of the double-mutant cells suggested that Trp53^ΔΔPten^Δ cells had higher basal GATA3 protein expression compared to corresponding WT cells and were resistant to T[H]1 polarization ([172]Fig. 4G). The Trp53^mutPten^Δ cells showed better growth in T[H]2 medium compared to other mouse model genotypes, especially in long-term culture (P < 0.05) (day 10) ([173]Fig. 4H). The observation that CD4+ T cells from Trp53^mut, Trp53^mutPten^Δ, Trp53^mutPten^ΔΔ, and Pten^ΔΔ had elevated GATA3 expression compared to WT when grown under T[H]2 conditions and reduced TBX21 expression compared to WT when grown under T[H]1 conditions ([174]Fig. 4F) is quite intriguing as CD4+ T cells from C57BL/6 mice preferentially produce T[H]1 cytokines with high interferon-γ (IFN-γ) and low IL-4 ([175]41). Thus, Trp53/Pten may regulate T[H] lineage plasticity and differentiation. CRISPR-edited human CD4+ T cells with aberrant TP53 and PTEN signaling favored T[H]2 differentiation We engineered human CD4+ T cells from four healthy donors to generate genotypes similar to the murine models, including TP53^ΔΔ, TP53^R175H, PTEN^ΔΔ, PTEN^Δ, and double-mutant (TP53^ΔΔPTEN^Δ and TP53^ΔΔPTEN^ΔΔ) using a CRISPR-Cas9 approach ([176]42) ([177]Fig. 5A and fig. S4A). TP53^ΔΔ CD4+ T cells exhibited multinucleated cells (fig. S4B). Loss of TP53 and down-regulation of target genes (e.g., CDKN1A/P21 and BAX) and knockout of PTEN and PI3K activation (i.e., p-AKT) due to PTEN loss were validated at the protein or mRNA level ([178]Fig. 5B and fig. S4C). CD4+ T cells with TP53^ΔΔ exhibited the ability to proliferate both under normal conditions (CD3/CD28+ IL-2) and in the absence of IL-2 (fig. S4D), but cells with PTEN^ΔΔ were difficult to maintain in culture and underwent rapid apoptosis upon TCR activation (fig. S4E), consistent with findings in murine CD4+ T cells, indicating that PI3K hyperactivation is disadvantageous. CD4+ T cells with TP53^ΔΔPTEN^Δ displayed the capability to grow in IL-2 stimulation alone, without dependence on CD3 and CD28 activation. Notably, when cultured in IL-2 with CD3 and CD28 stimulation, these cells did not proliferate well, whereas PI3K inhibition by duvelisib treatment improved their survival, indicating that regulation of PI3K activation is important for T cell survival (fig. S4D). Fig. 5. Generation and molecular analysis of human-edited CD4+ T cells. [179]Fig. 5. [180]Open in a new tab (A) Schematic representation showing an experimental approach for the CRISPR-Cas9 engineered human CD4+ T cells. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. (B) Validation of TP53 and/or PTEN knockout (KO) models. Reduced expression of PTEN and TP53 was confirmed by Western blot, and increased phosphorylation of PTEN target AKT was confirmed in PTEN^ΔΔ or PTEN^Δ CD4+ T cells. (C) Heatmap of the DEGs in human CD4+ T cell (modified cells). (D) GSEA of the CRISPR-modified cells versus their Cas9 controls. (E) Representative (of three) flow cytometry analyses for GATA3 and TBX21 expression in TP53^ΔΔ and TP53^R175H cells showed higher GATA3+ cells versus Cas9 WT control. (F) Validation of the TCR activation by Western blotting of TP53^ΔΔ CRISPR-modified cells. (G) TP53 ChIP-seq data (GSE #86164) show the enrichment of the region within the GATA3 gene (intron-3) in two cell lines upon treatment with Nutlin-3 (top). (H) ChIP-qPCR of the indicated regions in the GATA3 promoter and intron region show significant enrichment in the intron-3 region. The promoter region of CDKN1A (p21) was used as a control. Negative control, intron-3, and p21 fold enrichment was also determined in SMZ1, TP53^R175H CD4+ T cell knock-in, and TP53^∆∆ CD4+ T cell knockout, which showed inhibited or reduced enrichment compared to 293T (WT TP53). (I) Representative flow cytometry analysis of ectopic TP53 expression in 293T cells resulting in reduced GATA3 expression. GFP, green fluorescent protein. (J) Firefly and Renilla luciferase reporter assay with cotransfection of TP53 expression vectors showed repression of luciferase activity compared to the control vector. EV, empty vector; F.Luc, firefly luciferase; R.Luc, Renilla luciferase; RPM, reads per million/kb. We performed RNA-seq analysis of the edited human CD4+ T cells versus their corresponding control Cas9 cells. Unsupervised hierarchical clustering indicated that edited T cells primarily clustered by donor and culture time (fig. S4F), and TCR analysis showed restricted TCR clonality in the modified cells, which tended to show greater inclination toward oligoclonal evolution over time in culture (fig. S4G). We compared gene signature enrichment from the CRISPR-modified CD4+ T cells to their murine CD4+ T cell counterparts. Gene signatures enriched in TP53^ΔΔ and TP53^R175H CD4+ T cells showed similarities to each other and to their mouse counterparts, including T[H]2 enrichment and STAT activation ([181]Fig. 5, C and D). TP53^ΔΔ/PTEN^Δ human and mouse cells had enrichment of T[H]2, AKT, STAT, and IL-2 response-related signatures, whereas PTEN^Δ human and mouse cells had enrichment of T[H]2 and PI3K-responsive signatures. Human CD4+ T cells with TP53^ΔΔPTEN^ΔΔ and PTEN^ΔΔ were extremely difficult to expand in culture and were cultured in the absence of TCR engagement, demonstrating an overlap with their murine counterpart and had reduced expression of proliferation-related signatures such as MYC. In agreement with murine findings, and corroborating the GEP signature finding, a higher percentage of GATA3+ (%) T cells were observed in TP53^ΔΔ (66% versus 30%; P < 0.05) and TP53^R175H (48% versus 34%) compared to their Cas9 corresponding control cells ([182]Fig. 5E). We further evaluated the expression of the GATA3 target gene CCR4 by flow cytometry and observed higher expression of GATA3+/CCR4+ in TP53^ΔΔ (44%; P < 0.05) and TP53^R172H KI (33%) compared to Cas9 control cells (21%) (fig. S4H). We validated the GEP findings, [i.e., enhanced TCR activity (p-LCK (Py505) and p-ZAP70(pY319)], PI3K-p85(Y458), and mTORC1 (Thr^37/46) in TP53^ΔΔ by using Western blot or by flow cytometry ([183]Fig. 5F and fig. S4, I and J). Although CD4+ T cells with TP53^ΔΔ showed a growth advantage in T[H]2 conditional medium, their growth was inhibited in T[H]1 medium (fig. S4K). PTEN^ΔΔ or PTEN^Δ cells did not survive under any conditional media supplement, illustrating that hyperactivation of the PI3K pathway due to PTEN loss may be detrimental for in vitro proliferation. TP53 directly regulates GATA3 expression and thus T[H]2 differentiation Murine and human CD4+ T cells showed up-regulated GATA3 expression upon TP53 loss ([184]Figs. 4, F and G, and [185]5E). We examined TP53 chromatin immunoprecipitation sequencing (ChIP-seq) datasets (see Materials and Methods; [186]Fig. 5G) and observed potential TP53 binding regions in the GATA3 proximal (PP[1] and PP[2]: chr10:8,09,6687-7460) and distal promoter (DP; chr10:8,087,212-410) and intron-3 region (chr10:8102,082-325) [ChIP–quantitative PCR (qPCR)] ([187]Fig. 5G). In silico analysis of the TP53 binding site showed a single decamer TP53 binding site at the intron-3 locus of GATA3, lacking one nucleotide but functional in an earlier study ([188]43). The TP53 binding motif (RRRCWWGYYY) is present in GATA3 intron-3 using CentriMo Local Motif Enrichment Analysis ([189]43) but lacking the third “Y.” The motif investigated is the only TP53 motif with a MEME motif score of ≥5 in GATA3 promoter or intronic regions. To validate these findings further, TP53 was ectopically expressed in 293T and TP53 ChIP-qPCR was performed to interrogate the GATA3 locus. ChIP-qPCR results showed high (>5-fold) enrichment of p53 binding at the promoter regions (PP[1] and PP[2]) and more than 10-fold enrichment at intron-3 compared to isotype control ([190]Fig. 5H). Furthermore, TP53 expression in the 293T cells resulted in lower GATA3 expression ([191]Fig. 5I). Further validation in the SMZ1 T cell line carrying TP53^F113S showed >10-fold enrichment in the intron-3 locus ([192]Fig. 5H). This enrichment at intron-3 was not observed in CD4+ T cells with TP53^R175H or TP53^ΔΔ, which also have elevated GATA3 expression relative to CD4+ T cells with WT TP53 ([193]Fig. 5E). The positive control (CDKN1A/P21 promoter) did retain partial binding in TP53^R175H cells but not in TP53^ΔΔ ([194]Fig. 5H). We cloned three target regions [i.e., distal promoter, intron-3 region, and CDKN1A/P21 promoter region (positive TP53 binding control)] into a firefly luciferase reporter and cotransfected with TP53 expression vectors and SV40 Renilla luciferase reporter in 293T cells. The reporter assay showed repression of luciferase activity by TP53 overexpression in the intron-3 region compared to empty vector, suggesting that TP53 may directly regulate GATA3 expression ([195]Fig. 5J). In addition, a 50–base pair (bp) region of the GATA3 intron-3 containing TP53 binding motif was cloned into firefly luciferase reporters, and repression of luciferase activity similar to that of the full intron-3 construct was observed. Upon mutating the motif, the luciferase activity was increased by eightfold, suggesting direct TP53 binding and repression of GATA3 at this locus ([196]Fig. 5J). The m-TCL transcriptomic signature is prognostic in h-PTCL We next identified the overlapping mouse and human transcripts to compare the hPTCL-GATA3 tumors to the m-TCLs to evaluate their association with each other ([197]Fig. 6A). We first assessed the expression of the human GATA3 classifier signature genes ([198]5) in the murine models. Whereas the PTCL-GATA3-(up) signature genes were either significantly enriched in Trp53^mutPten^Δ (P = 0.03) or trending Trp53^ΔΔPten^Δ (P = 0.07) m-TCL by GSEA, the PTCL-GATA3-(down) showed low expression in Trp53^mutPten^Δ, Trp53^ΔΔPten^Δ, Trp53^mutPten^ΔΔ, and Pten^ΔΔ ([199]Fig. 6B), suggesting a similarity between the m-TCL and hPTCL-GATA3, with a frequent codeletion of TP53 and PTEN. Fig. 6. Integrative transcriptomic analysis of human (h-PTCLs) and murine (m-TCLs). [200]Fig. 6. [201]Open in a new tab (A) Schematic representation of the ortholog analysis of h-PTCL and m-TCL transcriptomics. (B) Heatmap depicting the human GATA3 classifier ([202]5) in the m-TCLs. TP53^mutPten^Δ tumors notably show enriched PTCL-GATA3-high signature genes (P = 0.03), resembling hPTCL-GATA3. (C) Consensus DEGs between mouse TP53^mutPten^Δ tumors and hPTCL-GATA3 cases with TP53 and PTEN loss. (D) Consensus DEGs between mouse TP53^ΔΔPten^Δ tumors and hPTCL-GATA3 cases with TP53 and PTEN loss. (E) Consensus DEGs between mouse TP53^mutPten^ΔΔ tumors and hPTCL-GATA3 cases with TP53 and PTEN loss were identified and OS in another PTCL-NOS cohort on the basis of the expression of the signature. (F) Consensus DEGs between mouse Pten^ΔΔ tumors and hPTCL-GATA3 cases with PTEN loss were identified. (G) Consensus DEGs between mouse Trp53^mut tumors and hPTCL-GATA3 cases with TP53 loss were identified. (H) The consensus DE signatures were evaluated in another PTCL-NOS cohort and cases were separated into halves on the basis of differential signature expression and evaluated for OS. The bar plot shows the PTCL-subtype composition of the cases separated by low and high signature expression, indicating that PTCL-GATA3 cases were enriched in high expression signature. Next, we identified the differentially expressed genes (DEGs) in each m-TCL or the preneoplastic CD4+ T cells from each genotype, compared to either corresponding WT controls, and PCA of these DEG signatures in hPTCL-NOS cases indicated that these signatures were able to separate the PTCL-GATA3 cases into a cluster distinct from PTCL-TBX21 cases (fig. S5, A to K). Furthermore, we generated consensus gene signatures between the m-TCL genotypes and PTCL-GATA3 cases carrying the corresponding TP53/PTEN deletion ([203]Fig. 6, C to G) and evaluated whether these refined transcriptomic signatures can demonstrate diagnostic or prognostic significance in independent PTCL-NOS cohort from earlier GEP studies ([204]5, [205]6, [206]44). We observed that the gene signatures were able to subclassify PTCL-NOS cases into PTCL-GATA3 and PTCL-TBX21 subtypes (P < 0.05; [207]Fig. 6H, left), suggesting that m-TCLs recapitulate molecular pathogenesis of the PTCL-GATA3 subtype. Furthermore, the signatures demonstrated prognostic significance in this independent cohort as cases with higher differential signature expression of the Trp53^mutPten^Δ, Trp53^ΔΔPten^Δ, and the Trp53^mutPten^ΔΔ consensus signatures trended toward worse outcomes ([208]Fig. 6H, right). m-TCLs with combined TP53 and PTEN loss are therapeutically targetable We allografted Trp53^mutPten^ΔΔ and Trp53^ΔΔPten^Δ neoplastic splenocytes (P[0]) subcutaneously into nonobese diabetic severe combined immunodeficient gamma (NSG) mice to develop a cohort of tumors for preclinical assessment ([209]Fig. 7A). Tumor development in NSG mice was observed within a week at the site of inoculation, and tumors emerged in the secondary lymphoid organs around 10 days, requiring euthanasia by 30 days (P[1]). The P[1] tumor cells were adoptively transferred to generate P[2] tumors in NSG mice, and P[2] tumor formation was highly aggressive, with a 2-week survival time. We confirmed the Trp53 mutation and Pten deletion in secondary passages and confirmed the clonal relationship of the tumors from the P[0] and P[1], by Trp53 mutation and Pten deletion genotyping using Trp53/Pten rearrangement primers (table S3 and fig. S6A). In addition, IHC and analysis by flow cytometry showed consistent immunophenotype, i.e., CD3+, CD4+, CD8−, p-STAT3, and TDT− ([210]Fig. 7B). We evaluated GATA3 expression in successive generations ([211]Fig. 7C). Fig. 7. Preclinical assessment of Pten/Trp53 loss-derived tumors with PI3K inhibitors. [212]Fig. 7. [213]Open in a new tab (A) Schematic of Trp53^ΔΔPten^ΔΔ and Trp53^mutPten^ΔΔ tumors in NSG mice (P[1] and P[2]) using tumor splenocytes from parental tumor (P[0]). P1 tumors appeared in 4 to 6 weeks post-P[0] engraftment, whereas P[2] tumors appeared in 2 weeks post-P1 engraftment in NSG mice. (B) IHC analysis of the P[0], P[1], and P[2] Trp53^mutPten^ΔΔ and Trp53^ΔΔPten^Δ tumors reaffirm the expression of CD3, CD4, and CD8 in tumor cells and no TDT expression. (C) Representative flow cytometry analysis showed that GATA3 expression was elevated compared to WT-NSG (n = 3) in P[0] and P[1] Trp53^mutPten^ΔΔ (n = 6) and Trp53^ΔΔPten^Δ (n = 3) samples. (D) Kaplan-Meier plot displaying the OS advantage in P2 mice treated with duvelisib compared to vehicle control. (E) Duvelisib-treated mice had smaller observed spleens than vehicle control mice. (F) Kaplan-Meier plot displaying the OS advantage in P2 mice treated with copanlisib compared to vehicle control. (G) Copanlisib-treated mice had smaller observed spleens than vehicle control mice. To test the effectiveness in vivo, P[2] mice generated from Trp53^mutPten^ΔΔ and Trp53^ΔΔPten^Δ were each randomized into two groups (n = 6 each) and treated with clinical-grade duvelisib (10 mg/kg) and copanlisib (10 mg/kg) compared with vehicle treatment control. We observed a survival advantage with reduced splenomegaly in mice treated with either duvelisib (P < 0.001) ([214]Fig. 7, D and E) or copanlisib (P = 0.005) ([215]Fig. 7, F and G). Although the two inhibitors target different PI3K components, we analyzed a few spleens after treatment by IHC and observed decreased p-STAT3 expression (fig. S6, B and C). These data suggest that aberrant p53 signaling, a hard-to-target pathway, is vulnerable to PI3K inhibition in preclinical models. DISCUSSION Recent multiomics studies have delineated two biological subtypes within PTCL-NOS, with the PTCL-GATA3 subtype frequently associated with codeletion of TP53 and PTEN and a poor outcome ([216]2, [217]5, [218]6, [219]44). GATA3, known for its role in T[H] cell differentiation, also regulates T[H]2 cell growth and proliferation and has been also shown to have a proto-oncogenic role in T cell neoplasms ([220]10, [221]45). We generated CD4+ T cell conditional murine models with hotspot mutation Trp53^R172H or Trp53 loss (Trp53^mut or Trp53^ΔΔ) combined with Pten loss (Pten^Δ) and observed CD4+ T cell m-TCLs ([222]37) with GATA3 immunophenotype and enriched with T[H]2-like GEP. Trp53^mutPten^Δ promoted lymphoma development with increased thymic or splenic cellularity and a significantly higher number of CD4+ T cells, B cells, and myeloid cells. The altered cellularity with a significant increase in B cells in the thymus may be attributed to depletion or negative selection of CD4+ thymocytes ([223]38), as noted in Pten-deficient HY-TCR transgenic mice ([224]46). Trp53^mutPten^ΔΔ showed accelerated lymphoma development with significant enrichment of CD4+ T cells in the lymphoid organs, consistent with previous findings ([225]47). Similar histological and molecular observations were noted in Trp53^ΔΔPten^Δ, thus reaffirming the cooperative role of the two genes in lymphomagenesis. Trp53^mutPten^Δ and Trp53^ΔΔPten^Δ mice also developed m-TCLs with including DP CD4+CD8+ immunophenotype. The DN (CD4^−CD8^−) to DP (CD4^+CD8^+) transition during T cell maturation is dependent on the pre-TCR expression and negatively regulated by Trp53 ([226]48); thus, enriched DP immunophenotype in some neoplasms is expected and reaffirming its role in the maturation phase. These neoplasms were further characterized with RNA-seq, indicating a T[H]2-like transcriptomic program, characterized by high Gata3 mRNA and protein expression and enrichment of its target genes. These neoplasms also demonstrated activated TCR, STAT3 signaling, and MYC-regulated transcriptome, where the loss of TP53 regulation could be driving this phenotype ([227]49, [228]50). TP53-directed STAT3-MYC oncogenic signaling has been noted earlier ([229]51), and genetic and transcriptomic analysis showed that MYC contributed to the molecular mechanisms underlying tumorigenesis in these murine models. CN gains in MYC are frequent in PTCL-GATA3 ([230]12), but the loss of TP53 also allows MYC expression to be up-regulated due to the disruption of direct TP53 regulation at the MYC promoter, which could play a role in breaking senescence during transformation ([231]52) rather than MYC being the primary driver of lymphomagenesis. Whether the MYC deregulation is caused by secondary genetic changes is something that needs to be investigated further. To understand the functional properties of the CD4+ T cells from the murine models, in vitro culture demonstrated that loss of p53 signaling promotes TCR-independent growth. However, Pten loss resulted in rapid apoptosis in mature human and murine CD4+ T cells upon TCR engagement but is ameliorated in the presence of Trp53 mutation under low serum/stress conditions, suggesting that a hyper-PI3K signal is highly detrimental to mature T cell survival. This may explain the observation of the rarity of homozygous PTEN loss in PTCL-GATA3 as PTEN expression levels and the associated PI3K signaling are needed to be maintained within a certain range for T cell growth and survival. The Trp53^mutPten^Δ or Trp53^ΔΔPten^Δ CD4+ T cells adapted to better survival under low IL-2 conditions, and this advantage disappeared when cultured with high IL-2. These cells may have a selective advantage under in vivo conditions due to their ability to grow even in limited IL-2 concentration, and pro-survival adaptation under stress conditions as observed in serum/nutrient depletion assays, thus adapting to changes for tumor expansion under in vivo conditions, although this was not evident with engraftment of preneoplastic cells from the same genotype. These findings with murine models were largely concordant in human CD4^+ T cells and suggest that alterations of several signaling pathways (e.g., cell cycle transition, proliferation, T cell activation, metabolism, and T[H]1 differentiation block), mediated by TP53 and PI3K deficiencies, are central events in T[H]2 cell lymphoma initiation. As noted, TP53 deletion facilitates T cell activation and survival in in vitro human CD4+ T cells. Whereas Pten^ΔΔTP53^Δ or Pten^ΔΔ were challenging to grow under in vitro conditions for a longer period, TP53^Δ and TP53^mut survived for long-term cell cultures and demonstrated higher TCR activity and GATA3 expression. In addition, we observed that ectopic expression of TP53 in human cell lines down-regulated GATA3 expression. ChIP-seq and ChIP-qPCR validated a TP53 binding site at the intron-3 locus of GATA3 involved in the transcriptional regulation of GATA3, and luciferase reporter assays showed that inclusion of the intron-3 region in luciferase vector transduction reduced luciferase transcriptional activity with increased p53 expression compared to baseline levels. Inducing a mutation at a proposed “half” p53 binding motif located in this region resulted in increased luciferase expression, reversing the inhibition observed with the WT motif. The mechanistic link between TP53 and GATA3 is strongly evident from the ChIP-qPCR, motif enrichment analysis, and functional luciferase assays, thus establishing the role of p53 in regulating GATA3 expression. Together, this suggests that p53 signaling is involved in regulation of GATA3 and T[H]2 differentiation at this locus, beyond its canonical function of cell cycle regulation. Although the current understanding of T[H] lineage fate decisions point to extrinsic factors that drive naïve CD4+ T cell differentiation ([232]53, [233]54), we demonstrated that the intrinsic mechanisms including p53 and PI3K signaling may cooperate to polarize naïve CD4+ T cells toward a T[H]2 differentiation and block T[H]1 differentiation, as observed in polarization assays. The aberrations may result in the activation of STAT3 and MYC signaling and is a hardwired response due to PTEN and TP53 loss, consistent with an earlier finding that STAT3 signaling promotes T[H]2 differentiation ([234]33). We performed comparative transcriptomic analysis on the hPTCL-NOS and m-TCLs to establish the relevance of murine models to hPTCLs. We observed that PTCL-GATA3 signatures are significantly associated with Trp53^mutPten^Δ and Trp53^ΔΔPten^Δ m-TCLs, suggesting that murine models may recapitulate PTCL-GATA3 lymphomagenesis. Integrative analysis of m-TCLs with hPTCLs carrying del-TP53 and del-PTEN defined molecular distinction within PTCL-NOS (i.e., PTCL-GATA3 and PTCL-TBX21). The consensus mRNA signatures were enriched with a T[H]2-like gene expression program, PI3K, STAT3, and MYC-mediated transcriptional program and showed association with prognosis within PTCL-NOS. MYC expression is commonly increased in PTCL-GATA3 and our biological models. The cooperative loss of TP53 and PTEN in PTCL-GATA3 also presents an unprecedented opportunity for therapeutic vulnerability by blocking PI3K activation. To demonstrate therapeutic relevance, we generated preclinical orthotropic models engrafted with Trp53^mutPten^ΔΔ or Trp53^ΔΔPten^Δ m-TCLs in NSG mice and further passaged in NSG mice to generate a preclinical cohort. These cohorts were randomized and tested a clinical-grade PI3K inhibitor (i.e., duvelisib or copanlisib), and PI3K inhibitor treatment led to a survival advantage and reduced PI3K activation. Although the use of PI3K inhibition itself is not novel, we determined that its use in PTCL-GATA3 has a targeted value due to the dependency frequent up-regulation of the PI3K/AKT pathway. These data provide a rationale for the hard-to-target pathway (e.g., aberrant p53 signaling), which are vulnerable to PI3K inhibition in preclinical m-TCL models. Although there are no standard treatments available for PTCLs, the role of PI3K inhibitors can be logically extended to mature PTCLs, including PTCL-GATA3 or other PI3K-dependent T cell malignancies. Although PI3K inhibitors (e.g., duvelisib and copanlisib) are used in clinical settings for PTCL with T[FH] immunophenotype ([235]55, [236]56), our findings suggest that PTCL-GATA3 subtype with TP53 and PTEN alterations may show increased sensitivity to PI3K blockade. The therapeutic responses observed in preclinical Trp53^mutPten^ΔΔ and Trp53^ΔΔPten^Δ murine models supports the effectiveness of PI3K inhibitors. Although dysregulated TP53 and PTEN signaling represents a frequent genetic alteration in PTCL-GATA3, there are other targetable pathways in this recently identified PTCL entity. FISH and/or next-generation sequencing could be implemented clinically to identify TP53 and PTEN aberrations as our study has highlighted the prognostic importance, as well as identifying pathways that could be therapeutically targeted, like the PI3K/AKT pathway (PI3K inhibitors) or IL-2–dependent signaling (anti-CD25 therapy). Nevertheless, translational success may require additional preclinical patient-derived xenograft (PDX) models followed up by phase 1 clinical trials to evaluate other drug combinations for clinical improvement. In summary, our in-depth molecular and cellular characterization using distinct T cell murine models and innovative genetically edited human CD4+ T cell culture systems showed that the frequently observed TP53 and PTEN alterations promoted GATA3 expression and T[H]2 differentiation and activation of diverse oncogenic pathways promoted T cell transformation. These observations provide direction for future clinical-translational studies in hard-to-diagnose and hard-to-treat PTCL subtypes. MATERIALS AND METHODS Detailed materials and methods are included in Supplementary Materials and Methods. Experimental design The objective of the study was to generate a mouse model representative of PTCL-GATA3. We generated six mouse models with Trp53 and Pten abnormalities: Pten-heterozygous (Pten^Δ:Cd4-Cre^+/−;Pten^fl/+), Pten-homozygous (Pten^ΔΔ:Cd4-Cre^+/−;Pten^fl/fl), Trp53-mutant (Trp53^mut:Cd4-Cre^+/−;Trp53^LSL-R172H+/−), Trp53-mutant; Pten-heterozygous (Trp53^mut Pten^Δ: Cd4-Cre^+/−; Trp53^LSL-R172H+/−;Pten^fl/+), and Trp53-mutant; Pten-homozygous (Trp53^mut Pten^ΔΔ: Cd4-Cre^+/−;Trp53^LSL-R172H+/−; Pten^fl/fl) andTrp53-knockout; Pten-heterozygous (Trp53^ΔΔPten^Δ: Cd4-Cre^+/− Trp53^fl/fl;Pten^fl/+) and compared them to WT (Cd4-Cre^−/−) for phenotype and functional assessments. CRISPR-edited human CD4+ T cells with TP53 and PTEN abnormalities were generated to validate findings from murine CD4+ T cells. Preclinical evaluation of PI3Kγ/δ inhibitors duvelisb and copanlisib was completed using Trp53^mutPten^ΔΔ and Trp53^ΔΔPten^Δ allografts into NSG mice. Patient information The basic clinical and pathological characteristics of PTCL-NOS cases are included in table S1. PTCL-NOS cases with expression of two or more T follicular helper (T[FH]) markers were excluded, and the remaining cases were subclassified into PTCL-GATA3 and PTCL-TBX21 using transcriptomic-based molecular diagnostic signatures ([237]5) or a recently defined algorithm on the basis of IHC ([238]11). The research protocol was approved by the UNMC and COH-MC Institutional Review Board under protocol number 0543-09-EP. Human CD4+ T cell culture and genetic editing Human CD4+ T cells were isolated from peripheral blood mononuclear cells (PBMCs) of healthy donors using the EasySep Human CD4+ T cell Isolation Kit (STEMCELL Technologies Inc., Vancouver, Canada, #17952) according to the manufacturer’s instructions. Specific sequences of CRISPR RNA (crRNA) and genetic editing methods can be found in the Supplementary Materials. Generation of murine models All mouse models generated were on a C57BL/6 background, bred, and monitored at the UNMC animal facility under the supervision of trained veterinarians and analyzed per the UNMC animal ethics committee regulations under the approved Institutional Animal Care and Use Committee (IACUC) protocols 16-130-12-FC and 16-131-12-FC. Specifics into the acquisition/breeding methods for all murine models can be found in Supplementary Materials and Methods. Mouse genotype analysis was performed by using 2 x M-PCR OPTI Mix (B4001), per the manufacturer’s protocol. To confirm genotypes and gene allelic rearrangement in the murine models, gene-specific PCR was used on isolated CD4+ T cells, and the detailed primers are shown in table S3. Histological, IHC, and flow cytometry analysis of murine tumors Mice were monitored regularly and euthanized in accordance with IACUC guidelines. Necropsy was performed when indicated, and tissues were collected for histology, immunophenotyping, PCR, and Western blotting. Thymus, spleen, lymph nodes (axillary, brachial, and inguinal), and tumor-affected infiltrated organs of lymphoma-bearing animals were dissected, weighed, and fixed in 10% buffered formalin for 24 hours before being paraffin embedded at the Tissue Science Facility (UNMC) for histological analysis. Tissue sections and flow cytometry staining details can be found in tables S4 and S5 and Supplementary Materials and Methods. The cellularity of the tumors was evaluated using flow cytometry (ACEA NovoCyte), and data analysis was performed with NovoExpress software. Murine CD4+ T cell isolation and cellular assays Spleens were dissected from all the genotypes of murine models and age-matched WT mice, and single-cell suspensions were prepared after red blood cell lysis and final 1x phosphate-buffered saline (PBS) wash. CD4+ T cells were isolated from the total splenocytes using the EasySep Mouse CD4+ T Cell Isolation Kit (STEMCELL Technologies). Further details on proliferation, apoptosis, polarization, and Western blotting can be found in tables S6 and S7 and the Supplementary Materials. RNA-seq and WES analysis for murine cells or neoplasm RNA-seq libraries were prepared with 175 ng of RNA [RNA integrity number (RIN) value > 7 to 9] extracted from either CD4+ T cells or tumor tissue following the Illumina TruSeq RNA Sample Preparation v2 protocol (Illumina). The samples were sequenced on a NextSeq500 instrument to obtain 20 million, 75-bp paired-end reads. The sequencing reads were aligned using the STAR aligner (v2.5.3). Counts per gene were assessed using HTSeq (v0.9.1). HTSeq counts normalized by DESeq2 and imported into BRB-ArrayTools for analysis. TCR and B cell receptor (BCR) clonotypes were analyzed using MiXCR (v3.0.7). For WES, libraries were captured (SureSelect XT Mouse All Exon Kit, Agilent), amplified, and sequenced on an Illumina sequencer as described earlier ([239]12). Read quality was assessed with FASTQC (v. 0.11). High-quality reads were mapped to the mouse genome (mm10) using BWA-mem (v.0 0.7.17-r1188). Duplicates were marked with Picard (v. 2.9.0), followed by base recalibration by GATK (v.4-4.1.8.1). Variant calling was done with Varscan2 (v2.4.4) and variant annotation with ANNOVAR ([240]57). ChIP and luciferase assay ChIP and luciferase assay protocol specifics can be found in Supplementary Materials and Methods. The primer sequences for GATA3 regions in both assays are provided in table S3. These regions were preselected from earlier ChIP-seq data ([241]43) as potential TP53 binding sites. Luciferase assay was performed per the manufacturer’s instructions with the Dual-Luciferase Reporter Assay System (Promega Corp. Inc.). Drug treatment and monitoring of subcutaneously inoculated tumor cells Trp53^mutPten^ΔΔ or Trp53^ΔΔPten^Δ tumor cells (1 × 10^6) were injected subcutaneously in NSG mice (4 to 6 weeks old), and tumor growth was assessed every other day. Imaging details can be found in the Supplementary Materials. When engrafted tumors reached a volume of ~100 mm^3, drug treatments were started. Mice were split into two groups: vehicle control [10% PEG-400 (polyethylene glycol, molecular weight 400) + 90% PBS] and duvelisib (10 mg/kg) or copanlisib (10 mg/kg). Statistical analysis Comparisons between the two groups were conducted using a two-tailed Student’s t test. P values < 0.05 were considered significant. The data analysis for this manuscript was conducted using R ([242]https://r-project.org/). Graphs were generated using the R packages ggplot2 and survival and Circos ([243]http://circos.ca). The Kaplan-Meier method was used to estimate the OS distributions using the R survminer package. OS times were calculated as the time from diagnosis to the date of death or last contact. Patients who were alive at the last contact were treated as censored for the OS analysis. The log-rank test was used to compare survival distributions. All statistical tests are two sided, and P values less than 0.05 are statistically significant unless otherwise specified. P values were considered significantly different among groups, with *P < 0.05, **P < 0.001, and ***P < 0.0001, unless otherwise indicated in the figures. Acknowledgments