Cross-Species Comparison of Acquired Genetic Changes In T Cell Malignancy.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1192-1192
Author(s):  
Lynnie A. Rudner ◽  
Kim H. Brown ◽  
Kimberly P. Dobrinski ◽  
Diana F. Bradley ◽  
Jonathan Downie ◽  
...  

Abstract Abstract 1192 T cell acute lymphoblastic leukemia (T-ALL) is a challenging clinical entity. Over half of adult, and about 20% of pediatric, T-ALL patients either relapse or fail to achieve remission, and despite salvage attempts outcomes are poor. To discover new acquired genetic changes that occur in T-ALL, as well as those that contribute to disease relapse, we studied zebrafish (Danio rerio) T-ALL samples using array comparative genomic hybridization (aCGH). Five different D. rerio T-ALL models have been described, and all 5 develop neoplasias that clinically and molecularly resemble human T-ALL. We performed aCGH on 14 de novo T-ALLs representing 4 of these models. To evaluate possible similarities between human and zebrafish copy number aberrations (CNAs) in T-ALL, we compared all D. rerio genes found in any CNA in our 14 zebrafish cancers to a cohort comprised of 61 published primary human T-ALLs analyzed by aCGH. In these D. rerio CNAs, we identified 764 genes with human homologues. Of these, we found significant overlap (62%) with genes in CNAs from the human T-ALL dataset. In addition, 10 genes recurrently altered (>3 samples) in zebrafish T-ALL were also seen in CNAs from 5 or more human T-ALL cases, suggesting a conserved role for these loci in T-ALL transformation across vertebrate species. In addition to studying primary T-ALL, we also conducted iterative allo-transplantations with 3 zebrafish malignancies. This technique selects for particularly aggressive disease, increasing engraftment rates and also resulting in shorter time to death of recipient animals in successive transplant rounds. Because these passaged cancers show more malignant behavior in vivo, this procedure models refractory and relapsed T-ALL. In these 3 serially-passaged cancers, 55% of the original CNAs were preserved after iterative transplantation, demonstrating the clonal relationship between the primary and passaged leukemia. In addition, 101 CNAs were acquired during passaging of the 3 T-ALLs. Genes in these loci may underlie the enhanced malignant behavior of these neoplasias. We also compared genes found in CNAs from passaged zebrafish malignancies to human aCGH results from 50 T-ALL patients who failed induction, went on to relapse, or had already relapsed. Again, many genes (n=76) were present in both datasets. Four genes altered in 2 of 3 D. rerio samples were also found in >4 cases from the human dataset. Collectively, these results suggest that zebrafish and human T-ALL are similar at the genomic level, with several homologous genes commonly gained or lost by cancers from both organisms. Genes from recurring CNAs in disease samples from both species may have particular relevance, as these candidates likely have conserved roles in T cell oncogenesis or T-ALL disease progression. As the comprehensive list of genes in CNAs from human T-ALL cases is vast and heterogeneous (comprising 15,724 unique genes from just 75 clinical samples), zebrafish T-ALL models may permit a more expedient prioritization of these candidates for further investigation. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 810-810 ◽  
Author(s):  
Haiying Qin ◽  
Sang M Nguyen ◽  
Sneha Ramakrishna ◽  
Samiksha Tarun ◽  
Lila Yang ◽  
...  

Abstract Treatment of pre-B cell acute lymphoblastic leukemia (ALL) using chimeric antigen receptor expressing T cells (CART) targeting CD19 have demonstrated impressive clinical results in children and young adults with up to 70-90% complete remission rate in multiple clinical trials. However, about 30% of patients relapse due to loss of the targeted epitope on CD19 or CART failure. Our CD22-targeted CAR trial has generated promising results in relapsed/refractory ALL, including CD19 antigen negative ALL, but relapse associated with decreased CD22 site density has occurred. Thus, developing strategies to prevent relapses due to changes in antigen expression have the potential to increase the likelihood of durable remissions. In addition, dual targeting of both CD19 and CD22 on pre-B ALL may be synergistic compared to targeting a single antigen, a potential approach to improve efficacy in patients with heterogeneous expression of CD19 and CD22 on leukemic blasts. We describe the systematic development and comparison of the structure and therapeutic function of three different types (over 15 different constructs) of novel CARs targeting both CD19 and CD22: (1) Bivalent Tandem CAR, (2) Bivalent Loop CAR, and (3) Bicistronic CAR. These dual CARs were assembled using CD19- and CD22-binding single chain fragment variable (scFv) regions derived from clinically validated single antigen targeted CARs. They are structurally different in design: both tandem and loop CARs have the CD19 and CD22 scFv covalently linked in the same CAR in different orders, whereas, bicistronic CARs have 2 complete CAR constructs connected with a cleavable linker. The surface expression on the transduced T cell of the CD19/CD22 dual CARs was detected with CD22 Fc and anti-idiotype of CD19 and compared to single CD19 or CD22 CARs. Activities of dual CARs to either CD19 or CD22 were evaluated in vitro with cytotoxicity assays or killing assays against K562 cells expressing either CD19 or CD22 or both antigens and also tested against a leukemia CD19+/CD22+ cell line, NALM6, and NALM6 with CRISPER/CAS9 knockout of CD19 or CD22 or both antigens. Therapeutic function of the top candidates of the dual CARs was then validated in vivo against these NALM6 leukemia lines. Some of these dual CARs were also further tested against patient-derived xenografts. Finally, we tested the dual targeting CARs in an artificial relapse model in which mice were co-injected with a mix of CD19 knockout and CD22 knockout NALM6 leukemia lines. From these studies, we established that the order of the scFv, size of the linker, type of leader sequence, and co-stimulatory domain in the CAR constructs all impact the efficacy of the dual targeting CARs. Tandem, Loop, and Bicistronic CARs all demonstrate some levels of in vitro and in vivo activities, but the bicistronic CAR was most effective at clearing leukemia and preventing relapse. In the CD19+/CD22+ NALM6 model, bicistronic CAR treated mice remain disease free while CD19 CAR or CD22 CAR treated mice already died or relapsed on day 27. In the relapse model, as expected, CD19 or CD22 single CAR T cell treatment resulted in progression of the corresponding antigen-negative NALM6. Treatment with dual targeted bicistronic CARs resulted in clearance of both CD19 and CD22 negative ALL with durable remission. In summary, we described novel CD19/CD22 dual targeting CARs with robust pre-clinical activity against pre-B cell ALL, and validated this approach in the prevention of resistance to single-antigen targeted CARs in preclinical models. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1283-1283
Author(s):  
Scott McComb ◽  
Julia Aguadé-Gorgorió ◽  
Blerim Marovca ◽  
Lena Harder ◽  
Gunnar Cario ◽  
...  

Abstract Dysregulation of apoptotic pathways provides an indiscriminate mechanism for refractory acute lymphoblastic leukemia (ALL) to escape cell death induced by many chemotherapeutic compounds. Here we have assessed the potential of SMAC mimetic (SM) compounds to short circuit cell death resistance by blocking the pro-survival cellular inhibitor of apoptosis (cIAP) proteins. By screening a large set of patient-derived precursor B-cell ALL samples in an ex vivo model of the leukemia microenvironment we detect exquisite sensitivity to two different SM compounds, Birinapant and LCL161, in about one third of ALL samples. Strong ex vivo SM activity correlated with potent in vivo anti-leukemic efficacy against de novo refractory and relapsed ALL xenografts. Intriguingly, we find that although SM-sensitivity is independent of TNF and TNFR1 levels, expression of TNFR2 is highly predictive of response to SM in two separate cohorts of ALL samples, suggesting that TNFR2 expression may represent a promising biomarker for identifying SM-sensitive cells. Downstream, we employ a novel and powerful multi-colour Lenti-CRISPR approach to show that simultaneous disruption of both apoptotic and necroptotic genes is necessary to block SM-induced death. In contrast, disruption of RIP1 alone was adequate to block SM-induced apoptosis and necroptosis. Surprisingly, RIP1 loss had no significant impact on response to standard anti-leukemic therapies, supporting a view that the RIP1-dependent death pathway is not likely to be selected against in leukemia cells that have failed to respond to front line therapy. These results provide the first evidence that SM compounds can circumvent apoptotic escape in drug-refractory ALL through parallel activation of both RIP1-dependent apoptosis and necroptosis. Furthermore, our data strongly support further development of SM as anti-leukemic agents for treatment in resistant disease. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5058-5058
Author(s):  
Jianda Hu ◽  
Yanxin Chen ◽  
Zhengjun Wu ◽  
Lingyan Wang ◽  
Jingjing Wen ◽  
...  

Chemotherapy resistance is considered to be the principal cause of ineffective treatment in acute lymphoblastic leukemia (ALL). Nucleolin (NCL) is high expression andplays oncogenic roles in most cancers. However, less research on the role of NCL in hematologic malignancies was noted. Our previous studies have showed that overexpression of NCL was associated with worse prognosis in the patients with acute leukemia and NCL expressionwashigher in resistant HL-60/ADR than in sensitive HL-60 cells. The potential mechanisms of NCL in chemotherapy resistance have yet to be revealed. Here we presented that expression of NCL was associated positively with chemotherapy resistance and poor prognosis in ALL. Overexpressed NCL at both mRNA and protein level was relevant to a poorer overall survival (OS) and relapse free survival (RFS), indicating NCL as an independent prognostic marker in ALL. mRNA level of NCL in de novo ALL was quantitatively higher than in complete remission(CR) status, and refractory/relapse ALL had the highest level. Upon above clinical data, we further investigated the mechanism(s) by which NCL regulated drug resistance in ALL cells. Remarkably, NCL expression was higher in resistant ALL cells relative to sensitive parental cells. When treated with ADM, NCL level was decreased in sensitive parental cells while unchanged in resistant cells. Overexpressing NCL suppressed drug sensitivity, altered drug effluxion and decreased intracellular drug accumulation, while inhibition of NCL led to a completely reversed appearance, more intracellular Adriamycin(ADM) mean fluorescence intensity (MFI) and percentage of ADM accumulated cells population. Overexpression of NCL increased significantly the IC50 of ADM. The IC50 of ADM on Jurkat-NCL-overexpression(OE), Jurkat-NCL-knockdown(KD), Molt-4-NCL-OE, Molt-4-NCL-KD, Nalm-6-NCL-OE, Nalm-6-NCL-KD were 1.362±0.271μg/ml, 0.077±0.010μg/ml, 4.863±0.733μg/ml, 0.081±0.018μg/ml, 0.237±0.042μg/ml and 0.046±0.002μg/ml, respectively (P <0.05). Involvement of ATP-binding cassette (ABC) transporters was proved in NCL mediated drug resistance. Silencing NCL resulted in a decrease of P-gp, MRP1, LRP and BCRP in ALL cells, and NCL overexpression increased the MRP1, LRP and BCRP. The Akt/mTOR and ERK signaling pathways were involved in this procedure. Notably, co-IP assays confirmed the NCL-Ras, NCL-ERK and NCL-BCRP interaction. For intervention study, aptamer AS1411, a NCL inhibitor, could reduce drug resistance in ALL cell lines and primary ALL cells.Moreover, AS1411 treatment decreased BCRP protein expression. Furthermore, the ALL leukemia models that nude mice engrafted with Nalm-6 cells and NCG mice engrafted with Luc+ Nalm-6 cells were established, then treated with ADM plus AS1411 or control CRO for comparison drug sensitivity and survival. Growth of subcutaneous xenograft tumors was inhibited in those treated with AS1411 or ADM, compared to their respective controls treated with CRO or PBS. The stronger inhibition effect was observed in those treated with AS1411 combined with ADM. For Luc+Nalm-6 derived ALL model, leukemia progression was suppressed in mice treated with AS1411 and AS1411 combined with ADM. AS1411and ADM, especially combination of AS1411 and ADM, could improve survival of the leukemic mice compared to those treated with PBS. The results showed that NCL targeted by AS1411 sensitized ADM treatment and prolonged survival in vivo. In summary, our findings revealed NCL as a survival predictor and the novel role of NCL in ALL chemo-resistance. NCL may be a potential target for improving outcome in ALL. Disclosures No relevant conflicts of interest to declare.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 6580-6580
Author(s):  
Ofelia Crombet Ramos ◽  
Claudia Hernandez ◽  
Kevin Morrow ◽  
John T. Cole ◽  
Paulo Rodriguez

6580 Background: Advances in therapies have resulted in an overall complete remission rate of approximately 85% for childhood acute lymphoblastic leukemia (ALL). In contrast, the overall remission rate of adults with leukemia continues to be poor, only about 40% in cases of T cell-ALL (T-ALL). Therefore, it is imperative to generate new therapies that alone or in combination with other treatments could potentially increase the percentages of complete responders or be used to treat the refractory ALL population. Our published results show that a pegylated form of human arginase I (peg-Arg I) prevented T-ALL cell proliferation in vitro and in vivo through the induction of tumor cell apoptosis. Interestingly, the anti-leukemic effects induced by peg-Arg I did not affect the anti-tumor activity of normal T cells, suggesting an anti-tumor specific effect. Our hypothesis states that peg-Arg I has an anti-tumoral effect on B-ALL and T-ALL cells in vitro and that the sensitivity of ALL cells to peg-Arg I depends on their expression of argininosuccinate synthase (ASS) and their ability to produce L-arginine de novo from citrulline. Methods: Malignant T cell proliferation was tested using nonradioactive cell proliferation yellow tretrazolium salt kit. Apoptosis studies were based on the expression of annexin V. Western blot assays were conducted to determine enzymatic expression in different cell lines. Results: The results of our in vitro experiments showed that peg-Arg I had a pro-apoptotic and anti-proliferattive effect on B-ALL cells similar to the one previously seen on T-ALL cells. These effects can be overcome in cell lines able that express ASS and therefore to produce L-arginine de novo. Conclusions: Our results suggest the role of ASS in the ALL-apoptosis induced by peg-Arg-I. Our next steps include: _Understand why ASS-expressing ALL cells do not undergo apoptosis when cultured with peg-Arg-I_Determine the role of ASS in the anti-leukemic effect induced by peg-Arg-I in vivo. Completion of this research is expected to lead to a better understanding of how peg-Arg-I kills ALL cells and could provide the foundation for a novel therapy for ALL patients.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 783-783
Author(s):  
Yuqing Sun ◽  
Hongzhi Miao ◽  
Zhenhua Zou ◽  
Bo Zhou ◽  
Kai Ge ◽  
...  

Abstract HOXA9 is a homeodomain-containing transcription factor that regulates hematopoietic stem cell renewal and differentiation and is commonly over expressed in acute leukemia, including acute myeloid leukemia (AML), and T- and B-precursor acute lymphoblastic leukemia (B-ALL and T-ALL). Together with its co-binding factor MEIS1, HOXA9 has been shown to play a causal role in leukemic transformation; however, the mechanism through which HOXA9 promotes leukemogenesis is poorly understood. Previously, we showed that HOXA9 primarily binds to promoter-distal regions of the genome that show histone H3 lysine 4 (H3K4) monomethylation and histone H3 and H4 acetylation, epigenetic signatures indicative of active enhancers. HOXA9 cobinds with other lineage specific transcription factors such as C/EBPα, which we previously showed to be essential for leukemic transformation. This suggests that HOXA9 functions in a multi-subunit complex including lineage-specific transcription factors as well as chromatin modulators, but the role of HOXA9 in promoting the formation of these "enhanceosomes" and how HOXA9 alters the enhancer landscape remains unknown. In these studies, we found that in both myeloid and lymphoid murine leukemia models, HOXA9 alters the enhancer landscape through creation of de novo enhancers, many of which are active in other cell lineages in early embryogenesis. RNA expression analysis revealed that these de novo enhancers drive a leukemia-specific transcription program, whose up regulation is significantly impaired upon either HOXA9 inactivation or CRISPR-mediated deletion of specific HOXA9-bound enhancer sequences. Protein and chromatin immunoprecipitation studies showed that HOXA9 physically interacts with the MLL3/MLL4 histone methyltransferase complex and colocalizes with MLL3/MLL4 at many sites in vivo . HOXA9 is required for the recruitment of C/EBPα, the MLL3/MLL4 complex and histone H3 lysine 4 monomethylation at de novo enhancers. This activity of HOXA9 is essential for the activation of genes regulated by de novo enhancers and is associated with increased interaction of these enhancers with promoters as assessed by chromosome conformation capture (4C). In contrast, HOXA9 is dispensable for both C/EBPα and MLL3/MLL4 binding and H3K4 monomethylation at enhancers active in normal hematopoietic cells. Genetic disruption of components of the MLL3/MLL4 complex abrogates the active epigenetic profile of de novo enhancer regions, and significantly delays leukemia progression driven by HOXA9/MEIS1 in vivo . Together these findings show that HOXA9 reprograms the enhancer landscape of hematopoietic progenitors in leukemic cells, including formation of many de novo enhancers active during early embryonic development. This mechanism involves HOXA9-dependent recruitment of MLL3/MLL4 methyltransferase complexes, suggesting that targeting this methyltransferase complex could be an effective strategy for malignancies associated with HOX deregulation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1322-1322
Author(s):  
Manabu Kusakabe ◽  
Ann Chong Sun ◽  
Kateryna Tyshchenko ◽  
Rachel Wong ◽  
Aastha Nanda ◽  
...  

Abstract Mechanistic studies in human cancer have relied heavily on established cell lines and genetically engineered mouse models, but these are limited by in vitro adaptation and species context issues, respectively. More recent efforts have utilized patient-derived xenografts (PDX); however, as an experimental model these are hampered by their variable genetic background, logistic challenges in establishing and distributing diverse collections, and the fact they cannot be independently reproduced. We report here a completely synthetic, efficient, and highly reproducible means for generating T-cell acute lymphoblastic leukemia (T-ALL) de novo by lentiviral transduction of normal CD34+ human cord blood (CB) derived hematopoietic progenitors with a combination of known T-ALL oncogenes. Transduced CB cells exhibit differentiation arrest and multi-log expansion when cultured in vitro on OP9-DL1 feeders, and generate serially transplantable, aggressive leukemia when injected into immunodeficient NSG mice with latencies as short as 80 days (median 161 days, range 79-321 days). RNA-seq analysis of synthetic CB leukemias confirmed their reproducibility and similarity to PDX tumors, while whole exome sequencing revealed ongoing clonal evolution in vivo with acquisition of secondary mutations that are seen recurrently in natural human disease. The in vitro component of this synthetic system affords direct access to "pre-leukemia" cells undergoing the very first molecular changes as they are redirected from normal to malignant developmental trajectories. Accordingly, we performed RNA-seq and modified histone ChIP-seq on nascently transduced CB cells harvested from the first 2-3 weeks in culture. We identified coordinate upregulation of multiple anterior HOXB genes (HOXB2-B5) with contiguous H3K27 demethylation/acetylation as a striking feature in these early pre-leukemia cells. Interestingly, we also found coordinate upregulation of these same HOXB genes in a cohort of 264 patient T-ALLs (COG TARGET study) and that they defined a subset of patients with significantly poorer event-free survival (Log-rank p-value = 0.0132). Patients in the "HOXB high" subgroup are distinct from those with ETP-ALL, but are enriched within TAL1, NKX2-1, and "unknown" transcription factor genetic subgroups. We further show by shRNA-mediated knockdown that HOXB gene expression confers growth advantage in nascently transduced CB cells, established synthetic CB leukemias, and a subset of established human T-ALL cell lines. Of note, while there is prior literature on the role of HOXA genes in AML and T-ALL, and of HOXB genes in normal HSC expansion, this is the first report to our knowledge of a role for HOXB genes in human T-ALL despite over 2 decades of studies relying mostly on mouse leukemia and cell line models. The synthetic approach we have taken here allows investigation of both early and late events in human leukemogenesis and delivers an efficient and reproducible experimental platform that can support functional testing of individual genetic variants necessary for precision medicine efforts and targeted drug screening/validation. Further, since all tumors including PDXs continue to evolve during serial propagation in vivo, synthetic tumors represent perhaps the only means by which we can explore early events in cellular transformation and segregate their biology from confounding effects of multiple and varied secondary events that accumulate in highly "evolved" samples. Disclosures Steidl: Seattle Genetics: Consultancy; Tioma: Research Funding; Bristol-Myers Squibb: Research Funding; Roche: Consultancy; Juno Therapeutics: Consultancy; Nanostring: Patents & Royalties: patent holding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5272-5272
Author(s):  
Gisele O. L. Rodrigues ◽  
Wenqing Li ◽  
Sarah Cramer ◽  
Livia W. Campos ◽  
Priscila P. Zenatti ◽  
...  

Abstract The IL7/IL7R mediated signaling is essential for normal development and homeostasis of T cell precursors. Early studies have shown that around 10% of patients with Acute lymphoblastic leukemia T cell (T-ALL) have mutations in the alpha chain of the receptor for IL7 (IL-7Ralpha) driving constitutive signaling via JAK1 and independent of IL-7, gamma-chain or JAK3. Some genetic changes are important factors to initiate leukemia, but in many cases these changes are insufficient to achieve the complete leukemic phenotype, suggesting that collaborative oncogenic mutations may be present. To identify candidate mutations that work in collaboration with the oncogenic IL7R, we performed exome sequencing and SNP-CNV-Array assay on a group of eight primary T-ALL samples carrying the IL7R mutation (T-ALL-IL7Rmut). The microarray was performed using Cytoscan HD - Affymetrix and CNVs were detected by ChAs software, version 2.0.1.2. For exome sequencing we used Illumina Hiseq2000 platform and Agilent SureSelect V4 51M Capture kit (mean sequencing depths of 80X / 50X for leukemia and remission samples, respectively). Somatic Single Nucleotide Variants (SNVs) and Small Insertion/Deletion (InDels) were detected using VarScan, and mutations were functionally annotated using ANNOVAR. All somatic mutations detected were manually curated. We found 17 genes recurrently mutated (in ≥ 2 cases) and chose five of them for further analyses due to their previous involvement in ALL (PHF6, RB1, CTCF, SGK223 and DNM2). Ongoing experiments are being conducted to determine whether these recurrent mutations can collaborate functionally with mutIL7R by co-transfection into immature murine thymocytes, transplanting into mice and determining incidence of leukemia. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1136-1136
Author(s):  
André De Almeida ◽  
Tim Pieters ◽  
Sara T'Sas ◽  
Steven Goossens ◽  
Pieter Van Vlierberghe

Abstract Background: T-lineage acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy that accounts for 10%-15% of pediatric and 25% of adult ALL cases. Although the prognosis of T-ALL has been improving over time, the outcome of T- ALL patients with primary resistant or relapsed leukemia remains poor. Thus, further advances in the treatment of T-ALL require the identification of new targets for the development of highly specific molecularly targeted drugs. SRY-related HMG-box 11 (SOX11) is a member of the SOXC family, which also includes SOX4 and SOX12. SOX11 is single-exon gene that encodes a transcription factor, but its molecular properties and functions remain incompletely understood. Expression of SOX11 is strongly associated with neurogenic activity, and it is confined to the central nervous system. Aberrant SOX11 upregulation has been observed in several cancer types, such as medulloblastoma, glioma, and B- and T-cell lymphomas. Under physiological conditions, SOX11 is absent in the T-cell lineage. However, SOX11 expression is consistently found in a subset of TAL/LMO T-ALL cases characterized by an abundancy of PTEN mutations and an underrepresentation of NOTCH mutations. Since the expression of SOX11 is restricted to a malignant context, we postulate that SOX11 might act as an oncogene in T-ALL. Aims: We want to assess SOX11's potential to promote a (pre)leukemic state, and to identify the role of SOX11 during oncogenic transformation in vivo. Methods: We generated a R26-SOX11 knock-in mouse model that allows conditional SOX11 expression in thymocytes, by using Lck-Cre (SL). Using this model, we analyzed the (immuno)phenotype and transcriptome of T-cell populations in 8- to 12-weeks old mice. In the future, we will also perform a syngeneic transplantation of SL thymocytes into sublethally irradiated mice to assess their colonizing potential. Additionally, we have set up an aging cohort of SL mice to investigate whether expression of SOX11 suffices to induce malignant transformation. Since the expression of SOX11 is associated with the TAL/LMO subgroup, we also crossed SL mice with a well-established murine T-ALL model, CD2-Lmo2, to give rise to mice that express both transgenes in their thymocytes (SCL). An aging cohort of this T-ALL model was followed, and we generated a survival curve and collected biological samples. These tumor samples are being analyzed, and their transcriptomic profile will be compared with that of mice overexpressing Lmo2 only. Lastly, we will lentivirally transduce human T-ALL cell lines from the TAL/LMO subgroup using a vector for SOX11 expression. These cell-lines will be used to study the consequences of expressing this transcription factor in a pre-established malignant setting. Results: In order to determine the effects of SOX11 expression in T-cells, we collected and analyzed blood, spleen, and thymus of 12-weeks old SL mice and their Cre-negative littermates (WT). To validate this model, we performed real-time PCR and immunoblotting to confirm the overexpression of SOX11 in T-cells. Interestingly, we observed a significant reduction in the number of lymphocytes in the peripheral blood and in the spleen of SL mice. Furthermore, significant changes were found in the proportions of all thymic populations, featuring an increase in double negative (DN) T-cells (Fig. 1A), particularly the DN3 subpopulation (Fig. 1B), and a decreased double positive CD4/CD8 fraction (DP). Based on that finding, we decided to sort out DN3 and DP cells from WT and SL thymi, and performed RNA-sequencing to further characterize the effect of SOX11 in these cells. In addition, we are following a cohort of SL mice, but so far no tumor formation was observed (average age of the cohort=300-days old). On the other hand, simultaneous overexpression of SOX11 and Lmo2 greatly accelerated the development of T-cell malignancies in SCL mice when compared with CD2-Lmo2 mice (Fig. 1C; median survival of 177 days versus 307; p&lt;0.0001), revealing a synergistic effect upon combination of both genetic events. Not surprisingly, most tumors were composed of immature T-cells, mainly DN cells, and no noteworthy differences were found between the composition and location of SCL and CD2-Lmo2 tumors. Conclusion: We showed that SOX11 skews the normal development of thymocytes in vivo. Additionally, we developed a murine SOX11 T-ALL model, thus confirming an oncogenic role for SOX11. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5083-5083 ◽  
Author(s):  
Vincenzo Giambra ◽  
Sam Gusscott ◽  
Deanne Gracias ◽  
Raymond Song ◽  
Andrew P. Weng

Abstract T-cell acute lymphoblastic leukemia (T-ALL) is a malignancy characterized by an uncontrolled proliferation of immature T-cells. While current therapies cure ~80% of pediatric patients, adults fare more poorly with ~40% overall survival. Refractory cases and relapses are presumably due to the ineffective targeting of leukemia stem cells (LSC), previously described in human and in mouse models of T-ALL, and thought to be resistant to standard treatments. Recently, we have reported that active signaling through the Wnt/β-catenin pathway is a defining feature of LSC in T-ALL, and that interruption of this signaling pathway abrogates disease propagation in vivo. Using an integrated, real-time reporter of Wnt/β-catenin signaling (7TGC; composed of 7 Tcf/Lef-binding sites upstream of a minimal promoter and GFP marker), we identified leukemia-initiating cell (LIC) activity to reside asymmetrically within the minor proportion of Wnt-active, GFP+ cells in primary mouse NOTCH1-induced T-cell leukemias. Moreover, inducible deletion of β-catenin in this context eliminated LIC activity. Here, we report that LIC activity in this Wnt-active subpopulation is dependent on Lef1. Using Lef1loxP/loxP animals, we show that inducible Cre-mediated deletion of Lef1 in established leukemias extinguishes both Wnt/GFP reporter expression and LIC activity. To explore mechanisms underlying asymmetry of LIC activity within the tumor population, we have also investigated the differential expression of various Lef1 protein isoforms in Wnt-active (GFP+) vs. Wnt-inactive (GFP-) leukemic subsets and assessed their function in supporting LIC activity. These results suggest that β-catenin acts via Lef1 to support asymmetric LIC activity within the Wnt-active subset of leukemia cells. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (16) ◽  
pp. 1837-1850 ◽  
Author(s):  
Aram Lyu ◽  
Todd A. Triplett ◽  
Seo Hee Nam ◽  
Zicheng Hu ◽  
Dhivya Arasappan ◽  
...  

Abstract Despite harboring mutations in oncogenes and tumor suppressors that promote cancer growth, T-cell acute lymphoblastic leukemia (T-ALL) cells require exogenous cells or signals to survive in culture. We previously reported that myeloid cells, particularly dendritic cells, from the thymic tumor microenvironment support the survival and proliferation of primary mouse T-ALL cells in vitro. Thus, we hypothesized that tumor-associated myeloid cells would support T-ALL in vivo. Consistent with this possibility, in vivo depletion of myeloid cells results in a significant reduction in leukemia burden in multiple organs in 2 distinct mouse models of T-ALL and prolongs survival. The impact of the myeloid compartment on T-ALL growth is not dependent on suppression of antitumor T-cell responses. Instead, myeloid cells provide signals that directly support T-ALL cells. Transcriptional profiling, functional assays, and acute in vivo myeloid-depletion experiments identify activation of IGF1R as a critical component of myeloid-mediated T-ALL growth and survival. We identify several myeloid subsets that have the capacity to directly support survival of T-ALL cells. Consistent with mouse models, myeloid cells derived from human peripheral blood monocytes activate IGF1R and directly support survival of primary patient T-ALL cells in vitro. Furthermore, enriched macrophage gene signatures in published clinical samples correlate with inferior outcomes for pediatric T-ALL patients. Collectively, these data reveal that tumor-associated myeloid cells provide signals critical for T-ALL growth in multiple organs in vivo and implicate tumor-associated myeloid cells and associated signals as potential therapeutic targets.


Sign in / Sign up

Export Citation Format

Share Document