GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • American Society of Hematology  (24)
  • 1
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3959-3959
    Abstract: Abstract 3959 Background: Entinostat (ENT) is an oral, class 1 isoform selective HDACi. In vitro in HL cell lines, ENT decreases proliferation and induces cell death in a dose dependent manner with an IC50 of 0.4 uM. As an epigenetic agent, ENT modulates immune pathways from a TH2 to a TH1 cytokine/chemokine profile and can upregulate the expression of cancer testis antigens, thus acting as an immunomodulator. This phase 2 study was initiated to define ENT single agent antitumor activity and safety and tolerability. Methods: The trial is a Simon 2-stage design that has completed enrollment in 2 dosing schedules with 24 evaluable patients at 6 sites, 6 patients remain active; enrollment is continuing in an alternate dosing schedule. ENT was administered at a dose of 10 mg every 14 days (d), in a 28 d cycle (C) for the first stage of the study, for the second stage the dose was escalated to 15 mg every 14 d beginning C1d15. Subjects ongoing from the first stage were also allowed to escalate. CT/PET scans are conducted every 2 cycles. Blood samples for correlative study analysis are obtained pre-treatment, C1D8, C1D15, and C3D15. Results: As of August 6, 2010, demographic data are available on 23 evaluable subjects, median age is 28 years (19-53), median prior regimens is 3 (2-10), 13 (56%) had prior autologous transplant, 3 (13%) had prior allogeneic and 3 (13%) had prior autologous and allogeneic transplant. 23 patients have 〉 1 post baseline result available, 65% have disease control (CR+PR+SD), 35% had PD and 2 patients with SD discontinued due to AE (Pericarditis and thrombocytopenia). Two (9%) responses (PR) are reported with time on study 14.7 months and one 〉 6 months. An additional 4 (17%) patients have tumor reduction (25-49%) for at least 4 cycles. Six (26%) patients completed ≥ 6 cycles. Common G1/2 AEs including all causalities were gastrointestinal, fatigue, and pyrexia. Of the 32 patients evaluated for safety, G 3/4 thrombocytopenia occurred in 19 (59.4%) patients, G 3/4 neutropenia in 9 (28.1%) patients, and G 3/4 anemia in 11 (34.4%) patients, including all causalities. Profiling of cytokine, chemokine and growth factor levels in patient serum samples provides support for the biological activity of entinostat as an immunomodulatory agent with ongoing evaluation demonstrating a reduction of IL-13 and TNFα levels within one week of therapy. Conclusions: ENT as a single agent in HL was well tolerated and demonstrated encouraging activity in this heavily pretreated patient population. Further dose intensity is currently being tested; updated results on the immunomodulatory effects will be presented. Disclosures: Off Label Use: Given the CD20 positivity of nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) rituximab has been evaluated previously for relapsed NLPHL and was shown to be efficacious. Rituximab however is not FDA approved for NLPHL. This is a retrospective study that evaluates the use of R-CHOP and other therapies for NLPHL. Current NCCN guidelines support consideration of R-CHOP for NLPHL treatment, and given the rarity of the disease there is no one defined preferred chemotherapy regimen. This information will be disclosed to the audience.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3718-3718
    Abstract: Abstract 3718 Based on preclinical experiments that demonstrated in vitro synergism between pan-deacetylase (DAC) inhibitor Panobinostat (LBH589) with mTOR inhibitor Everolimus (RAD001) in Hodgkin and non-Hodgkin lymphoma cell lines, we conducted a phase I/II study to determine the safety and efficacy of this novel regimen. Patients were eligible if they had relapsed or refractory Hodgkin or non-Hodgkin lymphoma regardless of the number of prior regimens, including autologous and allogeneic transplantation. Everolimus was self administered orally daily and Panobinostat three times weekly on 4 dose escalation levels [Table 1]. To date, a total of 30 patients have been treated. The histologic subtypes include small lymphocytic (n=1), follicular (n=2), mantle cell (n=3), hodgkin (n=12), diffuse large B-cell (n=7), T-cell (n=3), one discordant Hodgkin/marginal zone lymphoma and one discordant Hodgkin/large cell lymphoma. The median number of prior therapies is 3 with 11 patients receiving prior autologous transplantation. 30 patients received at least 1 dose and are evaluable for safety and 28 are evaluable for response. The DLT was thrombocytopenia observed in the 4th cohort with a dose of 30mg Panobinostat and 10mg of Everolimus. Therefore, 6 patients were treated at the lower dose level of 20mg Panobinostat and 10mg Everolimus which was determined to be the maximum tolerated dose (MTD) and starting dose in the phase II portion of the study. To date, 16 patients have been treated in the phase II portion of the study. Treatment side effects are manageable with the following grade 3/4 adverse events most commonly observed: thrombocytopenia 48%, neutropenia 48% and anemia 20%. One death occurred on study, possibly related to pulmonary embolus. 20 out of 28 evaluable patients (71%) had tumor reduction ranging from -12% to -72% of whom 50% had PR or CR. Our data demonstrate safety and promising clinical activity of this novel combination in a variety of lymphomas. The phase II portion continues to enroll patients to determine the efficacy of this novel regimen. Dose Level Everolimus Panobinostat N 1 5mg 10mg 3 2 5mg 20mg 3 3 (MTD) 10mg 20mg 6 4 10mg 30mg 2 Phase II 10mg 20mg 16 Disclosures: Neelapu: celgene: Research Funding. Pro:Celgene: Consultancy, Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3735-3735
    Abstract: Abstract 3735 Poster Board III-671 Recent clinical trials demonstrated a promising clinical activity of the HDAC inhibitor MGCD0103 in heavily pretreated patients with relapsed HL. However, the mechanisms underlying MGCD0103 activity in HL remains unclear. Here we show that MGCD0103 preferentially inhibited HDACs1, 2 with no effect on HDAC6. Using three HL-derived cell lines, the IC50 for MGCD0103 after 72 hour of incubation was in the submicromolar concentrations, and was more effective than vorinostat in inducing cell death. Using gene expression profiling (GEP) studies, pathway PCR array, and western blot analysis, we identified several pathways that are modulated by MGCD0103. MGCD0103 downregulated the X-linked inhibitor of apoptosis (XIAP) protein, activated the intrinsic caspase pathway, and synergized with TRAIL agonistic antibodies. MGCD0103 downregulated CD30 mRNA and surface protein expression in a dose dependent manner, but had no significant effect on B cell-associated proteins, including CD19 and CD20, or on the costimulatory receptors CD40 and CD80. MGCD0103 induced TNF-alfa expression and secretion, which was associated with NF-kB activation, upregulation of the silencer of cytokine signaling (SOCS)-3, downregulated STAT6, and upregulation of STAT1 and 2. Selective inhibition of TNF-alfa expression by short interfering mRNA enhanced MGCD0103-induced cell death. Similarly, inhibition of NF-kB by proteasome inhibitors also potentiated the effect of MGCD0103, thus demonstrating synergy independent of HDAC6 inhibition. These findings demonstrate that MGCD0103 has a potent anti lymphoma activity by modulating the expression of a variety of survival proteins, and provides mechanistic rationale for combining class-I HDAC inhibitors with proteasome inhibitors and TRAIL. Disclosures: Mamidipudi: Celgene: Employment. Heise:Celgene Corporation: Employment, Equity Ownership. Besterman:Methylgene: Employment. Martell:Methylgene: Employment. MacBeth:Celgene Corporation: Employment, Equity Ownership. Younes:MethylGene: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 12-12
    Abstract: Background: The serine/threonine kinase AKT, a major downstream PI3K target, promotes continued cell growth and metabolism by increasing glucose uptake, stimulating glycolysis and ATP production. It has been proposed that cancer cells with a high level of constitutively active AKT depend on glucose for survival. Recent data in transgenic models demonstrated that AKT activation results in mTOR dependent transcriptional upregulation of the glycolytic enzyne HK2 and glucose transporter Glut1 via induction of HIF1-α (Nature Med2004;10:594). The purpose of this study was to determine the frequency and prognostic significance of increased expression of HK2 in patients with ALL, to determine if proglycolytic conditions, high glucose (HG) or hypoxia, promote chemoresistance in ALL blast cells, and if chemoresistance is affected by mTOR inhibition. Methods: HK2 mRNA expression was measured by quantitative Taqman PCR in 28 primary ALL samples (14 newly diagnosed and 14 relapsed). Primary bone marrow samples (N=5) were co-cultured on either MS-5 or HS-5 stroma supplemented with 4 or 14 mM glucose for 24–48 hours. Jurkat cells were cultured in serum-free medium supplemented with 4 or 14 mM glucose under hypoxic (3% O2) or normoxic (21% O2) conditions. All samples were treated with doxorubicin (DOX - 25–50 ng/mL) +/− RAD001 (10–20 nM) or CCI779 (2.5–5 mg/mL). Results: Increased expression of HK2 (≥ 2-fold relative to normal samples) occurred in 5/28 samples (18%). Patients with HK2 levels above the median (0.43) had a worse failure free survival (FFS) (HR 2.95; CI 0.89–9.81; P=0.08) and a significantly worse overall survival (OS) (HR 5.01; CI 1.21–20.7; P=0.03). In 4/5 (80%) of primary ALL samples, exposure to HG (14 mM) resulted in decreased apoptosis with DOX compared to normal glucose (4 mM) (mean % Ann+ 32.0 vs 42.0; P=0.047). This effect was reversed with addition of RAD001 or CCI779 (mean % Ann+ 49.36 vs 54.02; P=0.11). Jurkat cells cultured under hypoxic conditions and treated with DOX demonstrated decreased apoptosis and increased cell viability compared to normoxic conditions [mean % Ann (+) cells 39.85 (Hypoxic) vs 55.00 (Normoxic); P=0.016). This effect was reversed with addition of RAD001 under hypoxic conditions [mean % Ann (+) cells 39.85 (DOX) vs 51.92 (DOX+RAD); P=0.009] in association with decreased expression of HK2 mRNA (RE to DMSO mean 62 vs 47; P=0.26). Conclusions: Overexpression of HK2 is associated with worse OS in pts with newly diagnosed or relapsed ALL. Exposure of ALL blast cells to high glucose or hypoxic environment results in chemoresistance to DOX. This effect is abrogated by mTOR inhibition in association with decreased expression of HK2 mRNA, suggesting that upregulation of glycolysis via the AKT/mTOR/HIF1a pathway may be an important mechanism of chemoresistance in ALL and that mTOR inhibition or glucose normalization may play an important role in chemosensitization and improved outcomes in ALL.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 1833-1833
    Abstract: Cancer cells exhibit a high rate of glycolysis, which leads to a stringent dependence on extracellular glucose for maintaining intracellular ATP levels. Leukemic cells infiltrating bone marrow have been shown to be markedly hypoxic compared to normal bone marrow cells, and stimulation of glycolysis is thought to be an adaptive mechanism to hypoxia. The serine/threonine kinase AKT promotes continued cell growth and metabolism by increasing glucose uptake, stimulating glycolysis and ATP production, at least in part via mTOR-dependent stabilization of HIF1-α. The purpose of this study was to investigate the role of mTOR signaling in the regulation of glycolytic activity of leukemic cells growing under high glucose or hypoxic conditions. Culture of Jurkat cells or primary ALL lymphoblasts in hypoxia (1% O2) resulted in robust induction of HIF-1α protein level which was significantly higher in high glucose conditions. This induction was blocked by inhibition of mTOR signaling, in association with decreased HK-2 mRNA and Glut-1 mRNA/protein expression. Likewise, hypoxia induced lactic acid production, a surrogate marker of glycolysis, and this induction was prevented by RAD001 (p=0.01). This data suggest that PI3K/AKT/mTOR pathway critically controls HIF-1α-dependent upregulation of glucose transport and glycolysis under in leukemic cells growing under pro-glycolytic conditions. We next examined the relationship between enhanced glycolytic activity and chemosensitivity to Doxorubicin. Doxorubicin (25ng/ml) efficiently inhibited growth of Jurkat cells under normoxic conditions supplemented with 4 mM glucose (equivalent of 72mg/dl). In contrast, Doxorubicin was approximately 50% less effective when cells were cultured either in high (14mM, equivalent of 252mg/dl) glucose or under hypoxia with low- or high-glucose supplementation. Inhibition of mTOR signaling strikingly enhanced effects of Doxorubicin resulting in complete inhibition of growth, and this effect was apparent only under high glucose conditions. We next examined effects of RAD001 in samples from 11 patients’ samples from newly diagnosed B-ALL patients growing under high/low glucose in hypoxic or normoxic conditions. RAD001 alone did not induce apoptosis in ALL cells (p 〉 0.05). In contrast, inhibition of mTOR signaling significantly enhanced Doxorubicin-induced apoptosis in 8 samples, with highest sensitization observed in high glucose environment (specific apoptosis, Doxorubicin alone, 5.6%, RAD001 7.7%, RAD001+Doxorubicin 22.4%, p 〈 0.02). In conclusion, blockade of mTOR-dependent HIF-1α and its downstream target Glut-1 efficiently promotes chemosensitivity of ALL cells under pro-glycolytic conditions. Hence, mTOR inhibition or blockade of HIF-1α-mediated glycolysis may play an important role in chemosensitization and improved outcomes in ALL.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 112, No. 4 ( 2008-08-15), p. 1424-1433
    Abstract: Epigenetic changes have been implicated in silencing several B-cell genes in Hodgkin and Reed-Sternberg cells (HRS) of Hodgkin lymphoma (HL), and this mechanism has been proposed to promote HRS survival and escape from immunosurveillance. However, the molecular and functional consequences of histone deacetylase (HDAC) inhibition in HL have not been previously described. In this study, we report that the HDAC inhibitor vorinostat induced p21 expression and decreased Bcl-xL levels causing cell-cycle arrest and apoptosis. Furthermore, vorinostat inhibited STAT6 phosphorylation and decreased its mRNA levels in a dose- and time-dependent manner, which was associated with a decrease in the expression and secretion of Thymus and Activation-Regulated Chemokine (TARC/CCL17) and interleukin (IL)–5 and an increase in IP-10 levels. Moreover, vorino-stat inhibited TARC secretion by dendritic cells that were activated by the thymic stromal lymphopoietin (TSLP). Collectively, these data suggest that pharmacologic HDAC inhibition in HL may induce favorable antitumor activity by a direct antiproliferative effect on HRS cells, and possibly by an immune mediated effect by altering cytokine and chemokines secretion in the microenvironment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 4981-4981
    Abstract: Alemtuzumab, a monoclonal anti-CD52 antibody, has shown high efficacy against lymphoproliferative disorders such as B-cell chronic lymphocytic leukemia (B-CLL). However, its use results in a profound immunosuppression with decrease of T lymphocytes leading to increased susceptibility to infections. The aim of our study was to assess the in vitro cytotoxic effect of alemtuzumab on normal T and neoplastic B lymphocytes obtained from B-CLL patients. Peripheral blood mononuclear cells (PBMC) from 12 B-CLL patients (5 at diagnosis and 7 previously treated) were collected and treated in vitro with alemtuzumab (10 μg/ml) and autologous serum in the culture as complement source. Spontaneous and alemtuzumab-induced apoptosis were quantified in T (CD3+) and B (CD20+) lymphocytes after 24 hours using an annexin-V flow cytometry based asssay. Alemtuzumab was able to induce apoptosis on both B and T lymphocytes of CLL patients. However, the cytotoxic activity on neoplastic B cells was comparable in all cases analyzed, irrespective of stage or previous treatments. Spontaneous apoptosis of B CLL cells was also comparable in all studied samples. On the contrary, the activity of alemtuzumab on normal T lymphocytes varied according to stage of disease and previous treatment. In early stages (0–I) alemtuzumab induced apoptosis in 19 % of T cells vs 60% of advanced stages (II–IV) (p=0,009). Differences were also relevant when patients were split by treatment: in fact, alemtuzumab induced apoptosis in 15% of T cells of untreated patients vs 52% of T cells of previously treated patients (p=0,005). Moreover, spontaneous apoptosis of T cells was more pronounced in treated vs. untreated patients (15% vs 2%, p=0.03) while the difference was not statistically significant in early vs advanced stage (p=0.06).Our in vitro data confirm that alemtuzumab is active against B-CLL cells in all stage of disease. However, in advanced stages and in previously treated patients, T cell compartment seems to be more fragile and susceptible to both spontaneous and alemtuzumab-induced apoptosis. This observation supports the hypothesis that using Alemtuzumab earlier in the treatment of B-CLL might result in less immunosuppression. The study is still ongoing with accrual of more patients samples.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 2566-2566
    Abstract: Purpose: HL patients (pts) with recurrent refractory disease after stem cell transplant have poor prognosis and are considered not curable with currently available salvage therapy. Novel therapeutic agents are needed for this pt population. MGCD0103 is an oral isotype-selective inhibitor of histone deacetylases (HDACS) with significant biological activity in preclinical models of hematopoietic cancers. Thymus- and activation-regulated chemokine (TARC) which is highly expressed by Reed-Sternberg cells in HL was one of the biomarkers assessed in this study. Methods: A phase II trial of MGCD0103 is ongoing in pts with relapsed/refractory classical HL. MGCD0103 was dosed at 110 mg 3×/week in 4-week cycles, with dose reductions to 85 and 60 mg in case of toxicities. Eligibility criteria included prior treatment with autologous and/or allogeneic stem cell transplant, at least one target lesion ≥ 2 cm, ECOG performance status of 0–1 and platelet counts of at least 25,000/mL. Tumor responses were determined every 8 weeks. Serum TARC levels were determined by ELISA using blood samples that were obtained prior to starting therapy and 8 days after therapy. Results: To date, 27 pts of a planned 12–35 have been enrolled (median age, 29 yrs; range, 20–62 yrs). Among 20 evaluable pts, 2 (10%) had CRs and 6 (30%) had PRs, for an OR rate of 40% (median time to response, 2 cycles). In addition, one pt (5%) had SD ( & lt;50% reduction) for ≥ 6 m. The rate of disease control (CR + PR + SD ≥ 6 m) was 45%. As assessed by CT scans, 15 pts (75%) exhibited tumor reductions: 12 (60%) had reductions of & gt; 30%, and 8 (40%) had reductions of ≥ 50%. Serum TARC levels were determined in 15 paired samples. After one week of therapy serum TARC levels were decreased by at least 40% in 5 pts, and all achieved major clinical responses (PR+CR). Seventeen of 25 pts required dose reductions for management of toxicities. The most common drug-related non-hematological toxicities were nausea (11/25), fatigue (10/25), vomiting (8/25), diarrhea (7/25), anorexia (4/25), pneumonia (3/25), abdominal pain (3/25) and weight loss (3/25). There were 2 deaths on study, both in heavily pretreated pts, one of unknown cause in a woman with h/o mantle XRT, BMT, suffering from significant GI AEs and the other of neutropenic fever, pneumonia and sepsis in a man with severe marrow compromise at baseline. Six pts experienced grade 3 or 4 toxicity. Of these, only pneumonia occurred in & gt; 1 pt. Dose modification was effective in managing most toxicities. HDAC inhibition was observed in PBMCs from the majority of evaluable pts. By using microarray expression analysis, transcriptional profiles of PBMCs from limited pts with/without clinical response were compared. Conclusions: Interim results from this ongoing trial suggest that single-agent MGCD0103 demonstrates significant anti-tumor activity in relapsed/refractory classical HL and is well tolerated, with dose modifications used as necessary to manage toxicities. Preliminary data also suggests that early decrease in serum TARC levels may predict response to therapy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1562-1562
    Abstract: Abstract 1562 Poster Board I-585 Purpose SNDX-275 is an oral, class 1 isoform selective HDACi. Phase 1 studies in leukemia demonstrated the agent has a long half-life and that weekly or every other week dosing is sufficient for antitumor activity. Based on recent favorable in vitro and in vivo activity of several HDAC inhibitors in HL, we investigated the in vitro activity of SNDX275 in HL-derived cell lines. Methods For apoptosis and gene expression analysis 05 × 106 cells were incubated with 0.1-2 μM of SNDX-275 for 24-72 hours before they were examined for proliferation and cell death by the MTS assay and the annexin-PI and FACS analysis. For combination studies, cells were incubated with 0.1-2 uM of SNDX-275 and 1-20 nM of either gemcitabine or bortezomib for 48-72 hours. Gene and protein expression were measured by RT-PCR, western blot, and immunohistochemistry. SNDX-275 effects on a panel of 30 cytokines and chemokines was assayed on 05 × 106 cells after incubation of 48 hrs using a multiplex assay. Results SNDX-275 induced cell death in a dose and time dependent manner with an IC50 of 0.4 μM. At the molecular level, SNDX-275 increased H3 acetylation, up-regulated p21 protein expression, and activated the intrinsic apoptosis pathway by down-regulating the anti-apoptotic X-linked inhibitor or apoptosis (XIAP) protein, which was associated with activation of caspase 9 and 3. Combination studies demonstrated that SNDX-275 had synergistic effects when combined with gemcitabine and bortezomib. To further investigate the potential for SNDX-275 activity in HL we measured the effect of SNDX-275 on pathways that may contribute to an anti-tumor immune response. Dysregulated cytokine/chemokine production has been shown to contribute to HL pathology, including immune tolerance of the cancer cells. SNDX-275 increased IL12 p40-70, IP10, and RANTES, and decreased the level of IL13 and IL4, thus favoring Th1-type cytokines/chemokines. In addition, recent data has demonstrated that a variety of epigenetic-modulating drugs may up-regulate the expression of cancer testis tumor associated antigens, leading to a favorable immune response. None of the lines expressed the CTAs without induction. SNDX275 was able to induce CTA expression of SSX2 in L428 but not HDLM2 whereas MAGE-A was induced in both HL cell lines. NY-ESO expression was not induced. Conclusions Our studies demonstrate that SNDS-275 has dual effect on apoptotic and immunomodulatory pathways in HL. Furthermore, this data demonstrates that SNDX-275 may upregulate CTAs suggesting that this treatment may render the tumor more immunogeneic and susceptible to immune mediated killing with tumor-specific cytotoxic T lymphocytes. The selectivity profile of SNDX-275 also suggests that HDAC1 and 2 are the primary targets for HDAC inhibition in these cells. Phase 2 studies with SNDX-275 in HL are ongoing. Disclosures Younes: MethylGene: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2009
    In:  Blood Vol. 114, No. 22 ( 2009-11-20), p. 1692-1692
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1692-1692
    Abstract: Abstract 1692 Poster Board I-718 Transforming growth factor-b-activated kinase 1 (TAK1) is a key regulator of NF-kB activation. TAK1 can be activated by a variety of pro-inflammatory cytokines and T and B cell receptors. Recent experiments demonstrated that deletion of TAK1 results in inactivation of both JNK and NF-kB signaling resulting in massive apoptotic death of hematopoietic cells in mice. In this study, we examined the expression pattern of TAK1 and its role as a potential therapeutic target for lymphoma. First, we examined TAK1 expression in a panel of lymphoid cell lines by western blot, and found it to be highly expressed in mantle cell lymphoma cell lines (Mino, SP53, and Jeko-1). These lines expressed relatively low levels of the tumor suppressor protein A20. Mino and SP53 expressed high level of p-p38. Subsequently, we investigated the in vitro activity of the novel TAK1 small molecule inhibitor AZ-Tak1 in these cell lines. AZ-Tak1 is a potent and a relatively selective inhibitor of TAK1 kinase activity, with an IC50 of 0.009 mM. It also inhibits Jak2 but at a much higher concentration (IC50=0.18 mM). AZ-Tak1 treatment decreased the level of p38 and ERK in mantle cell lymphoma cells, and induced apoptosis in a dose and time dependent manner, with an IC50 of 0.1-0.5 mM. Using the annexin-V and PI staining and FACS analysis, After 48 hours of incubation, AZ-Tak1 (0.1 mM) induced apoptosis in 28%, 34% and 86% of Mino, SP53, and Jeko cells, respectively, which was increased to 32%, 42%, and 86% when 0.5 mM concentration was used. Similar activity was also observed when primary mantle cell lymphoma cells were examined. Using pathway-specific protein arrays focusing on apoptosis, kinases, and transcription factors, AZ-Tak1 (0.5 mM) altered the level of several proteins that regulate cell growth and survival, especially members of the inhibitors of apoptosis (IAP) family. Specifically, AZ-Tak1 decreased the level of SMAC/DIABOLO and cytochrome –C in the mitochondria, which was associated with a decrease in the level of the anti-apoptotic protein X-linked IAP (XIAP) and activation of the intrinsic apoptotic pathway as evident by activation of caspase 9, cleavage of caspase 3, and induction of apoptosis. Furthermore, and consistant with its ability to inhibit Jak2 activity, AZ-Tak1 reduced STAT2 and STAT6 levels. AZ-Tak1 demonstrated no significant effect on bcl-2 family members. Finally, co-treatment with HDAC inhibitors demonstrated synergistic effect with low concentrations of AZ-Tak1. Collectively, our data demonstrate that targeting TAK1 by the small molecule inhibitor AZ-Tak1 induces cell death in mantle cell lymphoma by activating the intrinsic apoptosis pathway, suggesting that targeting TAK1 may have a therapeutic value for the treatment of mantle cell lymphoma. Disclosures Palakurthi: Astra Zeneca: Employment. Byth:Astra Zeneca : Employment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...