Inhibitors Of Farnesyltransferase and Everolimus Act Synergistically In Growth Inhibition Of Human T-NHL Cells By Involvement Of AMPK

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3076-3076 ◽  
Author(s):  
Wolfgang Melchinger ◽  
Leonidas Zierock ◽  
Bettina Wehrle ◽  
Reinhard Marks

Abstract Introduction The outcome of patients with T cell lymphoma treated with standard chemoterapeutic substances remain poor, making the search for new active substances a highly medical need in this hematologic neoplasia. Recent phase II clinical trials showed very promising activity of farnesyltransferaseinhibitors (FTI) in relapsed/refractory T-NHL patients (Witzig et al. 2011). Regarding the molecular mechanisms behind this therapeutic effect, conflicting data regarding Ras as the initially proposed intracellular target of FTI and the involvement of MAP kinases in cellular effects of FTI in T cells exist (Marks et al. 2007, Ding et al. 2011). Together with observations in breast and ovarian cancer cells suggesting the GTPase Rheb as target for inhibition of farnesylation (Basso et al. 2005), the targets of FTI might vary according to the examined cell type. Interestingly, in breast cancer cells FTI mediatied inhibition of Rheb action resulted in reduced mTOR signaling. Nevertheless, as a putative additional targeted treatment approach in T-NHL, incubation with mTOR inhibitors showed not only substantial antiproliferative effects in normal T cells but also in malignant human T cell lymphoma lines in vitro (Huang et al. 2010). Since further clinical trials with both substances did not show severe side effects, adding everolimus as combination partner might even enhance clinical activity of FTI in T cell lymphomas. Therefore, in order to test this hypothesis and to analyse if both substances differ in their molecular mechanisms of action, FTI and everolimus were tested in vitro in T cell lymphoma lines (Karpas, Derl-2, Jurkat) to evaluate potential synergistic modes of action. Methods and Results Incubation of human T cell lymphoma lines Karpas and Derl-2 with the FTI SCH66336 (lonafarnib) or the mTOR inhibitor everolimus showed a reduction in proliferation in a dose dependent manner (EC50 for everolimus: 0.1nM, EC50 for lonafarnib: 0.5 µM). Combining both drugs resulted in synergistic inhibition of proliferation. This inhibitory effect correlated with increased p27KIP1 expression. In our experiments, Rheb appeared to be highly expressed in all examined T cell lymphoma lines with even additional increase of protein expression in Karpas cells after FTI incubation. Comparing FTI action to inhibition of mTOR by everolimus on a molecular level, in our experiments lonafarnib treatment of Karpas cells resulted in an unexpected reduction in AMPK-phosphorylation, implicating involvement of this metabolic pathway in FTI mediated inhibition of proliferation in malignant T cells. This effect could not be observed in everolimus treated Karpas cells. In contrast, naive human CD4+ T cells showed very little Rheb protein expression, which could be significantly increased after TCR stimulation by induction of Rheb mRNA transcription. While everolimus treatment of TCR-activated normal human CD4+ T cells resulted in AKT-hyperphosphorylation, FTI did not induce any changes in AKT. Contrary to the malignant T cells, FTI treatment had no impact of AMPK phosphorylation in activated T cells. Actually, naive T cells treated with FTI showed an hyperphosphorylated AMPK status. Conclusion Lonafarnib and everolimus show synergistic antiproliferative effects in T cell lymphoma lines, most likely by interfering with mTOR and AMPK signalling, making this combination therapy interesting for clinical trials. In contrast, FTI does not mediate AMPK in activated normal T cells. This observations are in accordance with a differential targeting of Rheb by FTI in malignant or normal human T cells. Disclosures: No relevant conflicts of interest to declare.

2020 ◽  
Vol 3 (1) ◽  
Author(s):  
Babu Gajendran ◽  
Krishnapriya M. Varier ◽  
Wuling Liu ◽  
Chunlin Wang ◽  
Klarke M. Sample ◽  
...  

AbstractThe SIN3 repressor complex and the NAD-dependent deacetylase SIRT3 control cell growth, and development as well as malignant transformation. Even then, a little known about cross-talks between these two chromatin modifiers or whether their interaction explored therapeutically. Here we describe the identification of a C21-steroidal derivative compound, 3-O-chloroacetyl-gagamine, A671, which potently suppresses the growth of mouse and human T-cell lymphoma and erythroleukemia in vitro and preclinical models. A671 exerts its anti-neoplastic effects by direct interaction with Histone deacetylase complex subunit SAP18, a component of the SIN3 suppressor complex. This interaction stabilizes and activates SAP18, leading to transcriptional suppression of SIRT3, consequently to inhibition of proliferation and cell death. The resistance of cancer cells to A671 correlated with diminished SAP18 activation and sustained SIRT3 expression. These results uncover the SAP18-SIN3-SIRT3 axis that can be pharmacologically targeted by a C21-steroidal agent to suppress T-cell lymphoma and other malignancies.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2767-2767
Author(s):  
Waseem Lone ◽  
Alyssa Bouska ◽  
Tyler Herek ◽  
Catalina Amador ◽  
Mallick Saumyaranajn ◽  
...  

Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of non-Hodgkin lymphomas and approximately 30% of PTCLs are designated as not-otherwise specified (PTCL-NOS). Gene expression profiling (GEP) identified molecular classifiers for PTCL entities and identified 2 novel biological subgroups within PTCL-NOS (PTCL-GATA3 and PTCL-TBX21), associated with T-cell differentiation subsets. To further investigate molecular oncogenesis, we performed microRNA expression profiling (miR-EP) in several molecular subtypes of PTCL including angioimmunoblastic T-cell lymphoma (AITL), PTCL-GATA3 and PTCL-TBX21 using formalin fixed paraffin embedded tissues. We also performed miR-EP of normal T-cell subsets polarized to represent different differentiation stages (TFH, TH1 and TH2). We performed miR-EP on 102 PTCL cases using either quantitative real time PCR (ABI, Biosystem) or ultra-sensitive direct miRNA counting (nCounter, NanoString). Corresponding GEP (mRNA) were available for 67 PTCL cases. Normal T-cells were polarized in-vitro with different cytokine milieu and examined by flow cytometry. We observed distinct miRNA profiles, with miRNA being uniquely expressed in TFH polarized cells (miR-26a-5p, miR-17-5p, miR-30d-5p, miR-22-3p, miR-222-3p, miR-142-3p, let-7i-5p and miR-29b-3p). In contrast, the TH1 lineage was enriched for expression of miR-155-5p, miR-146a-5p, miR-1246, miR-93-5p, miR-16-5p, miR-21-5p, miR-363-3p, miR-1260a, miR-186-5p, miR-148a-3p and miR-579-3p, whereas TH2 polarized cells expressed miR-181a-5p, let-7a-5p, miR-191-5p, miR-15b-5p, let-7d-5p, let-7b-5p, miR-140-5p, miR-98-5p, miR-423-5p and miR-630. Several of these miRNA expressed in the T-cells subsets showed corresponding expression in their respective PTCL entity such as miR-142-3p, let7i-5p, miR-21-5p and miR-29b-3p with AITL, miR-146-5p, miR-155-5p and miR-16-5p in PTCL-TBX21 and miR-181a-5p, miR-630 and let7a-5p in PTCL-GATA3. We also performed the MiRNA Enrichment Analysis and Annotation (miEAA) for miRNA signatures and observed an enrichment of miRNA regulating epigenetic modifications in TFH cells (p=0.028), whereas TH1 showed an enrichment of miRNA regulating IFN-g signaling (p=0.0024), and miRNA signatures in TH2 showed negative regulation of TGF-b signaling (p=0.023). Supervised analysis (p=0.05) of the miRNA profiles identified significant association of miR-126, miR-145, and let-7c-5p with AITL, when compared to other PTCLs. Similarly, miR-92a, miR-25, miR-636, miR-210, miR-222 and miR-491-5p significantly associated with PTCL-GATA3 and miRNA 126-3p, 145-5p, miR-26a-5p and miR-34a-5p associated with PTCL-TBX21. The miEAA for tumor miRNA signatures revealed enrichment of miRNAs regulating histone methylation (h3 k4 methylation) and chemokine receptor signaling in AITL, whereas miRNA regulating T-cell receptor were enriched in PTCL-TBX21 and TP53 signaling pathway in PTCL-GATA3. We validated the expression of miR-126 in AITL by qRT-PCR and also observed its increased expression in IL21 stimulated CD4+ T-cells. Ectopic expression of miR-126 resulted in a ~3 fold increased expression in T-cell lines and led to reduced proliferation and increased apoptosis with expression of T-cell exhaustion makers PD1 and TIM3. Computational algorithmic programs identified relevant biological targets of miR-126, including p85/PIK3R2, S1PR2 and DNMT3A that were further validated in-vitro. We observed an inverse correlation of miR-126 expression with S1PR2 expression (r=-0.64). S1PR2 is a crucial G protein-coupled receptor regulating B and T-cell migration in the germinal center (GC) reaction. Migration assays demonstrated significant decreases in T to B-cell migration, when B-cells (Raji) were co-cultured with Jurkat cells with ectopic expression of miR-126. With the GC reaction holding an important role in AITL, we investigated the biological significance of miRNA-126 in the context of the AITL microenvironment. High expression of miRNA-126 significantly associated with inferior survival in AITL (p=0.008) and significant differences in tumor microenvironment signatures. We identified distinct miRNA signatures for AITL and molecular subgroups of PTCL-NOS. Furthermore, elevated expression of miR-126 may contribute to the dysregulation and the homing of TFH cells in GC reaction through S1PR2 and warrants further mechanistic investigation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4870-4870
Author(s):  
Leonidas Zierock ◽  
Wolfgang Melchinger ◽  
Bettina Wehrle ◽  
Juergen Finke ◽  
Reinhard Marks

Abstract Abstract 4870 Since the succesful treatment of T cell lymphoma remains to be problematic, identification of new pharmacological targets in this malignancies are desperately needed. The AMPK-Rheb-mTOR signaling pathway plays an important role in regulating processes such as proliferation and proteinsynthesis according to energy and nutrient levels in normal and malignant T cells. Inhibitors of mTOR have shown promising results in clinical trials in several lymphoma types. Similarly, recent data could prove inhibitors of farnesyltransferase (FTI) to be effective as a single agent in certain subtypes of T cell lymphoma. Despite divergent data regarding the molecular target of FTI action, recently published work suggest inhibition of prenylation of the GTPase Rheb as putative mechanism for the antineoplastic effects of FTI (Basso et al., J Biol Chem, 2005). Therefore, combining inhibition of mTOR and Rheb might result in increased inhibition of T cell lymphoma proliferation. To investigate this hypothesis, human T cell lymphoma cell lines DERL-2 (originated from hepatosplenic gamma-delta T cell lymphoma), Karpas-299 (originated from anaplastic large cell T cell lymphoma) and normal human CD4+ T cells were incubated with a combination of everolimus as mTOR inhibitor and FTI (lonafarnib, SCH-66336) or the single agents. While both substances showed an additive combined anti-proliferative effect in DERL-2 cells, proliferation of Karpas cells were more susceptible to inhibition by FTI. On a molecular level, despite substantial growth inhibition in both cell lines by everolimus alone, phosphorylation of 4EBP1 and p70S6K remained unaffected, while FTI mediated reduction of Karpas cell proliferation was associated with a substantial decrease in AMPK phosphorylation together with an overexpression of p27kip, which could not be observed in DERL-2 cells. In contrast, incubation of stimulated human CD4+ T cells with the drugs alone or in combination did not result in changes in the phosporylation status of AMPK. Nevertheless, in contrast to everolimus, FTI induced a reduction of total protein expression of AMPK and other proteins, e.g. AKT. In addition, contrary to the observations in the malignant T cells, FTI treatment of unstimulated human CD4+ T cells resulted even in an increase of AMPK-phosphorylation. A hint for the explanation of these conflicting data came from analyses of Rheb expression in the examined cell types. While Rheb was easily detectable in the malignant T cell lines and the stimulated CD4+ T cells, it was almost absent in unstimulated CD4+ T cells. A model derived from this findings is that FTI effects depend on different targets available for inhibition of prenylation according to the activation or differentiation status of the T cells. While Rheb might be the target in malignant or activated T cells, another target, e.g. phosphatases, might be responsible for the FTI effect in resting T cells where Rheb is not available. In Karpas cells a particular connection between Rheb and AMPK might exist, as described for other cell lines (Lacher et al., Oncogene, 2010). Inhibition of this Rheb-AMPK axis might explain the particular gowth inhibiting effect of FTI in this model of anaplastic large T cell lymphoma. Nevertherless, the presented data show a combined effect of mTOR inhibitors and FTI for the potent treatment of T cell lymphoma involving different molecular mechanisms according to the lymphoma subtype. Disclosures: Finke: Fresenius Biotech GmbH: Honoraria, Research Funding.


1981 ◽  
Vol 154 (6) ◽  
pp. 1957-1964 ◽  
Author(s):  
M Robert-Guroff ◽  
F W Ruscetti ◽  
L E Posner ◽  
B J Poiesz ◽  
R C Gallo

A monoclonal antibody specific for the internal p19 protein of a type-C retrovirus (HTLV) isolated from human neoplastic T cells has been developed. Its specificity has been shown by radioimmune precipitation and by affinity chromatography of iodinated HTLV proteins. By indirect immune fluorescence this antibody recognizes only HTLV-producing cells. Examination of cells from patients with cutaneous T cell lymphomas and leukemias and with other types of lymphomas and leukemias indicated that HTLV p19 expression is rare. The monoclonal antibody will be useful in determining the natural reservoir of HTLV, possibly in a subset of mature T cell neoplasias.


Blood ◽  
1984 ◽  
Vol 63 (2) ◽  
pp. 477-481 ◽  
Author(s):  
F Miedema ◽  
FG Terpstra ◽  
JW Smit ◽  
S Daenen ◽  
W Gerrits ◽  
...  

Abstract The neoplastic T cells from five patients with adult T cell lymphoma/leukemia (ATLL), born in the Caribbean, were studied with respect to immunoregulatory activity on pokeweed mitogen (PWM) driven immunoglobulin (Ig) synthesis as well as surface-marker phenotypes with monoclonal antibodies. The neoplastic T cells in all patients had an OKT1+4+8–11+M1-I1–3A1- phenotype, but differed in the reactivity with OKT3. None of the patients' cells exerted helper activity on PWM- induced Ig synthesis. The neoplastic cells of three patients had suppressor activity on PWM-induced Ig synthesis. All patients were positive for human T cell leukemia/lymphoma virus (HTLV) or had antibodies against HTLV antigens. It has previously been shown that the neoplastic cells in Japanese ATLL patients and in patients from the Caribbean are indistinguishable by morphology and marker phenotype. We now show them to be also similar with respect to their functional properties.


Cancers ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 280
Author(s):  
Chella Krishna Vadivel ◽  
Maria Gluud ◽  
Sara Torres-Rusillo ◽  
Lasse Boding ◽  
Andreas Willerslev-Olsen ◽  
...  

Perturbation in JAK-STAT signaling has been reported in the pathogenesis of cutaneous T cell lymphoma (CTCL). JAK3 is predominantly associated with the intra-cytoplasmic part of IL-2Rγc located in the plasma membrane of hematopoietic cells. Here we demonstrate that JAK3 is also ectopically expressed in the nucleus of malignant T cells. We detected nuclear JAK3 in various CTCL cell lines and primary malignant T cells from patients with Sézary syndrome, a leukemic variant of CTCL. Nuclear localization of JAK3 was independent of its kinase activity whereas STAT3 had a modest effect on nuclear JAK3 expression. Moreover, JAK3 nuclear localization was only weakly affected by blockage of nuclear export. An inhibitor of the nuclear export protein CRM1, Leptomycin B, induced an increased expression of SOCS3 in the nucleus, but only a weak increase in nuclear JAK3. Importantly, immunoprecipitation experiments indicated that JAK3 interacts with the nuclear protein POLR2A, the catalytic subunit of RNA Polymerase II. Kinase assays showed tyrosine phosphorylation of recombinant human Histone H3 by JAK3 in vitro—an effect which was blocked by the JAK inhibitor (Tofacitinib citrate). In conclusion, we provide the first evidence of nuclear localization of JAK3 in malignant T cells. Our findings suggest that JAK3 may have a cytokine-receptor independent function in the nucleus of malignant T cells, and thus a novel non-canonical role in CTCL.


Blood ◽  
1984 ◽  
Vol 63 (2) ◽  
pp. 477-481
Author(s):  
F Miedema ◽  
FG Terpstra ◽  
JW Smit ◽  
S Daenen ◽  
W Gerrits ◽  
...  

The neoplastic T cells from five patients with adult T cell lymphoma/leukemia (ATLL), born in the Caribbean, were studied with respect to immunoregulatory activity on pokeweed mitogen (PWM) driven immunoglobulin (Ig) synthesis as well as surface-marker phenotypes with monoclonal antibodies. The neoplastic T cells in all patients had an OKT1+4+8–11+M1-I1–3A1- phenotype, but differed in the reactivity with OKT3. None of the patients' cells exerted helper activity on PWM- induced Ig synthesis. The neoplastic cells of three patients had suppressor activity on PWM-induced Ig synthesis. All patients were positive for human T cell leukemia/lymphoma virus (HTLV) or had antibodies against HTLV antigens. It has previously been shown that the neoplastic cells in Japanese ATLL patients and in patients from the Caribbean are indistinguishable by morphology and marker phenotype. We now show them to be also similar with respect to their functional properties.


Sign in / Sign up

Export Citation Format

Share Document