Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 8, No. 1 ( 2018-02-07)
    Abstract: CD8 + T-cell expansions are the primary manifestation of T-cell large granular lymphocytic leukemia (T-LGLL), which is frequently accompanied by neutropenia and rheumatoid arthritis, and also occur as a secondary phenomenon in leukemia patients treated with dasatinib, notably in association with various drug-induced side-effects. However, the mechanisms that underlie the genesis and maintenance of expanded CD8 + T-cell receptor (TCR)-Vβ + populations in these patient groups have yet to be fully defined. In this study, we performed a comprehensive phenotypic and clonotypic assessment of expanded (TCR-Vβ + ) and residual (TCR-Vβ − ) CD8 + T-cell populations in T-LGLL and dasatinib-treated chronic myelogenous leukemia (CML) patients. The dominant CD8 + TCR-Vβ + expansions in T-LGLL patients were largely monoclonal and highly differentiated, whereas the dominant CD8 + TCR-Vβ + expansions in dasatinib-treated CML patients were oligoclonal or polyclonal, and displayed a broad range of memory phenotypes. These contrasting features suggest divergent roles for antigenic drive in the immunopathogenesis of primary versus dasatinib-associated CD8 + TCR-Vβ + expansions.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2615211-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 128, No. 20 ( 2016-11-17), p. 2465-2468
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3828-3828
    Abstract: T-PLL is a rare mature post-thymic T-cell neoplasm with an aggressive clinical course and median overall survival of less than one year. Almost 75% of T-PLL cases harbor chromosome 14 translocations involving the T-cell receptor A/D locus resulting in aberrant activation of the proto-oncogenes TCL1A or MTCP1. T-PLL patients are difficult to treat as the leukemic cells are often resistant to most available chemotherapeutic drugs. Due to the rareness and aggressive nature of the disease, large clinical trials are difficult to execute. We therefore aimed to discover novel potential therapeutic targets using a high-throughput ex vivo drug sensitivity and resistance testing (DSRT) platform covering 306 approved and investigational oncology drugs. Methods Primary T-PLL cells were available from two patients. The first patient had a double positive CD4+CD8+CD3+ Vβ.14+ T-cell phenotype (patient 1) and cells underwent DSRT twice during a 5-month time-period (no treatment during that time). The second patient had a CD4+CD3+ phenotype (patient 2) and the cells were assayed once by DSRT. Fresh blood mononuclear cells (MNCs) were separated by Ficoll centrifugation from the patient samples (over 85 % leukemic cells in the MNC fraction) and healthy controls. Cells were seeded in 384-well plates and 306 active substances were tested using a 10,000-fold concentration range resulting in a dose-response curve for each compound. Cell viability was measured after 72 h incubation and differential drug sensitivity scores (DSS), representing leukemia-specific responses, were calculated by comparing patient samples with those obtained from healthy donors. In addition, both exome and RNA sequencing was performed from T-PLL cells (patient 1). Results Both patient samples showed high sensitivity to small molecule BCL2-inhibitors navitoclax (EC50 values 44nM and 10nM) and ABT-199 (EC50 23nM and 20nM) (Fig. 1 and 2). HDAC-inhibitors (quisinostat, belinostat and panobinostat) also showed high sensitivity in both patients in low nM concentrations (EC50 values 1-80nM). As AKT1/mTOR pathway is activated in most T-PLL patients due to the TCL1 oncoprotein, it was interesting to observe that neither of the patient samples showed any response to an AKT1 inhibitor (MK-2206 EC50 values 〉 1000 nM) nor to mTOR inhibitors (temsirolimus and everolimus)(Fig. 1). Furthermore, T-PLL cells were resistant to corticosteroids such as prednisolone and methylprednisolone. To further elucidate the molecular mechanism behind the drug responses, exome and RNA sequencing was performed from T-PLL cells (patient 1). No deletion was found in the ATM gene, but instead a homozygous missense mutation K2413Q was detected. This particular mutation is in the region coding for the FAT domain and while it has not been described earlier in T-PLL, it is in a cancer mutation hotspot region of ATM, suggesting that it is inactivating. No mutations directly linked to the BCL2-family genes were observed. In the RNA sequencing analysis, TCL1A was overexpressed when compared to the healthy CD4+ cells as expected. Similarly, AKT1 was overexpressed. The expression of BCL-2 and BCL-XL did not differ from those observed in healthy CD4+ cells while pro-apoptotic BCL-2 family members BID and BAD were elevated compared to the healthy control. Conclusions Primary T-PLL cells showed sensitivity to BCL-2 and HDAC inhibitors in a systematic high-throughput ex vivo drug sensitivity testing across a range of clinical and investigational drugs. The BCL-2 inhibitor sensitivity was not related to increased BCL-2 expression or activating mutations in the BCL-2 family genes, and further studies are needed to clarify the mechanism of action. However, the results suggest that BCL-2 inhibitors could be a novel promising candidate drug for T-PLL-patients and warrant further clinical development in this group of patients. In contrast, inhibitors of AKT and mTOR, kinases known to be activated by TCL1, showed no efficacy ex vivo in this assay. Disclosures: Porkka: BMS: Consultancy, Research Funding, Speakers Bureau; Novartis: Consultancy, Research Funding, Speakers Bureau. Mustjoki:Novartis: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: New England Journal of Medicine, Massachusetts Medical Society, Vol. 366, No. 20 ( 2012-05-17), p. 1905-1913
    Type of Medium: Online Resource
    ISSN: 0028-4793 , 1533-4406
    RVK:
    Language: English
    Publisher: Massachusetts Medical Society
    Publication Date: 2012
    detail.hit.zdb_id: 1468837-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Biomarker Research, Springer Science and Business Media LLC, Vol. 8, No. 1 ( 2020-12)
    Abstract: T-cell prolymphocytic leukemia (T-PLL) is a poor prognostic disease with very limited options of efficient therapies. Most patients are refractory to chemotherapies and despite high response rates after alemtuzumab, virtually all patients relapse. Therefore, there is an unmet medical need for novel therapies in T-PLL. As the chemokine receptor CCR7 is a molecule expressed in a wide range of malignancies and relevant in many tumor processes, the present study addressed the biologic role of this receptor in T-PLL. Furthermore, we elucidated the mechanisms of action mediated by an anti-CCR7 monoclonal antibody (mAb) and evaluated whether its anti-tumor activity would warrant development towards clinical applications in T-PLL. Our results demonstrate that CCR7 is a prognostic biomarker for overall survival in T-PLL patients and a functional receptor involved in the migration, invasion, and survival of leukemic cells. Targeting CCR7 with a mAb inhibited ligand-mediated signaling pathways and induced tumor cell killing in primary samples. In addition, directing antibodies against CCR7 was highly effective in T-cell leukemia xenograft models. Together, these findings make CCR7 an attractive molecule for novel mAb-based therapeutic applications in T-PLL, a disease where recent drug screen efforts and studies addressing new compounds have focused on chemotherapy or small molecules.
    Type of Medium: Online Resource
    ISSN: 2050-7771
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2699926-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4117-4117
    Abstract: Background: Large granular lymphocyte (LGL) leukemia is a rare disease characterized by a clonal persistence of cytotoxic T cells or natural killer (NK) cells. Patients usually suffer from cytopenias and other organ-related autoimmune phenomena. These are putatively mediated by the cytotoxic LGL cells constitutively activated following an antigen-driven immune response. In addition to gain-of-function mutations in the STAT3 gene, which occur in 40-50% of patients, recurrent alterations only in the STAT5b and TNFAIP3 tumor suppressor genes have been described thus far. However, based on gene expression analyses, JAK/STAT pathway activation and deregulation of several pro-apoptotic (sphingolipid and FAS/FAS ligand) and pro-survival signaling pathways (PI3K/AKT and RAS) are common features of LGL leukemia. In this project, we aimed to characterize the genomic landscape of LGL leukemia using exome sequencing and systems genetics approaches in a patient cohort including both T- and NK-LGL cases and patients without known STAT mutations. Methods: The study cohort included 19 patients diagnosed with LGL leukemia that underwent exome sequencing analysis with matched germline controls. 13 patients had CD8+ T LGL and 3 patients CD4+ T LGL phenotype and 3 patients were NK LGL cases. From the T LGL leukemia cases CD8+ or CD4+ T cells were sorted (according to the dominant phenotype) and used as the tumor sample. In NK LGL leukemias, sorted CD3neg,CD16/56+ NK cells constituted the tumor fraction that underwent exome sequencing. Polyclonal blood lymphocytes depleted from LGL cells were used as germline controls. The exome was captured with the Nimblegen SeqCap EZ Exome Library v2.0 and the sequencing was performed with the Illumina HiSeq2000 sequencing platform. All bioinformatics steps were carried out using a custom bioinformatics pipeline. Putative somatic variants were identified by subtracting, for each patient, the ones called in the normal samples from those found in the tumor sample. After filtering by call quality and allele frequency in ExAC database, somatic variants were prioritized according to the predicted impact from the SnpEff software. Genes hit by variants putatively altering their function were finally mapped to Kegg and Reactome to generate pathway-derived meta gene networks for the identification of affected functional components. Results: 4 patients had STAT3 mutations and 4 additional cases had STAT5B mutations. In addition to STAT mutations, a number of novel somatic variants, which were recurrently mutated were discovered. These included the tumor suppressor gene FAT4, the epigenetic regulator KMT2D, as well as genes involved in the control of cell proliferation (CDC27 and ARL13B). With the systems genetics approach based on integration of pathway-derived mutated gene network topologies for identification of connected components we were able to discover affected functional modules. The main network component included key genes, which either directly interact (such as the FLT3 tyrosine kinase) or are functionally connected (such as ADCY3, ANGPT2, CD40LG, PRKCD, PTK2, KRAS, and RAB12 genes) with STAT proteins. Additional modules with putative pathogenetic relevance in LGL leukemia and mutated in the absence of STAT mutations were cell cycle control (CDC27, PLK1, CDC25B, RAD21), Notch signaling (NOTCH2, NOTCH3 and MAML3) and epigenetic regulation through histone-lysine methyltransferase activity (KMT2D and ASH1L). The comparison of various LGL leukemia subtypes revealed that the mutation burden was especially high among the CD4+ T LGL leukemia cases. Part of the genes and modules affected were shared between the different subtypes of LGL leukemia, but for example KIR2DL1 mutations were only found in CD8+ and NK LGL leukemia cases. Conclusions: With the exome sequencing and systems genetic approach we were able to discover specific gene networks, which are recurrently mutated in LGL leukemia and particularly in patients without STAT mutations. As several mutated genes are directly or indirectly connected with the STAT pathway, the data strengthen the key role of JAK/STAT signaling in LGL leukemia. The novel identified pathway modules beyond STAT networks provide intriguing insights into the pathobiology of LGL leukemia. Disclosures Maciejewski: Apellis Pharmaceuticals Inc: Membership on an entity's Board of Directors or advisory committees; Alexion Pharmaceuticals Inc: Consultancy, Honoraria, Speakers Bureau; Celgene: Consultancy, Honoraria, Speakers Bureau. Mustjoki:Pfizer: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, Research Funding; Ariad: Research Funding; Novartis: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 871-871
    Abstract: Abstract 871 Background: Large granular lymphocytic (LGL) leukemia is a rare lymphoproliferative disease, characterized by the clonal expansion of cytotoxic CD3+CD8+ T-cells or CD3-CD16/56+ natural killer (NK)-cells. It is often associated with autoimmune phenomena (e.g. cytopenias, rheumatoid arthritis). We recently identified somatic mutations in the STAT3 gene in 40% of monoclonal T-LGL cases (Koskela et al, NEJM, 2012). Here, we report the discovery and functional analysis of novel STAT5b mutations as well as small subclones of STAT3 mutations in other LGL patients, expanding the evidence implicating STAT activation in LGL. METHODS: In order to find novel LGL-leukemia associated mutations, exome sequencing was done from untreated STAT3 mutation negative T-LGL leukemia patients using CD8+ LGL-leukemic cells and matched CD4+ control cells. Samples from 158 T-LGL and 40 NK-LGL leukemia patients were further analyzed using both targeted Sanger sequencing and ultra-deep targeted next-gen amplicon sequencing with up to 10,000x coverage (MiSeq, Illumina). Functional analysis of mutated proteins was carried out in Hela cells by Western analysis and luciferase reporter assays. RESULTS: Exome sequencing revealed a novel somatic missense mutation Y665F in the STAT5b gene in two T-LGL patients diagnosed with WHO2009 criteria with a large CD8+ T-LGL clone ( 〉 90%). Only wild-type STAT5b was seen in the matched CD4+ control cells of these patients. Amplicon sequencing of exon 16 of STAT5b (corresponding to the Y665F site) in 158 T-LGL and 40 NK-LGL patients revealed an additional mutation (N642H) in one T-LGL and one NK-LGL patient, resulting in the 2% total frequency (4/198) of STAT5b mutations across all patients. The N642H and Y665F mutations were both located in the Src homology 2 (SH2) domain of STAT5b, which mediates dimerization and activation by trans-phosphotyrosine binding. STAT3 mutations previously reported in T-LGL patients were located in the corresponding domain. The transcriptional activity of wild-type and mutant STAT5b proteins (N642H and Y665F) was assayed in cells carrying a luciferase reporter with STAT5 binding elements. Luciferase activity of Hela cells transfected with the mutated STAT5b constructs was significantly increased compared to wild-type STAT5b. Furthermore, both the N642H and Y665F variants of STAT5b exhibited higher levels of tyrosine (Y694) phosphorylation than the wild type protein. The exon 21 in the SH2 domain of the STAT3 gene was also screened by ultra-deep next-gen amplicon sequencing, both from the original T-LGL patient cohort (n=77, patients with monoclonal disease) and 142 additional monoclonal/oligoclonal LGL patients. In the monoclonal cohort, a total of nine new STAT3 mutation positive patients were detected by amplicon sequencing, raising the total number of positive cases to 41 (53%) from the 32 identified by Sanger sequencing. Concomitant to the previously described Y640F, N647I, K658N, D661H, D661V and D661Y STAT3 mutations, several novel mutations in this gene were found: I659L, Q643H, G656C, K658H, K658R and D661I. In the oligoclonal LGL cohort, the mutation frequency was lower (31/142, 22%), suggesting that it may also include patients with reactive polyclonal LGL proliferation. CONCLUSIONS: Our mutational and functional data affirm that STAT family transcription factors play a critical role in the pathogenesis of LGL leukemia. In addition to the previously identified mutations in the STAT3 gene, we found recurrent somatic STAT5b mutations in LGL leukemia. Furthermore, our ultra-deep (10,000x) next-gen sequencing revealed small subclones of STAT3 mutations in patients with oligoclonal LGL. Both STAT3 and STAT5b mutations increased the phosphorylation and transcriptional activity of corresponding proteins. The detection of STAT mutations should be included in the diagnostic assessment of LGL leukemia. Disclosures: Koskela: Novartis: Honoraria; BMS: Honoraria; Janssen-Cilag: Honoraria. Kallioniemi:TEKES-FiDiPro: Research Funding. Porkka:Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria, Research Funding. Maciejewski:NIH: Research Funding; Aplastic Anemia & MDS International Foundation: Research Funding. Mustjoki:Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 1771-1771
    Abstract: Introduction: Constitutive hyperactivation of the STAT3 and STAT5B transcription factors is often observed in cancer. Lately, activating STAT3 mutations have been identified in hematological malignancies including large granular lymphocytic (LGL) leukemia (prevalence 40%), aplastic anemia (7%) and CD30+ T-cell lymphoma (17%). Furthermore, recent studies highlight the importance of STAT5B mutations in the pathogenesis and prognosis of T-cell malignancies such as T-cell prolymphocytic leukemia (36%), T-cell acute lymphoblastic leukemia (8%) and hepatosplenic T-cell lymphoma (33%). While STAT3 and STAT5B mutations lead to constitutive STAT3/STAT5B signaling, several other known gene mutations and mechanisms may also cause JAK/STAT-pathway activation. These findings indicate that inhibiting the JAK/STAT pathway with targeted drugs could be used as a treatment option. Here, we aimed to identify drugs that inhibit STAT3 or STAT5B function and determine if mutant STAT3/5B and wild type STAT3/5B cells respond differently to the tested drugs. In addition, we wished to ascertain if STAT3 inhibition is sufficient to induce apoptosis in patient derived LGL cells with constitutively active STAT3. Methods: High-throughput drug sensitivity testing was performed with a compound collection containing over 300 approved and investigational oncology drugs including many kinase inhibitors (such as those targeting JAK, SRC, VEGFR, mTOR, MEK, and CHK) and small molecule STAT3 inhibitors (Stattic, LLL12, Sta-21). All drugs were tested in 5-8 different concentrations over a 10,000-fold concentration range. Mutant STAT3 (Y640F) and mutant STAT5B (N642H) transformed Ba/F3 cells as well as HEK293 cells containing a STAT5 (pGL4.52[luc2P/STAT5 RE/Hygro]) or STAT3 specific luciferase reporter gene element (HEK-SIE) were used in the screens. In addition, drug sensitivities of five LGL leukemia patient samples were also assessed. Primary patient cells and the Ba/F3 cells were incubated in 384-well plates for three days with the drugs after which cell viability was measured with CellTiter-Glo. STAT3/5B induced luciferase activity in the HEK cells was analyzed after the cells were incubated for 6 or 24 hours with the drugs using the ONE-Glo luciferase assay system. Results: A significant decrease in luciferase activity was detected in STAT3 mutant Y640F, STAT5B mutant N642H and wild type STAT3 transfected HEK-SIE cells in the presence of PI3K/mTOR inhibitors such as PF-04691502 and INK128. In addition, PI3K/mTOR inhibitors significantly decreased the viability of mutant STAT3 and STAT5B transformed Ba/F3 cells compared to wild type cells. Interestingly, JAK inhibitors (e.g. ruxolitinib, gandotinib) did not inhibit mutant STAT3 activity in the HEK-SIE cells, whereas IL6-induced wild type STAT3 activity was completely blocked. A BET family inhibitor (JQ1+) and glucocorticoids (e.g. dexamethasone, methylprednisolone) showed specific and strong cytotoxicity to mutant STAT3 and STAT5B transformed Ba/F3 cells. Although JQ1+ inhibited luciferase activity of STAT5B N642H cells, no effect on the luciferase activity of STAT3 Y640F transfected HEK cells was detected, suggesting that JQ1+ may have a direct effect on mutant STAT5B function while the effect on mutant STAT3 transformed cells may be indirect. Cells from LGL leukemia patients showed high sensitivity against glucocorticoids, the histone deacetylase inhibitor quisinostat, JQ1+ and PF-04691502 when compared to healthy CD8+ T-cells. However, no increase in apoptosis was observed with JAK or other mTOR inhibitors. Conclusions: Our results suggest that JAK inhibitors lack efficacy in STAT3 mutated diseases. However, our ex vivo and in vitro drug screens highlight some other promising agents including PF-04691502 (PI3K/mTOR inhibitor) and JQ1+ (BET family inhibitor) that inhibited mutant STAT5B and STAT3 activity in the cell line models and were effective against primary LGL patient cells. Additional experiments are ongoing to determine how these drugs function to block STAT signaling and induce cell death. As the STAT3 and STAT5 pathways are activated in many other cancer types as well, the results may be applicable to a variety of different malignancies. Figure 1 Figure 1. Table 1. Drug sensitivity scores of the different cell line models and patient samples. DSS value range 0-50. (0 = no drug response with any conc., 50 = maximal drug response with every conc.) Disclosures Mustjoki: Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 315-315
    Abstract: T-cell prolymphocytic leukemia (T-PLL) is a rare disease with an aggressive clinical course and a median overall survival of less than three years. Although almost 75% of T-PLL patients are reported to harbor translocations causing the activation of the proto-oncogene TCL1A, T-PLL is genetically heterogenous: most T-PLL patients also have mutations or deletions in the ATM gene and the genes involved in the JAK-STAT pathway are mutated in 76% of cases. There is an urgent need for more rational based therapies, but clinical trials are difficult to perform due to the rareness of the disease. Here, we systematically explored the diversity of drug responses in T-PLL patient samples ex vivo using a drug sensitivity and resistance testing (DSRT) system including 306 oncology drugs (approved or investigational). We also aimed to determine any associations between the genetic aberrations and drug sensitivities in T-PLL patients. Primary mononuclear cells were gathered from 30 T-PLL patients for drug testing. Cells were plated in 384-well plates and subjected to the 306 substances using a 10,000-fold concentration range. After 72 hours, cell viabilities were measured, the results were depicted as dose-response curves for each compound, and differential drug sensitivity scores (sDSS), representing leukemia-specific responses, were computed by comparing patient samples to healthy donors. Drug response profiles across patients were clustered and visualized by hierarchical clustering. The subgroups resulting from the clustering were statistically compared using a two-sample t-test to understand which drug classes were driving the grouping. To delineate heterogeneous pathway dependencies, drug sensitivities were correlated with somatic genetic variants and recurrent chromosomal aberrations. Genetic characterization was performed by targeted amplicon sequencing of tumor cells to profile known recurrent genetic variants (STAT5b, IL2RG, JAK1, JAK3, ATM). Information on chromosomal aberrations (TCL1A translocations, ATM deletions) was derived from parallel clinicopathologic databases. Amplicon sequencing revealed that 70% of T-PLL patients (21/30) harbored a mutation in genes involved in the JAK-STAT pathway (JAK1, JAK3, STAT5b or IL2RG). The most prevalent mutation led to an M511I amino acid exchange in the JAK3 protein (26% of patients). Interestingly, the STAT5b mutations (5/30) did not coexist with any of the JAK mutations in our cohort. Based on DSRT analysis, all T-PLL samples were sensitive to the CDK-inhibitor SNS-032 and the anti-cancer antibiotic actinomycin D. Next, we clustered patients using sDSS values for all 306 different substances, and this showed that patient samples could be divided into 3 main groups, based on their drug responses (Figure). According to two-sample t-test, the grouping was driven by the selective sensitivities of Groups II and III to HDAC inhibitors (belinostat, panobinostat, quisinostat, CUDC-101 and vorinostat) and the selective sensitivity of Group III to PI3K/AKT/mTOR inhibitors (AZD-8055, MK-2206, apitolisib, dactolisib, PF-04691502, ZSTK474, and omipalisib), HSP90 inhibitors (BIIB021, luminespib, alvespimycin, and tanespimycin) as well as JAK inhibitors (ruxolitinib, momelotinib, tofacitinib, gandotinib). Group I samples were on the other hand relatively resistant to these classes of drugs. Surprisingly, despite the prevalence of the signature event of activation of TCL1 (the established AKT coactivator) in nearly all cases, only a subset of cases (group III) responded to PI3K/AKT/mTOR inhibitors. Strikingly, the grouping of selective responses to HDAC, JAK, PI3K/mTOR/Akt and HSP90 inhibitors did not link to the presence of JAK/STAT mutations, TCL1A translocations, or ATM deletion status. Ex vivo drug screening of primary T-PLL samples revealed heterogenous selective drug responses in specific drug classes (such as HDAC-, JAK-, HSP90- and PI3K/Akt/mTOR-inhibitors). Surprisingly, the drug response patterns did not correlate with known recurrent genetic aberrations suggesting that sequencing for recurrent genetic biomarkers cannot easily be turned into effective therapeutic strategies in T-PLL, and that further elucidation of the biological pathways driving T-PLL is needed. Figure 1. Mutation status and clustering of HDAC-, PI3K/mTOR/Akt-, HSP90-, and JAK-inhibitor responses in PLL patients based on sDSS values Figure 1. Mutation status and clustering of HDAC-, PI3K/mTOR/Akt-, HSP90-, and JAK-inhibitor responses in PLL patients based on sDSS values Disclosures Koschmieder: Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel reimbursement for scientific conferences, Research Funding; Novartis Foundation: Research Funding; Baxalta/CTI: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Janssen Cilag: Other: Travel reimbursement for scientific conferences ; Pfizer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel reimbursement for scientific conferences. Wennerberg:Pfizer: Honoraria, Research Funding. Ding:Merek: Research Funding. Mustjoki:Pfizer: Honoraria, Research Funding; Bristol Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 121, No. 22 ( 2013-05-30), p. 4541-4550
    Abstract: Somatic mutations were discovered for the first time in the SH2 domain of the STAT5b gene in LGL leukemia. The mutations are activating and lead to increased phosphorylation and transcriptional activity of STAT5b.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages