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
Material
Language
  • 1
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 302-302
    Abstract: Abstract 302 Background: Marizomib has a novel, non-peptide based, bicyclic structure and compared to other proteasome inhibitors, unique properties of clinical relevance. Specifically, marizomib produces rapid, broad and prolonged inhibition of all 3 20S proteasome catalytic activities, and markedly different safety and efficacy profiles, including activity against MM resistant to bortezomib (BZ) both in vitro and in vivo. Materials and Methods: Marizomib was given IV over 1–120 minutes on days 1, 4, 8 and 11 of 21-day cycles in 2 separate and parallel dose escalation studies performed in Australia and the United States in patients with relapsed and refractory MM. In addition to standard safety and efficacy monitoring, pharmacokinetics (PK) and proteasome inhibition as part of pharmacodynamics (PD) were assessed. Dexamethasone (20 mg) was given the day prior to and day of treatment in one study and could be added for patients who did not achieve a minimal response (MR) or better after 2 cycles in the other study. Toxicity evaluation was performed using CTCAE v3.0 and response was assessed by modified European Group for Blood and Marrow Transplantation (EBMT) and Uniform Criteria (UC). Results: 34 patients (16 men and 18 women) have been treated at doses of 0.075 to 0.6 mg/m2/dose BIW with a median age of 62.5 years, in both studies. Patients received a median of 6 prior regimens; 30 patients (88%) had been exposed to prior BZ, including 24 (71%) who were BZ -refractory. The maximum tolerated dose of marizomib was found to be 0.4 mg/m2 over a 60 minute infusion time and 0.5 mg/m2 over a 120 minute infusion. Dose limiting toxicities included transient hallucinations, cognitive changes and loss of balance, all of which proved reversible. The most common drug-related adverse events were fatigue, nausea, vomiting, dizziness, headache, diarrhea, constipation, insomnia, anorexia, and dyspnea, which proved manageable with supportive care and/or dose reduction. Importantly, marizomib did not appear to induce myelosuppression, peripheral neuropathy (PN) or thrombocytopenia. PK analysis demonstrated a rapid elimination half-life ( 〈 20 minutes) and large volume of distribution, with PD analyses of packed whole blood (PWB) and peripheral blood mononuclear cells (PBMC) confirming dose dependent proteasome inhibition. At interim analysis, of 22 patients with evaluable disease for best response to marizomib +/− dexamethasone, 3 had achieved partial response (PR) by EBMT/UC (14%). In the active dose range of 0.4–0.6 mg/m2, 15 pts were evaluable with PR in 3 pts (20%), all of whom were refractory to prior BZ. Median time on treatment was 1.5 months, with stable disease or better documented in 16 pts (73%). Conclusions: The safety profile of marizomib clearly differs from BZ, without significant treatment–emergent PN or myelosuppression described. Preliminary results suggest anti-myeloma activity, with responses seen in patients in whom BZ had previously failed, as well as interesting PK/PD characteristics and tissue distribution supporting a possible role in patients with different disease characteristics (such as extramedullary spread). The efficacy and safety of 0.5 mg/m2 of marizomib given twice weekly, alone or with low dose dexamethasone, warrants further study, and continues to be investigated. Future directions will include combination approaches with lenalidomide and dexamethasone. Disclosures: Richardson: Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Membership on an entity's Board of Directors or advisory committees. Cannell:Nereus Pharmaceuticals: Investigator. Harrison:Nereus Pharmaceuticals: Research Funding. Jakubowiak:Ortho Biotech: Consultancy, Honoraria, Speakers Bureau; Celgene: Consultancy, Honoraria, Speakers Bureau; Millennium Pharmaceuticals: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Onyx Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Exelixis: Consultancy, Honoraria. Palladino:Nereus Pharmaceuticals, Inc: Employment, Equity Ownership. Longenecker:Nereus Pharmaceuticals, Inc: Employment, Equity Ownership. Lay:Nereus Pharmaceuticals, Inc: Employment, Equity Ownership. Lloyd:Nereus Pharmaceuticals, Inc: Employment, Equity Ownership. Hannah:Nereus Pharmaceuticals, Inc.: Consultancy. Reich:Nereus Pharmaceuticals: Consultancy. Spear:Nereus Pharmaceuticals, Inc: Employment, Equity Ownership. Anderson:Onyx: Consultancy; Merck: Consultancy; Bristol Myers Squibb: Consultancy; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Acetylon: Founder; Nereus Pharmaceuticals, Inc: Consultancy; Millennium: Consultancy; Celgene: Consultancy.
    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 ...
  • 2
    In: Journal of Medicinal Chemistry, American Chemical Society (ACS), Vol. 39, No. 17 ( 1996-01-01), p. 3235-3237
    Type of Medium: Online Resource
    ISSN: 0022-2623 , 1520-4804
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 1996
    detail.hit.zdb_id: 1491411-6
    SSG: 15,3
    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. 431-431
    Abstract: Abstract 431 Background: NPI-0052 has a novel, non-peptide based, bicyclic structure resulting in a unique proteasome inhibition and safety profile. In contrast to other proteasome inhibitors, NPI-0052 produces rapid, broad and prolonged inhibition of all 3 catalytic activities. Preclinical data subsequently suggested improvements in toxicology and efficacy, including activity MM resistant to bortezomib (BZ) and other agents (Chauhan et al, Cancer Cell 2005), thus this Phase 1 dose escalation trial in patients (pts) with relapsed/refractory MM was initiated. Materials and Methods: Patients (pts) were treated with NPI-0052 IV weekly for 3 weeks in 4-week cycles. Measurable disease by EBMT criteria was not required. The dose of NPI-0052 was escalated using a combination of accelerated titration and 3+3 design. PK and proteasome inhibition (blood and PBMCs) were assayed after the first and third doses. Preliminary Results: 27 pts have been treated at doses ranging from 0.025 to 0.7 mg/m2; median age is 62; 18 males/9 females; IgG/IgA/light chain/non-secretory 14/4/2/6; median of 4 prior regimens and 27% refractory to prior bortezomib. Reversible DLT was observed in two out of eight patients treated at 0.7 mg/m2 (Grade 3 fatigue; Grade 3 mental status changes and loss of balance), with 2 additional pts undergoing dose reductions in Cycle 1 (nausea and vomiting; vertigo and confusion/word-finding difficulties). Prophylactic anti-emetics have been instituted with a decrease in infusion-related nausea; similarly, pts with dizziness/vertigo have been administered meclizine with symptomatic improvement. Other drug-related adverse events have consisted principally of mild-to-moderate fatigue, nausea, vomiting, dizziness, headache and diarrhea; interestingly, myelosuppression, neuropathy and thrombosis do not appear to be elicited by NPI-0052. PK assessment demonstrates a rapid elimination half-life ( 〈 20 minutes) and relatively large Vz. NPI-0052 produces dose dependent proteasome inhibitions. At 0.7 mg/m2, Day 1/Day 15 inhibition of chymotrypsin-like activity in whole blood is 73% and 99%, respectively (the value for bortezomib at 1.3 mg/m2 is 65%). One patient with IgA MM (4 prior regimens plus ASCT; relapsed after prior BZ, not refractory) had a 71% decrease in M-protein (unconfirmed PR; off study after 3 cycles). A second pt with non-secretory disease (4 prior regimens;relapsed after prior BZ, not refractory) had a nearly 50% reduction in involved light chain; this pt remains active on study at 5+ months. In addition, 8 pts with relapsed/refractory MM remained on study for between 6-15 months (3 pts were on-study for over one year) with stable disease and no significant toxicity; 2 of these pts were BZ-refractory. Conclusions: Tolerability of 0.7 mg/m2 continues to be investigated in pts with MM, with prophylactic antiemetics and meclizine to reduce common drug-related toxicities of nausea and dizziness. The safety profile of NPI-0052 is importantly different from bortezomib in spite of higher and more durable proteasome inhibition; peripheral neuropathy and thrombocytopenia were not seen. Accrual continues to expand upon these results and assess the new lyophile formulation of NPI-0052. Disclosures: Richardson: Millennium: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Keryx: Membership on an entity's Board of Directors or advisory committees; Gentium: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; Merck: Membership on an entity's Board of Directors or advisory committees. Off Label Use: Vorinostat is a histone deacetylase (HDAC) inhibitor that was approved in the FDA in October 2006 for the treatment of cutaneous manifestations in patients with cutaneous T-cell lymphoma (CTCL) who have progressive, persistent, or recurrent disease on or following two systemic therapies. Jakubowiak:Celgene: Consultancy, Honoraria; Millennium: Consultancy, Honoraria; Centocor Ortho Biotech: Consultancy, Honoraria; Exelixis: Consultancy, Honoraria; Bristol-Myers-Squibb: Consultancy, Honoraria. Spear:Nereus Pharmaceuticals: Employment, Equity Ownership. Palladino:Nereus Pharmaceuticals: Employment, Equity Ownership. Longenecker:Nereus Pharmaceuticals: Employment, Equity Ownership. Neuteboom:Nereus Pharmaceuticals: Employment, Equity Ownership. Cropp:Nereus Pharmaceuticals: Consultancy. Lloyd:Nereus Pharmaceuticals: Employment, Equity Ownership. Hannah:Nereus Pharmaceuticals: Consultancy. Anderson:Nereus Pharmaceuticals: Consultancy, 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. 112, No. 11 ( 2008-11-16), p. 3665-3665
    Abstract: Background: We recently characterized a novel proteasome inhibitor NPI-0052, a small molecule derived from the fermentation of a marine gram-positive actinomycete Salinispora tropica. NPI-0052 induces apoptosis in multiple myeloma (MM) cells resistant to conventional and bortezomib therapies. Importantly, NPI-0052 is distinct from bortezomib in its chemical structure, proteasome inhibition profiles, and mechanisms of action. In the present study, we utilized a human plasmacytoma xenograft mouse model to examine the effect of NPI-0052 on proteasome activity profiles in selected organs and tumors. Our results demonstrate that NPI-0052 rapidly leaves the vascular compartment in an active form after intravenous (IV) administration and inhibits the proteasome in extra-vascular tumors and other organs, excluding brain. NPI-0052 triggers a more sustained proteasome inhibition in tumors than in other organs examined. Importantly, we also confirmed the anti-tumor efficacy of NPI-0052. Methods and Model: Animal studies were approved by the DFCI Institutional Animal Care and Use Committee. Sixty CB-17 SCID-male mice were inoculated with 5.0 × 106 MM.1S cells in 100ul of serum free RPMI-1640 medium. The mice were divided into three different groups: Groups 1 and 2 (25 mice EA group) for pharmacodynamic studies (time course) and Group 3 (10 mice) for drug efficacy study. Tumor size was measured every third day in two dimensions using calipers, and tumor volume was calculated using the formula V = 0.5 a × b2, where a and b are the long and short diameter of the tumor respectively. When tumors were ~250 mm3 (~three-four weeks after injection), mice were treated with 0.15 mg/kg of NPI-0052 (IV) or vehicle control. Proteasome inhibition was assessed after either single NPI-0052 treatment (given at Day1) or three treatments (given at Day1, Day4 and Day8). Mice were euthanized at 10 mins, 1h, 4h, and 24h; and packed whole blood (PWB), liver, spleen, lung, kidney, brain and tumors were analyzed for chymotrypsin-like (CT-L), Caspase-like (C-L), and Trypsin-like (T-L) proteasome activities. For efficacy studies mice were treated with NPI-0052 twice a week for three weeks. Mice were sacrificed when their tumors reached ~1.5 cm3. NPI-0052 was dissolved in 100% DMSO to generate a 10 mg/ml stock solution, aliquoted, and stored frozen at − 80°C. The stock solution was serially diluted with 100% DMSO and for injection with 5% Solutol (Solutol HS, polyethylene glycol 660, 12 hydroxystearate; BASF, Shreveport, LA) yielding a final concentration of 2% DMSO and 98% (5% Solutol). The vehicle control was 2% DMSO and 98% (5% Solutol). The pH of the dosing solutions is between 6–7. Results: Inhibition of all three proteasome activities after a single treatment of NPI-0052 was detectable as early as 10 mins in the liver, lung, spleen, kidney and PWB; Within 24h after either a single or three IV treatments of NPI-0052, proteasome activity recovered in liver, lung, spleen and kidney, but not in tumor or PWB; No significant proteasome inhibition was noted in brain up to 24h after either a single or three IV treatments with NPI-0052; CT-L activity was inhibited within 1h post first dose, and 24h exposure triggered marked inhibition of CT-L, C-L and T-L activities in vivo in the xenografted MM.1S tumors. For example, in 1h CT-L activity was inhibited 34%, T-L activity 6% and C-L activity 16%. After 24h hours, CT-L activity was inhibited 60%, T-L activity 24% and C-L activity 49%; and finally, 6) Inhibition of CT-L, C-L and T-L activities increased in the tumor after the third NPI-0052 treatment compared to the first treatment. For example, at 1h post third dose all three activities were inhibited approximately 70–80%. Additionally, the anti-MM activity of NPI-0052 was associated with significant proteasome inhibition in tumors (P 〈 0.005). Conclusions: Our findings show that NPI-0052 induces a prolonged inhibition ( 〉 24h) of all three-20S proteasome activities in established MM.1S tumor xenografts which correlated with marked anti-tumor activity. In contrast, proteasome inhibition in normal tissues including liver, spleen, kidney and lung markedly recovered within 24h of administration after a single or three treatments with NPI-0052. In addition, no significant inhibition of proteasome activities was detectable in the brain after either treatment schedule.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2770-2770
    Abstract: Background: NPI-0052 is a novel proteasome inhibitor that produces prolonged inhibition of all three catalytic activities (C-L, T-L, CT-L) of the 20S proteasome. Preclinical data suggest NPI-0052 may demonstrate an improved therapeutic ratio, with significant activity in hematologic and solid tumor malignancies including multiple myeloma (MM) resistant to bortezomib and other agents (Chauhan et al, Blood 2006). Studies have therefore been initiated in patients with multiple myeloma, lymphoma, leukemia and solid tumors. Materials and Methods: This Phase 1 dose escalation study evaluated NPI-0052 monotherapy in patients with relapsed and relapsed/refractory MM, including those that have received bortezomib and/or lenalidomide. Patients were assessed for safety, pharmacodynamics (including ex vivo proteasome inhibition), plasma pharmacokinetics (PK), and clinical activity (with response assessed by modified EBMT criteria). Patients were treated with NPI-0052 administered as a weekly IV injection on Days 1, 8 and 15 every 4 weeks with concomitant hydration. The dose of NPI-0052 was escalated in cohorts of 3 patients dependent on observed adverse events utilizing a 3+3 design. Proteasome inhibition and PK were assayed after the 1st and 3rd dose and upon any intra-patient dose escalation. Proteasome inhibition was also assessed in CD138 positive cells isolated from bone marrow aspirates obtained at baseline and after the 3rd dose in a subset of patients. Preliminary Results: To date, 10 patients have been treated at doses ranging from 0.025 mg/m2 to 0.075 mg/m2 without reaching an MTD. One patient experienced reversible elevation in serum creatinine that responded to drug cessation and steroids; this event may have been related to progression of his underlying light chain nephropathy (as interval worsening of renal function was noted prior to enrollment). Drug-related adverse events have been otherwise unremarkable at all dose levels tested. PK data demonstrate a rapid elimination half life (estimated to be 3–4 minutes) with clearance between 8–21 mL/min and Vz of 44–99L; no change in PK has been observed comparing the 1st and 3rd injection. Proteasome inhibition in whole blood suggests drug-dependent CT-L inhibition, with inhibition up to 28% observed (inhibition up to 100% at doses of up to 0.7 mg/m2 has been observed in other clinical trials with NPI-0052 without producing the profile of toxicity reported with standard doses of bortezomib). Whilst no responses have been confirmed, two patients with relapse/refractory MM remained on study for over 6 months and one year, respectively, with stable disease and no significant toxicity. Importantly, no peripheral neuropathy or myelosuppression has been seen in 41 treatment cycles in patients to date. Conclusions: In patients with relapsed and relapsed, refractory MM, NPI-0052 affects parameters relevant to pharmacodynamics, PK and demonstrated potential clinical benefit at doses well below the MTD anticipated from Phase 1 clinical trials with NPI-0052 in lymphoma and solid tumors. Drug administration to date has been well tolerated. Dose escalation continues to define DLT and MTD, and to recommend a phase 2 dose for further study in patients with advanced MM.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3842-3842
    Abstract: Abstract 3842 Poster Board III-778 Background and Rationale Vascular disrupting agents (VDAs) act via selectively disrupting established tumor vasculature and have shown remarkable clinical success as anti-cancer therapies. NPI-2358 is a novel VDA with a distinct structure and mechanism of action from other available VDAs. NPI-2358 binds to the colchicine-binding site of beta-tubulin preventing polymerization and disrupting the cytoplasmic microtubule network, thereby causing loss of vascular endothelial cytoskeletal function, and inducing cytotoxicity in cancer cells. Here, we examined the anti-angiogenic and anti-tumor activity of NPI-2358 in multiple myeloma (MM) cells using both in vitro and in vivo model systems. Material and Methods We utilized MM.1S, MM.1R, RPMI-8226, U266, and INA-6 human MM cell lines, as well as purified tumor cells from MM patients relapsing after prior anti-MM therapies. Cell viability/apoptosis assays were performed using MTT, trypan blue exclusion, and Annexin V/PI staining. Angiogenesis was measured in vitro using Matrigel capillary-like tube structure formation assays: Since human vascular endothelial cells (HUVECs) plated onto Matrigel differentiate and form capillary-like tube structures similar to in vivo neovascularization, this assay measures anti-angiogenic effects of drugs/agents. Migration assays were performed using transwell insert assays. Immunoblot analysis was performed using antibodies to caspase-8, caspase-9, caspase-3, PARP, Bcl-2, Bax, pJNK and GAPDH. Statistical significance was determined using a Student t test. Results Treatment of MM.1S, RPMI-8226, MM.1R, INA-6, and KMS-12BM with NPI-2358 for 24h induces a dose-dependent significant (P 〈 0.005) decrease in viability of all cell lines (IC50 range: 5-8 nM; n=3). To determine whether NPI-2358-induced decrease in viability is due to apoptosis, MM cell lines were treated with NPI-2358 for 24h; harvested, and analyzed for apoptosis using Annexin V/PI staining. A significant increase in NPI-2358-induced apoptosis was observed in all MM cell lines (% Annexin V+/PI- apoptotic cells: MM.1S, 48 ± 2.3%; MM.1R, 46.6 ± 3.1%; RPMI-8226, 61.7 ± 4.5%; and INA-6, 59.9 ± 3.2%; P 〈 0.05; n=3). Importantly, NPI-2358 decreased viability of freshly isolated MM cells from patients (IC50 range: 3-7 nM; P 〈 0.005), without affecting the viability of normal peripheral blood mononuclear cells, suggesting specific anti-MM activity and a favorable therapeutic index for NPI-2358. Examination of in vitro angiogenesis using capillary-like tube structure formation assay showed that even low doses of NPI-2358 (7 nM treatment for 12h; IC50: 20 nM at 24h) significantly decreased tubule formation in HUVECs (70-80% decrease; P 〈 0.05). Transwell insert assays showed a marked reduction in serum-dependent migration of NPI-2358-treated MM cells (42 ± 2.1% inhibition in NPI-2358-treated vs. control; P 〈 0.05). NPI-2358 at the concentrations tested (5 nM for 12h) in the migration assays did not affect survival of MM cells ( 〉 95% viable cells). A similar anti-migration activity of NPI-2358 was noted against HUVEC cells (48 ± 1.7% decrease in migration; P 〈 0.05). Mechanistic studies showed that NPI-2358-induced apoptosis was associated with activation of caspase-8, caspase-9, caspase-3 and PARP. Importantly, treatment of MM.1S cells with NPI-2358 (5 nM) triggered phosphorylation of c-Jun amino-terminal kinase (JNK), a classical stress response protein, without affecting Bcl-2 family members Bax and Bcl-2. Blockade of JNK using dominant negative strategy markedly abrogated NPI-2358-induced apoptosis. Conclusion Our preclinical data provide evidence for remarkable anti-angiogenic and anti-tumor activity of NPI-2358 against MM cells, without significant toxicity in normal cells. Ongoing studies are examining in vivo anti-MM activity of NPI-2358 in animal models. Importantly, a Phase-1 study of NPI-2358 as a single agent in patients with advanced malignancies (lung, prostrate and colon cancer) has already established a favorable pharmacokinetic, pharmacodynamic and safety profile; and, a Phase-2 study of the combination of NPI-2358 and docetaxel in non-small cell lung cancer showed encouraging safety, pharmacokinetic and activity data. These findings, coupled with our preclinical studies, provide the framework for the development of NPI-2358-based novel therapies to improve patient outcome in MM. Disclosures: Chauhan: Nereus Pharmaceuticals, Inc: Consultancy. Lloyd:Nereus Pharmaceuticals, In: Employment. Palladino:Nereus Pharmaceuticals, Inc: Employment. Anderson:Nereus Pharmaceuticals, Inc: Consultancy.
    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 ...
  • 7
    In: ChemBioChem, Wiley, Vol. 6, No. 1 ( 2005-01-07), p. 133-144
    Abstract: The synthesis and the biological evaluation of a new family diterpenes are presented. The synthetic studies were inspired by the structural framework of acanthoic acid ( 1 ) and yielded a family of compounds that were evaluated as anti‐inflammatory agents. Among them, compounds 2 , 10 , 12 , and 16 exhibited a very low nonspecific cytotoxicity and inhibited the synthesis of TNF‐α with greater than 65 % efficacy at low micromolar concentrations. Cytokine‐specificity studies revealed that these compounds also inhibited the synthesis of the proinflammatory cytokines IL‐1β and IL‐6, while inhibition of IL‐1ra and IL‐8 synthesis was marginal and only occurred at high concentrations. Further studies, through EMSA and Western blot analyses, indicated that these compounds decreased the extent of phosphorylation of I κB α; this suggests that they exert their anti‐inflammatory profile by inhibiting NF‐ κB ‐mediated cytokine synthesis. These findings imply that these diterpenes represent promising leads for the development of novel anti‐inflammatory agents.
    Type of Medium: Online Resource
    ISSN: 1439-4227 , 1439-7633
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2005
    detail.hit.zdb_id: 2020469-3
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2015
    In:  Journal of Clinical Oncology Vol. 33, No. 15_suppl ( 2015-05-20), p. e12644-e12644
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 33, No. 15_suppl ( 2015-05-20), p. e12644-e12644
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2015
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Anti-Cancer Drugs, Ovid Technologies (Wolters Kluwer Health), Vol. 17, No. 1 ( 2006-01), p. 25-31
    Type of Medium: Online Resource
    ISSN: 0959-4973
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2006
    detail.hit.zdb_id: 2025803-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Frontiers in Oncology, Frontiers Media SA, Vol. 11 ( 2021-3-3)
    Abstract: Reprogramming tumor infiltrating myeloid cells to elicit pro-inflammatory responses is an exciting therapeutic maneouver to improve anti-tumor responses. We recently demonstrated that a distinct microtubule-targeting drug, plinabulin—a clinical-stage novel agent—modulates dendritic cell maturation and enhances anti-tumor immunity. Here, we investigated the effects of plinabulin on macrophage polarization in vitro and in vivo . Plinabulin monotherapy induced significant tumor growth inhibition in mice bearing subcutaneous MC38 colon cancer. Importantly, the regressing tumors were characterized by an increase in M1-like/M2-like tumor-associated macrophages (TAM) ratio. The efficacy of plinabulin remained unaltered in T cell-deficient Rag2 −/− mice, suggesting an important role of macrophages in driving the drug's anti-tumor effect. Exposure of murine and healthy human macrophages to plinabulin induced polarization toward the M1 phenotype, including increased expression of co-stimulatory molecules CD80, CD86 and pro-inflammatory cytokines IL-1β, IL-6, and IL-12. M2-associated immunosuppressive cytokines IL-10 and IL-4 were reduced. This pro-inflammatory M1-like skewing of TAMs in response to plinabulin was dependent on the JNK pathway. Functionally, plinabulin-polarized human M1 macrophages directly killed HuT 78 tumor cells in vitro . Importantly, plinabulin induced a functional M1-like polarization of tumor infiltrating macrophages in murine tumors as well as in tumor samples from ovarian cancer patients, by preferentially triggering M1 proliferation. Our study uncovers a novel immunomodulatory effect of plinabulin in directly triggering M1 polarization and proliferation as well as promoting TAM anti-tumoral effector functions.
    Type of Medium: Online Resource
    ISSN: 2234-943X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2021
    detail.hit.zdb_id: 2649216-7
    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...